Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 104: 105170, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38823088

ABSTRACT

BACKGROUND: Ebola virus disease (EVD) survivors experience ocular sequelae including retinal lesions, cataracts, and vision loss. While monoclonal antibodies targeting the Ebola virus glycoprotein (EBOV-GP) have shown promise in improving prognosis, their effectiveness in mitigating ocular sequelae remains uncertain. METHODS: We developed and characterized a BSL-2-compatible immunocompetent mouse model to evaluate therapeutics targeting EBOV-GP by inoculating neonatal mice with vesicular stomatitis virus expressing EBOV-GP (VSV-EBOV). To examine the impact of anti-EBOV-GP antibody treatment on acute retinitis and ocular sequelae, VSV-EBOV-infected mice were treated with polyclonal antibodies or monoclonal antibody preparations with antibody-dependent cellular cytotoxicity (ADCC-mAb) or neutralizing activity (NEUT-mAb). FINDINGS: Treatment with all anti-EBOV-GP antibodies tested dramatically reduced viremia and improved survival. Further, all treatments reduced the incidence of cataracts. However, NEUT-mAb alone or in combination with ADCC-mAb reduced viral load in the eyes, downregulated the ocular immune and inflammatory responses, and minimized retinal damage more effectively. INTERPRETATION: Anti-EBOV-GP antibodies can improve survival among EVD patients, but improved therapeutics are needed to reduce life altering sequelae. This animal model offers a new platform to examine the acute and long-term effect of the virus in the eye and the relative impact of therapeutic candidates targeting EBOV-GP. Results indicate that even antibodies that improve systemic viral clearance and survival can differ in their capacity to reduce acute ocular inflammation, and long-term retinal pathology and corneal degeneration. FUNDING: This study was partly supported by Postgraduate Research Fellowship Awards from ORISE through an interagency agreement between the US DOE and the US FDA.


Subject(s)
Antibodies, Viral , Disease Models, Animal , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Mice , Ebolavirus/immunology , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/immunology , Antibodies, Viral/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology , Humans , Viral Load , Glycoproteins/immunology , Glycoproteins/metabolism , Viral Envelope Proteins/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Antibody-Dependent Cell Cytotoxicity
2.
J Autoimmun ; 102: 65-76, 2019 08.
Article in English | MEDLINE | ID: mdl-31080013

ABSTRACT

IL-22 has opposing effects in different tissues, from pro-inflammatory (skin, joints) to protective (liver, intestine) but little is known about its effects on neuroinflammation. We examined the effect of IL-22 on retinal tissue by using the model of experimental autoimmune uveitis (EAU) in IL-22-/- mice, as well as by intraocular injections of recombinant IL-22 or anti-IL-22 antibodies in wild type animals. During EAU, IL-22 was produced in the eye by CD4+ eye-infiltrating T cells. EAU-challenged IL-22-/- mice, as well as WT mice treated systemically or intraocularly with anti-IL-22 antibodies during the expression phase of disease, developed exacerbated retinal damage. Furthermore, IL-22-/- mice were more susceptible than WT controls to glutamate-induced neurotoxicity, whereas local IL-22 supplementation was protective, suggesting direct or indirect neuroprotective effects. Mechanistic studies revealed that retinal glial Müller cells express IL-22rα1 in vivo, and in vitro IL-22 enhanced their ability to suppress proliferation of effector T cells. Finally, IL-22 injected into the eye concurrently with IL-1, inhibited the (IL-1-induced) expression of multiple proinflammatory and proapoptotic genes in retinal tissue. These findings suggest that IL-22 can function locally within the retina to reduce inflammatory damage and provide neuroprotection by affecting multiple molecular and cellular pathways.


Subject(s)
Autoimmunity , Central Nervous System/immunology , Central Nervous System/metabolism , Disease Susceptibility , Interleukins/metabolism , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Autoimmunity/genetics , Central Nervous System/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Ependymoglial Cells/immunology , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Interleukins/genetics , Interleukins/pharmacology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neuroprotection/genetics , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Uveitis/etiology , Uveitis/metabolism , Uveitis/pathology , Interleukin-22
3.
Cell Rep ; 26(7): 1718-1726.e4, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30759384

ABSTRACT

Zaire Ebola virus (ZEBOV) survivors experience visual and CNS sequelae that suggests the ZEBOV glycoprotein can mediate neurotropism. Replication-competent rVSVΔG-ZEBOV-GP vaccine candidate is generally well tolerated; however, its potential neurotropism requires careful study. Here, we show that a single inoculation of rVSVΔG-ZEBOV-GP virus in neonatal C57BL/6 mice results in transient viremia, neurological symptoms, high viral titers in eyes and brains, and death. rVSVΔG-ZEBOV-GP infects the inner layers of the retina, causing severe retinitis. In the cerebellum, rVSVΔG-ZEBOV-GP infects neurons in the granular and Purkinje layers, resulting in progressive foci of apoptosis and neurodegeneration. The susceptibility to infection is not due to impaired type I IFN responses, although MDA5-/-, IFNß-/-, and IFNAR1-/- mice have accelerated mortality. However, boosting interferon levels by co-administering poly(I:C) reduces viral titers in CNS and improves survival. Although these data should not be directly extrapolated to humans, they challenge the hypothesis that VSV-based vaccines are non-neurotropic.


Subject(s)
Central Nervous System/pathology , Neurodegenerative Diseases/genetics , Retina/pathology , Animals , Animals, Newborn , Apoptosis , Humans , Mice , Neurons
4.
Emerg Microbes Infect ; 7(1): 96, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29802245

ABSTRACT

Zika-infected patients can have eye involvement ranging from mild conjunctivitis to severe chorioretinal lesions, however the possible long-term sequelae of infection and timeline to recovery remain unknown. Here we describe the partial recovery of chorioretinal lesions in an immunocompetent patient diagnosed with bilateral posterior uveitis associated with Zika infection and show that some lesions resolved with focal atrophy evident as pigmentary changes on funduscopy. To better understand the progression of the lesions and correlate the changes in fundus imaging with local viral load, immune responses, and retinal damage, we developed a symptomatic mouse model of ocular Zika virus infection. Imaging of the fundus revealed multiple hypopigmentary patches indicative of chorioretinal degeneration as well as thinning of the retina that mirror the lesions in patients. Microscopically, the virus primarily infected the optic nerve, retinal ganglion cells, and inner nuclear layer cells, showing thinning of the outer plexiform layer. During acute infection, the eyes showed retinal layer disorganization, retinitis, vitritis, and focal choroiditis, with mild cellular infiltration and increased expression of tumor necrosis factor, interferon-γ, granzyme B, and perforin. Focal areas of gliosis and retinal degeneration persisted 60 dpi. The model recapitulates features of ZIKA infections in patients and should help elucidate the mechanisms underlying the damage to the eyes and aid in the development of effective therapeutics.


Subject(s)
Chorioretinitis/virology , Retina/virology , Uveitis, Posterior/virology , Zika Virus Infection/pathology , Zika Virus/isolation & purification , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Conjunctivitis, Viral/virology , Humans , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Optic Nerve/virology , Retinal Ganglion Cells/virology
5.
Exp Eye Res ; 166: 116-119, 2018 01.
Article in English | MEDLINE | ID: mdl-29074386

ABSTRACT

In this study we compared polarized mouse T-helper (Th) lymphocytes of four populations, sensitized against an ocular antigen, for their patterns of migration and induction of inflammatory processes in recipient mouse eyes expressing the target antigen. Th1, Th2, Th9 and Th17 cells transgenically expressing T-cell receptor (TCR) specific against hen egg lysozyme (HEL) were adoptively transferred to recipient mice expressing HEL in their eyes. Recipient eyes collected 4 or 7 days post injection were analyzed for histopathological changes. Th1 and Th17 cells induced moderate to severe intraocular inflammation in the recipient mouse eyes, but essentially did not migrate into the conjunctiva. In contrast, Th2 and Th9 cells invaded minimally the intraocular space of recipient eyes, but accumulated in the limbus and migrated into the conjunctiva of the recipient mice and initiated allergy-like inflammatory responses, as indicated by remarkable eosinophil involvement. These data thus shed new light on the differences between the migration patterns and ocular pathogenic processes mediated by Th1/Th17 and by Th2/Th9 populations.


Subject(s)
Cell Movement , Conjunctiva/pathology , Eosinophilia/pathology , Limbus Corneae/parasitology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Disease Models, Animal , Lens, Crystalline/metabolism , Mice , Muramidase , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
6.
J Immunol ; 196(3): 1013-25, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26712943

ABSTRACT

During chronic inflammation, tertiary lymphoid tissue (TLT) can form within an inflamed organ, including the CNS. However, little is known about TLT formation in the neuroretina. In a novel spontaneous autoimmune mouse model of uveitis (R161H), we identified well-organized lymphoid aggregates in the retina and examined them for TLT characteristics. Presence of immune cells, tissue-specific markers, and gene expression patterns typically associated with germinal centers and T follicular helper cells were examined using immunohistochemistry and gene analysis of laser capture microdissected retina. Our data revealed the retinal lymphoid structures contained CD4(+) T cells and B cells in well-defined zonal areas that expressed classic germinal center markers, peanut lectin (agglutinin) and GL-7. Gene expression analysis showed upregulation of T follicular helper cell markers, most notably CXCR5 and its ligand CXCL13, and immunohistochemical analysis confirmed CXCR5 expression, typically associated with CD4(+) T follicular helper cells. Highly organized stromal cell networks, a hallmark of organized lymphoid tissue, were also present. Positive staining for phospho-Zap70 in retina-specific T cells indicated CD4(+) T cells were being activated within these lymphoid structures. CD138(+)/B220(+) plasma cells were detected, suggesting the retinal lymphoid aggregates give rise to functional germinal centers, which produce Abs. Interestingly, eyes with lymphoid aggregates exhibited lower inflammatory scores by fundus examination and a slower initial rate of loss of visual function by electroretinography, compared with eyes without these structures. Our findings suggest that the lymphoid aggregates in the retina of R161H mice represent organized TLT, which impact the course of chronic uveitis.


Subject(s)
Autoimmune Diseases/pathology , Lymphoid Tissue/pathology , Uveitis/pathology , Animals , Autoimmune Diseases/immunology , Disease Models, Animal , Electroretinography , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Laser Capture Microdissection , Lymphoid Tissue/immunology , Mice , Mice, Transgenic , Microscopy, Confocal , Polymerase Chain Reaction , Transcriptome , Uveitis/immunology , Vision, Ocular/physiology
7.
Invest Ophthalmol Vis Sci ; 56(9): 5439-49, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26284549

ABSTRACT

PURPOSE: Experimental autoimmune uveitis (EAU) induced in mice using the retinal antigen interphotoreceptor retinoid binding protein (IRBP) is an animal model for posterior uveitis in humans. However, EAU induced by native IRBP protein or its widely used epitope amino acid residues 1 to 20 of human IRBP (hIRBP1-20) is inconsistent, often showing low scores and incidence. We found an urgent need to identify a better pathogenic epitope for the C57BL/6 strain. METHODS: Mice were immunized with uveitogenic peptides or with native bovine IRBP. Clinical and histological disease and associated immunological responses were evaluated. Truncated and substituted peptides, as well as bioinformatic analyses, were used to identify critical major histocompatibility complex (MHC)/T cell receptor (TCR) contact residues and the minimal core epitope. RESULTS: The new uveitogenic epitope of IRBP, amino acid residues 651 to 670 of human IRBP (LAQGAYRTAVDLESLASQLT [hIRBP651-670]) is uveitogenic for mice of the H-2b haplotype and elicits EAU with a higher severity and incidence in C57BL/6 mice than the previously characterized hIRBP1-20 epitope. Using truncated and substituted peptides, as well as bioinformatic analysis, we identified the critical contact residues with MHC/TCR and defined the minimal core epitope. This made it possible to design MHC tetramers and use them to detect epitope-specific T cells in the uveitic eye and in lymphoid organs of hIRBP651-670-immunized mice. CONCLUSIONS: Data suggest that hIRBP651-670 is an epitope naturally processed from a conserved region of native IRBP, potentially explaining its relatively high uveitogenicity. This epitope should be useful for basic and preclinical studies of uveitis in the C57BL/6 model and gives access to genetically engineered mice available on this background.


Subject(s)
Autoimmune Diseases/immunology , Eye Proteins/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Cellular , Retinitis/immunology , Retinol-Binding Proteins/immunology , T-Lymphocytes/immunology , Uveitis, Posterior/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cattle , Cells, Cultured , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Eye Proteins/metabolism , Haplotypes , Histocompatibility Antigens Class II/metabolism , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Retinitis/metabolism , Retinitis/pathology , Retinol-Binding Proteins/metabolism , Severity of Illness Index , Uveitis, Posterior/metabolism , Uveitis, Posterior/pathology
8.
Immunity ; 43(2): 343-53, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26287682

ABSTRACT

Activated retina-specific T cells that have acquired the ability to break through the blood-retinal barrier are thought to be causally involved in autoimmune uveitis, a major cause of human blindness. It is unclear where these autoreactive T cells first become activated, given that their cognate antigens are sequestered within the immune-privileged eye. We demonstrate in a novel mouse model of spontaneous uveitis that activation of retina-specific T cells is dependent on gut commensal microbiota. Retina-specific T cell activation involved signaling through the autoreactive T cell receptor (TCR) in response to non-cognate antigen in the intestine and was independent of the endogenous retinal autoantigen. Our findings not only have implications for the etiology of human uveitis, but also raise the possibility that activation of autoreactive TCRs by commensal microbes might be a more common trigger of autoimmune diseases than is currently appreciated.


Subject(s)
Intestines/immunology , Microbiota/immunology , Retina/immunology , T-Lymphocytes/immunology , Uveitis/immunology , Animals , Antigens, Bacterial/administration & dosage , Autoantigens/immunology , Autoimmunity , Blood-Retinal Barrier/immunology , Cells, Cultured , Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/immunology , Eye Proteins/metabolism , Immune Tolerance , Intestines/microbiology , Lymphocyte Activation , Mice, Inbred Strains , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/metabolism , Uveitis/microbiology
9.
Growth Horm IGF Res ; 23(3): 45-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23578754

ABSTRACT

IGFs and their binding proteins have been shown to exhibit both protective and deleterious effects in ocular disease. Recent studies have characterized the expression patterns of different IGFBPs in retinal layers and within the vitreous. IGFBP-3 has roles in vascular protection stimulating proliferation, migration, and differentiation of vascular progenitor cells to sites of injury. IGFBP-3 increases pericyte ensheathment and shows anti-inflammatory effects by reducing microglia activation in diabetes. IGFBP-5 has recently been linked to mediating fibrosis in proliferative vitreoretinopathy but also reduces neovascularization. Thus, the regulatory balance between IGF and IGFBPs can have profound impact on target tissues. This review discusses recent findings of IGF and IGFBP expression in the eye with relevance to different retinopathies.


Subject(s)
Eye Diseases/pathology , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Eye Diseases/metabolism , Humans
10.
PLoS One ; 7(7): e39398, 2012.
Article in English | MEDLINE | ID: mdl-22792172

ABSTRACT

Previously, we showed that insulin growth factor (IGF)-1 binding protein-3 (IGFBP-3), independent of IGF-1, reduces pathological angiogenesis in a mouse model of the oxygen-induced retinopathy (OIR). The current study evaluates novel endothelium-dependent functions of IGFBP-3 including blood retinal barrier (BRB) integrity and vasorelaxation. To evaluate vascular barrier function, either plasmid expressing IGFBP-3 under the regulation of an endothelial-specific promoter or a control plasmid was injected into the vitreous humor of mouse pups (P1) and compared to the non-injected eyes of the same pups undergoing standard OIR protocol. Prior to sacrifice, the mice were given an injection of horseradish peroxidase (HRP). IGFBP-3 plasmid-injected eyes displayed near-normal vessel morphology and enhanced vascular barrier function. Further, in vitro IGFBP-3 protects retinal endothelial cells from VEGF-induced loss of junctional integrity by antagonizing the dissociation of the junctional complexes. To assess the vasodilatory effects of IGFBP-3, rat posterior cerebral arteries were examined in vitro. Intraluminal IGFBP-3 decreased both pressure- and serotonin-induced constrictions by stimulating nitric oxide (NO) release that were blocked by L-NAME or scavenger receptor-B1 neutralizing antibody (SRB1-Ab). Both wild-type and IGF-1-nonbinding mutant IGFBP-3 (IGFBP-3NB) stimulated eNOS activity/NO release to a similar extent in human microvascular endothelial cells (HMVECs). NO release was neither associated with an increase in intracellular calcium nor decreased by Ca(2+)/calmodulin-dependent protein kinase II (CamKII) blockade; however, dephosphorylation of eNOS-Thr(495) was observed. Phosphatidylinositol 3-kinase (PI3K) activity and Akt-Ser(473) phosphorylation were both increased by IGFBP-3 and selectively blocked by the SRB1-Ab or PI3K blocker LY294002. In conclusion, IGFBP-3 mediates protective effects on BRB integrity and mediates robust NO release to stimulate vasorelaxation via activation of SRB1. This response is IGF-1- and calcium-independent, but requires PI3K/Akt activation, suggesting that IGFBP-3 has novel protective effects on retinal and systemic vasculature and may be a therapeutic candidate for ocular complications such as diabetic retinopathy.


Subject(s)
Blood-Retinal Barrier/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Animals , Blood-Retinal Barrier/pathology , Cadherins/genetics , Cadherins/metabolism , Calcium/metabolism , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fura-2 , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor I/metabolism , Intercellular Junctions/drug effects , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Male , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Nitric Oxide , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retinal Diseases/genetics , Retinal Diseases/metabolism , Retinal Vessels/drug effects , Retinal Vessels/metabolism , Scavenger Receptors, Class B/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology
11.
Blood ; 119(2): 629-36, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-22028476

ABSTRACT

The ovarian peptide hormone, relaxin, circulates during pregnancy, contributing to profound maternal vasodilation through endothelial and nitric oxide (NO)-dependent mechanisms. Circulating numbers of bone marrow-derived endothelial cells (BMDECs), which facilitate angiogenesis and contribute to repair of vascular endothelium, increase during pregnancy. Thus, we hypothesized that relaxin enhances BMDEC NO production, circulating numbers, and function. Recombinant human relaxin-2 (rhRLX) stimulated PI3K/Akt B-dependent NO production in human BMDECs within minutes, and activated BMDEC migration that was inhibited by L-N(G)-nitroarginine methyl ester. In BMDECs isolated from relaxin/insulin-like family peptide receptor 2 gene (Rxfp2) knockout and wild-type mice, but not Rxfp1 knockout mice, rhRLX rapidly increased NO production. Similarly, rhRLX increased circulating BMDEC number in Rxfp2 knockout and wild-type mice, but not Rxfp1 knockout mice as assessed by colony formation and flow cytometry. Taken together, these results indicate that relaxin effects BMDEC function through the RXFP1 receptor. Finally, both vascularization and incorporation of GFP-labeled BMDECs were stimulated in rhRLX-impregnated Matrigel pellets implanted in mice. To conclude, relaxin is a novel regulator of BMDECs number and function, which has implications for angiogenesis and vascular remodeling in pregnancy, as well as therapeutic potential in vascular disease.


Subject(s)
Cell Movement , Endothelium, Vascular/cytology , Neovascularization, Physiologic , Nitric Oxide/metabolism , Receptors, G-Protein-Coupled/physiology , Relaxin/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cells, Cultured , Endothelium, Vascular/metabolism , Female , Flow Cytometry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vasodilation
12.
Invest Ophthalmol Vis Sci ; 52(11): 8278-86, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21931131

ABSTRACT

PURPOSE: To examine the effect of free insulin-like growth factor (IGF) binding protein-3 (IGFBP-3), independent of the effect of insulin-like growth factors, in modulating retinal vascular permeability. METHODS: We assessed the ability of a form of IGFBP-3 that does not bind IGF-1 (IGFBP-3NB), to regulate the blood retinal barrier (BRB) using two distinct experimental mouse models, laser-induced retinal vessel injury and vascular endothelial growth factor (VEGF)-induced retinal vascular permeability. Additionally, in vitro studies were conducted. In the animal models, BRB permeability was quantified by intravenous injection of fluorescein labeled serum albumin followed by digital confocal image analysis of retinal flat-mounts. Claudin-5 and vascular endothelial-cadherin (VE-cadherin) localization at interendothelial junctions was studied by immunofluorescence. In vitro changes in transendothelial electrical resistance (TEER) and flux of fluorescent dextran in bovine retinal endothelial monolayers (BREC) were measured after IGFBP-3NB treatment. Acid (ASMase) and neutral (NSMase) sphingomyelinase mRNA levels and activity were measured in mouse retinas. RESULTS: Four days postinjury, laser-injured mouse retinas injected with IGFBP-3NB plasmid demonstrated reduced vascular permeability compared with retinas of laser-injured mouse retinas injected with control plasmid. IGFBP-3NB administration resulted in a significant decrease in laser injury-associated increases in ASMase and NSMase mRNA and activity when compared with laser alone treated mice. In vivo, intravitreal injection of IGFBP-3NB reduced vascular leakage associated with intravitreal VEGF injection. IGFBP-3NB partially restored VEGF-induced in vivo permeability and dissociation of claudin-5 and VE-cadherin at junctional complexes. When IGFBP-3NB was applied basally to bovine retinal endothelial cells (BREC) in vitro, TEER increased and macromolecular flux decreased. CONCLUSIONS: Intravitreal administration of IGFBP-3NB preserves junctional integrity in the presence of VEGF or laser injury by reducing BRB permeability in part by modulating sphingomyelinase levels.


Subject(s)
Blood-Retinal Barrier/drug effects , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Retina/injuries , Retina/metabolism , Retinal Vessels/drug effects , Sphingomyelin Phosphodiesterase/metabolism , Adherens Junctions/metabolism , Animals , Blood-Retinal Barrier/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cattle , Disease Models, Animal , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Lasers/adverse effects , Light Coagulation , Mice , Mice, Inbred C57BL , Plasmids/pharmacology , Recombinant Proteins/pharmacology , Retina/drug effects , Retinal Vessels/metabolism , Sphingomyelin Phosphodiesterase/genetics , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
13.
Am J Pathol ; 178(4): 1517-28, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21435441

ABSTRACT

This study was conducted to determine the perivascular cell responses to increased endothelial cell expression of insulin-like growth factor binding protein-3 (IGFBP-3) in mouse retina. The contribution of bone marrow cells in the IGFBP-3-mediated response was examined using green fluorescent protein-positive (GFP(+)) adult chimeric mice subjected to laser-induced retinal vessel occlusion injury. Intravitreal injection of an endothelial-specific IGFBP-3-expressing plasmid resulted in increased differentiation of GFP(+) hematopoietic stem cells (HSCs) into pericytes and astrocytes as determined by immunohistochemical analysis. Administration of IGFBP-3 plasmid to mouse pups that underwent the oxygen-induced retinopathy model resulted in increased pericyte ensheathment and reduced pericyte apoptosis in the developing retina. Increased IGFBP-3 expression reduced the number of activated microglial cells and decreased apoptosis of neuronal cells in the oxygen-induced retinopathy model. In summary, IGFBP-3 increased differentiation of GFP(+) HSCs into pericytes and astrocytes while increasing vascular ensheathment of pericytes and decreasing apoptosis of pericytes and retinal neurons. All of these cytoprotective effects exhibited by IGFBP-3 overexpression can result in a more stable retinal vascular bed. Thus, endothelial expression of IGFBP-3 may represent a physiologic response to injury and may represent a therapeutic strategy for the treatment of ischemic vascular eye diseases, such as diabetic retinopathy and retinopathy of prematurity.


Subject(s)
Apoptosis , Insulin-Like Growth Factor Binding Protein 3/metabolism , Ischemia/pathology , Microglia/metabolism , Neurons/metabolism , Pericytes/metabolism , Retina/injuries , Animals , Astrocytes/cytology , Cell Death , Cell Differentiation , Eye Diseases/pathology , Female , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence/methods , Pericytes/cytology , Retina/metabolism
14.
Microvasc Res ; 79(3): 207-16, 2010 May.
Article in English | MEDLINE | ID: mdl-20188747

ABSTRACT

Bone-marrow-derived endothelial progenitor cells (EPCs) contribute to angiogenesis-mediated pathological neovascularization, and recent studies have begun to recognize the biological significance of this contribution. This review will discuss the ability of EPCs to contribute to neovascularization in both physiological and pathological conditions. Circulating EPCs were originally identified in 1997 by Asahara as CD34(+) VEGFR2(+) mononuclear cells. These cells differentiated into an endothelial phenotype, expressed endothelial markers, and incorporated into neovessels at sites of ischemia (Asahara et al., 1997). EPCs provide both instructive (release of pro-angiogenic cytokines) and structural (vessel incorporation and stabilization) functions that contribute to the initiation of neo-angiogenesis. EPC populations can be characterized based on surface markers of freshly isolated cells, or they can be described by their in vitro characteristics once placed in culture. However, a major stumbling block to progress in the field has been the lack of consensus among investigators as to the optimal characterization of EPCs. This review intends to address the role of both EPC classes and evaluate how they interact in the setting of pathological angiogenesis. Since the EPCs may be responsible for turning on the "angiogenic switch," strategies have been employed to keep this switch in the "off" position for diseases like cancer, retinopathy, and wet AMD. The expectation is that EPCs will evolve into clinically useful prognostic and predictive tools in cancer and in ocular diseases associated with pathological neovascularization and that targeting this cell type is a key to successful management of patients suffering from diseases associated with pathological neovascularization.


Subject(s)
Endothelial Cells/pathology , Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Stem Cells/pathology , Angiogenesis Inhibitors/therapeutic use , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Endothelial Cells/drug effects , Humans , Neoplasms/drug therapy , Neovascularization, Pathologic/physiopathology , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic , Phenotype , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Stem Cell Niche , Stem Cells/drug effects
15.
J Exp Med ; 206(13): 2897-906, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-19934019

ABSTRACT

The present epidemic of diabetes is resulting in a worldwide increase in cardiovascular and microvascular complications including retinopathy. Current thinking has focused on local influences in the retina as being responsible for development of this diabetic complication. However, the contribution of circulating cells in maintenance, repair, and dysfunction of the vasculature is now becoming appreciated. Diabetic individuals have fewer endothelial progenitor cells (EPCs) in their circulation and these cells have diminished migratory potential, which contributes to their decreased reparative capacity. Using a rat model of type 2 diabetes, we show that the decrease in EPC release from diabetic bone marrow is caused by bone marrow neuropathy and that these changes precede the development of diabetic retinopathy. In rats that had diabetes for 4 mo, we observed a dramatic reduction in the number of nerve terminal endings in the bone marrow. Denervation was accompanied by increased numbers of EPCs within the bone marrow but decreased numbers in circulation. Furthermore, denervation was accompanied by a loss of circadian release of EPCs and a marked reduction in clock gene expression in the retina and in EPCs themselves. This reduction in the circadian peak of EPC release led to diminished reparative capacity, resulting in the development of the hallmark feature of diabetic retinopathy, acellular retinal capillaries. Thus, for the first time, diabetic retinopathy is related to neuropathy of the bone marrow. This novel finding shows that bone marrow denervation represents a new therapeutic target for treatment of diabetic vascular complications.


Subject(s)
Bone Marrow/innervation , CLOCK Proteins/genetics , Diabetic Neuropathies/complications , Diabetic Retinopathy/etiology , Animals , Circadian Rhythm , Denervation , Female , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Norepinephrine/blood , Rats , Sympathetic Nervous System/physiology
16.
Circ Res ; 105(9): 897-905, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19762684

ABSTRACT

RATIONALE: Insulin-like growth factor binding protein (IGFBP)-3 modulates vascular development by regulating endothelial progenitor cell (EPC) behavior, specifically stimulating EPC cell migration. This study was undertaken to investigate the mechanism of IGFBP-3 effects on EPC function and how IGFBP-3 mediates cytoprotection following vascular injury. OBJECTIVE: To examine the mechanism of IGFBP-3-mediated repair following vascular injury. METHODS AND RESULTS: We used 2 complementary vascular injury models: laser occlusion of retinal vessels in adult green fluorescent protein (GFP) chimeric mice and oxygen-induced retinopathy in mouse pups. Intravitreal injection of IGFBP-3-expressing plasmid into lasered GFP chimeric mice stimulated homing of EPCs, whereas reversing ischemia induced increases in macrophage infiltration. IGFBP-3 also reduced the retinal ceramide/sphingomyelin ratio that was increased following laser injury. In the OIR model, IGFBP-3 prevented cell death of resident vascular endothelial cells and EPCs, while simultaneously increasing astrocytic ensheathment of vessels. For EPCs to orchestrate repair, these cells must migrate into ischemic tissue. This migratory ability is mediated, in part, by endogenous NO generation. Thus, we asked whether the migratory effects of IGFBP-3 were attributable to stimulation of NO generation. IGFBP-3 increased endothelial NO synthase expression in human EPCs leading to NO generation. IGFBP-3 exposure also led to the redistribution of vasodilator-stimulated phosphoprotein, an NO regulated protein critical for cell migration. IGFBP-3-mediated NO generation required high-density lipoprotein receptor activation and stimulation of phosphatidylinositol 3-kinase/Akt pathway. CONCLUSION: These studies support consideration of IGFBP-3 as a novel agent to restore the function of injured vasculature and restore NO generation.


Subject(s)
Cell Movement , Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Nitric Oxide/metabolism , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Cell Adhesion Molecules/metabolism , Cell Death , Cell Proliferation , Cells, Cultured , Ceramides/metabolism , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Cytoprotection , Disease Models, Animal , Endothelial Cells/pathology , Female , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Humans , Infant, Newborn , Insulin-Like Growth Factor Binding Protein 3/genetics , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/pathology , Retinal Neovascularization/physiopathology , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/physiopathology , Scavenger Receptors, Class B/metabolism , Signal Transduction , Sphingomyelins/metabolism , Stem Cells/pathology , Vasodilation
17.
Mol Ther ; 17(9): 1594-604, 2009 09.
Article in English | MEDLINE | ID: mdl-19584817

ABSTRACT

The ability to control the differentiation of adult hematopoietic stem cells (HSCs) would promote development of new cell-based therapies to treat multiple degenerative diseases. Systemic injection of NaIO(3) was used to ablate the retinal pigment epithelial (RPE) layer in C57Bl6 mice and initiate neural retinal degeneration. HSCs infected ex vivo with lentiviral vector expressing the RPE-specific gene RPE65 restored a functional RPE layer, with typical RPE phenotype including coexpression of another RPE-specific marker, CRALBP, and photoreceptor outer segment phagocytosis. Retinal degeneration was prevented and visual function, as measured by electroretinography (ERG), was restored to levels similar to that found in normal animals. None of the controls (no HSCs, HSCs alone and HSCs infected with lentiviral vector expressing LacZ) showed these effects. In vitro gene array studies demonstrated that infection of HSC with RPE65 increased adenylate cyclase mRNA. In vitro exposure of HSCs to a pharmacological agonist of adenylate cyclase also led to in vitro differentiation of HSCs to RPE-like cells expressing pigment granules and the RPE-specific marker, CRALBP. Our data confirm that expression of the cell-specific gene RPE65 promoted fate determination of HSCs toward RPE for targeted tissue repair, and did so in part by activation of adenylate cyclase signaling pathways. Expression by HSCs of single genes unique to a differentiated cell may represent a novel experimental paradigm to influence HSC plasticity, force selective differentiation, and ultimately lead to identification of pharmacological alternatives to viral gene delivery.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Differentiation , Cells, Cultured , Electroretinography , Eye Proteins/genetics , Eye Proteins/physiology , Female , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Humans , Immunohistochemistry , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Degeneration/therapy , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/ultrastructure , cis-trans-Isomerases
18.
Exp Eye Res ; 88(3): 367-77, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18723015

ABSTRACT

The efficacy of novel monoclonal antibodies that neutralize the pro-angiogenic mediator, sphingosine-1-phosphate (S1P), were tested using in vitro and in vivo angiogenesis models, including choroidal neovascularization (CNV) induced by laser disruption of Bruch's membrane. S1P receptor levels in human brain choroid plexus endothelial cells (CPEC), human lung microvascular endothelial cells, human retinal vascular endothelial cells, and circulating endothelial progenitor cells were examined by semi-quantitative PCR. The ability of murine or humanized anti-S1P monoclonal antibodies (mAbs) to inhibit S1P-mediated microvessel tube formation by CPEC on Matrigel was evaluated and capillary density in subcutaneous growth factor-loaded Matrigel plugs was determined following anti-S1P treatment. S1P promoted in vitro capillary tube formation in CPEC consistent with the presence of cognate S1P(1-5) receptor expression by these cells and the S1P antibody induced a dose-dependent reduction in microvessel tube formation. In a murine model of laser-induced rupture of Bruch's membrane, S1P was detected in posterior cups of mice receiving laser injury, but not in uninjured controls. Intravitreous injection of anti-S1P mAbs dramatically inhibited CNV formation and sub-retinal collagen deposition in all treatment groups (p<0.05 compared to controls), thereby identifying S1P as a previously unrecognized mediator of angiogenesis and subretinal fibrosis in this model. These findings suggest that neutralizing S1P with anti-S1P mAbs may be a novel method of treating patients with exudative age-related macular degeneration by reducing angiogenesis and sub-retinal fibrosis, which are responsible for visual acuity loss in this disease.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Choroidal Neovascularization/prevention & control , Lysophospholipids/immunology , Sphingosine/analogs & derivatives , Angiogenesis Inhibitors/pharmacology , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Collagen , Disease Models, Animal , Drug Combinations , Drug Evaluation, Preclinical/methods , Female , Fibrosis/prevention & control , Gene Expression , Laminin , Lasers , Lysophospholipids/analysis , Lysophospholipids/pharmacology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Proteoglycans , RNA, Messenger/genetics , Rabbits , Receptors, Lysosphingolipid/biosynthesis , Receptors, Lysosphingolipid/genetics , Retina/pathology , Reverse Transcriptase Polymerase Chain Reaction/methods , Sphingosine/analysis , Sphingosine/immunology , Sphingosine/pharmacology , Vitreous Body/chemistry
19.
Invest Ophthalmol Vis Sci ; 48(4): 1674-82, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17389499

ABSTRACT

PURPOSE: To isolate and characterize progenitor cells derived from adult mammalian ciliary body. METHODS: The authors isolated progenitor cells from the ciliary body of adult mice, rats, and human cadaver eyes and determined quantitative growth characteristics of groups of progenitor cells called neurosphere (NS) cells, including individual cell diameter, NS diameter, percentage of NS-forming cells, and cell number per eye in mouse, rat, and human eyes. The immunolabeling and ultrastructure of NS cells were investigated by confocal and transmission electron microscopy. RESULTS: Average diameters of individual cells and neurospheres after 1 week in culture were similar in mice, rats, and humans (cell diameters: 22 +/- 1.1, 21 +/- 0.3, 25 +/- 0.4 mum; NS diameters: 139 +/- 22, 137 +/- 9, 141 +/- 11 mum, respectively). Mean numbers of cells per NS were estimated to be 1183 in mice, 5360 in rats, and 685 in humans. Molecules that were identified by immunolabeling in NS cells included nestin, Chx-10, vimentin, GFAP, and Pax-6. Thy-1 was expressed in some NS cells. Ultrastructurally, NS cells displayed abundant rough endoplasmic reticulum and many cellular processes but no characteristics of mature retinal neurons or glia. CONCLUSIONS: Progenitor cells from adult mammalian ciliary body have significant, but limited, proliferation potential and express markers characteristic of other progenitor cells and seen during early retinal development. The ciliary body could be a source of cells for transplantation in experimental rodent eyes and for autotransplantation in human eyes.


Subject(s)
Ciliary Body/cytology , Stem Cells/cytology , Aged , Aged, 80 and over , Animals , Biomarkers/metabolism , Cell Count , Cell Culture Techniques , Cell Proliferation , Cell Separation , Ciliary Body/physiology , Eye Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Homeodomain Proteins/metabolism , Humans , Intermediate Filament Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Nerve Tissue Proteins/metabolism , Nestin , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Rats , Rats, Wistar , Repressor Proteins/metabolism , Stem Cells/physiology , Thy-1 Antigens/metabolism , Transcription Factors/metabolism , Vimentin/metabolism
20.
Exp Eye Res ; 83(2): 255-62, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16546168

ABSTRACT

Acute intraocular pressure (IOP) elevation causes accumulation of retrogradely-transported brain derived neurotrophic factor and its receptor at the optic nerve head (ONH) in rats and monkeys. Obstruction of axonal transport may therefore be involved in glaucoma pathogenesis, but it is unknown if obstruction is specific to certain transported factors or represents a generalized failure of retrograde axonal transport. The dynein motor complex mediates retrograde axonal transport in retinal ganglion cells (RGC). Our hypothesis was that elevated IOP interferes with dynein-mediated axonal transport. We studied the distribution of dynein subunits in the retina and optic nerve after acute and chronic experimental IOP elevation in the rat. IOP was elevated unilaterally in 54 rats. Dynein subunit distribution was compared in treated and control eyes by immunohistochemistry and Western blotting at 1 day (n=12), 3 days (n=4), 1 week (n=15), 2 weeks (n=12) and 4 weeks (n=11). For immunohistochemistry, sections through the ONH were probed with an anti-dynein heavy chain (HC) antibody and graded semi-quantitatively by masked observers. Other freshly enucleated eyes were microdissected for separate Western blot quantification of dynein intermediate complex (IC) in myelinated and unmyelinated optic nerve, ONH and retina. Immunohistochemistry showed accumulation of dynein HC at the ONH in IOP elevation eyes compared to controls (P<0.001, Wilcoxon paired sign-rank test, n=29). ONH dynein IC was elevated by 46.5% in chronic IOP elevation eyes compared to controls by Western blotting (P<0.001, 95% CI=25.9% to 67.8%, n=17). The maximum increase in ONH dynein IC was 78.7% after 1 week (P<0.05, n=5), but significant increases were also detected after 4 h and 4 weeks of IOP elevation (P<0.05, n=4 rats per group). Total retinal dynein IC was increased by 8.7% in chronic IOP elevation eyes compared to controls (P<0.03, 95% CI 1.4% to 16.1%, n=24). In the retina, IOP elevation particularly affected the 72 kD subunit of dynein IC, which was 100.7% higher in chronic IOP elevation eyes compared to controls (P<0.00001, 95% CI 71.0% to 130.4%, n=21). Dynein IC changes in myelinated and unmyelinated optic nerve were not significant (P>0.05). We conclude that dynein accumulates at the ONH with experimental IOP elevation in the rat, supporting the hypothesis that disrupted axonal transport in RGC may be involved in the pathogenesis of glaucoma. The effect of IOP elevation on other motor proteins deserves further investigation in the future.


Subject(s)
Dyneins/metabolism , Glaucoma/metabolism , Intraocular Pressure/physiology , Optic Disk/metabolism , Animals , Axonal Transport/physiology , Axons/metabolism , Blotting, Western/methods , Disease Models, Animal , Dyneins/analysis , Immunohistochemistry/methods , Male , Rats , Rats, Wistar , Retinal Ganglion Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...