Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 222
1.
Cells ; 10(8)2021 08 03.
Article En | MEDLINE | ID: mdl-34440742

Glaucoma is a multifactorial disease resulting in progressive vision loss due to retinal ganglion cell (RGC) dysfunction and death. Early events in the pathobiology of the disease include oxidative, metabolic, or mechanical stress that acts upon RGC, causing these to rapidly release danger signals, including extracellular ATP, resulting in micro- and macroglial activation and neuroinflammation. Danger signaling also leads to the formation of inflammasomes in the retina that enable maturation of proinflammatory cytokines such IL-1ß and IL-18. Chronic neuroinflammation can have directly damaging effects on RGC, but it also creates a proinflammatory environment and compromises the immune privilege of the retina. In particular, continuous synthesis of proinflammatory mediators such as TNFα, IL-1ß, and anaphylatoxins weakens the blood-retina barrier and recruits or activates T-cells. Recent data have demonstrated that adaptive immune responses strongly exacerbate RGC loss in animal models of the disease as T-cells appear to target heat shock proteins displayed on the surface of stressed RGC to cause their apoptotic death. It is possible that dysregulation of these immune responses contributes to the continued loss of RGC in some patients.


Glaucoma/pathology , Retinal Ganglion Cells/metabolism , Adenosine Triphosphate/metabolism , Cytokines/metabolism , Glaucoma/immunology , Glaucoma/metabolism , Humans , Immunity, Innate , Inflammasomes/metabolism , Signal Transduction
2.
Life Sci ; 282: 119796, 2021 Oct 01.
Article En | MEDLINE | ID: mdl-34245774

Adenosine, an endogenous purine nucleoside, is a well-known actor of the immune system and the inflammatory response both in physiologic and pathologic conditions. By acting upon particular, G-protein coupled adenosine receptors, i.e., A1, A2- a & b, and A3 receptors mediate a variety of intracellular and immunomodulatory actions. Several studies have elucidated Adenosine's effect and its up-and downstream molecules and enzymes on the anti-tumor response against several types of cancers. We have also targeted a couple of molecules to manipulate this pathway and get the immune system's desired response in our previous experiences. Besides, the outgrowth of the studies on ocular Adenosine in recent years has significantly enhanced the knowledge about Adenosine and its role in ocular immunology and the inflammatory response of the eye. Glaucoma is the second leading cause of blindness globally, and the recent application of Adenosine and its derivatives has shown the critical role of the adenosine pathway in its pathophysiology. However, despite a very promising background, the phase III clinical trial of Trabodenoson failed to achieve the non-inferiority goals of the study. In this review, we discuss different aspects of the abovementioned pathway in ophthalmology and ocular immunology; following a brief evaluation of the current immunotherapeutic strategies, we try to elucidate the links between cancer immunotherapy and glaucoma in order to introduce novel therapeutic targets for glaucoma.


Adenosine/immunology , Glaucoma/immunology , Neoplasms/immunology , Animals , Eye/immunology , Glaucoma/therapy , Humans , Immunity , Immunotherapy , Neoplasms/therapy
3.
Front Immunol ; 12: 803485, 2021.
Article En | MEDLINE | ID: mdl-34975917

Glaucoma as the leading neurodegenerative disease leads to blindness in 3.6 million people aged 50 years and older worldwide. For many decades, glaucoma therapy has primarily focused on controlling intraocular pressure (IOP) and sound evidence supports its role in delaying the progress of retinal ganglial cell (RGC) damage and protecting patients from vision loss. Meanwhile, accumulating data point to the immune-mediated attack of the neural retina as the underlying pathological process behind glaucoma that may come independent of raised IOP. Recently, some scholars have suggested autoimmune aspects in glaucoma, with autoreactive T cells mediating the chief pathogenic process. This autoimmune process, as well as the pathological features of glaucoma, largely overlaps with other neurodegenerative diseases in the central nervous system (CNS), including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. In addition, immune modulation therapy, which is regarded as a potential solution for glaucoma, has been boosted in trials in some CNS neurodegenerative diseases. Thus, novel insights into the T cell-mediated immunity and treatment in CNS neurodegenerative diseases may serve as valuable inspirations for ophthalmologists. This review focuses on the role of T cell-mediated immunity in the pathogenesis of glaucoma and discusses potential applications of relevant findings of CNS neurodegenerative diseases in future glaucoma research.


Autoimmunity , Glaucoma/immunology , Immunity, Cellular , Nerve Degeneration , Neuroglia/immunology , Retinal Neurons/immunology , T-Lymphocytes/immunology , Animals , Bacteria/immunology , Bacteria/metabolism , Chemotaxis, Leukocyte , Dysbiosis , Gastrointestinal Microbiome , Glaucoma/metabolism , Glaucoma/microbiology , Glaucoma/pathology , Gliosis , Host-Pathogen Interactions , Humans , Neuroglia/metabolism , Neuroglia/pathology , Retinal Neurons/metabolism , Retinal Neurons/pathology , T-Lymphocytes/metabolism
4.
Invest Ophthalmol Vis Sci ; 61(14): 18, 2020 12 01.
Article En | MEDLINE | ID: mdl-33320171

Purpose: We previously demonstrated that passive transfer of lymphocytes from glaucomatous mice induces retinal ganglion cell (RGC) damage in recipient animals, suggesting a role for immune responses in the multifactorial pathophysiology of glaucoma. Here we evaluate whether absence of an adaptive immune response reduces RGC loss in glaucoma. Methods: Elevated intraocular pressure (IOP) was induced in one eye of C57BL/6J (B6) or T- and B-cell-deficient Rag1-/- knockout mice. After 16 weeks RGC density was determined in both the induced and the normotensive contralateral eyes. Data were compared to mice having received injections of "empty" vector (controls). The number of extravascular CD3+ cells in the retinas was determined using FACS. Results: Retinas of eyes with elevated IOP contain significantly more extravasated CD3+ cells than control retinas (46.0 vs. 27.1, P = 0.025). After 16 weeks of elevated IOP the average RGC density in B6 mice decreased by 20.7% (P = 1.9 × 10-4). In contrast, RGC loss in Rag1-/- eyes with elevated IOP was significantly lower (10.3%, P = 0.006 vs. B6). RGC loss was also observed in the contralateral eyes of B6 mice, despite the absence of elevated IOP in those eyes (10.1%; P = 0.008). In RAG1-/- loss in the contralateral eyes was minimal (3.1%) and significantly below that detected in B6 (P = 0.02). Conclusions: Our findings demonstrate that T Rag1-/- mice are significantly protected from glaucomatous RGC loss. In this model, lymphocyte activity contributes to approximately half of all RGC loss in eyes with elevated IOP and to essentially all loss observed in normotensive contralateral eyes.


B-Lymphocytes/pathology , Glaucoma/immunology , Homeodomain Proteins/physiology , Retinal Ganglion Cells/pathology , T-Lymphocytes/pathology , Animals , B-Lymphocytes/immunology , Cell Count , Disease Models, Animal , Female , Flow Cytometry , Glaucoma/pathology , Intraocular Pressure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
5.
Front Immunol ; 11: 566279, 2020.
Article En | MEDLINE | ID: mdl-33162981

Previous studies demonstrated that retinal damage correlates with a massive remodeling of extracellular matrix (ECM) molecules and reactive gliosis. However, the functional significance of the ECM in retinal neurodegeneration is still unknown. In the present study, we used an intraocular pressure (IOP) independent experimental autoimmune glaucoma (EAG) mouse model to examine the role of the ECM glycoprotein tenascin-C (Tnc). Wild type (WT ONA) and Tnc knockout (KO ONA) mice were immunized with an optic nerve antigen (ONA) homogenate and control groups (CO) obtained sodium chloride (WT CO, KO CO). IOP was measured weekly and electroretinographies were recorded at the end of the study. Ten weeks after immunization, we analyzed retinal ganglion cells (RGCs), glial cells, and the expression of different cytokines in retina and optic nerve tissue in all four groups. IOP and retinal function were comparable in all groups. Although RGC loss was less severe in KO ONA, WT as well as KO mice displayed a significant cell loss after immunization. Compared to KO ONA, less ßIII-tubulin+ axons, and downregulated oligodendrocyte markers were noted in WT ONA optic nerves. In retina and optic nerve, we found an enhanced GFAP+ staining area of astrocytes in immunized WT. A significantly higher number of retinal Iba1+ microglia was found in WT ONA, while a lower number of Iba1+ cells was observed in KO ONA. Furthermore, an increased expression of the glial markers Gfap, Iba1, Nos2, and Cd68 was detected in retinal and optic nerve tissue of WT ONA, whereas comparable levels were observed in KO ONA. In addition, pro-inflammatory Tnfa expression was upregulated in WT ONA, but downregulated in KO ONA. Vice versa, a significantly increased anti-inflammatory Tgfb1 expression was measured in KO ONA animals. We conclude that Tnc plays an important role in glial and inflammatory response during retinal neurodegeneration. Our results provide evidence that Tnc is involved in glaucomatous damage by regulating retinal glial activation and cytokine release. Thus, this transgenic EAG mouse model for the first time offers the possibility to investigate IOP-independent glaucomatous damage in direct relation to ECM remodeling.


Autoimmune Diseases/immunology , Glaucoma/immunology , Tenascin/immunology , Animals , Antigens/administration & dosage , Cytokines/immunology , Disease Models, Animal , Extracellular Matrix , Female , Gliosis/immunology , Immunization , Male , Mice, Knockout , Retinal Ganglion Cells/immunology , Tenascin/genetics
6.
Front Immunol ; 11: 573955, 2020.
Article En | MEDLINE | ID: mdl-33154752

Patients with chronic anterior uveitis are at particularly high risk of developing secondary glaucoma when corticosteroids [e.g., dexamethasone (Dex)] are used or when inflammatory activity has regressed. Macrophage migration into the eye increases when secondary glaucoma develops and may play an important role in the development of secondary glaucoma. Our aim was to evaluate in vitro if increased hydrostatic pressure and corticosteroids could induce changes in macrophages phenotype. By using a pressure chamber cell culture system, we assessed the effect of increased hydrostatic pressure (HP), inflammation, and immunosuppression (Dex) on the M1/M2 phenotype of macrophages. Bone marrow-derived macrophages (BMDMs) were stimulated with medium, lipopolysaccharide (LPS, 100 ng/ml), Dex (200 ng/ml), or LPS + Dex and incubated with different HP (0, 20, or 60 mmHg) for 2 or 7 days. The numbers of CD86+/CD206- (M1 phenotype), CD86-/CD206+ (M2 phenotype), CD86+/CD206+ (intermediate phenotype), F4/80+/TNF-α+, and F4/80+/IL-10+ macrophages were determined by flow cytometry. TNF-α and IL-10 levels in cell culture supernatants were quantified by ELISA. TNF-α, IL-10, fibronectin, and collagen IV expression in BMDMs were detected by immunofluorescence microscopy. Higher HP polarizes macrophages primarily to an M1 phenotype (LPS, 60 vs. 0 mmHg, d2: p = 0.0034) with less extra cellular matrix (ECM) production and secondary to an M2 phenotype (medium, 60 vs. 0 mmHg, d7: p = 0.0089) (medium, 60 vs. 20 mmHg, d7: p = 0.0433) with enhanced ECM production. Dex induces an M2 phenotype (Dex, medium vs. Dex, d2: p < 0.0001; d7: p < 0.0001) with more ECM production. Higher HP further increased M2 polarization of Dex-treated macrophages (Dex, 60 vs. 0 mmHg, d2: p = 0.0417; d7: p = 0.0454). These changes in the M1/M2 phenotype by high HP or Dex treatment may play a role in the pathogenesis of secondary uveitic glaucoma- or glucocorticoid (GC)-induced glaucoma.


Hydrostatic Pressure/adverse effects , Macrophages/immunology , Animals , Cell Survival , Cells, Cultured , Collagen Type IV/metabolism , Dexamethasone/pharmacology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Glaucoma/etiology , Glaucoma/immunology , Inflammation , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Phenotype , Tumor Necrosis Factor-alpha/metabolism
7.
Prog Brain Res ; 256(1): 151-188, 2020.
Article En | MEDLINE | ID: mdl-32958211

Glaucoma is a chronic neurodegenerative disease characterized by retinal ganglion cell loss. Although significant advances in ophthalmologic knowledge and practice have been made, some glaucoma mechanisms are not yet understood, therefore, up to now there is no effective treatment able to ensure healing. Indeed, either pharmacological or surgical approaches to this disease aim in lowering intraocular pressure, which is considered the only modifiable risk factor. However, it is well known that several factors and metabolites are equally (if not more) involved in glaucoma. Oxidative stress, for instance, plays a pivotal role in both glaucoma onset and progression because it is responsible for the trabecular meshwork cell damage and, consequently, for intraocular pressure increase as well as for glaucomatous damage cascade. This review at first shows accurately the molecular-derived dysfunctions in antioxidant system and in mitochondria homeostasis which due to both oxidative stress and aging, lead to a chronic inflammation state, the trabecular meshwork damage as well as the glaucoma neurodegeneration. Therefore, the main molecular events triggered by oxidative stress up to the proapoptotic signals that promote the ganglion cell death have been highlighted. The second part of this review, instead, describes some of neuroprotective agents such as polyphenols or polyunsaturated fatty acids as possible therapeutic source against the propagation of glaucomatous damage.


Fatty Acids, Omega-6/therapeutic use , Glaucoma , Neuroprotective Agents/therapeutic use , Polyphenols/therapeutic use , Retinal Ganglion Cells , Trabecular Meshwork , Glaucoma/drug therapy , Glaucoma/immunology , Glaucoma/metabolism , Glaucoma/pathology , Humans , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Trabecular Meshwork/drug effects , Trabecular Meshwork/immunology , Trabecular Meshwork/metabolism , Trabecular Meshwork/pathology
8.
Prog Brain Res ; 256(1): 125-149, 2020.
Article En | MEDLINE | ID: mdl-32958210

Glaucoma is an age-related neurodegenerative disease that begins at the onset of aging. In this disease, there is an involvement of the immune system and therefore of the microglia. The purpose of this study is to evaluate the microglial activation using a mouse model of ocular hypertension (OHT) at the onset of aging. For this purpose, we used both naive and ocular hypertensives of 15-month-old mice (early stage of aging). In the latter, we analyzed the OHT eyes and the eyes contralateral to them to compare them with their aged controls. In the eyes of aged naive, aged OHT and aged contralateral eyes, microglial changes were observed compared to the young mice, including: (i) aged naive vs young naive: An increased soma size and vertical processes; (ii) aged OHT eyes vs young OHT eyes: A decrease in the area of the retina occupied by Iba-1 cells and in vertical processes; and (iii) aged contralateral vs young contralateral: A decrease in the soma size and arbor area and an increase in the number of microglia in the outer segment layer. Aged OHT eyes and the eyes contralateral to them showed an up-regulation of the CD68 expression in the branched microglia and a down-regulation in the MHCII and P2RY12 expression with respect to the eyes of young OHT mice. Conclusion: in the early phase of aging, morphological microglial changes along with changes in the expression of MHCII, CD68 and P2RY12, in both naive and OHT mice. These changes appear in aged OHT eyes and the eyes contralateral to them eyes.


Aging , Calcium-Binding Proteins , Glaucoma , Inflammation , Microfilament Proteins , Microglia , Retina , Aging/immunology , Aging/metabolism , Aging/pathology , Animals , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Glaucoma/immunology , Glaucoma/metabolism , Glaucoma/pathology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Microfilament Proteins/metabolism , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Retina/immunology , Retina/metabolism , Retina/pathology
9.
Prog Brain Res ; 256(1): 49-77, 2020.
Article En | MEDLINE | ID: mdl-32958215

Glaucoma is a complex neurodegenerative disease involving RGC axons, somas, and synapses at dendrites and axon terminals. Recent research advancements in the field have revealed a bigger picture of glaucomatous neurodegeneration that encompasses multiple stressors, multiple injury sites, multiple cell types, and multiple signaling pathways for asynchronous degeneration of RGCs during a chronic disease period. Optic nerve head is commonly viewed as the critical site of injury in glaucoma, where early injurious insults initiate distal and proximal signaling for axonal and somatic degeneration. Despite compartmentalized processes for degeneration of RGC axons and somas, there are intricate interactions between the two compartments and mechanistic overlaps between the molecular pathways that mediate degeneration in axonal and somatic compartments. This review summarizes the recent progress in the molecular understanding of RGC degeneration in glaucoma and highlights various etiological paths with biomechanical, metabolic, oxidative, and inflammatory components. Through this growing body of knowledge, the glaucoma community moves closer toward causative treatment of this blinding disease.


Glaucoma , Inflammation , Nerve Degeneration , Optic Nerve , Retinal Ganglion Cells , Animals , Glaucoma/immunology , Glaucoma/metabolism , Glaucoma/pathology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Optic Nerve/immunology , Optic Nerve/metabolism , Optic Nerve/pathology , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
10.
Prog Brain Res ; 256(1): 79-97, 2020.
Article En | MEDLINE | ID: mdl-32958216

Over the last decade, new evidence has become increasingly more compelling that commensal microflora profoundly influences the maturation and function of resident immune cells in host physiology. The concept of gut-retina axis is actively being explored. Studies have revealed a critical role of commensal microbes linked with neuronal stress, immune responses, and neurodegeneration in the retina. Microbial dysbiosis changes the blood-retina barrier permeability and modulates T cell-mediated autoimmunity to contribute to the pathogenesis of retinal diseases, such as glaucoma. Heat shock proteins (HSPs), which are evolutionarily conserved, are thought to function both as neuroprotectant and pathogenic antigens of T cells contributing to cell protection and tissue damage, respectively. Activated microglia recruit and interact with T cells during this process. Glaucoma, characterized by the progressive loss of retinal ganglion cells, is the leading cause of irreversible blindness. With nearly 70 million people suffering glaucoma worldwide, which doubles the number of patients with Alzheimer's disease, it represents the most frequent neurodegenerative disease of the central nervous system (CNS). Thus, understanding the mechanism of neurodegeneration in glaucoma and its association with the function of commensal microflora may help unveil the secrets of many neurodegenerative disorders in the CNS and develop novel therapeutic interventions.


Blood-Retinal Barrier , Gastrointestinal Microbiome , Glaucoma , Heat-Shock Proteins , Nerve Degeneration , Retina , T-Lymphocytes , Animals , Blood-Retinal Barrier/immunology , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/microbiology , Blood-Retinal Barrier/pathology , Glaucoma/immunology , Glaucoma/metabolism , Glaucoma/microbiology , Glaucoma/pathology , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Humans , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Degeneration/microbiology , Nerve Degeneration/pathology , Retina/immunology , Retina/metabolism , Retina/microbiology , Retina/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Prog Brain Res ; 256(1): 99-124, 2020.
Article En | MEDLINE | ID: mdl-32958217

The chapter is a review enclosed in the volume "Glaucoma: A pancitopatia of the retina and beyond." No cure exists for glaucoma. Knowledge on the molecular and cellular alterations underlying glaucoma neurodegeneration (GL-ND) includes innovative and path-breaking research on neuroinflammation and neuroprotection. A series of events involving immune response (IR), oxidative stress and gene expression are occurring during the glaucoma course. Uveitic glaucoma (UG) is a prevalent acute/chronic complication, in the setting of chronic anterior chamber inflammation. Managing the disease requires a team approach to guarantee better results for eyes and vision. Advances in biomedicine/biotechnology are driving a tremendous revolution in ophthalmology and ophthalmic research. New diagnostic and imaging modalities, constantly refined, enable outstanding criteria for delimiting glaucomatous neurodegeneration. Moreover, biotherapies that may modulate or inhibit the IR must be considered among the first-line for glaucoma neuroprotection. This review offers the readers useful and practical information on the latest updates in this regard.


Artificial Intelligence , Biological Therapy , Glaucoma , Inflammation , Nerve Degeneration , Uveitis , Glaucoma/diagnostic imaging , Glaucoma/immunology , Glaucoma/metabolism , Glaucoma/therapy , Humans , Inflammation/diagnostic imaging , Inflammation/immunology , Inflammation/metabolism , Inflammation/therapy , Nerve Degeneration/diagnostic imaging , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , Nerve Degeneration/therapy , Uveitis/diagnostic imaging , Uveitis/immunology , Uveitis/metabolism , Uveitis/therapy
13.
Int J Mol Sci ; 21(19)2020 Sep 23.
Article En | MEDLINE | ID: mdl-32977518

Glaucoma is identified by an irreversible retinal ganglion cell (RGC) loss and optic nerve damage. Over the past few years, the immune system gained importance in its genesis. In a glaucoma-like animal model with intraocular S100B injection, RGC death occurs at 14 days. In an experimental autoimmune glaucoma model with systemic S100B immunization, a loss of RGCs is accompanied by a decreased synaptic signal at 28 days. Here, we aimed to study synaptic alterations in these two models. In one group, rats received a systemic S100B immunization (n = 7/group), while in the other group, S100B was injected intraocularly (n = 6-7/group). Both groups were compared to appropriate controls and investigated after 14 days. While inhibitory post-synapses remained unchanged in both models, excitatory post-synapses degenerated in animals with intraocular S100B injection (p = 0.03). Excitatory pre-synapses tendentially increased in animals with systemic S100B immunization (p = 0.08) and significantly decreased in intraocular ones (p = 0.04). Significantly more n-methyl-d-aspartate (NMDA) receptors (both p ≤ 0.04) as well as gamma-aminobutyric acid (GABA) receptors (both p < 0.03) were observed in S100B animals in both models. We assume that an upregulation of these receptors causes the interacting synapse types to degenerate. Heightened levels of excitatory pre-synapses could be explained by remodeling followed by degeneration.


Autoimmune Diseases/immunology , Glaucoma/immunology , Receptors, GABA/immunology , Receptors, N-Methyl-D-Aspartate/immunology , S100 Calcium Binding Protein beta Subunit/toxicity , Synapses/immunology , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/pathology , Disease Models, Animal , Glaucoma/chemically induced , Glaucoma/pathology , Intraocular Pressure/drug effects , Male , Optic Nerve/immunology , Optic Nerve/pathology , Rats , Rats, Inbred Lew , Rats, Wistar , Retinal Ganglion Cells/immunology , Retinal Ganglion Cells/pathology , S100 Calcium Binding Protein beta Subunit/immunology , Synapses/pathology
14.
Autoimmun Rev ; 19(6): 102535, 2020 Jun.
Article En | MEDLINE | ID: mdl-32234407

Glaucoma is characterized by retinal ganglion cell (RGC) neurodegeneration. Elevated intraocular pressure (IOP) is a major risk factor however, mechanisms independent of IOP play a role in RGC pathology. Both antibodies and CD4 T-cells as well as microbiota take part in the pathogenesis of both glaucoma and rheumatoid arteritis (RA).Heat shock proteins (HSPs) which originate in bacteria cross-react with RCG epitopes and were involved in rat model of retinal injury. Enhanced expression of HSPs in the retina was associated with glaucoma-like neuropathology and previous studies have also suggested a pathogenic role for HSPs in RA. In view of these data we suggest that glaucoma should be included in the spectrum of autoimmune diseases and that proven medications for RA should be adopted as an innovative IOP -independent therapeutic strategy for glaucoma.


Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Glaucoma/immunology , Glaucoma/pathology , Animals , Disease Models, Animal , Heat-Shock Proteins , Humans , Intraocular Pressure , Retinal Ganglion Cells/pathology
15.
Int Immunopharmacol ; 83: 106395, 2020 Jun.
Article En | MEDLINE | ID: mdl-32199351

Glaucoma is a kind of blind-causing disease with structural damages of optic nerve and defection of visual field. It is believed that the death of retinal ganglion cell (RGC) is a consequential event of over-reactive immune orchestral cells such as microglia. Previous evidences in animal and clinical studies show the innate immunity plays a pivotal role in neuro-inflammation of glaucoma. Toll-like receptor 4 (TLR4) is expressed on microglia and mediates many neuroinflammatory diseases. We aimed to explore the impacts of high intraocular pressure (IOP) on rat microglia in retina and the regulation of TLR4/NF-κB signaling pathway in scratched microglia cells. In our study, we successfully established chronic high IOP rat model by episcleral vein cauterization (EVC) which behaved like the chronic glaucoma. Besides, we set up an in vitro scratch-induced injury model in rat microglia cells. We found the level of activated microglia cells were significantly increased in the retina of chronic high IOP groups. Moreover, the inhibition of TLR4/NF-κB signaling pathway suppressed the expression of TLR4 protein and mRNA levels of P50, IL-6 and TNF-α. Our original study provided a theoretical basis on targeting TLR4/NF-κB to suppress pro-inflammatory factors releasing in activated microglia and it might be a good treatment target to prevent glaucoma from progressing.


Glaucoma/immunology , Microglia/immunology , NF-kappa B/metabolism , Ocular Hypertension/immunology , Optic Nerve/pathology , Retinal Ganglion Cells/pathology , Toll-Like Receptor 4/metabolism , Animals , Cautery , Cell Death , Cell Line , Disease Models, Animal , Humans , Interleukin-6/metabolism , Male , Neurogenic Inflammation , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha
16.
Front Immunol ; 11: 4, 2020.
Article En | MEDLINE | ID: mdl-32117217

Patients with Sturge-Weber syndrome (SWS) are susceptible to ocular complications, and among them, glaucoma is one of the most frequent forms. In current study, we utilized multiplex human cytokine antibody array to simultaneously measure the concentration of 40 cytokines in aqueous humor (AH) of patients with SWS-induced glaucoma (SG), or from patients with senile cataract as controls. Compared with the control group, levels of interleukin (IL)-12p40, macrophage inflammatory protein (MIP)-1d, tumor necrosis factor-alpha (TNF-a), IL-5, IL-7, interleukin-6 receptor (IL-6R), and B lymphocyte chemoattractant (BLC) in AH were significantly higher in SG group. Samples from SG patients displayed significantly lower levels of MIP-1b, IL-6, MIP-1a, and monocyte chemoattractant protein (MCP)-1 than controls. Further analysis showed that IL-7, MIP-1a, TNF-a were positively correlated with intraocular pressure (IOP) in patients with early-onset SG. Moreover, IL-12p40 was negatively correlated with age in patients with SG. These cytokines may make contributions to the immunopathogenesis or progression of glaucoma in patients with SWS.


Aqueous Humor/metabolism , Cytokines/metabolism , Glaucoma/etiology , Glaucoma/immunology , Signal Transduction/immunology , Sturge-Weber Syndrome/complications , Sturge-Weber Syndrome/immunology , Aged , Aged, 80 and over , Case-Control Studies , Cataract/immunology , Child, Preschool , Cluster Analysis , Cross-Sectional Studies , Female , Humans , Infant , Intraocular Pressure , Male
17.
Front Immunol ; 11: 65, 2020.
Article En | MEDLINE | ID: mdl-32117239

Glaucoma is a globally unmet medical challenge and the most prevalent neurodegenerative disease, which permanently damages the optic nerve and retinal ganglion cells (RGCs), leading to irreversible blindness. Present therapies target solely at lowering intraocular ocular pressure (IOP), a major risk factor of the disease; however, elevated IOP is neither necessary nor sufficient to cause glaucoma. Glaucomatous RGC and nerve fiber loss also occur in individuals with normal IOP. Recent studies have provided evidence indicating a link between elevated IOP and T cell-mediated autoimmune responses, particularly that are specific to heat shock proteins (HSPs), underlying the pathogenesis of neurodegeneration in glaucoma. Reactive glial responses and low-grade inflammation may initially represent an adaptive reaction of the retina to primary stress stimuli; whereas, sustained and excessive glial reactions lead to expanded immune responses, including adaptive immunity, that contribute to progressive neural damage in glaucoma. Emerging data suggest a similar mechanism in play in causing neurodegeneration of other forms of optic neuropathy, such as that resulted from acute ischemia and traumatic injuries. These studies may lead to the paradigm shift and offer a new basis for the development of novel mechanism-based diagnosis, therapy, and preventive interventions for glaucoma. As HSPs are induced under various conditions of neural stress and damage in the brain and spinal cord, these findings may have broader implications for our elucidating of the etiology of other neurodegenerative disorders in the central nervous system.


Adaptive Immunity , Glaucoma/immunology , Neurodegenerative Diseases/immunology , Animals , Glaucoma/metabolism , Heat-Shock Proteins/immunology , Humans , Intraocular Pressure , Neurodegenerative Diseases/metabolism , Retina , T-Lymphocytes/immunology
18.
Int J Mol Sci ; 21(3)2020 Jan 30.
Article En | MEDLINE | ID: mdl-32019187

Diabetic retinopathy (DR) is an ocular complication of diabetes mellitus (DM). International Diabetic Federations (IDF) estimates up to 629 million people with DM by the year 2045 worldwide. Nearly 50% of DM patients will show evidence of diabetic-related eye problems. Therapeutic interventions for DR are limited and mostly involve surgical intervention at the late-stages of the disease. The lack of early-stage diagnostic tools and therapies, especially in DR, demands a better understanding of the biological processes involved in the etiology of disease progression. The recent surge in literature associated with NOD-like receptors (NLRs) has gained massive attraction due to their involvement in mediating the innate immune response and perpetuating inflammatory pathways, a central phenomenon found in the pathogenesis of ocular diseases including DR. The NLR family of receptors are expressed in different eye tissues during pathological conditions suggesting their potential roles in dry eye, ocular infection, retinal ischemia, cataract, glaucoma, age-related macular degeneration (AMD), diabetic macular edema (DME) and DR. Our group is interested in studying the critical early components involved in the immune cell infiltration and inflammatory pathways involved in the progression of DR. Recently, we reported that NLRP3 inflammasome might play a pivotal role in the pathogenesis of DR. This comprehensive review summarizes the findings of NLRs expression in the ocular tissues with special emphasis on its presence in the retinal microglia and DR pathogenesis.


Diabetic Retinopathy/immunology , Glaucoma/immunology , Inflammasomes/immunology , Macular Degeneration/immunology , Macular Edema/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Proteins/immunology , Eye/immunology , Humans , Immunity, Innate
19.
Curr Eye Res ; 45(9): 1124-1135, 2020 09.
Article En | MEDLINE | ID: mdl-31935132

PURPOSE: In glaucoma, an apoptotic death of retinal ganglion cells (RGCs) has been shown. However, little is known about other cell death mechanisms, like autophagy or necrosis. Therefore, we investigated these mechanisms in addition to antibody deposits in an experimental autoimmune glaucoma model. METHODS: Rats were immunized with a retinal ganglion cell-layer homogenate (RGA), while controls received sodium chloride. Untreated rats served as natїve group. After seven weeks, retinal cross-sections were stained with antibodies against RGCs (Brn-3a), apoptosis (cleaved caspase 2, cleaved caspase 3 as well as caspase 3, 8, and 9), autophagy (LC3BII and LAMP1), and necrosis (RIPK3) followed by cell counts. Autophagy was additionally visualized via transmission electron microscopy on retinal sections. Antibody deposits were also analyzed. RESULTS: We noted a RGC loss after RGA immunization compared to both control groups. Also, significantly more cleaved caspase 2+ RGCs were observed in RGA animals. More caspase 3 and 8 signals were noted in RGA retinas compared to both controls, while no changes were seen in regard to caspase 9. Furthermore, significantly more cleaved caspase 3+ cells were detected in RGA animals. We noted an increase of LC3BII+ and LAMP1+ autophagic cells in the RGA group, while no alterations were seen regarding necrotic RIPK3+ cells. Autophagic vesicles were observed via transmission electron microscopy. IgG staining revealed significant differences between the RGA group and controls concerning IgG deposits in the ganglion cell layer. CONCLUSIONS: Due to the novel results from this study, we conclude that IgG antibodies are involved in RGC loss in this model leading to apoptotic and autophagic cell loss. These results could help to develop new therapy strategies for glaucoma patients.


Apoptosis/immunology , Autoantigens/immunology , Autoimmune Diseases/pathology , Autophagy/immunology , Disease Models, Animal , Glaucoma/pathology , Retinal Ganglion Cells/pathology , Animals , Autoantibodies/blood , Autoimmune Diseases/immunology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Glaucoma/immunology , Immunoglobulin G/blood , Lysosomal Membrane Proteins/metabolism , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Rats , Rats, Inbred Lew , Retinal Ganglion Cells/immunology
20.
Clin Exp Pharmacol Physiol ; 46(10): 875-882, 2019 10.
Article En | MEDLINE | ID: mdl-31330064

Elevated expression of autoantibodies is a hallmark of immune dysregulation in glaucoma and may cause retinal ganglion cell apoptosis and immune-mediated nerve damage, thus contributing to the development of blindness. The cause of autoantibody upregulation remains unclear. Th17 cells are shown to promote autoimmunity and Ig production. Here, we demonstrate that the serum levels of interleukin (IL)-17A and IL-21 are comparable between glaucoma patients and non-glaucoma controls. However, the levels of Th17-promoting cytokines, such as tumour necrosis factor (TNF) IL-6, are higher in glaucoma patients than in controls. Subsequently, we demonstrate that glaucoma patients present upregulated levels of Th17 cells that are quiescent directly ex vivo. Interestingly, compared to the Th17 cells from non-glaucoma subjects, the Th17 cells from glaucoma patients present similar IL-17A production capacity but significantly higher IL-21 production capacity. Given that IL-21 is also described as a specific cytokine of follicular helper T cells, the Ig production by B cells following co-incubation with circulating Th17 cells is investigated. Th17 cells from glaucoma patients present significantly enhanced potential to promote Ig production than the Th17 cells from controls. Both glaucoma patient Th17 cells and control Th17 cells require IL-17A and IL-21 for Ig production. Overall, results from this study suggest that Th17 cells from glaucoma patients present elevated capacity to stimulate Ig production.


Glaucoma/blood , Glaucoma/metabolism , Immunoglobulins/biosynthesis , Interleukin-17/metabolism , Interleukins/metabolism , Th17 Cells/metabolism , Adult , Female , Glaucoma/immunology , Humans , Male , Middle Aged
...