Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Eye Res ; 146: 269-275, 2016 05.
Article in English | MEDLINE | ID: mdl-27060376

ABSTRACT

Increased histone deacetylase (HDAC) activity and the resulting dysregulation of protein acetylation is an integral event in retinal degenerations associated with ischemia and ocular hypertension. This study investigates the role of preconditioning on the process of acetylation in ischemic retinal injury. Rat eyes were unilaterally subjected to retinal injury by 45 min of acute ischemia, and retinal neuroprotection induced by 5 min of an ischemic preconditioning (IPC) event. HDAC activity was evaluated by a fluorometric enzymatic assay with selective isoform inhibitors. Retinal localization of acetylated histone-H3 was determined by immunohistochemistry on retina cross sections. Cleaved caspase-3 level was evaluated by Western blots. Electroretinogram (ERG) analyses were used to assess differences in retinal function seven days following ischemic injury. In control eyes, analysis of HDAC isoforms demonstrated that HDAC1/2 accounted for 28.4 ± 1.6%, HDAC3 for 42.4 ± 1.5% and HDAC6 activity 27.3 ± 3.5% of total activity. Following ischemia, total Class-I HDAC activity increased by 21.2 ± 6.2%, and this increase resulted solely from a rise in HDAC1/2 activity. No change in HDAC3 activity was measured. Activity of Class-II HDACs and HDAC8 was negligible. IPC stimulus prior to ischemic injury also suppressed the rise in Class-I HDAC activity, cleaved caspase-3 levels, and increased acetylated histone-H3 in the retina. In control animals 7 days post ischemia, ERG a- and b-wave amplitudes were significantly reduced by 34.9 ± 3.1% and 42.4 ± 6.3%, respectively. In rats receiving an IPC stimulus, the ischemia-induced decline in ERG a- and b-wave amplitudes was blocked. Although multiple HDACs were detected in the retina, these studies provide evidence that hypoacetylation associated with ischemic injury results from the selective rise in HDAC1/2 activity and that neuroprotection induced by IPC is mediated in part by suppressing HDAC activity.


Subject(s)
Histone Deacetylases/metabolism , Ischemic Preconditioning , Neuroprotection/physiology , Retina/metabolism , Acetylation , Analysis of Variance , Animals , Blotting, Western , Caspase 3/metabolism , Disease Models, Animal , Electroretinography , Histones/metabolism , Immunohistochemistry , Male , Rats
2.
Invest Ophthalmol Vis Sci ; 55(11): 7486-93, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25358731

ABSTRACT

PURPOSE: The current studies investigate if the histone deacetylase (HDAC) inhibitor, valproic acid (VPA), can limit retinal ganglion cell (RGC) degeneration in an ocular-hypertensive rat model. METHODS: Intraocular pressure (IOP) was elevated unilaterally in Brown Norway rats by hypertonic saline injection. Rats received either vehicle or VPA (100 mg/kg) treatment for 28 days. Retinal ganglion cell function and number were assessed by pattern electroretinogram (pERG) and retrograde FluoroGold labeling. Western blotting and a fluorescence assay were used for determination of histone H3 acetylation and HDAC activity, respectively, at 3-day, 1-week, and 2-week time points. RESULTS: Hypertonic saline injections increased IOPs by 7 to 14 mm Hg. In vehicle-treated animals, ocular hypertension resulted in a 29.1% and 39.4% decrease in pERG amplitudes at 2 and 4 weeks, respectively, and a 42.9% decrease in mean RGC density at 4 weeks. In comparison, VPA treatment yielded significant amplitude preservation at 2 and 4 weeks and showed significant RGC density preservation at 4 weeks. No significant difference in RGC densities or IOPs was measured between control eyes of vehicle- and VPA-treated rats. In ocular-hypertensive eyes, class I HDAC activity was significantly elevated within 1 week (13.3 ± 2.2%) and histone H3 acetylation was significantly reduced within 2 weeks following the induction of ocular hypertension. CONCLUSIONS: Increase in HDAC activity is a relatively early retinal event induced by elevated IOP, and suppressing HDAC activity can protect RGCs from ocular-hypertensive stress. Together these data provide a basis for developing HDAC inhibitors for the treatment of optic neuropathies.


Subject(s)
Intraocular Pressure , Ocular Hypertension/metabolism , Retinal Ganglion Cells/metabolism , Acetylation , Acetyltransferases/metabolism , Animals , Blotting, Western , Chronic Disease , Disease Models, Animal , Electroretinography , Enzyme Inhibitors/pharmacology , Female , Histone Deacetylase Inhibitors/pharmacology , Immunohistochemistry , Male , Ocular Hypertension/drug therapy , Ocular Hypertension/physiopathology , Rats , Rats, Inbred BN , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Valproic Acid/pharmacology
3.
Exp Eye Res ; 127: 124-31, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25064603

ABSTRACT

Neuroretinal ischemic injury contributes to several degenerative diseases in the eye and the resulting pathogenic processes involving a series of necrotic and apoptotic events. This study investigates the time and extent of changes in acetylation, and whether this influences function and survival of neuroretinal cells following injury. Studies evaluated the time course of changes in histone deacetylase (HDAC) activity, histone-H3 acetylation and caspase-3 activation levels as well as retinal morphology and function (electroretinography) following ischemia. In addition, the effect of two HDAC inhibitors, trichostatin-A and valproic acid were also investigated. In normal eyes, retinal ischemia produced a significant increase in HDAC activity within 2 h that was followed by a corresponding significant decrease in protein acetylation by 4 h. Activated caspase-3 levels were significantly elevated by 24 h. Treatment with HDAC inhibitors blocked the early decrease in protein acetylation and activation of caspase-3. Retinal immunohistochemistry demonstrated that systemic administration of trichostatin-A or valproic acid, resulted in hyperacetylation of all retinal layers after systemic treatment. In addition, HDAC inhibitors provided a significant functional and structural neuroprotection at seven days following injury relative to vehicle-treated eyes. These results provide evidence that increases in HDAC activity is an early event following retinal ischemia, and are accompanied by corresponding decreases in acetylation in advance of caspase-3 activation. In addition to preserving acetylation status, the administration of HDAC inhibitors suppressed caspase activation and provided structural and functional neuroprotection in model of ischemic retinal injury. Taken together these data provide evidence that decrease in retinal acetylation status is a central event in ischemic retinal injury, and the hyperacetylation induced by HDAC inhibition can provide acute neuroprotection.


Subject(s)
Caspase 3/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Lysine/metabolism , Reperfusion Injury/prevention & control , Retinal Degeneration/prevention & control , Acetylation , Animals , Blotting, Western , Cell Survival , Electroretinography , Female , Hydroxamic Acids/pharmacology , Male , Neuroprotective Agents/pharmacology , Rats, Inbred BN , Reperfusion Injury/enzymology , Reperfusion Injury/physiopathology , Retinal Degeneration/enzymology , Retinal Degeneration/physiopathology , Retinal Neurons/drug effects , Retinal Neurons/enzymology , Retinal Neurons/pathology , Valproic Acid/pharmacology
4.
Invest Ophthalmol Vis Sci ; 55(4): 2269-75, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24550368

ABSTRACT

PURPOSE: Retinal edema, the accumulation of extracellular fluid in the retina is usually attributed to inner blood retina barrier (BRB) leakage. Vascular endothelial growth factor plays an important role in this process. The effects of VEGF on the outer BRB, the RPE, however, have received limited attention. Here, we present a methodology to assess how VEGF modulates the integrity of the RPE barrier in vivo. METHODS: Control subretinal blebs (1-5 µL) and blebs containing VEGF (1-100 µg/mL), placental growth factor (PlGF; 100 µg/mL), or albumin (100-1000 µg/mL) were injected into New Zealand White or Dutch Belted rabbits with IOP maintained at 10, 15, or 20 mm Hg. One-hour intravitreal pretreatment with ZM323881 (10 µM/L) was used to inhibit the VEGF response. Fluid resorption was followed by optical coherence tomography for 1 hour. Retinal pigment epithelium leakage was assessed by fluorescein angiography. RESULTS: Increasing IOP resulted in an elevated rate of bleb resorption, while increasing albumin concentration in the bleb decreased the rate of resorption. Vascular endothelial growth factor, but not PlGF, caused a significant, concentration-dependent decrease in the rate of fluid resorption, which was reversed by ZM323881. Compared with albumin-filled blebs, VEGF-filled blebs showed accelerated early-phase leakage from the choroid. CONCLUSIONS: Consistent with a localized modulation of RPE function, VEGF induced a significant reduction in fluid resorption and an increase in hydraulic conductivity. Our results establish VEGF as a major cytokine regulating RPE barrier properties in vivo and indicate that the RPE is a principal factor in the pathogenesis of retinal edema.


Subject(s)
Blood-Retinal Barrier/physiology , Papilledema/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Disease Models, Animal , Fluorescein Angiography , Fundus Oculi , Papilledema/pathology , Rabbits , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence
5.
Invest Ophthalmol Vis Sci ; 54(6): 4072-80, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23696608

ABSTRACT

PURPOSE: Protein acetylation is an essential mechanism in regulating transcriptional and inflammatory events. Studies have shown that nonselective histone deacetylase (HDAC) inhibitors can protect the retina from ischemic injury in rats. However, the role of specific HDAC isoforms in retinal degenerative processes remains obscure. The purpose of this study was to investigate the role of HDAC2 isoform in a mouse model of ischemic retinal injury. METHODS: Localization of HDAC2 in mice retinas was evaluated by immunohistochemical analyses. To investigate whether selective reduction in HDAC2 activity can protect the retina from ischemic injury, Hdac2⁺/⁻ mice were utilized. Electroretinographic (ERG) and morphometric analyses were used to assess retinal function and morphology. RESULTS: Our results demonstrated that HDAC2 is primarily localized in nuclei in inner nuclear and retinal ganglion cell layers, and HDAC2 activity accounted for approximately 35% of the total activities of HDAC1, 2, 3, and 6 in the retina. In wild-type mice, ERG a- and b-waves from ischemic eyes were significantly reduced when compared to pre-ischemia baseline values. Morphometric examination of these eyes revealed significant degeneration of inner retinal layers. In Hdac2⁺/⁻ mice, ERG a- and b-waves from ischemic eyes were significantly greater than those measured in ischemic eyes from wild-type mice. Morphologic measurements demonstrated that Hdac2⁺/⁻ mice exhibit significantly less retinal degeneration than wild-type mice. CONCLUSIONS: This study demonstrated that suppressing HDAC2 expression can effectively reduce ischemic retinal injury. Our results support the idea that the development of selective HDAC2 inhibitors may provide an efficacious treatment for ischemic retinal injury.


Subject(s)
Disease Models, Animal , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Reperfusion Injury/prevention & control , Retina/drug effects , Retinal Degeneration/prevention & control , Animals , Electroretinography , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Hydroxamic Acids/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/enzymology , Retina/metabolism , Retina/physiology , Retinal Degeneration/enzymology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/physiology
6.
J Pharmacol Exp Ther ; 344(1): 96-102, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23086231

ABSTRACT

In diabetic retinopathy, vision loss is usually secondary to macular edema, which is thought to depend on the functional integrity of the blood-retina barrier. The levels of advanced glycation end products in the vitreous correlate with the progression of diabetic retinopathy. Natriuretic peptides (NP) are expressed in the eye and their receptors are present in the retinal pigment epithelium (RPE). Here, we investigated the effect of glycated-albumin (Glyc-alb), an advanced glycation end product model, on RPE-barrier function and the ability of NP to suppress this response. Transepithelial electrical resistance (TEER) measurements were used to assess the barrier function of ARPE-19 and human fetal RPE (hfRPE) monolayers. The monolayers were treated with 0.1-100 µg/ml Glyc-alb in the absence or presence of 1 pM to 100 nM apical atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), or C-type natriuretic peptide (CNP). Glyc-alb induced a significant reduction in TEER within 2 hours. This response was concentration-dependent (EC(50)= 2.3 µg/ml) with a maximal reduction of 40 ± 2% for ARPE-19 and 27 ± 7% for hfRPE at 100 µg/ml 6 hours post-treatment. One hour pretreatment with ANP, BNP, or CNP blocked the reduction in TEER induced by Glyc-alb (100 µg/ml). The suppression of the Glyc-alb response by NP was dependent on the generation of cyclic guanosine monophosphate and exhibited a rank order of agonist potency consistent with the activation of natriuretic-peptide-receptor-2 (NPR2) subtype (CNP >> BNP ≥ ANP). Our data demonstrate that Glyc-alb is effective in reducing RPE-barrier function, and this response is suppressed by NP. Moreover, these studies support the idea that NPR2 agonists can be potential candidates for treating retinal edema in diabetic patients.


Subject(s)
Blood-Retinal Barrier/drug effects , Glycation End Products, Advanced/antagonists & inhibitors , Glycation End Products, Advanced/toxicity , Natriuretic Peptide, C-Type/pharmacology , Retinal Pigment Epithelium/drug effects , Atrial Natriuretic Factor/pharmacology , Blotting, Western , Cell Line , Cyclic AMP/physiology , Cyclic GMP/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Natriuretic Peptide, Brain/pharmacology , Serum Albumin/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Glycated Serum Albumin
7.
Invest Ophthalmol Vis Sci ; 51(7): 3639-45, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20164449

ABSTRACT

PURPOSE. The pathogenesis of retinal ischemia results from a series of events involving changes in gene expression and inflammatory cytokines. Protein acetylation is an essential mechanism in regulating transcriptional and inflammatory events. The purpose of this study was to investigate the neuroprotective action of the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) in a retinal ischemic model. METHODS. To investigate whether HDAC inhibition can reduce ischemic injury, rats were treated with TSA (2.5 mg/kg intraperitoneally) twice daily on days 0, 1, 2, and 3. Seven days after ischemic injury, morphometric and electroretinographic (ERG) analyses were used to assess retinal structure and function. Western blot and immunohistochemical analyses were used to evaluate TSA-induced changes in histone-H3 acetylation and MMP secretion. RESULTS. In vehicle-treated animals, ERG a- and b-waves from ischemic eyes were significantly reduced compared with contralateral responses. In addition, histologic examination of these eyes revealed significant degeneration of inner retinal layers. In rats treated with TSA, amplitudes of ERG a- and b-waves from ischemic eyes were significantly increased, and normal inner retina morphology was preserved. Ischemia also increased the levels of retinal TNF-alpha, which was blocked by TSA treatment. In astrocyte cultures, the addition of TNF-alpha (10 ng/mL) stimulated the secretion of MMP-1 and MMP-3, which were blocked by TSA (100 nM). CONCLUSIONS. These studies provide the first evidence that suppressing HDAC activity can protect the retina from ischemic injury. This neuroprotective response is associated with the suppression of retinal TNF-alpha expression and signaling. The use of HDAC inhibitors may provide a novel treatment for ischemic retinal injury.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Reperfusion Injury/prevention & control , Retina/drug effects , Retinal Diseases/prevention & control , Acetylation , Animals , Blotting, Western , Electroretinography , Female , Histones/metabolism , Immunohistochemistry , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Inbred BN , Reperfusion Injury/enzymology , Retinal Diseases/enzymology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...