Network


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

Hotspot


Dive into the research topics where Ahmed Elsherbini is active.

Publication


Featured researches published by Ahmed Elsherbini.


BioMed Research International | 2015

MicroRNA-146b-3p Regulates Retinal Inflammation by Suppressing Adenosine Deaminase-2 in Diabetes

Sadanand Fulzele; Ahmed Elsherbini; Saif Ahmad; Rajnikumar Sangani; Suraporn Matragoon; Azza B. El-Remessy; Reshmitha Radhakrishnan; Gregory I. Liou

Hyperglycemia- (HG-) Amadori-glycated albumin- (AGA-) induced activation of microglia and monocytes and their adherence to retinal vascular endothelial cells contribute to retinal inflammation leading to diabetic retinopathy (DR). There is a great need for early detection of DR before demonstrable tissue damages become irreversible. Extracellular adenosine, required for endogenous anti-inflammation, is regulated by the interplay of equilibrative nucleoside transporter with adenosine deaminase (ADA) and adenosine kinase. ADA, including ADA1 and ADA2, exists in all organisms. However, because ADA2 gene has not been identified in mouse genome, how diabetes alters adenosine-dependent anti-inflammation remains unclear. Studies of pig retinal microglia and human macrophages revealed a causal role of ADA2 in inflammation. Database search suggested miR-146b-3p recognition sites in the 3′-UTR of ADA2 mRNA. Coexpression of miR-146b-3p, but not miR-146-5p or nontargeting miRNA, with 3′-UTR of the ADA2 gene was necessary to suppress a linked reporter gene. In the vitreous of diabetic patients, decreased miR-146b-3p is associated with increased ADA2 activity. Ectopic expression of miR-146b-3p suppressed ADA2 expression, activity, and TNF-α release in the AGA-treated human macrophages. These results suggest a regulatory role of miR-146b-3p in diabetes related retinal inflammation by suppressing ADA2.


Life Sciences | 2013

ABT-702, an adenosine kinase inhibitor, attenuates inflammation in diabetic retinopathy

Nehal M. Elsherbiny; Saif Ahmad; Mohammad Naime; Ahmed Elsherbini; Sadanand Fulzele; Mohammed M.H. Al-Gayyar; Laila A. Eissa; Mamdouh M. El-Shishtawy; Gregory I. Liou

AIMS This study was undertaken to determine the effect of an adenosine kinase inhibitor (AKI) in diabetic retinopathy (DR). We have shown previously that adenosine signaling via A2A receptors (A2AAR) is involved in retinal protection from diabetes-induced inflammation. Here we demonstrate that AKI-enhanced adenosine signaling provides protection from DR in mice. MAIN METHODS We targeted AK, the key enzyme in adenosine metabolism, using a treatment regime with the selective AKI, ABT-702 (1.5mg/kg intraperitoneally twice a week) commencing at the beginning of streptozotocin-induced diabetes at the age of eight weeks. This treatment, previously demonstrated to increase free adenosine levels in vivo, was maintained until the age of 16 weeks. Retinal inflammation was evaluated using Western blot, Real-Time PCR and immuno-staining analyses. Role of A2AAR signaling in the anti-inflammation effect of ABT-702 was analyzed in Amadori-glycated-albumin (AGA)-treated microglial cells. KEY FINDINGS At 16 weeks, when diabetic mice exhibit significant signs of retinal inflammation including up-regulation of oxidative/nitrosative stress, A2AAR, ENT1, Iba1, TNF-α, ICAM1, retinal cell death, and down-regulation of AK, the ABT-702 treated group showed lower signs of inflammation compared to control animals receiving the vehicle. The involvement of adenosine signaling in the anti-inflammation effect of ABT-702 was supported by the TNF-α release blocking effect of A2AAR antagonist in AGA-treated microglial cells. SIGNIFICANCE These results suggest a role for AK in regulating adenosine receptor signaling in the retina. Inhibition of AK potentially amplifies the therapeutic effects of site- and event-specific accumulation of extracellular adenosine, which is of highly translational impact.


Biochemical and Biophysical Research Communications | 2013

Potential roles of adenosine deaminase-2 in diabetic retinopathy

Nehal M. Elsherbiny; Mohammad Naime; Saif Ahmad; Ahmed Elsherbini; Shuaib Mohammad; Sadanand Fulzele; Azza B. El-Remessy; Mohammed M.H. Al-Gayyar; Laila A. Eissa; Mamdouh M. El-Shishtawy; Guichun Han; Richard E. White; Toque Flores Haroldo; Gregory I. Liou

The early activation of microglia that induces retinal inflammation in DR may serve as a target for therapeutic intervention of DR. Our demonstration that retinal inflammation is attenuated via adenosine receptor A(2A)AR supports the hypothesis that a mechanism to maintain extracellular concentrations of adenosine important in normal physiology is impaired in DR. Extracellular concentrations of adenosine are regulated by the interplay of equiliberative nucleoside transporter (ENT)s with enzymes of adenosine metabolism including adenosine deaminase-1 (ADA1), adenosine kinase (AK) and CD73. In the vertebrates but not rodents, a macrophage-associated ADA2 is identified. The role of ADA2 is, therefore, understudied as the sequencing probes or antibodies to mouse ADA2 are not available. We identified increased ADA2 expression and activity in human and porcine retinas with diabetes, and in Amadori glycated albumin (AGA)- or hyperglycemia-treated porcine and human microglia. In rodent as well as porcine cells, modulation of TNF-α release is mediated by A(2A)AR. Quantitative analysis of normal and diabetic porcine retinas reveals that while the expression levels of ADA2, A2AAR, ENT1, TNF-α and MMP9 are increased, the levels of AK are reduced during inflammation as an endogenous protective mechanism. To determine the role of ADA2, we found that AGA induces ADA2 expression, ADA2 activity and TNF-α release, and that TNF-α release is blocked by ADA2-neutralizing antibody or ADA2 siRNA, but not by scrambled siRNA. These results suggest that retinal inflammation in DR is mediated by ADA2, and that the anti-inflammatory activity of A(2A)AR signaling is impaired in diabetes due to increased ADA2 activity.


Journal of Neuroimmunology | 2013

Potential role of A2A adenosine receptor in traumatic optic neuropathy.

Saif Ahmad; Nadeem Fatteh; Nehal M. Elsherbiny; Mohammad Naime; Ahmed S. Ibrahim; Ahmed Elsherbini; Sally Elshafey; Sohail Khan; Sadanand Fulzele; Joyce Gonzales; Gregory I. Liou

In traumatic optic neuropathy (TON), apoptosis of retinal ganglion cells is closely related to the local production of reactive oxygen species and inflammatory mediators from activated microglial cells. Adenosine receptor A2A (A2AAR) has been shown to possess anti-inflammatory properties that have not been studied in TON. In the present study, we examined the role of A2AAR in retinal complications associated with TON. Initial studies in wild-type mice revealed that treatment with the A2AAR agonist resulted in marked decreases in the TON-induced microglial activation, retinal cell death and releases of reactive oxygen species and pro-inflammatory cytokines TNF-α and IL-6. To further assess the role of A2AAR in TON, we studied the effects of A2AAR ablation on the TON-induced retinal abnormalities. A2AAR-/- mice with TON showed a significantly higher mRNA level of TNF-α, Iba1-1 in retinal tissue, and ICAM-1 expression in retinal sections compared with wild-type mice with TON. To explore a potential mechanism by which A2AAR-signaling regulates inflammation in TON, we performed additional studies using hypoxia- or LPS-treated microglial cells as an in vitro model for TON. Activation of A2AAR attenuates hypoxia or LPS-induced TNF-α release and significantly repressed the inflammatory signaling, ERK in the activated microglia. Collectively, this work provides pharmacological and genetic evidence for A2AAR signaling as a control point of cell death in TON and suggests that the retinal protective effect of A2AAR is mediated by attenuating the inflammatory response that occurs in microglia via interaction with MAPKinase pathway.


Journal of Neuroimmunology | 2014

Inhibition of adenosine kinase attenuates inflammation and neurotoxicity in traumatic optic neuropathy.

Saif Ahmad; Nehal M. Elsherbiny; Kanchan Bhatia; Ahmed Elsherbini; Sadanand Fulzele; Gregory I. Liou

Traumatic optic neuropathy (TON) is associated with apoptosis of retinal ganglion cells. Local productions of reactive oxygen species and inflammatory mediators from activated microglial cells have been hypothesized to underlie apoptotic processes. We previously demonstrated that the anti-inflammatory effect of adenosine, through A2A receptor activation had profound protective influence against retinal injury in traumatic optic neuropathy. This protective effect is limited due to rapid cellular re-uptake of adenosine by equilibrative nucleotside transporter-1 (ENT1) or break down by adenosine kinase (AK), the key enzyme in adenosine clearance pathway. Further, the use of adenosine receptors agonists are limited by systemic side effects. Therefore, we seek to investigate the potential role of amplifying the endogenous ambient level of adenosine by pharmacological inhibition of AK. We tested our hypothesis by comparing TON-induced retinal injury in mice with and without ABT-702 treatment, a selective AK inhibitor (AKI). The retinal-protective effect of ABT-702 was demonstrated by significant reduction of Iba-1, ENT1, TNF-α, IL-6, and iNOS/nNOS protein or mRNA expression in TON as revealed by western blot and real time PCR. TON-induced superoxide anion generation and nitrotyrosine expression were reduced in ABT-702 treated mice retinal sections as determined by immunoflourescence. In addition, ABT-702 attenuated p-ERK1/2 and p-P38 activation in LPS induced activated mouse microglia cells. The results of the present investigation suggested that ABT-702 had a protective role against marked TON-induced retinal inflammation and damage by augmenting the endogenous therapeutic effects of site- and event-specific accumulation of extracellular adenosine.


Journal of Orthopaedic Research | 2017

Mechanical loading disrupts osteocyte plasma membranes which initiates mechanosensation events in bone

Kanglun Yu; David P. Sellman; Anoosh Bahraini; Mackenzie L. Hagan; Ahmed Elsherbini; Kayce T. Vanpelt; Peyton L. Marshall; Mark W. Hamrick; Anna K. McNeil; Paul L. McNeil; Meghan E. McGee-Lawrence

Osteocytes sense loading in bone, but their mechanosensation mechanisms remain poorly understood. Plasma membrane disruptions (PMD) develop with loading under physiological conditions in many cell types (e.g., myocytes, endothelial cells). These PMD foster molecular flux across cell membranes that promotes tissue adaptation, but this mechanosensation mechanism had not been explored in osteocytes. Our goal was to investigate whether PMD occur and initiate consequent mechanotransduction in osteocytes during physiological loading. We found that osteocytes experience PMD during in vitro (fluid flow) and in vivo (treadmill exercise) mechanical loading, in proportion to the level of stress experienced. In fluid flow studies, osteocyte PMD preferentially formed with rapid as compared to gradual application of loading. In treadmill studies, osteocyte PMD increased with loading in weight bearing locations (tibia), but this trend was not seen in non‐weight bearing locations (skull). PMD initiated osteocyte mechanotransduction including calcium signaling and expression of c‐fos, and repair rates of these PMD could be enhanced or inhibited pharmacologically to alter downstream mechanotransduction and osteocyte survival. PMD may represent a novel mechanosensation pathway in bone and a target for modifying skeletal adaptation signaling in osteocytes.


Cancer Research | 2016

Abstract 4672: Targeting MEK/MAPK1/2 in vivo to eradicate antiestrogen resistance

Jason Conger; Matthew Manning; Kingsley Anosike; Ahmed Elsherbini; Brandon Ware; Emily Bass; Mathusamy Thangaraju

Tamoxifen (TAM), an antiestrogen and selective estrogen receptor modifier (SERM) has become a widely utilized therapeutic drug for treating ER+ breast cancer, however many cases develop TAM resistance. In previous in vitro studies, TAM combined with the selective Mitogen/Extracellular signal-regulated Kinase pathway (MEK) inhibitor U0126 (U) led to increased levels of the proapoptotic protein BimEL. This combined treatment was more effective at killing estrogen receptor expressing (ER+) breast cancer cells than either agent alone via a Bim-dependent mechanism [Periyasamy-Thandavan el al., Breast Cancer Res. 14(2), 2012]. In this study, we aimed to recapitulate the superior efficacy of this combined treatment strategy with the clinically relevant MEK inhibitor Selumetinib (SEL) using the TAM sensitive, ER+ MCF-7 breast cancer cell line. In addition, we utilized MCF7-TAM resistant cells (TR5), created in vitro through exposures to incremental increases in TAM concentration(100ng to 5.0μM). MCF-7 cells that were treated with TAM, SEL, U, or combination therapies in vitro demonstrated a similar efficacy between U and SEL with the lowest cell viability resulting from TAM plus U or SEL compared to TAM, U, or SEL as single agents. Western blot analysis of U- or SEL-treated MCF7 cells showed a similar robust upregulation of BimEL in the presence or absence of TAM. Comparison of the TR5 TAM resistant cells to parent MCF-7 cells showed elevated levels of active MAPK1/2 protein (p-MAPK) that mediated the phosphorylation and subsequent proteasomal degradation of BimEL. Importantly, inhibition of MEK/MAPK1/2 by SEL or U in TR5 cells in vitro significantly increased BimEL levels and TAM induced apoptosis as evidenced by increased levels of cleaved PARP and/or lamin A. In a pilot in vivo study, we further demonstrated that TR5 cells were highly tumorigenic with the xenografts being estrogen independent and insensitive to TAM inhibition. In contrast, the TR5 xenografts were sensitive to SEL when delivered in the presence or absence of TAM. Further, tissue analyzed from SEL-treated TR5 xenografts showed significantly higher levels of BimEL than tissue analyzed from TAM- or E2-treated xenografts. These initial studies provide strong data that MEK1/MAPK1/2 inhibition will be required in vivo to raise BimEL levels to negatively affect breast cancer growth. These studies also support the concept of combining SEL with antiestrogen treatment (as a combined adjuvant therapy) to reduce the emergence of antiestrogen resistant cells and to include SEL as part of a regimen to treat antiestrogen resistant breast cancer. Citation Format: Jason Conger, Matthew Manning, Kingsley Anosike, Ahmed Elsherbini, Brandon Ware, Emily Bass, Mathusamy Thangaraju, Darren Browning, Patricia V. Schoenlein. Targeting MEK/MAPK1/2 in vivo to eradicate antiestrogen resistance. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4672.


BMC Genomics | 2014

Adenosine kinase as a therapeutic target in traumatic optic neuropathy

Saif Ahmad; Nehal M. Elsherbiny; Ahmed Elsherbini; Sadan Fulzele; Gregory I. Liou

Background The purpose of this study is to understand the mechanism of traumatic optic neuropathy (TON) in order to prevent vision loss. Following traumatic insults to the optic nerve, retinal microglia cells are activated through MAP Kinase pathways and increased cytotoxic activity that causes retinal ganglion cell death [1]. Under stress condition, extracellular concentration of adenosine is likely to increase and activates an anti-inflammatory pathway through A2A adenosine receptor. But in TON, the accumulated extracellular adenosine is then transported intracellular through equilibrative nucleoside transporters (ENTs) which further gets converted into AMP by Adenosine kinase (AK), which results in low extracellular adenosine concentration. We have demonstrated that microglia activation and TNF-a release was inhibited by AK inhibitor. Based on these findings, we hypothesize that an imbalance in adenosine formation and metabolism in the retinal microglia participated by AK may contribute significantly to retinal complications in the setting of TON.


Investigative Ophthalmology & Visual Science | 2014

The micro-RNA miR-146b-3p controls diabetic retinal inflammation by suppressing adenosine deaminase-2

Ahmed Elsherbini; Sadanand Fulzele; Saif Ahmad; Rajini Sangani; Suraporn Matragoon; Azza B. El-Remessy; Gregory I. Liou


Archive | 2013

Role of Adenosine Receptor A2A in Traumatic Optic Neuropathies

Gregory I. Liou; Saif Ahmad; Ahmed Elsherbini

Collaboration


Dive into the Ahmed Elsherbini's collaboration.

Top Co-Authors

Avatar

Saif Ahmad

King Abdulaziz University

View shared research outputs
Top Co-Authors

Avatar

Gregory I. Liou

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar

Sadanand Fulzele

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mohammad Naime

Georgia Regents University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge