Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 61(3): 5, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32150247

ABSTRACT

Purpose: Activating the cell survival modulator sigma 1 receptor (Sig1R) delays cone photoreceptor cell loss in Pde6ßrd10/J (rd10) mice, a model of retinitis pigmentosa. Beneficial effects are abrogated in rd10 mice lacking NRF2, implicating NRF2 as essential to Sig1R-mediated cone neuroprotection. Here we asked whether activation of NRF2 alone is sufficient to rescue cones in rd10 mice. Methods: Expression of antioxidant genes was evaluated in 661W cells and in mouse retinas after treatment with monomethylfumarate (MMF), a potent NRF2 activator. Rd10 mice were administered MMF (50 mg/kg) or the Sig1R ligand (+)-pentazocine (PTZ; 0.5 mg/kg) intraperitoneally (every other day, P14-42). Mice were evaluated for visual acuity (optokinetic tracking response), retinal function (electroretinography) and architecture (SD-OCT); histologic retinal sections were evaluated morphometrically. Results: MMF treatment increased Nrf2, Nqo1, Cat, Sod1, and Hmox1 expression in vitro and in vivo. Visual acuity of (+)-PTZ-treated rd10 mice was similar to wild-type mice; however, MMF treatment did not alter acuity compared with nontreated rd10 mice. Cone electroretinography b-wave amplitudes were greater in PTZ-treated than nontreated or MMF-treated rd10 mice. SD-OCT assessment of retinal thickness was greater in (+)-PTZ-treated mice versus nontreated or MMF-treated rd10 mice. Morphometric assessment of the outer nuclear layer revealed approximately 18 cells/100 µm retinal length in (+)-PTZ-treated rd10 mice, but only approximately 10 to 12 cells/100 µm in MMF-treated and nontreated rd10 retinas. Conclusions: Activation of NRF2 using MMF, at least at our dosing regimen, is insufficient to attenuate catastrophic photoreceptor damage characteristic of rd10 mice. The data prompt investigation of additional mechanisms involved in Sig1R-mediated retinal neuroprotection.


Subject(s)
Fumarates/therapeutic use , Maleates/therapeutic use , Neuroprotective Agents/therapeutic use , Receptors, sigma/physiology , Retinitis Pigmentosa/prevention & control , Animals , Antioxidants/metabolism , Disease Models, Animal , Electroretinography/methods , Fumarates/pharmacology , Hydroquinones/pharmacology , Maleates/pharmacology , Mice, Knockout , NF-E2-Related Factor 2/physiology , Neuroprotection/physiology , Neuroprotective Agents/pharmacology , Pentazocine/pharmacology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/physiology , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/pathology , Retinitis Pigmentosa/physiopathology , Tomography, Optical Coherence/methods , Up-Regulation/drug effects , Visual Acuity/drug effects , Sigma-1 Receptor
2.
J Clin Med ; 9(2)2020 Jan 25.
Article in English | MEDLINE | ID: mdl-31991839

ABSTRACT

Diabetic retinopathy (DR) is a significant cause of blindness in working-age adults worldwide. Lack of effective strategies to prevent or reduce vision loss is a major problem. Since the degeneration of retinal neurons is an early event in the diabetic retina, studies to characterize the molecular mechanisms of diabetes-induced retinal neuronal damage and dysfunction are of high significance. We have demonstrated that spermine oxidase (SMOX), a mediator of polyamine oxidation is critically involved in causing neurovascular damage in the retina. The involvement of SMOX in diabetes-induced retinal neuronal damage is completely unknown. Utilizing the streptozotocin-induced mouse model of diabetes, the impact of the SMOX inhibitor, MDL 72527, on neuronal damage and dysfunction in the diabetic retina was investigated. Retinal function was assessed by electroretinography (ERG) and retinal architecture was evaluated using spectral domain-optical coherence tomography. Retinal cryosections were prepared for immunolabeling of inner retinal neurons and retinal lysates were used for Western blotting. We observed a marked decrease in retinal function in diabetic mice compared to the non-diabetic controls. Treatment with MDL 72527 significantly improved the ERG responses in diabetic retinas. Diabetes-induced retinal thinning was also inhibited by the MDL 72527 treatment. Our analysis further showed that diabetes-induced retinal ganglion cell damage and neurodegeneration were markedly attenuated by MDL 72527 treatment. These results strongly implicate SMOX in diabetes-induced retinal neurodegeneration and visual dysfunction.

3.
J Clin Med ; 9(2)2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31979105

ABSTRACT

Western diet-induced obesity is linked to the development of metabolic dysfunctions, including type 2 diabetes and complications that include retinopathy, a leading cause of blindness. Aberrant activation of the inflammasome cascade leads to the progression of obesity-induced pathologies. Our lab showed the critical role of arginase 2 (A2), the mitochondrial isoform of this ureahydrolase, in obesity-induced metabolic dysfunction and inflammation. A2 deletion also has been shown to be protective against retinal inflammation in models of ischemic retinopathy and multiple sclerosis. We investigated the effect of A2 deletion on western diet-induced retinopathy. Wild-type mice fed a high-fat, high-sucrose western diet for 16 weeks exhibited elevated retinal expression of A2, markers of the inflammasome pathway, oxidative stress, and activation of microglia/macrophages. Western diet feeding induced exaggerated retinal light responses without affecting visual acuity or retinal morphology. These effects were reduced or absent in mice with global A2 deletion. Exposure of retinal endothelial cells to palmitate and high glucose, a mimic of the obese state, increased expression of A2 and inflammatory mediators and induced cell death. These effects, except for A2, were prevented by pretreatment with an arginase inhibitor. Collectively, our study demonstrated a substantial role of A2 in early manifestations of diabetic retinopathy.

4.
Invest Ophthalmol Vis Sci ; 60(13): 4397-4407, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31639826

ABSTRACT

Purpose: Retinitis pigmentosa (RP), a retinal photoreceptor degeneration, typically affects rod function and subsequently cones. Activation of sigma 1 receptor (Sig1R) has been shown to preserve cone function through 6 weeks in the rd10 mouse model of RP, when mice were treated systemically with the Sig1R ligand (+)-pentazocine (PTZ). This study determined the extent to which cone function is preserved in rd10 mice when Sig1R is activated. Methods: Rd10 mice were administered (+)-PTZ (alternate days beginning at postnatal day [P]14) over a period of 180 days. Mouse visual function and structure were measured in vivo using optokinetic tracking response, scotopic and photopic electroretinography plus photopic assessment using "natural" noise stimuli, and optical coherence tomography (OCT). Immunofluorescent methods were used to detect cones in retinal cryosections. Results: Visual acuity was maintained in rd10(+)-PTZ-treated mice through P56, whereas rd10 nontreated mice showed marked decline by P28. Cone responses were detected in (+)-PTZ-treated mice through P60, which were more robust when tested with natural noise stimuli; cone responses were minimal in nontreated rd10 mice. OCT revealed significantly thicker retinas in (+)-PTZ-treated rd10 mice through P60 compared to nontreated mice. Cones were detected by immunofluorescence in (+)-PTZ-treated rd10 retinas through P120. Conclusions: The extent to which cone rescue could be sustained in (+)-PTZ-treated rd10 mice was evaluated comprehensively, showing that activation of Sig1R is associated with prolonged visual acuity, extended detection of cone function, and detection of cones in retinal histologic sections. The data reflect promising long-term neuroprotection when Sig1R is activated.


Subject(s)
Analgesics, Opioid/therapeutic use , Disease Models, Animal , Pentazocine/therapeutic use , Receptors, sigma/metabolism , Retinal Cone Photoreceptor Cells/physiology , Retinitis Pigmentosa/drug therapy , Visual Acuity/drug effects , Animals , Cell Survival/physiology , Color Vision/physiology , Electroretinography , Intraocular Pressure , Mice , Mice, Inbred C57BL , Mice, Knockout , Night Vision/physiology , Nystagmus, Optokinetic/physiology , Retina/metabolism , Retina/physiopathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/physiopathology , Tomography, Optical Coherence , Visual Acuity/physiology , Sigma-1 Receptor
5.
Redox Biol ; 24: 101199, 2019 06.
Article in English | MEDLINE | ID: mdl-31026769

ABSTRACT

Hyperhomocysteinemia (Hhcy), or increased levels of the excitatory amino acid homocysteine (Hcy), is implicated in glaucoma, a disease characterized by increased oxidative stress and loss of retinal ganglion cells (RGCs). Whether Hhcy is causative or merely a biomarker for RGC loss in glaucoma is unknown. Here we analyzed the role of NRF2, a master regulator of the antioxidant response, in Hhcy-induced RGC death in vivo and in vitro. By crossing Nrf2-/- mice and two mouse models of chronic Hhcy (Cbs+/- and Mthfr+/- mice), we generated Cbs+/-Nrf2-/- and Mthfr+/-Nrf2-/- mice and performed systematic analysis of retinal architecture and visual acuity followed by assessment of retinal morphometry and gliosis. We observed significant reduction of inner retinal layer thickness and reduced visual acuity in Hhcy mice lacking NRF2. These functional deficits were accompanied by fewer RGCs and increased gliosis. Given the key role of Müller glial cells in maintaining RGCs, we established an ex-vivo indirect co-culture system using primary RGCs and Müller cells. Hhcy-exposure decreased RGC viability, which was abrogated when cells were indirectly cultured with wildtype (WT) Müller cells, but not with Nrf2-/- Müller cells. Exposure of WT Müller cells to Hhcy yielded a robust mitochondrial and glycolytic response, which was not observed in Nrf2-/- Müller cells. Taken together, the in vivo and in vitro data suggest that deleterious effects of Hhcy on RGCs are likely dependent upon the health of retinal glial cells and the availability of an intact retinal antioxidant response mechanism.


Subject(s)
Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Retinal Ganglion Cells/metabolism , Animals , Biomarkers , Cell Count , Coculture Techniques , Disease Models, Animal , Electroretinography , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Glycolysis , Hyperhomocysteinemia/genetics , Intraocular Pressure , Mice , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Retina/diagnostic imaging , Retina/metabolism , Retinal Ganglion Cells/pathology
6.
Invest Ophthalmol Vis Sci ; 59(13): 5548-5557, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30480743

ABSTRACT

Purpose: Traumatic optic neuropathy (TON) is the most feared visual consequence of head and ocular trauma in both military and civilian communities, for which standard treatment does not exist. Animal models are critical for the development of novel TON therapies as well as the understanding of TON pathophysiology. However, the models currently used for TON have some limitations regarding consistency and mirroring the exact pathological progression of TON in closed ocular trauma. In this study, we modified the model of controlled cortical impact and adapted it for studying TON. Methods: We defined new standardized procedures to induce TON in mice, wherein the optic nerve is reproducibly exposed to a graded controlled impact of known velocity to produce a graded deficit in retinal ganglion cell (RGC) electrophysiological functions. Results: The key results of validating this newly modified model, "controlled orbital impact (COI)," included (1) the injury parameters (velocity as well as contusion depth and time), which were quantifiable and manageable to generate a wide range of TON severities; (2) a reproducible endpoint of diminished positive scotopic threshold response (pSTR) has been achieved without the interference of surgical variability and destruction of surrounding tissues; (3) the contralateral eyes showed no significant difference to the eyes of naïve mice, allowing them to be used as an internal control to minimize interindividual variability among mice; and (4) the occurrence of injury-associated mortality and/or ocular comorbidity was rare. Conclusions: Taken together, this model overcomes some limitations of prior TON mouse models and provides an innovative platform to identify therapeutic targets for neuroprotection and/or neurorestoration following traumatic ocular injury.


Subject(s)
Disease Models, Animal , Optic Nerve Injuries/physiopathology , Optic Nerve/physiopathology , Retina/physiopathology , Animals , Axons/pathology , Blotting, Western , Electroretinography , Mice , Mice, Inbred C57BL , Night Vision/physiology , Retinal Ganglion Cells/pathology
7.
Invest Ophthalmol Vis Sci ; 58(11): 4545-4558, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28877319

ABSTRACT

Purpose: Sigma 1 Receptor (Sig1R) is a novel therapeutic target in neurodegenerative diseases, including retinal disease. Sig1R-/- mice have late-onset retinal degeneration with ganglion cell loss that worsens under stress. Whether Sig1R plays a role in maintaining other retinal neurons is unknown, but was investigated here using rd10 mice, a model of severe photoreceptor degeneration. Methods: Wild-type, rd10, and rd10/Sig1R-/- mice were subjected to ERG and spectral-domain optical coherence tomography (SD-OCT) to assess visual function/structure in situ. Retinas imaged microscopically were subjected to morphometric analysis, immunodetection of cones, and analysis of gliosis. Oxidative and endoplasmic reticulum (ER) stress was evaluated at mRNA/protein levels. Results: Photopic ERG responses were reduced significantly in rd10/Sig1R-/- versus rd10 mice at P28 (31 ± 6 vs. 56 ± 7 µV), indicating accelerated cone loss when Sig1R was absent. At P28, SD-OCT revealed reduced retinal thickness in rd10/Sig1R-/- mice (60% of WT) versus rd10 (80% of WT). Morphometric analysis disclosed profound photoreceptor nuclei loss in rd10/Sig1R-/- versus rd10 mice. rd10/Sig1R-/- mice had 35% and 60% fewer photoreceptors, respectively, at P28 and P35, than rd10. Peanut agglutinin cone labeling decreased significantly; gliosis increased significantly in rd10/Sig1R-/- versus rd10 mice. At P21, NRF2 levels increased in rd10/Sig1R-/- mice versus rd10 and downstream antioxidants increased indicating oxidative stress. At P28, ER stress genes/proteins, especially XBP1, a potent transcriptional activator of the unfolded protein response and CHOP, a proapoptotic transcription factor, increased significantly in rd10/Sig1R-/- mice versus rd10. Conclusions: Photoreceptor cell degeneration accelerates and cone function diminishes much earlier in rd10/Sig1R-/- than rd10 mice emphasizing the importance of Sig1R as a modulator of retinal cell survival.


Subject(s)
Apoptosis , Disease Models, Animal , Receptors, sigma/physiology , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/pathology , Animals , Electroretinography , Female , Fluorescent Antibody Technique, Indirect , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Retina/physiology , Tomography, Optical Coherence , Sigma-1 Receptor
8.
Doc Ophthalmol ; 134(3): 195-203, 2017 06.
Article in English | MEDLINE | ID: mdl-28349282

ABSTRACT

PURPOSE: Previous work has suggested that the retinal degeneration mutant rd8 mouse lacks an electroretinographic (ERG) phenotype until about 9 months of age. We evaluated the ERG phenotype of these mice by measuring both conventional ERG responses and scotopic threshold responses. METHODS: Groups of 4-month-old wild-type (WT) and mutant (rd8) mice were anesthetized and tested for mass retinal responses (ERGs) to several types of visual stimuli. Scotopic threshold responses were accumulated with brief scotopic flashes at a series of very dim intensities. Dark-adapted (scotopic) and light-adapted (photopic) responses to brief flashes at a series of higher intensities were recorded, along with long flashes and random modulations of light levels under photopic conditions. RESULTS: Negative scotopic threshold responses (nSTRs) had lower amplitudes in rd8 mice compared to WTs. Positive scotopic threshold responses were similar in the two groups. With the more intense stimuli, a- and c-wave amplitudes were smaller in rd8 mice. Both scotopic and photopic b-wave amplitudes tended to be larger in rd8 mice, though generally not significantly. CONCLUSIONS: The striking decrease in nSTR amplitudes was surprising, given that the main retinal effects of the rd8 mutation occur in the outer retina, at the external limiting membrane. The primary source of nSTRs in mice is thought to be at the amacrine cell level in the inner retina. Investigation of how this mutation leads to inner retinal dysfunction might reveal unexpected aspects of retinal cell biology and circuitry.


Subject(s)
Electroretinography , Retina/physiopathology , Retinal Degeneration/physiopathology , Animals , Color Vision/physiology , Dark Adaptation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Sensory Thresholds/physiology
9.
Adv Exp Med Biol ; 964: 267-284, 2017.
Article in English | MEDLINE | ID: mdl-28315277

ABSTRACT

This review article focuses on studies of Sigma 1 Receptor (Sigma1R) and retina . It provides a brief overview of the earliest pharmacological studies performed in the late 1990s that provided evidence of the presence of Sigma1R in various ocular tissues. It then describes work from a number of labs concerning the location of Sigma1R in several retinal cell types including ganglion, Müller glia , and photoreceptors . The role of Sigma1R ligands in retinal neuroprotection is emphasized. Early studies performed in vitro clearly showed that targeting Sigma1R could attenuate stress-induced retinal cell loss. These studies were followed by in vivo experiments. Data about the usefulness of targeting Sigma1R to prevent ganglion cell loss associated with diabetic retinopathy are reviewed. Mechanisms of Sigma1R-mediated retinal neuroprotection involving Müller cells , especially in modulating oxidative stress are described along with information about the retinal phenotype of mice lacking Sigma1R (Sigma1R -/- mice). The retina develops normally in Sigma1R -/- mice, but after many months there is evidence of apoptosis in the optic nerve head, decreased ganglion cell function and eventual loss of these cells. Additional studies using the Sigma1R -/- mice provide strong evidence that in the retina, Sigma1R plays a key role in modulating cellular stress. Recent work has shown that targeting Sigma1R may extend beyond protection of ganglion cells to include photoreceptor cell degeneration as well.


Subject(s)
Receptors, sigma/metabolism , Retina/metabolism , Animals , Ependymoglial Cells/metabolism , Humans , Oxidative Stress/physiology , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Sigma-1 Receptor
10.
Cell Death Dis ; 7(11): e2483, 2016 11 24.
Article in English | MEDLINE | ID: mdl-27882947

ABSTRACT

Retinal ischemia is a major cause of visual impairment and blindness and is involved in various disorders including diabetic retinopathy, glaucoma, optic neuropathies and retinopathy of prematurity. Neurovascular degeneration is a common feature of these pathologies. Our lab has previously reported that the ureahydrolase arginase 2 (A2) is involved in ischemic retinopathies. Here, we are introducing A2 as a therapeutic target to prevent neurovascular injury after retinal ischemia/reperfusion (I/R) insult. Studies were performed with mice lacking both copies of A2 (A2-/-) and wild-type (WT) controls (C57BL6J). I/R insult was conducted on the right eye and the left eye was used as control. Retinas were collected for analysis at different times (3 h-4 week after injury). Neuronal and microvascular degeneration were evaluated using NeuN staining and vascular digests, respectively. Glial activation was evaluated by glial fibrillary acidic protein expression. Necrotic cell death was studied by propidium iodide labeling and western blot for RIP-3. Arginase expression was determined by western blot and quantitative RT-PCR. Retinal function was determined by electroretinography (ERG). A2 mRNA and protein levels were increased in WT I/R. A2 deletion significantly reduced ganglion cell loss and microvascular degeneration and preserved retinal morphology after I/R. Glial activation, reactive oxygen species formation and cell death by necroptosis were significantly reduced by A2 deletion. ERG showed improved positive scotopic threshold response with A2 deletion. This study shows for the first time that neurovascular injury after retinal I/R is mediated through increased expression of A2. Deletion of A2 was found to be beneficial in reducing neurovascular degeneration after I/R.


Subject(s)
Arginase/metabolism , Nerve Degeneration/enzymology , Nerve Degeneration/pathology , Reperfusion Injury/enzymology , Retinal Vessels/enzymology , Retinal Vessels/pathology , Animals , Arginase/genetics , Cell Death , Cell Survival , Gene Deletion , Mice, Inbred C57BL , Microvessels/pathology , Models, Biological , Neuroglia/pathology , Neurons/enzymology , Neurons/pathology , Neuroprotection , Oxidative Stress , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reperfusion Injury/pathology
11.
Antioxid Redox Signal ; 25(17): 921-935, 2016 12 10.
Article in English | MEDLINE | ID: mdl-27393735

ABSTRACT

AIMS: Sickle retinopathy (SR) is a major cause of blindness in sickle cell disease (SCD). The genetic mutation responsible for SCD is known, however; oxidative stress and inflammation also figure prominently in the development and progression of pathology. Development of therapies for SR is hampered by the lack of (a) animal models that accurately recapitulate human SR and (b) strategies for noninvasive yet effective retinal drug delivery. This study addressed both issues by validating the Townes humanized SCD mouse as a model of SR and demonstrating the efficacy of oral administration of the antioxidant fumaric acid ester monomethyl fumarate (MMF) in the disease. RESULTS: In vivo ophthalmic imaging, electroretinography, and postmortem histological RNA and protein analyses were used to monitor retinal health and function in normal (HbAA) and sickle (HbSS) hemoglobin-producing mice over a one-year period and in additional HbAA and HbSS mice treated with MMF (15 mg/ml) for 5 months. Functional and morphological abnormalities and molecular hallmarks of oxidative stress/inflammation were evident early in HbSS retinas and increased in number and severity with age. Treatment with MMF, a known inducer of Nrf2, induced γ-globin expression and fetal hemoglobin production, improved hematological profiles, and ameliorated SR-related pathology. Innovation and Conclusion: United States Food and Drug Administration-approved formulations in which MMF is the primary bioactive ingredient are currently available to treat multiple sclerosis; such drugs may be effective for treatment of ocular and systemic complications of SCD, and given the pleiotropic effects, other nonsickle-related diseases in which oxidative stress, inflammation, and retinal vascular pathology figure prominently. Antioxid. Redox Signal. 25, 921-935.


Subject(s)
Anemia, Sickle Cell/complications , Fumarates/administration & dosage , Retinal Diseases/etiology , Retinal Diseases/pathology , Administration, Oral , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/diagnosis , Animals , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA-Binding Proteins , Disease Models, Animal , Electroretinography , Gene Expression , Gene Expression Regulation , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Neovascularization, Pathologic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Repressor Proteins , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Diseases/drug therapy , Retinal Diseases/metabolism , Retinal Neurons/drug effects , Retinal Neurons/metabolism , Retinal Neurons/pathology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , gamma-Globulins/genetics , gamma-Globulins/metabolism
12.
Proc Natl Acad Sci U S A ; 113(26): E3764-72, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27298364

ABSTRACT

Retinal degenerative diseases are major causes of untreatable blindness, and novel approaches to treatment are being sought actively. Here we explored the activation of a unique protein, sigma 1 receptor (Sig1R), in the treatment of PRC loss because of its multifaceted role in cellular survival. We used Pde6ß(rd10) (rd10) mice, which harbor a mutation in the rod-specific phosphodiesterase gene Pde6ß and lose rod and cone photoreceptor cells (PRC) within the first 6 wk of life, as a model for severe retinal degeneration. Systemic administration of the high-affinity Sig1R ligand (+)-pentazocine [(+)-PTZ] to rd10 mice over several weeks led to the rescue of cone function as indicated by electroretinographic recordings using natural noise stimuli and preservation of cone cells upon spectral domain optical coherence tomography and retinal histological examination. The protective effect appears to result from the activation of Sig1R, because rd10/Sig1R(-/-) mice administered (+)-PTZ exhibited no cone preservation. (+)-PTZ treatment was associated with several beneficial cellular phenomena including attenuated reactive gliosis, reduced microglial activation, and decreased oxidative stress in mutant retinas. To our knowledge, this is the first report that activation of Sig1R attenuates inherited PRC loss. The findings may have far-reaching therapeutic implications for retinal neurodegenerative diseases.


Subject(s)
Receptors, sigma/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Animals , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Receptors, sigma/genetics , Retinal Degeneration/congenital , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics , Sigma-1 Receptor
13.
Rev. méd. hered ; 27(1): 50-59, ene.-jun. 2016. ilus, tab
Article in Spanish | LILACS, LIPECS | ID: lil-786611

ABSTRACT

El modelo biomédico tradicional centrado en la enfermedad, con su énfasis en la tecnología, en los últimos años viene siendo cuestionado; en su lugar, se propugna el Modelo biopsicosocial, más integral, sistémico y holístico, que se centra en la persona como un ser biológico, psicológico y social. Para responder adecuadamente las necesidades y expectativas de los pacientes surge el método clínico centrado en el paciente (MCCP) desarrollado por Stewart y Brown (1995). Para ejemplificarlo, en este artículo tomamos un caso clínico, desarrollando los cuatro componentes de la MCCP: la exploración de la dolencia, la enfermedad y la salud; el entendimiento de la persona como un todo, la búsqueda de un espacio común para definir problemas, metas y roles que se adoptarán en el encuentro de la consulta y finalmente el desarrollo de la relación médico paciente.


The traditional biomedical model centered in the disease with emphasis on technological advances is being questioned in recent years. Instead of this model, a bio-psycho-social more integral, systemic and holistic model that is centered in the patient is proposed. To better answer the needs and expectations of the patients, Stewart and Brown (1995) developed a patient-centered clinical method (PCCM). To describe this method, we present a clinical case and explain the components of the method: exploring the ailment, integration of the concept of disease and health, understanding the patient as a whole with search of common ground to define problems, aims and roles during medical consultation, and finally the physician-patient relationship.


Subject(s)
Humans , Male , Young Adult , Patient-Centered Care , Primary Health Care , Physician-Patient Relations
14.
Exp Eye Res ; 145: 164-172, 2016 04.
Article in English | MEDLINE | ID: mdl-26646559

ABSTRACT

Mutations in crumb homologue 1 (CRB1) in humans are associated with Leber's congenital amaurosis (LCA) and retinitis pigmentosa (RP). There is no clear genotype-phenotype correlation for human CRB1 mutations in RP and LCA. The high variability in clinical features observed in CRB1 mutations suggests that environmental factors or genetic modifiers influence severity of CRB1 related retinopathies. Retinal degeneration 8 (rd8) is a spontaneous mutation in the Crb1 gene (Crb1(rdr/rd8)). Crb1(rdr/rd8) mice present with focal disruption in the outer retina manifesting as white spots on fundus examination. Mild retinal dysfunction with decreased b-wave amplitude has been reported in Crb1(rdr/rd8) mice at 18 months. Methylene tetrahydrofolate reductase (MTHFR) is a crucial enzyme of homocysteine metabolism. MTHFR mutations are prevalent in humans and are linked to a broad spectrum of disorders including cardiovascular and neurodegenerative diseases. We recently reported the retinal phenotype in Mthfr-deficient (Mthfr(+/-)) heterozygous mice. At 24 weeks the mice showed decreased RGC function, thinner nerve fiber layer, focal areas of vascular leakage and 20% fewer cells in the ganglion cell layer (GCL). Considering the variability in CRB1-related retinopathies and the high occurrence of human MTHFR mutations we evaluated whether Mthfr deficiency influences rd8 retinal phenotype. Mthfr heterozygous mice with rd8 mutations (Mthfr(+/-)(rd8/rd8)) and Crb(rd8/rd8) mice (Mthfr(+/+rd8/rd8)) mice were subjected to comprehensive retinal evaluation using ERG, fundoscopy, fluorescein angiography (FA), morphometric and retinal flat mount immunostaining analyses of isolectin-B4 at 8-54 wks. Assessment of retinal function revealed a significant decrease in the a-, b- and c-wave amplitudes in Mthfr(+/-)(rd8/rd8) mice at 52 wks. Fundoscopic evaluation demonstrated the presence of signature rd8 spots in Mthfr(+/+rd8/rd8) mice and an increase in the extent of these rd8 spots in Mthfr(+/-)(rd8/rd8) mice at 24 weeks and beyond. FA revealed marked vascular leakage, ischemia and vascular tortuosity in Mthfr(+/-)(rd8/rd8) mice at 24 and 52 weeks. Retinal dysplasia was observed in ∼14-33% Mthfr(+/-)(rd8/rd8) mice by morphometric analysis. This was accompanied by a ∼20% reduction in cells of the GCL of Mthfr(+/-)(rd8/rd8) mice at 24 and 52 weeks. Retinal flat mount immunostaining with isolectin-B4 showed neovascularization and loss of blood vessel integrity in Mthfr(+/-)(rd8/rd8) mice in contrast to mild vasculopathy in Mthfr(+/+rd8/rd8) mice. Taken together, our data support an earlier onset and worsened retinal phenotype when Mthfr and rd8 mutations coexist. Our study sets the stage for future studies to investigate the role of MTHFR deficiency in human CRB1 retinopathies.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Nerve Tissue Proteins/genetics , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Ganglion Cells/metabolism , Animals , DNA/genetics , DNA Mutational Analysis , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Genetic Association Studies , Mice , Mice, Inbred C57BL , Mutation , Nerve Tissue Proteins/metabolism , Phenotype , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Ganglion Cells/pathology
15.
Vision (Basel) ; 1(1)2016 May 09.
Article in English | MEDLINE | ID: mdl-31740628

ABSTRACT

Releasing patients from the fixation task, and permitting them to view natural stimuli such as movies, would provide increased comfort, and potentially additional signs of retinal function, when recording multifocal electroretinograms (mfERGs). Techniques must be developed to handle the difficulties that arise from these alternative stimulation strategies. Multifocal stimuli were presented to volunteer human subjects with and without fixation. Retinocentric analyses were performed to deal with shifts of the stimulus across the retina in the presence of eye movements. Artificial scotomas that moved with the eyes to simulate local retinal defects were presented to assess whether such defects might be detectable in the presence of eye movements. Temporal and spatial correlations in the stimulus can be discounted, permitting retinal kernels to be measured in response to natural stimuli. Responses to temporally natural stimuli tend to have slightly stronger amplitudes because of the presence of low temporal frequencies in these stimuli. The effects of eye movement artifacts can be reduced, permitting similar kernels to be obtained in the absence and presence of eye movements. Convergence to stable kernels took slightly longer in the presence of temporal correlations or eye movements. Artificial scotomas can be localized with these methods. It may be possible to perform multifocal ERG recordings in the clinic using more flexible, natural techniques. However, work is needed to achieve results comparable to those routinely obtained with conventional methods.

16.
Invest Ophthalmol Vis Sci ; 56(4): 2684-95, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25766590

ABSTRACT

PURPOSE: Methylenetetrahydrofolate reductase (Mthfr) is a key enzyme in homocysteine-methionine metabolism. We investigated Mthfr expression in retina and asked whether mild hyperhomocysteinemia, due to Mthfr deficiency, alters retinal neurovascular structure and function. METHODS: Expression of Mthfr was investigated at the gene and protein level using quantitative (q) RT-PCR, in situ hybridization, immunoblotting, and immunohistochemistry (IHC). The Mthfr+/+ and Mthfr+/- mice were subjected to comprehensive evaluation using ERG, funduscopy, fluorescein angiography (FA), spectral-domain optical coherence tomography (SD-OCT), HPLC, and morphometric and IHC analysis of glial fibrillary acidic protein (GFAP) at 8 to 24 weeks. RESULTS: Gene and protein analyses disclosed widespread retinal expression of Mthfr. Electroretinography (ERG) revealed a significant decrease in positive scotopic threshold response in retinas of Mthfr+/- mice at 24 weeks. Fundus examination in mice from both groups was normal; FA revealed areas of focal vascular leakage in 20% of Mthfr+/- mice at 12 to 16 weeks and 60% by 24 weeks. The SD-OCT revealed a significant decrease in nerve fiber layer (NFL) thickness at 24 weeks in Mthfr+/- compared to Mthfr+/+ mice. There was a 2-fold elevation in retinal hcy at 24 weeks in Mthfr+/- mice by HPLC and IHC. Morphometric analysis revealed an approximately 20% reduction in cells in the ganglion cell layer of Mthfr+/- mice at 24 weeks. The IHC indicated significantly increased GFAP labeling suggestive of Müller cell activation. CONCLUSIONS: Mildly hyperhomocysteinemic Mthfr+/- mice demonstrate reduced ganglion cell function, thinner NFL, and mild vasculopathy by 24 weeks. The retinal phenotype is similar to that of hyperhomocysteinemic mice with deficiency of cystathionine-ß-synthase (Cbs) reported earlier. The data support the hypothesis that hyperhomocysteinemia may be causative in certain retinal neurovasculopathies.


Subject(s)
DNA/genetics , Gene Expression Regulation , Homocystinuria/pathology , Hyperhomocysteinemia/pathology , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity/pathology , Retinal Ganglion Cells/pathology , Animals , Disease Models, Animal , Electroretinography , Fluorescein Angiography , Fundus Oculi , Homocystinuria/genetics , Homocystinuria/metabolism , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Immunoblotting , Immunohistochemistry , Methylenetetrahydrofolate Reductase (NADPH2)/biosynthesis , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Mice , Mice, Transgenic , Muscle Spasticity/genetics , Muscle Spasticity/metabolism , Psychotic Disorders/genetics , Psychotic Disorders/metabolism , Psychotic Disorders/pathology , Real-Time Polymerase Chain Reaction , Tomography, Optical Coherence
17.
Mol Vis ; 18: 2860-70, 2012.
Article in English | MEDLINE | ID: mdl-23233788

ABSTRACT

PURPOSE: Sigma receptor 1 (σR1) is a non-opioid transmembrane protein that may act as a molecular chaperone at the endoplasmic reticulum-mitochondrial membrane. Ligands for σR1, such as (+)-pentazocine [(+)-PTZ], confer marked retinal neuroprotection in vivo and in vitro. Recently we analyzed the retinal phenotype of mice lacking σR1 (σR1 KO) and observed normal retinal morphology and function in young mice (5-30 weeks) but diminished negative scotopic threshold responses (nSTRs), retinal ganglion cell (RGC) loss, and disruption of optic nerve axons consistent with inner retinal dysfunction by 1 year. These data led us to test the hypothesis that σR1 may be critical in forestalling chronic retinal stress; diabetes was used as the model of chronic stress. METHODS: To determine whether σR1 is required for (+)-PTZ neuroprotective effects, primary RGCs isolated from wild-type (WT) and σR1 KO mice were exposed to xanthine-xanthine oxidase (10 µM:2 mU/ml) to induce oxidative stress in the presence or absence of (+)-PTZ. Cell death was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) analysis. To assess effects of chronic stress on RGC function, diabetes was induced in 3-week C57BL/6 (WT) and σR1 KO mice, using streptozotocin to yield four groups: WT nondiabetic (WT non-DB), WT diabetic (WT-DB), σR1 KO non-DB, and σR1 KO-DB. After 12 weeks of diabetes, when mice were 15-weeks old, intraocular pressure (IOP) was recorded, electrophysiologic testing was performed (including detection of nSTRs), and the number of RGCs was counted in retinal histological sections. RESULTS: In vitro studies showed that (+)-PTZ could not prevent oxidative stress-induced death of RGCs harvested from σR1 KO mice but afforded robust protection against death of RGCs harvested from WT mice. In the studies of chronic stress induced by diabetes, the IOP measured in the four mouse groups was within the normal range; however, there was a significant increase in the IOP of σR1 KO-DB mice (16 ± 0.5 mmHg) compared to the other groups tested (σR1 KO non-DB, WT non-DB, WT-DB: ~12 ± 0.6 mmHg). Regarding electrophysiologic testing, the nSTRs of σR1 KO non-DB mice were similar to WT non-DB mice at 15 weeks; however, they were significantly lower in σR1 KO-DB mice (5 ± 1 µV) compared to the other groups, including, notably, σR1 KO-nonDB (12±2 µV). As expected, the number of RGCs in σR1 KO non-DB mice was similar to WT non-DB mice at 15 weeks, but under chronic stress of diabetes there were fewer RGCs in retinas of σR1 KO-DB mice. CONCLUSIONS: This is the first report showing unequivocally that the neuroprotective effects of (+)-PTZ require σR1. σR1 KO mice show normal retinal structure and function at young ages; however, when subjected to the chronic stress of diabetes, there is an acceleration of retinal functional deficits in σR1 KO mice such that ganglion cell dysfunction is observed at a much earlier age than nondiabetic σR1 KO mice. The data support the hypothesis that σR1 plays a key role in modulating retinal stress and may be an important target for retinal disease.


Subject(s)
Aging , Diabetes Mellitus, Experimental/genetics , Diabetic Retinopathy/genetics , Receptors, sigma/genetics , Retinal Ganglion Cells/metabolism , Animals , Cell Death/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/complications , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Female , Gene Deletion , In Situ Nick-End Labeling , Intraocular Pressure , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Oxidative Stress , Pentazocine/pharmacology , Primary Cell Culture , Receptors, sigma/deficiency , Retinal Ganglion Cells/pathology , Tonometry, Ocular , Xanthine Oxidase/pharmacology , Sigma-1 Receptor
18.
Invest Ophthalmol Vis Sci ; 52(10): 7749-60, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21862648

ABSTRACT

PURPOSE: Sigma receptor 1 (σR1) is expressed abundantly in the eye, and several reports suggest that this putative molecular chaperone plays a role in lens cell survival, control of intraocular pressure (IOP), and retinal neuroprotection. The present study examined the consequence of the absence of σR1 on ocular development, structure, and function. METHODS: Wild-type (σR1⁺/⁺), heterozygous (σR1⁺/⁻), and homozygous (σR1⁻/⁻, knockout) mice aged 5 to 59 weeks were subjected to comprehensive electrophysiological testing and IOP measurement. The eyes were examined by light and electron microscopy and subjected to morphometric examination and detection of apoptosis. RESULTS: Cornea and lens of σR1⁻/⁻ mice were similar to wild-type mice in morphologic appearance at all ages examined, and IOP was within normal limits. Comprehensive ERG and morphometric analyses initially yielded normal findings in the σR1⁻/⁻ mice compared with those in the wild-type. By 12 months, however, significantly decreased ERG b-wave amplitudes and diminished negative scotopic threshold responses, consistent with inner retinal dysfunction, were detected in σR1⁻/⁻ mice. Concomitant with these late-onset changes were increased TUNEL- and active caspase 3-positive cells in the inner retina and significant loss of cells in the ganglion cell layer, particularly in the central retina. Before these functional and structural abnormalities, there was ultrastructural evidence of axonal disruption in the optic nerve head of σR1⁻/⁻ mice as early as 6 months of age, although there were no alterations observed in retinal vascularization in σR1⁻/⁻ mice. CONCLUSIONS: These data suggest that lack of σR1 leads to development of late-onset retinal dysfunction with similarities to optic neuropathy.


Subject(s)
Optic Nerve Diseases/physiopathology , Receptors, sigma/physiology , Retina/physiopathology , Retinal Diseases/physiopathology , Animals , Axons/metabolism , Axons/ultrastructure , Blotting, Western , Caspase 3/metabolism , Disease Models, Animal , Electroretinography , Fluorescent Antibody Technique, Indirect , Gene Expression , Genotype , In Situ Nick-End Labeling , Intraocular Pressure , Mice , Mice, Inbred C57BL , Mice, Knockout , Optic Disk/metabolism , Optic Disk/ultrastructure , Optic Nerve Diseases/metabolism , Optic Nerve Diseases/pathology , RNA, Messenger/metabolism , Receptors, sigma/deficiency , Retina/metabolism , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Sigma-1 Receptor
19.
PLoS One ; 6(7): e22460, 2011.
Article in English | MEDLINE | ID: mdl-21811615

ABSTRACT

BACKGROUND: Retinopathy of prematurity (ROP) is a major cause of vision impairment in low birth weight infants. While previous work has focused on defining the mechanisms of vascular injury leading to retinal neovascularization, recent studies show that neurons are also affected. This study was undertaken to determine the role of the mitochondrial arginine/ornithine regulating enzyme arginase 2 (A2) in retinal neuro-glial cell injury in the mouse model of ROP. METHODS AND FINDINGS: Studies were performed using wild type (WT) and A2 knockout (A2-/-) mice exposed to Oxygen Induced Retinopathy (OIR). Neuronal injury and apoptosis were assessed using immunohistochemistry, TUNEL (terminal deoxynucleotidyl transferase dUTP nick end) labeling and Western blotting. Electroretinography (ERG) was used to assess retinal function. Neuro-glial injury in WT ROP mice was evident by TUNEL labeling, retinal thinning, decreases in number of rod bipolar cells and glial cell activation as compared with room air controls. Significant reduction in numbers of TUNEL positive cells, inhibition of retinal thinning, preservation of the rod bipolar cells and prevention of glial activation were observed in the A2-/- retinas. Retinal function was markedly impaired in the WT OIR mice as shown by decreases in amplitude of the b-wave of the ERG. This defect was significantly reduced in A2-/- mice. Levels of the pro-apoptotic proteins p53, cleaved caspase 9, cytochrome C and the mitochondrial protein Bim were markedly increased in WT OIR retinas compared to controls, whereas the pro-survival Mitochondrial protein BCL-xl was reduced. These alterations were largely blocked in the A2-/- OIR retina. CONCLUSIONS: Our data implicate A2 in neurodegeneration during ROP. Deletion of A2 significantly improves neuronal survival and function, possibly through the regulation of mitochondrial membrane permeability mediated apoptosis during retinal ischemia. These molecular events are associated with decreased activation of glial cells, suggesting a rescue effect on macroglia as well.


Subject(s)
Arginase/metabolism , Gene Deletion , Neuroglia/pathology , Neurons/pathology , Retina/enzymology , Retina/physiology , Retinopathy of Prematurity/physiopathology , Animals , Apoptosis , Caspase 9/metabolism , Cytoprotection , Disease Models, Animal , Humans , Infant, Newborn , Mice , Neuroglia/metabolism , Neurons/metabolism , Oxygen , Retina/physiopathology , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/pathology , Retinal Degeneration/complications , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Retinopathy of Prematurity/complications , Retinopathy of Prematurity/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism
20.
Vis Neurosci ; 27(5-6): 171-81, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20932359

ABSTRACT

Even during active fixation, small eye movements persist that might be expected to interfere with vision. Numerous brain mechanisms probably contribute to discounting this jitter. Changes in the timing of responses in the visual thalamus associated with fixational saccades are considered in this study. Activity of single neurons in alert monkey lateral geniculate nucleus (LGN) was recorded during fixation while pseudorandom visual noise stimuli were presented. The position of the stimulus on the display monitor was adjusted based on eye position measurements to control for changes in retinal locations due to eye movements. A method for extracting nonstationary first-order response mechanisms was applied, so that changes around the times of saccades could be observed. Saccade-related changes were seen in both amplitude and timing of geniculate responses. Amplitudes were greatly reduced around saccades. Timing was retarded slightly during a window of about 200 ms around saccades. That is, responses became more sustained. These effects were found in both parvocellular and magnocellular neurons. Timing changes in LGN might play a role in maintaining cortical responses to visual stimuli in the presence of eye movements, compensating for the spatial shifts caused by saccades via these shifts in timing.


Subject(s)
Fixation, Ocular/physiology , Geniculate Bodies/physiology , Saccades/physiology , Acoustic Stimulation , Action Potentials/physiology , Algorithms , Animals , Artifacts , Cell Size , Color , Data Interpretation, Statistical , Geniculate Bodies/cytology , Macaca mulatta , Neurons/physiology , Neurons/ultrastructure , Photic Stimulation , Space Perception/physiology , Visual Fields
SELECTION OF CITATIONS
SEARCH DETAIL
...