Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
J Neuroinflammation ; 21(1): 145, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824526

ABSTRACT

BACKGROUND: Recent experimental studies of neuroinflammation in glaucoma pointed to cFLIP as a molecular switch for cell fate decisions, mainly regulating cell type-specific caspase-8 functions in cell death and inflammation. This study aimed to determine the importance of cFLIP for regulating astroglia-driven neuroinflammation in experimental glaucoma by analyzing the outcomes of astroglia-targeted transgenic deletion of cFLIP or cFLIPL. METHODS: Glaucoma was modeled by anterior chamber microbead injections to induce ocular hypertension in mouse lines with or without conditional deletion of cFLIP or cFLIPL in astroglia. Morphological analysis of astroglia responses assessed quantitative parameters in retinal whole mounts immunolabeled for GFAP and inflammatory molecules or assayed for TUNEL. The molecular analysis included 36-plexed immunoassays of the retina and optic nerve cytokines and chemokines, NanoString-based profiling of inflammation-related gene expression, and Western blot analysis of selected proteins in freshly isolated samples of astroglia. RESULTS: Immunoassays and immunolabeling of retina and optic nerve tissues presented reduced production of various proinflammatory cytokines, including TNFα, in GFAP/cFLIP and GFAP/cFLIPL relative to controls at 12 weeks of ocular hypertension with no detectable alteration in TUNEL. Besides presenting a similar trend of the proinflammatory versus anti-inflammatory molecules displayed by immunoassays, NanoString-based molecular profiling detected downregulated NF-κB/RelA and upregulated RelB expression of astroglia in ocular hypertensive samples of GFAP/cFLIP compared to ocular hypertensive controls. Analysis of protein expression also revealed decreased phospho-RelA and increased phospho-RelB in parallel with an increase in caspase-8 cleavage products. CONCLUSIONS: A prominent response limiting neuroinflammation in ocular hypertensive eyes with cFLIP-deletion in astroglia values the role of cFLIP in the molecular regulation of glia-driven neuroinflammation during glaucomatous neurodegeneration. The molecular responses accompanying the lessening of neurodegenerative inflammation also seem to maintain astroglia survival despite increased caspase-8 cleavage with cFLIP deletion. A transcriptional autoregulatory response, dampening RelA but boosting RelB for selective expression of NF-κB target genes, might reinforce cell survival in cFLIP-deleted astroglia.


Subject(s)
Astrocytes , CASP8 and FADD-Like Apoptosis Regulating Protein , Glaucoma , Neuroinflammatory Diseases , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Mice , Astrocytes/metabolism , Astrocytes/pathology , Glaucoma/metabolism , Glaucoma/pathology , Glaucoma/genetics , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice, Transgenic , Disease Models, Animal , Cytokines/metabolism , Retina/metabolism , Retina/pathology , Mice, Inbred C57BL , Optic Nerve/pathology , Optic Nerve/metabolism , Glial Fibrillary Acidic Protein/metabolism
3.
Int Ophthalmol ; 43(6): 1841-1848, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36417008

ABSTRACT

PURPOSE: To investigate the macular spectral domain optical coherence tomography (SD-OCT) measurements of the segmented inner retinal layers in patients with exfoliation syndrome (XFS), exfoliation glaucoma (XFG). METHODS: This prospective cross-sectional study included 28 eyes with XFS, 47 eyes with XFG, and 29 healthy controls. Thickness of the inner retinal layers, including retinal nerve fiber layer (RNFL), ganglion cell layer (GCL), and inner plexiform layer (IPL) was obtained from the horizontal SD-OCT scans. Functional correlation of structural parameters was analyzed using the mean sensitivity (MS) values on 10-2 visual fields. RESULTS: The RNFL, GCL, and IPL were thinnest in eyes with XFG. Among these retinal layers, IPL was significantly thinner in eyes with XFS than healthy controls (p = 0.02) and the IPL thickness was significantly correlated with the corresponding MS scores on 10-2 visual fields (r = 0.492, p = 0.02) in eyes with XFS. Neither GCL nor RNFL thickness values demonstrated significant correlations with functional parameters in eyes with XFS (r = 0.377, p = 0.08; r = 0.212, p = 0.34). In eyes with XFG, the IPL thickness correlated with the visual field MS scores (r = 0.572, p = 0.0007), similar to the correlation of GCL (r = 0.585, p = 0.0005) thickness with visual field scores. CONCLUSIONS: Segmented analysis of the macular IPL thickness presented a significant correlation with the 10-2 visual field scores not only in eyes with XFG but also in eyes with XFS. With respect to early dendritic/synaptic alterations in animal models, larger and longitudinal studies are encouraged to determine the predictive value of the IPL thickness for conversion of XFS to XFG.


Subject(s)
Exfoliation Syndrome , Humans , Exfoliation Syndrome/diagnosis , Retinal Ganglion Cells , Cross-Sectional Studies , Prospective Studies , Retina/diagnostic imaging , Tomography, Optical Coherence/methods
4.
Article in English | MEDLINE | ID: mdl-35020595

ABSTRACT

Elevated intraocular pressure (IOP) is the most prevalent risk factor for initiation and progression of neurodegeneration in glaucoma. Ocular hypertension results from increased resistance to aqueous fluid outflow caused by reduced porosity and increased stiffness of tissues of the outflow pathway. Acoustic activation and resulting bioeffects of the perfluorocarbon (PFC) nanodroplets (NDs) introduced into the anterior chamber (AC) of the eye could potentially represent a treatment for glaucoma by increasing permeability in the aqueous outflow track. To evaluate the potential of NDs to enter the outflow track, 100-nm diameter perfluoropentane (PFP) NDs with a lipid shell were injected into the AC of ex vivo pig eyes and in vivo rat eyes. The NDs were activated and imaged with 18- and 28-MHz linear arrays to assess their location and diffusion. NDs in the AC could also be visualized using optical coherence tomography (OCT). Because of their higher density with respect to aqueous humor, some NDs settled into the iridocorneal angle where they entered the outflow pathway. After acoustic activation of the NDs at the highest acoustic pressure, small gas bubbles were observed in the AC. After two days, no acoustic activation events were visible in the AC of the rats and their eyes showed no evidence of inflammation.


Subject(s)
Fluorocarbons , Glaucoma , Animals , Aqueous Humor/metabolism , Glaucoma/diagnostic imaging , Glaucoma/metabolism , Intraocular Pressure , Rats , Swine , Ultrasonography
5.
Prog Retin Eye Res ; 87: 100998, 2022 03.
Article in English | MEDLINE | ID: mdl-34348167

ABSTRACT

Neuroinflammation relying on the inflammatory responses of glial cells has emerged as an impactful component of the multifactorial etiology of neurodegeneration in glaucoma. It has become increasingly evident that despite early adaptive and reparative features of glial responses, prolonged reactivity of the resident glia, along with the peripheral immune cells, create widespread toxicity to retinal ganglion cell (RGC) axons, somas, and synapses. As much as the synchronized responses of astrocytes and microglia to glaucoma-related stress or neuron injury, their bi-directional interactions are critical to build and amplify neuroinflammation and to dictate the neurodegenerative outcome. Although distinct molecular programs regulate somatic and axonal degeneration in glaucoma, inhibition of neurodegenerative inflammation can provide a broadly beneficial treatment strategy to rescue RGC integrity and function. Since inflammatory toxicity and mitochondrial dysfunction are converging etiological paths that can boost each other and feed into a vicious cycle, anti-inflammatory treatments may also offer a multi-target potential. This review presents an overview of the current knowledge on neuroinflammation in glaucoma with particular emphasis on the cell-intrinsic and cell-extrinsic factors involved in the reciprocal regulation of glial responses, the interdependence between inflammatory and mitochondrial routes of neurodegeneration, and the research aspects inspiring for prospective immunomodulatory treatments. With the advent of powerful technologies, ongoing research on molecular and functional characteristics of glial responses is expected to accumulate more comprehensive and complementary information and to rapidly move the field forward to safe and effective modulation of the glial pro-inflammatory activities, while restoring or augmenting the glial immune-regulatory and neurosupport functions.


Subject(s)
Glaucoma , Neuroinflammatory Diseases , Axons , Glaucoma/drug therapy , Humans , Prospective Studies , Retinal Ganglion Cells/metabolism
6.
Expert Rev Proteomics ; 18(10): 911-924, 2021 10.
Article in English | MEDLINE | ID: mdl-34672220

ABSTRACT

INTRODUCTION: Glaucoma, a leading cause of irreversible blindness in the world, is a chronic neurodegenerative disease of multifactorial origin. Extensive research is ongoing to better understand, prevent, and treat progressive degeneration of retinal ganglion cells in glaucoma. While experimental models of glaucoma and postmortem tissues of human donors are analyzed for pathophysiological comprehension and improved treatment of this blinding disease, clinical samples of intraocular biofluids and blood collected from glaucoma patients are analyzed to identify predictive, diagnostic, and prognostic biomarkers. Multiplexing techniques for protein analysis offer a valuable approach for translational glaucoma research. AREAS COVERED: This review provides an overview of the increasing applications of multiplex protein analysis for glaucoma research and also highlights current research challenges in the field and expected solutions from emerging technological advances. EXPERT OPINION: Analytical techniques for multiplex analysis of proteins can help uncover neurodegenerative processes for enhanced treatment of glaucoma and can help identify molecular biomarkers for improved clinical testing and monitoring of this complex disease. This evolving field and continuously growing availability of new technologies are expected to broaden the comprehension of this complex neurodegenerative disease and speed up the progress toward new therapeutics and personalized patient care to prevent blindness from glaucoma.


Subject(s)
Glaucoma , Neurodegenerative Diseases , Biomarkers , Humans , Translational Research, Biomedical
7.
Cells ; 10(6)2021 06 02.
Article in English | MEDLINE | ID: mdl-34199494

ABSTRACT

Glaucoma is a chronic neurodegenerative disease characterized by apoptosis of retinal ganglion cell (RGC) somas, degeneration of axons, and loss of synapses at dendrites and axon terminals. Glaucomatous neurodegeneration encompasses multiple triggers, multiple cell types, and multiple molecular pathways through the etiological paths with biomechanical, vascular, metabolic, oxidative, and inflammatory components. As much as intrinsic responses of RGCs themselves, divergent responses and intricate interactions of the surrounding glia also play decisive roles for the cell fate. Seen from a broad perspective, multitarget treatment strategies have a compelling pathophysiological basis to more efficiently manipulate multiple pathogenic processes at multiple injury sites in such a multifactorial neurodegenerative disease. Despite distinct molecular programs for somatic and axonal degeneration, mitochondrial dysfunction and glia-driven neuroinflammation present interdependent processes with widespread impacts in the glaucomatous retina and optic nerve. Since dysfunctional mitochondria stimulate inflammatory responses and proinflammatory mediators impair mitochondria, mitochondrial restoration may be immunomodulatory, while anti-inflammatory treatments protect mitochondria. Manipulation of these converging routes may thus allow a unified treatment strategy to protect RGC axons, somas, and synapses. This review presents an overview of recent research advancements with emphasis on potential treatment targets to achieve the best treatment efficacy to preserve visual function in glaucoma.


Subject(s)
Axons/metabolism , Glaucoma , Nerve Degeneration , Neurodegenerative Diseases , Retinal Ganglion Cells/metabolism , Animals , Glaucoma/metabolism , Glaucoma/therapy , Humans , Nerve Degeneration/metabolism , Nerve Degeneration/therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy
8.
Exp Eye Res ; 207: 108606, 2021 06.
Article in English | MEDLINE | ID: mdl-33930396

ABSTRACT

Most studies of the effect of acute elevation of intraocular pressure (IOP) on ocular blood-flow have utilized optical coherence tomography (OCT) to characterize retinal and choroidal flow and vascular density. This study investigates the effect of acute IOP elevation on blood flow velocity in the retrobulbar arteries and veins supplying and draining the eye, which, unlike the retinal and choroidal vasculature, are not directly compressed as IOP is increased. By cannulation of the anterior chamber of 20 Sprague-Dawley rats, we increased IOP in 10 mmHg steps from 10 to 60 mmHg and returned to 10 mmHg. After 1 min at each IOP (and 3 min after return to 10 mmHg), we acquired 18 MHz plane-wave ultrasound data at 3000 compound images/sec for 1.5 s. We produced color-flow Doppler images by digital signal processing of the ultrasound data, identified retrobulbar arteries and veins, generated spectrograms depicting flow velocity over the cardiac cycle and characterized changes of vascular density and perfusion in the orbit overall. Systolic, diastolic and mean velocities and resistive and pulsatile indices were determined from arterial spectrograms at each IOP level. Baseline mean arterial and mean venous velocities averaged 30.9 ±â€¯10.8 and 8.5 ±â€¯3.3 mm/s, respectively. Arterial velocity progressively decreased and resistance indices increased at and above an IOP of 30 mmHg. Mean arterial velocity at 60 mmHg dropped by 55% with respect to baseline, while venous velocity decreased by 20%. Arterial and venous velocities and resistance returned to near baseline after IOP was restored to 10 mmHg. Both vascular density and orbital perfusion decreased with IOP, but while perfusion returned to near normal when IOP returned to 10 mmHg, density remained reduced. Our findings are consistent with OCT-based studies showing reduced perfusion of the retina at levels comparable to retrobulbar arterial flow velocity change with increased IOP. The lesser effect on venous flow is possibly attributable to partial collapse of the venous lumen as volumetric venous outflow decreased at high IOP. The continued reduction in orbital vascular density 3 min after restoration of IOP to 10 mmHg might be attributable to persisting narrowing of capillaries, but this needs to be verified in future studies.


Subject(s)
Intraocular Pressure/physiology , Ocular Hypertension/physiopathology , Orbit/blood supply , Animals , Blood Flow Velocity , Choroid/blood supply , Ciliary Arteries/physiology , Female , Male , Ophthalmic Artery/physiology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/physiology , Retinal Artery/physiology , Tonometry, Ocular
9.
PLoS One ; 16(2): e0247401, 2021.
Article in English | MEDLINE | ID: mdl-33630899

ABSTRACT

Glaucoma is a chronic neurodegenerative disease of the optic nerve and a leading cause of irreversible blindness, worldwide. While the experimental research using animal models provides growing information about cellular and molecular processes, parallel analysis of the clinical presentation of glaucoma accelerates the translational progress towards improved understanding, treatment, and clinical testing of glaucoma. Optic nerve axon injury triggers early alterations of retinal ganglion cell (RGC) synapses with function deficits prior to manifest RGC loss in animal models of glaucoma. For testing the clinical relevance of experimental observations, this study analyzed the functional correlation of localized alterations in the inner plexiform layer (IPL), where RGCs establish synaptic connections with retinal bipolar and amacrine cells. Participants of the study included a retrospective cohort of 36 eyes with glaucoma and a control group of 18 non-glaucomatous subjects followed for two-years. The IPL was analyzed on consecutively collected macular SD-OCT scans, and functional correlations with corresponding 10-2 visual field scores were tested using generalized estimating equations (GEE) models. The GEE-estimated rate of decrease in IPL thickness (R = 0.36, P<0.001) and IPL density (R = 0.36, P<0.001), as opposed to unchanged or increased IPL thickness or density, was significantly associated with visual field worsening at corresponding analysis locations. Based on multivariate logistic regression analysis, this association was independent from the patients' age, the baseline visual field scores, or the baseline thickness or alterations of retinal nerve fiber or RGC layers (P>0.05). These findings support early localized IPL alterations in correlation with progressing visual field defects in glaucomatous eyes. Considering the experimental data, glaucoma-related increase in IPL thickness/density might reflect dendritic remodeling, mitochondrial redistribution, and glial responses for synapse maintenance, but decreased IPL thickness/density might correspond to dendrite atrophy. The bridging of experimental data with clinical findings encourages further research along the translational path.


Subject(s)
Glaucoma, Open-Angle/pathology , Macula Lutea/pathology , Visual Fields/physiology , Aged , Aged, 80 and over , Amacrine Cells/pathology , Blindness/pathology , Female , Humans , Intraocular Pressure/physiology , Male , Middle Aged , Nerve Fibers/pathology , Optic Nerve/pathology , Retinal Ganglion Cells/pathology , Retrospective Studies , Tomography, Optical Coherence/methods , Visual Field Tests/methods
10.
Neurobiol Dis ; 150: 105258, 2021 03.
Article in English | MEDLINE | ID: mdl-33434617

ABSTRACT

Retinal ganglion cells (RGCs) expanding from the retina to the brain are primary victims of neurodegeneration in glaucoma, a leading cause of blindness; however, the neighboring astroglia survive the glaucoma-related stress and promote neuroinflammation. In light of diverse functions of caspase-8 in apoptosis, cell survival, and inflammation, this study investigated the importance of caspase-8 in different fates of glaucomatous RGCs and astroglia using two experimental approaches in parallel. In the first approach, cell type-specific responses of RGCs and astroglia to a caspase-8 cleavage-inhibiting pharmacological treatment were studied in rat eyes with or without experimentally induced glaucoma. The second approach utilized an experimental model of glaucoma in mice in which astroglial caspase-8 was conditionally deleted by cre/lox. Findings of these experiments revealed cell type-specific distinct processes that regulate caspase-8 functions in experimental glaucoma, which are involved in inducing the apoptosis of RGCs and promoting the survival and inflammatory responses of astroglia. Deletion of caspase-8 in astroglia protected RGCs against glia-driven inflammatory injury, while the inhibition of caspase-8 cleavage inhibited apoptosis in RGCs themselves. Various caspase-8 functions impacting both RGC apoptosis and astroglia-driven neuroinflammation may suggest the multi-target potential of caspase-8 regulation to provide neuroprotection and immunomodulation in glaucoma.


Subject(s)
Astrocytes/metabolism , Caspase 8/metabolism , Glaucoma/metabolism , Neuroinflammatory Diseases/metabolism , Retinal Ganglion Cells/metabolism , Animals , Apoptosis , Astrocytes/pathology , Axons , Cell Survival , Disease Models, Animal , Electroretinography , Glaucoma/pathology , Mice , Optic Nerve/pathology , Rats , Retinal Ganglion Cells/pathology
11.
Prog Brain Res ; 256(1): 49-77, 2020.
Article in English | MEDLINE | ID: mdl-32958215

ABSTRACT

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.


Subject(s)
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
12.
J Neuroinflammation ; 17(1): 252, 2020 Aug 28.
Article in English | MEDLINE | ID: mdl-32859212

ABSTRACT

BACKGROUND: Glia-driven neuroinflammation promotes neuron injury in glaucoma that is a chronic neurodegenerative disease of the optic nerve and a leading cause of irreversible blindness. Although therapeutic modulation of neuroinflammation is increasingly viewed as a logical strategy to avoid inflammatory neurotoxicity in glaucoma, current understanding of the molecular regulation of neuroinflammation is incomplete, and the molecular targets for immunomodulation remains unknown. Growing datasets pointed to nuclear factor-kappaB (NF-κB), a key transcriptional activator of inflammation, which was identified to be most affected in glaucomatous astroglia. Using a cell type-specific experimental approach, this study aimed to determine the value of astroglial NF-κB as a potential treatment target for immunomodulation in experimental mouse glaucoma. METHODS: Neuroinflammatory and neurodegenerative outcomes of experimental glaucoma were comparatively analyzed in mice with or without cre/lox-based conditional deletion of astroglial IκKß, which is the main activating kinase involved in IκB degradation through the canonical pathway of NF-κB activation. Glial responses and the inflammatory status of the retina and optic nerve were analyzed by cell morphology and cytokine profiling, and neuron structure and function were analyzed by counting retinal ganglion cell (RGC) axons and somas and recording pattern electroretinography (PERG) responses. RESULTS: Analysis of glial inflammatory responses showed immunomodulatory outcomes of the conditional transgenic deletion of IκKß in astroglia. Various pro-inflammatory cytokines known to be transcriptional targets for NF-κB exhibited decreased production in IκKß-deleted astroglia, which included TNF-α that can induce RGC apoptosis and axon degeneration during glaucomatous neurodegeneration. Indeed, transgenic modulation of inflammatory responses by astroglial IκKß deletion reduced neurodegeneration at different neuronal compartments, including both RGC axons and somas, and protected PERG responses. CONCLUSIONS: The findings of this study support a key role for astroglial NF-κB in neuroinflammatory and neurodegenerative outcomes of experimental glaucoma and the potential of this transcriptional regulator pathway as a glial treatment target to provide neuroprotection through immunomodulation. By pointing to a potential treatment strategy targeting the astroglia, these experimental findings are promising for future clinical translation through transgenic applications to improve the treatment of this blinding disease.


Subject(s)
Astrocytes/metabolism , Glaucoma/metabolism , NF-kappa B/metabolism , Nerve Degeneration/metabolism , Animals , Astrocytes/pathology , Axons/metabolism , Axons/pathology , Disease Models, Animal , Glaucoma/genetics , Glaucoma/pathology , Mice , Mice, Transgenic , NF-kappa B/genetics , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Retina/metabolism , Retina/pathology , Signal Transduction/physiology
13.
Exp Eye Res ; 193: 107986, 2020 04.
Article in English | MEDLINE | ID: mdl-32119869

ABSTRACT

Preclinical imaging, especially of rodent models, plays a major role in experimental ophthalmology. Our aim was to determine if ultrasound can be used to visualize and measure flow dynamics in the retrobulbar vessels supplying and draining the eye and the potential of contrast microbubbles to provide image and measurement enhancement. To accomplish this, we used a 128-element, 18 MHz linear array ultrasound probe and performed plane-wave imaging of the eyes of Sprague Dawley rats. Compound images were acquired by emitting unfocused wavefronts at multiple angles and combining echo data from all angles to form individual B-scans. Multiple imaging sequences were utilized, compounding up to six angles, with imaging rate of up to 3000 compound B-scans per second and sequence durations from 1.5 to 180 s. Data were acquired before and after intravenous introduction of contrast microbubbles. We found the total power of the Doppler signal in the image plane to increase approximately 20 fold after injection of contrast, followed by an exponential decay to baseline in about 90 s, The best-fit time constant of the decay averaged 41 s. While major vessels and the retinal/choroidal complex were evident pre-contrast, they were dramatically enhanced with contrast present, with details such as choroidal arterioles seen only with contrast. Ocular arteriovenous transit time determined from comparative enhancement curves in arteries and veins was approximately 0.2 s. In conclusion, plane wave ultrasound, especially with enhancement by contrast microbubbles, offers a means for the study of ocular hemodynamics using the rat eye as a model.


Subject(s)
Contrast Media/pharmacology , Ophthalmic Artery/physiology , Orbit/blood supply , Phantoms, Imaging , Regional Blood Flow/physiology , Ultrasonography/methods , Animals , Models, Animal , Ophthalmic Artery/diagnostic imaging , Rats , Rats, Sprague-Dawley
14.
Curr Ophthalmol Rep ; 7(2): 160-169, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31360618

ABSTRACT

PURPOSE OF REVIEW: This review aims to highlight the current knowledge about inflammatory mechanisms of neurodegeneration in glaucoma with emphasis on potential immunomodulation strategies. RECENT FINDINGS: Glaucomatous retina and optic nerve present multiple evidences of inflammatory responses of astroglia, microglia, and blood-born immune cells. Although adaptive/protective responses of resident or systemic immune cells can support neurons and promote tissue repair mechanisms after injurious insults, prolonged inflammatory processes can also produce neurotoxic mediators. Treatments targeting these neurodestructive outcomes may restore immune homeostasis and protect neurons from inflammatory injury. Due to widespread and chronic nature of neuroinflammation in glaucoma, immunomodulation offers a treatment strategy to protect different neuronal compartments of RGCs during the chronic and asynchronous course of neurodegeneration. Uncovering of distinct molecular responses and interactions of different immune cells that determine the neuroinflammatory phenotype and participate in neurodegenerative outcomes will be critical to develop effective strategies for immunomodulation in glaucoma. SUMMARY: Neuroinflammation has increasingly been recognized to play an important role in glaucomatous neurodegeneration, and its modulation appears to be a promising treatment strategy for neuroprotection.

15.
Invest Ophthalmol Vis Sci ; 60(4): 877-888, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30821813

ABSTRACT

Purpose: Besides glia-driven neuroinflammation, growing evidence from analysis of human blood samples, isolated autoantibodies, and postmortem tissues also support systemic immune responses during neurodegeneration in glaucoma patients. To explore the T-cell-mediated component of systemic immunity, this study analyzed T lymphocytes in patients' blood. Methods: Blood samples were collected from 32 patients with glaucoma and 21 nonglaucomatous controls, and mononuclear cells were isolated by Histopaque density gradient centrifugation. T-cell subset distribution was analyzed by multicolor flow cytometry after helper (Th) and cytotoxic fractions, and Th subpopulations, were stained with antibodies to CD4, CD8, or distinctive markers, such as IFN-γ (for Th1), IL-4 (for Th2), IL-17A (for Th17), and CD25/FoxP3 (for T regulatory cells [Tregs]). In addition, proliferative activity and cytokine secretion of T cells were analyzed after in vitro stimulation. Results: Analysis of T-cell subset distribution detected a glaucoma-related shift. Despite similar frequencies of CD4+ or CD8+ T cells, or Th1, Th2, or Th17 subsets in glaucoma and control groups, glaucomatous samples exhibited a trend toward decreased frequency of CD4+ (or CD8+)/CD25+/FoxP3+ Tregs within the entire CD4+ (or CD8+) population (P < 0.001). Furthermore, CD4+ T cells in glaucomatous samples presented a greater stimulation response (∼3-fold) as characterized by increased proliferation and proinflammatory cytokine secretion (P < 0.05). Conclusions: These findings suggest that the immunity activated in glaucoma may not be counterbalanced by an efficient immune suppression. More work is encouraged to determine whether shifted T-cell homeostasis may contribute to neurodegeneration in glaucoma, and/or whether T-cell subset imbalance may serve as a biomarker of autoimmune susceptibility.


Subject(s)
Glaucoma, Open-Angle/immunology , T-Lymphocyte Subsets/immunology , Aged , Aged, 80 and over , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Glaucoma, Open-Angle/diagnosis , Humans , Intraocular Pressure/physiology , Male , Middle Aged , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Tonometry, Ocular
16.
Clin Ophthalmol ; 12: 401-409, 2018.
Article in English | MEDLINE | ID: mdl-29520130

ABSTRACT

PURPOSE: This pilot cross-sectional study aimed to determine age-related changes of the retinal nerve fiber layer (RNFL) thickness in retinal periphery by swept-source optical coherence tomography-based analysis. METHODS: Forty eyes of 40 healthy subjects were studied in three age groups, group 1 (20-40 years, n=15), group 2 (41-60 years, n=14), and group 3 (≥61 years, n=11). Wide-angle swept-source optical coherence tomography scans, including the optic disc and macula, were montaged with the nasal peripheral optical coherence tomography images acquired with a contralateral gaze. The peripapillary and peripheral RNFL thickness values were obtained for nasal and temporal sides. The ratio of peripheral-to-peripapillary RNFL thickness was also calculated for these sectors. RESULTS: We detected a significantly thinner RNFL in older than younger subjects at a distance of 6 mm from the optic disc on nasal and temporal sides (P<0.001). An age-related reduction in peripheral-to-peripapillary RNFL thickness ratios (P<0.001 and P<0.02 for nasal and temporal sides, respectively) was also detected. CONCLUSION: The age-related decline should be taken into consideration when determining the glaucoma-related alterations in peripheral RNFL thickness. Continued analysis in patients with ocular hypertension and glaucoma should help determine whether RNFL in the periphery with lower nerve tissue reserve might be more susceptible to injury, whether injury to the peripheral RNFL might be easier to detect, and/or whether analysis of the peripheral RNFL thickness could improve clinical diagnosis and follow-up of glaucoma.

17.
Invest Ophthalmol Vis Sci ; 58(10): 4078-4088, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28820925

ABSTRACT

Purpose: Glaucoma-related molecular biomarkers can improve clinical testing to diagnose the disease early, predict its prognosis, and monitor treatment responses. Based on the evidence of increased oxidative stress in glaucomatous tissues, this study analyzed oxidative stress-related biomarker candidates in blood and aqueous humor samples with or without glaucoma. Methods: The blood and aqueous humor samples collected from carefully selected groups of 96 patients with glaucoma and 64 healthy subjects without glaucoma were included in the study. The samples were analyzed for protein carbonyls and advanced glycation end products (AGEs) through ELISA-based quantification assays. To allow proper comparisons, the Goldmann-Witmer coefficient that reflects the ratio of aqueous humor to blood values corrected to total protein concentration in individual samples was calculated. Results: Blood and aqueous humor levels of protein carbonyls and AGEs were found significantly higher in glaucomatous samples compared with age-matched nonglaucomatous controls (P < 0.001). The glaucoma-related increase in protein carbonyls and AGEs was more prominent in aqueous humor samples than blood samples (2.6-fold versus 1.9-fold for protein carbonyls, and 3.1-fold versus 1.9-fold for AGEs; P < 0.001). Comparison of the Goldmann-Witmer coefficients indicated greater values for protein carbonyls (1.37 ± 0.3 vs. 3.07 ± 0.8) and AGEs (1.2 ± 0.3 vs. 3.2 ± 1.1) in the glaucoma group (P < 0.001). Conclusions: Findings of this study encourage further validation studies of oxidative stress-related biomarkers in glaucoma. Analysis of protein carbonyls and AGEs in longitudinal studies of larger and heterogeneous patient cohorts should better assess the value of these promising candidates as molecular biomarkers of glaucoma for clinical predictions.


Subject(s)
Aqueous Humor/metabolism , Biomarkers/blood , Glaucoma, Open-Angle/blood , Glycation End Products, Advanced/blood , Oxidative Stress , Protein Carbonylation , Aged , Enzyme-Linked Immunosorbent Assay , Female , Glaucoma, Open-Angle/diagnosis , Gonioscopy , Healthy Volunteers , Humans , Intraocular Pressure , Male
19.
Invest Ophthalmol Vis Sci ; 57(4): 2344-54, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27127934

ABSTRACT

PURPOSE: Besides primary neurotoxicity, oxidative stress may compromise the glial immune regulation and shift the immune homeostasis toward neurodegenerative inflammation in glaucoma. We tested this hypothesis through the analysis of neuroinflammatory and neurodegenerative outcomes in mouse glaucoma using two experimental paradigms of decreased or increased oxidative stress. METHODS: The first experimental paradigm tested the effects of Tempol, a multifunctional antioxidant, given through osmotic mini-pumps for drug delivery by constant infusion. Following a 6-week treatment period after microbead/viscoelastic injection-induced ocular hypertension, retina and optic nerve samples were analyzed for markers of oxidative stress and cytokine profiles using specific bioassays. We also analyzed a redox-sensitive transcriptional regulator of neuroinflammation, namely NF-κB. The second paradigm included a similar analysis of the effects of overloaded oxidative stress on retina and optic nerve inflammation in mice knockout for a major antioxidant enzyme (SOD1(-/-)). RESULTS: Increased antioxidant capacity and decreased protein carbonyls and HNE adducts with Tempol treatment verified the drug delivery and biological function. Among a range of cytokines measured, proinflammatory cytokines, including IL-1, IL-2, IFN-γ, and TNF-α, exhibited more than 2-fold decreased titers in Tempol-treated ocular hypertensive eyes. Antioxidant treatment also resulted in a prominent decrease in NF-κB activation in the ocular hypertensive retina and optic nerve. Although pharmacological treatment limiting the oxidative stress resulted in decreased neuroinflammation, ocular hypertension-induced neuroinflammatory responses were increased in SOD1(-/-) mice with defective antioxidant response. CONCLUSIONS: These findings support the oxidative stress-related mechanisms of neuroinflammation and the potential of antioxidant treatment as an immunomodulation strategy for neuroprotection in glaucoma.


Subject(s)
Antioxidants/therapeutic use , Cyclic N-Oxides/therapeutic use , Glaucoma/drug therapy , Animals , Antioxidants/administration & dosage , Blotting, Western , Cyclic N-Oxides/administration & dosage , Cytokines/analysis , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Glaucoma/pathology , Inflammation/drug therapy , Inflammation/pathology , Infusion Pumps, Implantable , Intraocular Pressure/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/analysis , Ocular Hypertension/drug therapy , Optic Nerve/chemistry , Optic Nerve/pathology , Oxidative Stress/drug effects , Retina/chemistry , Retina/pathology , Spin Labels
20.
Mol Vis ; 22: 82-99, 2016.
Article in English | MEDLINE | ID: mdl-26900327

ABSTRACT

PURPOSE: To study the detailed cellular and molecular changes in the mouse sclera subjected to experimental glaucoma. METHODS: Three strains of mice underwent experimental bead-injection glaucoma and were euthanized at 3 days and 1, 3, and 6 weeks. Scleral protein expression was analyzed with liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) using (16)O/(18)O labeling for quantification in 1- and 6-week tissues. Sclera protein samples were also analyzed with immunoblotting with specific antibodies to selected proteins. The proportion of proliferating scleral fibroblasts was quantified with Ki67 and 4',6-diamidino-2-phenylindole (DAPI) labeling, and selected proteins were studied with immunohistochemistry. RESULTS: Proteomic analysis showed increases in molecules involved in integrin-linked kinase signaling and actin cytoskeleton signaling pathways at 1 and 6 weeks after experimental glaucoma. The peripapillary scleral region had more fibroblasts than equatorial sclera (p=0.001, n=217, multivariable regression models). There was a sixfold increase in proliferating fibroblasts in the experimental glaucoma sclera at 1 week and a threefold rise at 3 and 6 weeks (p=0.0005, univariate regression). Immunoblots confirmed increases for myosin, spectrin, and actinin at 1 week after glaucoma. Thrombospondin-1 (TSP-1), HINT1, vimentin, actinin, and α-smooth muscle actin were increased according to immunohistochemistry. CONCLUSIONS: Scleral fibroblasts in experimental mouse glaucoma show increases in actin cytoskeleton and integrin-related signaling, increases in cell division, and features compatible with myofibroblast transition.


Subject(s)
Disease Models, Animal , Fibroblasts/physiology , Glaucoma/physiopathology , Sclera/cytology , Actins/metabolism , Animals , Cell Proliferation/physiology , Chromatography, Liquid , Eye Proteins/metabolism , Fibroblasts/cytology , Glaucoma/metabolism , Immunoblotting , Indoles/metabolism , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Proteomics , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...