Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Oday Alsarraf is active.

Publication


Featured researches published by Oday Alsarraf.


Investigative Ophthalmology & Visual Science | 2010

Inhibition of Histone Deacetylase Protects the Retina from Ischemic Injury

Craig E. Crosson; Santhosh K. Mani; Shahid Husain; Oday Alsarraf; Donald R. Menick

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.


Investigative Ophthalmology & Visual Science | 2013

Inhibition of HDAC2 Protects the Retina From Ischemic Injury

Jie Fan; Oday Alsarraf; Mohammad Dahrouj; Kenneth A. Platt; C. James Chou; Dennis S. Rice; Craig E. Crosson

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.


Investigative Ophthalmology & Visual Science | 2014

Vascular Endothelial Growth Factor Modulates the Function of the Retinal Pigment Epithelium In Vivo

Mohammad Dahrouj; Oday Alsarraf; Jake C. McMillin; Yueying Liu; Craig E. Crosson; Zsolt Ablonczy

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.


Journal of Pharmacology and Experimental Therapeutics | 2013

C-type natriuretic peptide protects the retinal pigment epithelium against advanced glycation end-product-induced barrier dysfunction

Mohammad Dahrouj; Oday Alsarraf; Yueying Liu; Craig E. Crosson; Zsolt Ablonczy

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 (EC50= 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.


Investigative Ophthalmology & Visual Science | 2014

Acetylation preserves retinal ganglion cell structure and function in a chronic model of ocular hypertension.

Oday Alsarraf; Jie Fan; Mohammad Dahrouj; C. James Chou; Phillip W. Yates; Craig E. Crosson

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.


Experimental Eye Research | 2014

Acetylation: A lysine modification with neuroprotective effects in ischemic retinal degeneration

Oday Alsarraf; Jie Fan; Mohammad Dahrouj; C. James Chou; Donald R. Menick; Craig E. Crosson


Experimental Eye Research | 2016

Ischemic preconditioning, retinal neuroprotection and histone deacetylase activities

Jie Fan; Oday Alsarraf; C. James Chou; Phillip W. Yates; Nicole Cathleen Goodwin; Dennis S. Rice; Craig E. Crosson


Investigative Ophthalmology & Visual Science | 2014

Histone Deacetylases in Retinal Injury and Ischemic Preconditioning

Oday Alsarraf; Jie Fan; Phillip W. Yates; Craig E. Crosson


Investigative Ophthalmology & Visual Science | 2014

Acid sphingomyelinase activation suppresses the ischemia-induced production of TNF-α in the retina

Jie Fan; Oday Alsarraf; Craig E. Crosson


Investigative Ophthalmology & Visual Science | 2013

Localization and Activity of Histone Deacetylases in the Retina

Oday Alsarraf; Jie Fan; C. James Chou; Craig E. Crosson

Collaboration


Dive into the Oday Alsarraf's collaboration.

Top Co-Authors

Avatar

Craig E. Crosson

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Jie Fan

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

C. James Chou

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Mohammad Dahrouj

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Phillip W. Yates

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Donald R. Menick

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Santhosh K. Mani

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Yueying Liu

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Zsolt Ablonczy

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge