Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Med Sci (Paris) ; 36(8-9): 753-762, 2020.
Article in French | MEDLINE | ID: mdl-32821052

ABSTRACT

Macular edema is an increase in volume of the central area of the retina, responsible for visual acuity. Visual symptoms handicap the lives of millions of patients with macular edema secondary to chronic and sometimes acute retinal disease. Proteins that neutralize the vascular endothelial growth factor (VEGF) pathway or glucocorticoids, at the cost of repeated intraocular injections over years, limit visual symptoms. A better understanding of why and how edema forms and how therapeutic molecules exert an anti-edematous effect will help prevent this disabling and blinding retinal complication from occurring.


Title: Les œdèmes maculaires - Mieux comprendre leurs mécanismes pour mieux les traiter. Abstract: L'œdème maculaire est une augmentation de volume de la macula, zone centrale de la rétine, responsable de l'acuité visuelle. Des symptômes visuels handicapent la vie de millions de patients atteints d'œdème maculaire secondaire à une maladie chronique et parfois aiguë de la rétine. Les protéines qui neutralisent la voie du facteur de croissance de l'endothélium vasculaire (VEGF) ou les glucocorticoïdes, au prix d'injections intraoculaires répétées pendant des années, limitent les symptômes visuels. Mieux comprendre pourquoi et comment l'œdème se forme et comment les molécules thérapeutiques exercent un effet anti-œdémateux permettra de mieux prévenir la survenue de cette complication rétinienne handicapante et cécitante.


Subject(s)
Macular Edema/etiology , Macular Edema/therapy , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Glucocorticoids/therapeutic use , Humans , Signal Transduction/drug effects , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Visual Acuity/drug effects , Visual Acuity/physiology
2.
Prog Retin Eye Res ; 63: 20-68, 2018 03.
Article in English | MEDLINE | ID: mdl-29126927

ABSTRACT

Macular edema consists of intra- or subretinal fluid accumulation in the macular region. It occurs during the course of numerous retinal disorders and can cause severe impairment of central vision. Major causes of macular edema include diabetes, branch and central retinal vein occlusion, choroidal neovascularization, posterior uveitis, postoperative inflammation and central serous chorioretinopathy. The healthy retina is maintained in a relatively dehydrated, transparent state compatible with optimal light transmission by multiple active and passive systems. Fluid accumulation results from an imbalance between processes governing fluid entry and exit, and is driven by Starling equation when inner or outer blood-retinal barriers are disrupted. The multiple and intricate mechanisms involved in retinal hydro-ionic homeostasis, their molecular and cellular basis, and how their deregulation lead to retinal edema, are addressed in this review. Analyzing the distribution of junction proteins and water channels in the human macula, several hypotheses are raised to explain why edema forms specifically in the macular region. "Pure" clinical phenotypes of macular edema, that result presumably from a single causative mechanism, are detailed. Finally, diabetic macular edema is investigated, as a complex multifactorial pathogenic example. This comprehensive review on the current understanding of macular edema and its mechanisms opens perspectives to identify new preventive and therapeutic strategies for this sight-threatening condition.


Subject(s)
Macular Edema/physiopathology , Blood-Retinal Barrier , Central Serous Chorioretinopathy/complications , Central Serous Chorioretinopathy/physiopathology , Choroidal Neovascularization/complications , Choroidal Neovascularization/physiopathology , Diabetic Retinopathy/complications , Diabetic Retinopathy/physiopathology , Fluorescein Angiography , Humans , Macular Edema/diagnosis , Macular Edema/prevention & control , Retinal Vein Occlusion/complications , Retinal Vein Occlusion/physiopathology , Retinal Vessels/physiopathology , Subretinal Fluid , Tomography, Optical Coherence
3.
Am J Pathol ; 185(8): 2083-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26166807

ABSTRACT

Mast cells are important in the initiation of ocular inflammation, but the consequences of mast cell degranulation on ocular pathology remain uncharacterized. We induced mast cell degranulation by local subconjunctival injection of compound 48/80. Initial degranulation of mast cells was observed in the choroid 15 minutes after the injection and increased up to 3 hours after injection. Clinical signs of anterior segment inflammation paralleled mast cell degranulation. With the use of optical coherence tomography, dilation of choroidal vessels and serous retinal detachments (SRDs) were observed and confirmed by histology. Subconjunctival injection of disodium cromoglycate significantly reduced the rate of SRDs, demonstrating the involvement of mast cell degranulation in posterior segment disorders. The infiltration of polymorphonuclear and macrophage cells was associated with increased ocular media concentrations of tumor necrosis factor-α, CXCL1, IL-6, IL-5, chemokine ligand 2, and IL-1ß. Analysis of the amounts of vascular endothelial growth factor and IL-18 showed an opposite evolution of vascular endothelial growth factor compared with IL-18 concentrations, suggesting that they regulate each other's production. These findings suggest that the local degranulation of ocular mast cells provoked acute ocular inflammation, dilation, increased vascular permeability of choroidal vessels, and SRDs. The involvement of mast cells in retinal diseases should be further investigated. The pharmacologic inhibition of mast cell degranulation may be a potential target for intervention.


Subject(s)
Cell Degranulation/drug effects , Choroid/pathology , Mast Cells/pathology , Retina/pathology , Animals , Capillary Permeability/drug effects , Chemokines/metabolism , Choroid/drug effects , Choroid/metabolism , Cytokines/metabolism , Female , Mast Cells/drug effects , Mast Cells/metabolism , Rats , Rats, Inbred Lew , Retina/drug effects , Retina/metabolism , Tomography, Optical Coherence , p-Methoxy-N-methylphenethylamine/pharmacology
4.
Mol Vis ; 20: 908-20, 2014.
Article in English | MEDLINE | ID: mdl-24966662

ABSTRACT

PURPOSE: To evaluate whether anti-vascular endothelial growth factor (VEGF) neutralizing antibodies injected in the vitreous of rat eyes influence retinal microglia and macrophage activation. To dissociate the effect of anti-VEGF on microglia and macrophages subsequent to its antiangiogenic effect, we chose a model of acute intraocular inflammation. METHODS: Lewis rats were challenged with systemic lipopolysaccharide (LPS) injection and concomitantly received 5 µl of rat anti-VEGF-neutralizing antibody (1.5 mg/ml) in the vitreous. Rat immunoglobulin G (IgG) isotype was used as the control. The effect of anti-VEGF was evaluated at 24 and 48 h clinically (uveitis scores), biologically (cytokine multiplex analysis in ocular media), and histologically (inflammatory cell counts on eye sections). Microglia and macrophages were immunodetected with ionized calcium-binding adaptor molecule 1 (IBA1) staining and counted based on their differential shapes (round amoeboid or ramified dendritiform) on sections and flatmounted retinas using confocal imaging and automatic quantification. Activation of microglia was also evaluated with inducible nitric oxide synthase (iNOS) and IBA1 coimmunostaining. Coimmunolocalization of VEGF receptor 1 and 2 (VEGF-R1 and R2) with IBA1 was performed on eye sections with or without anti-VEGF treatment. RESULTS: Neutralizing rat anti-VEGF antibodies significantly decreased ocular VEGF levels but did not decrease the endotoxin-induced uveitis (EIU) clinical score or the number of infiltrating cells and cytokines in ocular media (interleukin [IL]-1ß, IL-6, tumor necrosis factor [TNF]-α, and monocyte chemoattractant protein [MCP]-1). Eyes treated with anti-VEGF showed a significantly decreased number of activated microglia and macrophages in the retina and the choroid and decreased iNOS-positive microglia. IBA1-positive cells expressed VEGF-R1 and R2 in the inflamed retina. CONCLUSIONS: Microglia and macrophages expressed VEGF receptors, and intravitreous anti-VEGF influenced the microglia and macrophage activation state. Taking into account that anti-VEGF drugs are repeatedly injected in the vitreous of patients with retinal diseases, part of their effects could result from unsuspected modulation of the microglia activation state. This should be further studied in other ocular pathogenic conditions and human pathology.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Macrophage Activation/drug effects , Microglia/pathology , Retina/pathology , Uveitis/drug therapy , Uveitis/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antibodies, Neutralizing/pharmacology , Calcium-Binding Proteins/metabolism , Cell Count , Disease Models, Animal , Humans , Lipopolysaccharides , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Neutralization Tests , Nitric Oxide Synthase Type II/metabolism , Rats , Receptors, Vascular Endothelial Growth Factor/metabolism , Retina/drug effects , Retina/enzymology , Vascular Endothelial Growth Factor A/metabolism
5.
PLoS One ; 7(11): e49036, 2012.
Article in English | MEDLINE | ID: mdl-23152847

ABSTRACT

We have previously shown that the eye is a mineralocorticoid-sensitive organ and we now question the role of mineralocorticoid receptor (MR) in ocular inflammation. The endotoxin-induced uveitis (EIU), a rat model of human intraocular inflammation, was induced by systemic administration of lipopolysaccharide (LPS). Evaluations were made 6 and 24 hours after intraocular injection of aldosterone (simultaneous to LPS injection). Three hours after onset of EIU, the MR and the glucocorticoid metabolizing enzyme 11-beta hydroxysteroid dehydrogenase type 2 (11ß-HSD2) expression were down-regulated in iris/ciliary body and the corticosterone concentration was increased in aqueous humor, altering the normal MR/glucocorticoid receptor (GR) balance. At 24 hours, the GR expression was also decreased. In EIU, aldosterone reduced the intensity of clinical inflammation in a dose-dependent manner. The clinical benefit of aldosterone was abrogated in the presence of the MR antagonist (RU26752) and only partially with the GR antagonist (RU38486). Aldosterone reduced the release of inflammatory mediators (6 and 24 hours: TNF-α, IFN-γ, MIP-1α) in aqueous humor and the number of activated microglia/macrophages. Aldosterone partly prevented the uveitis-induced MR down-regulation. These results suggest that MR expression and activation in iris/ciliary body could protect the ocular structures against damages induced by EIU.


Subject(s)
Anti-Inflammatory Agents/metabolism , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Uveitis/metabolism , Uveitis/pathology , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Aldosterone/administration & dosage , Aldosterone/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Aqueous Humor/drug effects , Aqueous Humor/metabolism , Chemokines/metabolism , Ciliary Body/enzymology , Ciliary Body/pathology , Down-Regulation/drug effects , Down-Regulation/genetics , Endotoxins , Female , Humans , Inflammation Mediators/metabolism , Intravitreal Injections , Iris/drug effects , Iris/enzymology , Iris/pathology , Lipopolysaccharides , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats , Rats, Inbred Lew , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Signal Transduction/drug effects , Spironolactone/administration & dosage , Spironolactone/pharmacology , Uveitis/chemically induced , Uveitis/drug therapy
6.
Am J Pathol ; 179(2): 942-53, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21712024

ABSTRACT

Diabetic retinopathy is associated with ocular inflammation, leading to retinal barrier breakdown, macular edema, and visual cell loss. We investigated the molecular mechanisms involved in microglia/macrophages trafficking in the retina and the role of protein kinase Cζ (PKCζ) in this process. Goto Kakizaki (GK) rats, a model for spontaneous type 2 diabetes were studied until 12 months of hyperglycemia. Up to 5 months, sparse microglia/macrophages were detected in the subretinal space, together with numerous pores in retinal pigment epithelial (RPE) cells, allowing inflammatory cell traffic between the retina and choroid. Intercellular adhesion molecule-1 (ICAM-1), caveolin-1 (CAV-1), and PKCζ were identified at the pore border. At 12 months of hyperglycemia, the significant reduction of pores density in RPE cell layer was associated with microglia/macrophages accumulation in the subretinal space together with vacuolization of RPE cells and disorganization of photoreceptors outer segments. The intraocular injection of a PKCζ inhibitor at 12 months reduced iNOS expression in microglia/macrophages and inhibited their migration through the retina, preventing their subretinal accumulation. We show here that a physiological transcellular pathway takes place through RPE cells and contributes to microglia/macrophages retinal trafficking. Chronic hyperglycemia causes alteration of this pathway and subsequent subretinal accumulation of activated microglia/macrophages.


Subject(s)
Diabetic Retinopathy/metabolism , Epithelial Cells/metabolism , Macrophages/metabolism , Microglia/metabolism , Protein Kinase C/metabolism , Retina/metabolism , Animals , Blood Glucose/metabolism , Cell Movement , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Lymphocytes/cytology , Microscopy, Confocal/methods , Rats , Rats, Wistar
7.
Invest Ophthalmol Vis Sci ; 52(8): 5414-23, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21666239

ABSTRACT

PURPOSE: To test the efficiency of locally administrated tresperimus in experimental autoimmune uveoretinitis (EAU). METHODS: EAU was induced in Lewis rats by S-antigen (S-Ag) immunization. Three intravitreal injections of tresperimus (prevention or prevention/treatment protocols) were performed at different time points after immunization. The pharmacokinetics of tresperimus was evaluated in the ocular tissues and plasma. The in vitro effect of tresperimus was evaluated on macrophages. EAU was graded clinically and histologically. Blood ocular barrier permeability was evaluated by protein concentration in ocular fluids. Immune response to S-Ag was examined by delayed type hypersensitivity, the expression of inflammatory cytokines in lymph nodes, ocular fluids and serum by multiplex ELISA, and in ocular cells by RT-PCR. RESULTS: In vitro, tresperimus significantly reduced the production of inflammatory cytokines by lipopolysaccharide-stimulated macrophages. In vivo, in the treatment protocol, efficient tresperimus levels were measured in the eye but not in the plasma up to 8 days after the last injection. Tresperimus efficiently reduced inflammation, retinal damage, and blood ocular barrier permeability breakdown. It inhibited nitric oxide synthase-2 and nuclear factor κBp65 expression in ocular macrophages. IL-2 and IL-17 were decreased in ocular media, while IL-18 was increased. By contrast, IL-2 and IL-17 levels were not modified in inguinal lymph nodes draining the immunization site. Moreover, cytokine levels in serum and delayed type hypersensitivity to S-Ag were not different in control and treated rats. In the prevention/treatment protocol, ocular immunosuppressive effects were also observed. CONCLUSIONS: Locally administered tresperimus appears to be a potential immunosuppressive agent in the management of intraocular inflammation.


Subject(s)
Autoimmune Diseases/prevention & control , Carbamates/administration & dosage , Disease Models, Animal , Immunosuppressive Agents/administration & dosage , Retinitis/prevention & control , Uveitis/prevention & control , Animals , Aqueous Humor/metabolism , Arrestin/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Blood-Retinal Barrier/drug effects , Capillary Permeability/drug effects , Carbamates/pharmacokinetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique, Indirect , Hypersensitivity, Delayed/immunology , Immunosuppressive Agents/pharmacokinetics , Intravitreal Injections , Lymph Nodes/immunology , Macrophages/drug effects , Macrophages/metabolism , RNA/isolation & purification , Rats , Rats, Inbred Lew , Retinitis/immunology , Retinitis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Uveitis/immunology , Uveitis/pathology , Vitreous Body/metabolism
8.
Invest Ophthalmol Vis Sci ; 51(9): 4683-93, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20393119

ABSTRACT

PURPOSE: To evaluate the effect of XG-102 (formerly D-JNKI1), a TAT-coupled dextrogyre peptide that selectively inhibits the c-Jun N-terminal kinase, in the treatment of endotoxin-induced uveitis (EIU). METHODS: EIU was induced in Lewis rats by LPS injection. XG-102 was administered at the time of LPS challenge. The ocular biodistribution of XG-102 was evaluated using immunodetection at 24 hours after either 20 microg/kg IV (IV) or 0.2 microg/injection intravitreous (IVT) administrations in healthy or uveitic eyes. The effect of XG-102 on EIU was evaluated using clinical scoring, infiltration cell quantification, inducible nitric oxide synthase (iNOS) expression and immunohistochemistry, and cytokines and chemokines kinetics at 6, 24, and 48 hours using multiplex analysis on ocular media. Control EIU eyes received vehicle injection IV or IVT. The effect of XG-102 on c-Jun phosphorylation in EIU was evaluated by Western blot in eye tissues. RESULTS: After IVT injection, XG-102 was internalized in epithelial cells from iris/ciliary body and retina and in glial and microglial cells in both healthy and uveitic eyes. After IV injection, XG-102 was concentrated primarily in inflammatory cells of uveitic eyes. Using both routes of administration, XG-102 significantly inhibited clinical signs of EIU, intraocular cell infiltration, and iNOS expression together with reduced phosphorylation of c-Jun. The anti-inflammatory effect of XG-102 was mediated by iNOS, IFN-gamma, IL-2, and IL-13. CONCLUSIONS: This is the first evidence that interfering with the JNK pathway can reduce intraocular inflammation. Local administration of XG-102, a clinically evaluated peptide, may have potential for treating uveitis.


Subject(s)
JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Peptides/pharmacology , Signal Transduction/drug effects , Uveitis/drug therapy , Animals , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Drug Combinations , Female , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/drug effects , Injections, Intraocular , Injections, Intravenous , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/toxicity , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oils , Peptides/pharmacokinetics , Phenols , Rats , Rats, Inbred Lew , Tissue Distribution , Uveitis/chemically induced , Uveitis/pathology , Vitreous Body
9.
Graefes Arch Clin Exp Ophthalmol ; 248(4): 573-84, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19956968

ABSTRACT

BACKGROUND: Patients with ocular toxoplasmosis (OT) develop autoreactivity to several retinal antigens, including retinal S-antigen. By establishing an experimental rabbit model of systemic and of primary and secondary ocular toxoplasmosis, we wished to investigate the onset and development of humoral response to retinal S-antigen. METHODS: Of twelve infection-naïve rabbits, six were left untreated, and the other six were infected subcutaneously with 5,000 tachyzoites of the highly virulent, non-cyst-forming BK-strain of Toxoplasma gondii. Three months later, the left eye of each animal was infected transvitreally with 5,000 tachyzoites of the same strain. The right eye of each rabbit served as an uninfected control. Blood and aqueous humor were collected prior to infection, and up to 90 days thereafter. Using the ELISA technique, all samples were analyzed in parallel for total IgG, and antibodies against toxoplasmic, bovine retinal S-antigen and peptide 35 from human S-antigen. RESULTS: In infection-naïve rabbits Toxoplasma-specific antibodies were detected 10 to 15 days after systemic and ocular infection. Serum antibodies against retinal S-antigen and peptide 35 were not detected in response to systemic Toxoplasma infection. After ocular challenge, aqueous-humour levels of antibodies against retinal S-antigen and peptide 35 in the infected eye began to rise 10 to 15 days later in infection-naïve, but not in infection-immunized animals. During the early post-infection period, the concentrations of anti-retinal antibodies in the infected eye correlated with the severity of inflammatory tissue destruction, but returned to baseline later even though the inflammatory response persisted. In the uninfected partner eye, concentrations of anti-retinal and toxoplasmic antibodies did not correlate with each other. CONCLUSION: Our data afford no evidence of similarities between toxoplasmic and retinal antigens, nor of infection-induced humoral autoimmunity. They indicate rather that retinal autoantigens are liberated in the context of inflammatory tissue destruction due to ocular toxoplasmosis.


Subject(s)
Antibodies, Protozoan/blood , Arrestin/immunology , Autoantibodies/blood , Chorioretinitis/immunology , Toxoplasma/immunology , Toxoplasmosis, Ocular/immunology , Animals , Aqueous Humor/immunology , Autoimmunity , Chorioretinitis/parasitology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Humoral , Immunoglobulin G/blood , Male , Rabbits , Toxoplasmosis, Ocular/parasitology
10.
J Control Release ; 139(1): 22-30, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19481575

ABSTRACT

We evaluated the benefits of a novel formulation of vasoactive intestinal peptide (VIP) based on the incorporation of VIP-loaded rhodamine-conjugated liposomes (VIP-Rh-Lip) within hyaluronic acid (HA) gel (Gel-VIP-Rh-Lip) for the treatment of endotoxin-induced uveitis (EIU) in comparison with VIP-Rh-Lip alone. In vitro release study and rheological analysis showed that interactions between HA chains and liposomes resulted in increased viscosity and reinforced elasticity of the gel. In vivo a single intravitreal injection of Gel-VIP-Rh-Lip was performed in rats 7 days prior to uveitis induction by subcutaneous lipopolysaccharide injection. The maximal ocular inflammation occurs within 16-24 h in controls (VIP-Rh-Lip, unloaded-Rh-Lip). Whereas intraocular injection of VIP-Rh-Lip had no effect on EIU severity compared with controls, Gel-VIP-Rh-Lip reduced significantly the clinical score and number of inflammatory cells infiltrating the eye. The fate of liposomes, VIP and HA in the eyes, regional and inguinal lymph nodes and spleen was analyzed by immunostaining and fluorescence microscopy. Retention of liposomes by HA gel was observed in vitro and in vivo. Inflammation severity seemed to impact on system stability resulting in the delayed release of VIP. Thus, HA gel containing VIP-Rh-Lip is an efficient strategy to obtain a sustained delivery of VIP in ocular and lymph node tissues.


Subject(s)
Hyaluronic Acid/chemistry , Uveitis/drug therapy , Vasoactive Intestinal Peptide/administration & dosage , Algorithms , Animals , Chemistry, Pharmaceutical , Drug Carriers , Elasticity , Endotoxins , Fluoresceins/chemistry , Gels , Injections , Liposomes , Male , Particle Size , Rats , Rats, Inbred Lew , Rheology , Uveitis/chemically induced , Vasoactive Intestinal Peptide/therapeutic use , Viscosity , Vitreous Body
11.
Invest Ophthalmol Vis Sci ; 50(4): 1761-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19321795

ABSTRACT

PURPOSE: Plasmid electrotransfer in the ciliary muscle allows the sustained release of therapeutic proteins within the eye. The aim of this study was to evaluate whether the ocular production of TNF-alpha soluble receptor, using this nonviral gene therapy method, could have a beneficial local effect in a model of experimental autoimmune uveoretinitis (EAU). METHODS: Injection of a plasmid encoding a TNF-alpha p55 receptor (30 microg) in the ciliary muscle, combined with electrotransfer (200 V/cm), was carried out in Lewis rat eyes 4 days before the induction of EAU by S-antigen. Control eyes received naked plasmid electrotransfer or simple injection of the therapeutic plasmid. The disease was evaluated clinically and histologically. Cytokines and chemokines were analyzed in the ocular media by multiplex assay performed 15 and 21 days after immunization. RESULTS: Ocular TNF-alpha blockade, resulting from the local secretion of soluble receptors, was associated with delayed and significantly less severe uveitis, together with a reduction of the retinal damages. Compared with the controls, treated eyes showed significantly lower levels of IL-1beta and MCP1, higher levels of IL-13 and IL-4, and reduced NOS-2 expression in infiltrating cells. Treatment did not influence TNF-alpha levels in inguinal lymph nodes. CONCLUSIONS: Taken together, these results indicate that local immunomodulation was achieved and that no systemic adverse effects of TNF-alpha blockade observed after systemic injection of TNF-alpha inhibitors should be expected.


Subject(s)
Autoimmune Diseases/therapy , Ciliary Body/metabolism , Genetic Therapy/methods , Muscle, Smooth/metabolism , Plasmids/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor Decoy Receptors/genetics , Uveitis, Posterior/therapy , Animals , Arrestin , Autoimmune Diseases/metabolism , Chemokines/metabolism , Disease Models, Animal , Electroporation/methods , Fluorescent Antibody Technique, Indirect , Gene Expression , Male , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Uveitis, Posterior/metabolism
12.
J Ocul Pharmacol Ther ; 25(1): 9-21, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19232006

ABSTRACT

PURPOSE: The aim of this study was to investigate the effect of a single intravitreal (i.v.t.) injection of vasoactive intestinal peptide (VIP) loaded in rhodamine-conjugated liposomes (VIP-Rh-Lip) on experimental autoimmune uveoretinitis (EAU). METHODS: An i.v.t. injection of VIP-Rh-Lip, saline, VIP, or empty-(E)-Rh-Lip was performed simultaneously, either 6 or 12 days after footpad immunization with retinal S-antigen in Lewis rats. Clinical and histologic scores were determined. Immunohistochemistry and cytokine quantification by multiplex enzyme-linked immunosorbent assay were performed in ocular tissues. Systemic immune response was determined at day 20 postimmunization by measuring proliferation and cytokine secretion of cells from inguinal lymph nodes (ILNs) draining the immunization site, specific delayed-type hypersensitivity (DTH), and the serum concentration of cytokines. Ocular and systemic biodistribution of VIP-Rh-Lip was studied in normal and EAU rats by immunofluorescence. RESULTS: The i.v.t. injection of VIP-Rh-Lip performed during the afferent, but not the efferent, phase of the disease reduced clinical EAU and protected against retinal damage. No effect was observed after saline, E-Rh-Lip, or VIP injection. VIP-Rh-Lip and VIP were detected in intraocular macrophages and in lymphoid organs. In VIP-Rh-Lip-treated eyes, macrophages expressed transforming growth factor-beta2, low levels of major histocompatibility complex class II, and nitric oxide synthase-2. T-cells showed activated caspase-3 with the preservation of photoreceptors. Intraocular levels of interleukin (IL)-2, interferon-gamma (IFN-gamma), IL-17, IL-4, GRO/KC, and CCL5 were reduced with increased IL-13. At the systemic level, treatment reduced retinal soluble autoantigen lymphocyte proliferation, decreased IL-2, and increased IL-10 in ILN cells, and diminished specific DTH and serum concentration of IL-12 and IFN-gamma. CONCLUSIONS: An i.v.t. injection of VIP-Rh-Lip, performed during the afferent stage of immune response, reduced EAU pathology through the immunomodulation of intraocular macrophages and deviant stimulation of T-cells in ILN. Thus, the encapsulation of VIP within liposomes appears as an effective strategy to deliver VIP into the eye and is an efficient means of the prevention of EAU severity.


Subject(s)
Autoimmune Diseases/prevention & control , Retinitis/prevention & control , Uveitis/prevention & control , Vasoactive Intestinal Peptide/administration & dosage , Animals , Arrestin/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Proliferation , Cytokines/biosynthesis , Disease Models, Animal , Injections , Liposomes , Lymph Nodes/metabolism , Lymph Nodes/pathology , Macrophages/immunology , Male , Rats , Rats, Inbred Lew , Retinitis/immunology , Retinitis/pathology , Rhodamines , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Uveitis/immunology , Uveitis/pathology , Vasoactive Intestinal Peptide/pharmacokinetics , Vitreous Body
14.
Ophthalmic Res ; 40(3-4): 175-80, 2008.
Article in English | MEDLINE | ID: mdl-18421235

ABSTRACT

PURPOSE: A major increased risk of developing birdshot chorioretinopathy is reported in humans who are HLA-A29-positive. To better characterize this disease, an animal model of HLA-A29-associated disease was developed and the pathology arising spontaneously in these transgenic mice was compared to animal models of autoimmune uveoretinitis and to human pathology. MATERIALS AND METHODS: HLA-A2902 cDNA (A29c) was obtained from a patient suffering from birdshot retinochoroidopathy and used for transgene construct to generate HLA-A29 transgenic mice. Histopathological examination of the animal cohort was performed up to 15 months of age. It was compared with the ocular pathology developed in C57BL/6 mice and in Lewis rats immunized with retinal autoantigens. RESULTS: Aging HLA-A29 transgenic mice spontaneously developed an ocular disease with resemblance to experimental retinal-Ag-induced autoimmune ocular disease and to human pathologies shown in birdshot retinochoroidopathy, Vogt-Koyanagi-Harada and sympathetic ophthalmia. Pathogenic mechanisms could possibly be shared by these conditions. CONCLUSION: Humanized models of ocular inflammation developed in HLA class I and class II transgenic mice will help better understand the mechanisms responsible for ocular inflammation. Local control of autoimmunity in HLA-A29-positive individuals would be an important option for new therapeutic strategies.


Subject(s)
Autoimmune Diseases/pathology , Autoimmunity/immunology , Disease Models, Animal , Retina/immunology , Uveitis/pathology , Animals , Autoimmune Diseases/immunology , Disease Progression , Female , Follow-Up Studies , HLA-A Antigens/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Rats, Inbred Lew , Severity of Illness Index , Uveitis/immunology , beta 2-Microglobulin/immunology
15.
Ophthalmic Res ; 40(3-4): 181-8, 2008.
Article in English | MEDLINE | ID: mdl-18421236

ABSTRACT

PURPOSE: We aimed to describe the clinical and angiographic changes in an experimental model of autoimmune uveoretinitis and vasculitis in primates. METHODS: Six cynomolgus monkeys received a single subcutaneous immunization with 100 microg of human S antigen with complete Freund's adjuvant. RESULTS: All the animals had a bilateral long-term disease occurring usually in 1 eye approximately 4 weeks after immunization, the second eye being involved 1-5 weeks later. A cyclic course of the disease could be demonstrated by repeated fundus fluorescein angiograms. The initial and principal manifestation consisted in retinal vascular sheathing affecting veins and venules. The more severe forms showed areas of posterior uveoretinitis, dense vitritis and anterior uveitis. CONCLUSION: A single systemic injection of pure human retinal S antigen could induce a chronic and recurrent ocular disease similar to human retinal vasculitis.


Subject(s)
Disease Models, Animal , Retina/pathology , Retinal Vasculitis/diagnosis , Retinal Vein/pathology , Retinitis/diagnosis , Uveitis/diagnosis , Vitreous Body/pathology , Adjuvants, Immunologic , Animals , Arrestin , Disease Progression , Fluorescein Angiography , Follow-Up Studies , Freund's Adjuvant , Fundus Oculi , Immunization/adverse effects , Laser Coagulation , Macaca fascicularis , Microscopy, Acoustic , Recurrence , Retina/diagnostic imaging , Retinal Vasculitis/chemically induced , Retinal Vasculitis/surgery , Retinitis/chemically induced , Retinitis/surgery , Severity of Illness Index , Uveitis/chemically induced , Uveitis/surgery , Vitreous Body/diagnostic imaging
16.
Ophthalmic Res ; 40(3-4): 208-11, 2008.
Article in English | MEDLINE | ID: mdl-18421241

ABSTRACT

Primary intraocular lymphoma is a high-grade non-Hodgkin lymphoma whose pathogenesis is still unclear. Few animal models exist in order to study this condition. Although intraocular lymphomas in humans are usually B cell lymphomas, most of these models are T cell lymphomas. Recently, a major step forward has been realized with the development of new models of intraocular B cell lymphoma. New therapeutic tools are being evaluated in these models of B cell lymphoma. We evaluate the contribution of the different animal models available to study intraocular lymphomas, and we discuss the new therapeutic strategies and their various targets in the tumor as well as in the environment, which are currently investigated through the development of these models.


Subject(s)
Disease Models, Animal , Eye Neoplasms/pathology , Lymphoma/pathology , Animals , Cytokines/immunology , Cytokines/metabolism , Eye Neoplasms/immunology , Eye Neoplasms/therapy , Lymphoma/immunology , Lymphoma/therapy , Neoplasms, Experimental
17.
Invest Ophthalmol Vis Sci ; 48(7): 3223-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591892

ABSTRACT

PURPOSE: Primary intraocular lymphoma is a high-grade non-Hodgkin lymphoma with a pathogenesis that is still unclear. Microenvironment is known to be crucial in controlling tumor growth and maintenance. To study the immune microenvironment in intraocular lymphomas and to characterize the cytokine polarization of infiltrating T-lymphocytes, a new murine model of intraocular B-cell lymphoma was developed. METHODS: Immunocompetent adult mice were injected intravitreally with a syngeneic lymphomatous B-cell line. Clinical, histologic, and flow cytometric analyses were performed to characterize the tumoral invasion and the immune infiltration. Cytokine production of ocular cells was investigated by RT-PCR and fluorescent immunoassay, with or without stimulation by anti-CD3(+) anti-CD28 antibodies. RESULTS: Intraocular lymphoma developed in eyes injected by lymphomatous B-cells. At day 19, the retina and the vitreous cavity were infiltrated by tumor cells. Up to 15% of living cells were T-lymphocytes. Cytokine profile analysis of the supernatant of ocular cells cultured ex vivo demonstrated the presence of IL10, IL6, IFNgamma, and TNFalpha. Stimulation of ocular cells with anti-CD3(+) anti-CD28 antibodies increased the IFNgamma level and led to the induction of IL2 production, completing the type 1 (Th1/Tc1-like) pattern of cytokine expression observed. IL12p70 and IL4, potent Th1 or Th2 differentiating factors, were undetectable, even after stimulation. CONCLUSIONS: The results suggest that T-cells from intraocular B-lymphomas are characterized by a Th1/Tc1-like profile that could be partially inhibited in vivo. These data raise the possibility of a T-cell immunostimulation to reactivate the Th1/Tc1-lymphocytes and improve intraocular antitumoral immunity.


Subject(s)
Cytokines/biosynthesis , Eye Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, B-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Vitreous Body/pathology , Animals , Cell Line , Cytokines/genetics , Disease Models, Animal , Eye Neoplasms/pathology , Female , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Lymphoma, B-Cell/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction , Transfection
18.
Invest Ophthalmol Vis Sci ; 48(7): 3230-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591893

ABSTRACT

PURPOSE: To reestablish the immunosuppressive microenvironment of the eye, disrupted by ocular inflammation during endotoxin-induced uveitis (EIU), by means of intravitreal injection of vasoactive intestinal peptide (VIP) in saline or encapsulated in liposomes, to increase its bioavailability and efficiency. METHODS: EIU was induced in Lewis rats by subcutaneous injection of lipopolysaccharide (LPS). Simultaneously, animals were intravitreally injected with saline, saline/VIP, VIP-loaded liposomes (VIP-Lip), or unloaded liposomes. EIU severity and cellular infiltration were assessed by clinical examination and specific immunostaining. VIP concentration was determined in ocular fluids by ELISA. Ocular expression of inflammatory cytokine and chemokine mRNAs was detected by semiquantitative RT-PCR. Biodistribution of rhodamine-conjugated liposomes (Rh-Lip) was analyzed by immunohistochemistry in eyes and regional cervical lymph nodes (LNs). RESULTS: Twenty-four hours after intravitreal injection of VIP-Lip, VIP concentration in ocular fluids was 15 times higher than after saline/VIP injection. At that time, EIU clinical severity, ocular infiltrating polymorphonuclear leukocytes (PMNs), and, to a lesser extent, ED1(+) macrophages, as well as inflammatory cytokine and chemokine mRNA expression, were significantly reduced in VIP-Lip-injected rats compared with rats injected with saline/VIP, unloaded liposomes, or saline. Rh-Lip was distributed in vitreous, ciliary body, conjunctiva, retina, and sclera. It was internalized by macrophages and PMNs, and VIP colocalized with liposomes at least up to 14 days after injection. In cervical LNs, resident macrophages internalized VIP-Rh-Lip, and some adjacent lymphocytes showed VIP expression. CONCLUSIONS: VIP was efficient at reducing EIU only when formulated in liposomes, which enhanced its immunosuppressive effect and controlled its delivery to all tissues affected by or involved in ocular inflammation.


Subject(s)
Lipopolysaccharides , Salmonella typhimurium , Uveitis/metabolism , Vasoactive Intestinal Peptide/administration & dosage , Vitreous Body/drug effects , Animals , Aqueous Humor/metabolism , Cytokines/genetics , Down-Regulation , Immunotherapy , Injections , Liposomes , Lymph Nodes/metabolism , Macrophages/immunology , Male , Microscopy, Confocal , Microscopy, Fluorescence , Neutrophils/immunology , Phosphatidylethanolamines , Polyethylene Glycols , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Uveitis/genetics , Vasoactive Intestinal Peptide/pharmacokinetics , Vitreous Body/metabolism
19.
Am J Pathol ; 170(4): 1241-57, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392164

ABSTRACT

We show that inhibitory effect of interleukin-13 on endotoxin-induced uveitis in the Lewis rat is dependent on signaling activity of protein kinase Czeta (PKCzeta). To understand the effect of interleukin-13 or PKCzeta inhibitor treatment, the activation status of rat bone marrow-derived macrophages was studied in vitro. At 6 hours, lipopolysaccharide-stimulated macrophages produced tumor necrosis factor-alpha (TNF-alpha) with nuclear factor kappaB (NF-kappaB)/p65 expression. Treatment led to absence of NF-kappaB/p65 expression and low levels of TNF-alpha, suggesting accelerated inactivation of macrophages. At 24 hours after lipopolysaccharide stimulation, nuclear NF-kappaB/p65 decreased and nuclear NF-kappaB/p50 increased, associated with nuclear BCL-3 and a low level of TNF-alpha, indicating onset of spontaneous resolution. Treatment limited PKCzeta cleavage, with expression of nuclear NF-kappaB/p50 and BCL-3 and low nuclear NF-kappaB/p65 promoting macrophage survival, as evidenced by Bcl-2 expression. At 24 hours, intraocular treatment decreased membranous expression of PKCzeta by ocular cells, reduced vascular leakage with low nitric-oxide synthase-2 expression in vascular endothelial cells, and limited inflammatory cell infiltration with decreased intraocular TNF-alpha, interleukin-6, and nitric-oxide synthase-2 mRNA. Importantly, treatment decreased nuclear NF-kappaB/p65, increased transforming growth factor-beta2, and reduced caspase 3 expression in infiltrating macrophages, implying a change of their phenotype within ocular microenvironment. Treatment accelerated endotoxin-induced uveitis resolution through premature apoptosis of neutrophils related to high expression of toll-like receptor 4 and caspase 3.


Subject(s)
Apoptosis/physiology , Inflammation/physiopathology , Protein Kinase C/metabolism , Uveitis/physiopathology , Animals , Apoptosis/drug effects , Blotting, Western , Caspase 3/metabolism , Cytokines/genetics , Cytokines/metabolism , Eye/drug effects , Eye/metabolism , Eye/pathology , Eye Proteins/metabolism , Immunohistochemistry , Inflammation/pathology , Inflammation/prevention & control , Interleukin-13/pharmacology , Lipopolysaccharides/toxicity , Macrophages/drug effects , Macrophages/metabolism , Male , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Uveitis/chemically induced , Uveitis/prevention & control
20.
J Immunol ; 176(12): 7171-9, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16751360

ABSTRACT

In many clinical cases, uveitis develops secondary to an infection. This could result from peripheral activation followed by ocular penetration and reactivation of T cells specific for microbial Ags expressed in the retina. To gain insights into the pathophysiology of uveitis, we developed a new mouse model based on stable retinal expression of influenza virus hemagglutinin (HA) neoantigen by adeno-associated virus-mediated gene transfer. One month thereafter, we adoptively transferred HA-specific T cells, which were activated in vitro or in vivo. Intraocular inflammation was clinically and histologically observed in all animals within 15 days. The ocular infiltrate was composed mostly of macrophages and HA-specific T cells with a proinflammatory cytokine profile. Depletion of CD4(+)CD25(+) regulatory T cells exacerbated the disease, whereas HA-specific CD4(+)CD25(+) T cells given i.v. controlled the disease. This novel model should allow to better study the pathophysiology and therapeutic of uveitis.


Subject(s)
Antigens, Viral/biosynthesis , Retina/immunology , Retinitis/immunology , Retinitis/prevention & control , T-Lymphocytes, Regulatory/transplantation , Th1 Cells/immunology , Uveitis/immunology , Uveitis/prevention & control , Animals , Antigens, Viral/genetics , Cell Movement/genetics , Cell Movement/immunology , Cytokines/biosynthesis , Dependovirus/genetics , Dependovirus/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Radiation Chimera , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Retina/pathology , Retina/virology , Retinitis/pathology , Retinitis/virology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/virology , Th1 Cells/pathology , Th1 Cells/virology , Uveitis/pathology , Uveitis/virology , Vitreous Body/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...