Network


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

Hotspot


Dive into the research topics where Hassan Y. Ebrahim is active.

Publication


Featured researches published by Hassan Y. Ebrahim.


Chemical Biology & Drug Design | 2015

Discovery, Optimization, and Pharmacophore Modeling of Oleanolic Acid and Analogues as Breast Cancer Cell Migration and Invasion Inhibitors Through Targeting Brk/Paxillin/Rac1 Axis

Heba E. Elsayed; Mohamed R. Akl; Hassan Y. Ebrahim; Asmaa A. Sallam; Eman G. Haggag; Amel M. Kamal; Khalid A. El Sayed

Bioassay‐guided fractionation of Terminalia bentzoe L. leaves methanol extract identified the known triterpene oleanolic acid (1) as its major breast cancer cell migration inhibitor. Further chemical optimization afforded five new (9–12 and 15) and seven known (4–8, 13, and 14) semisynthetic analogues. All compounds were tested for their ability to inhibit human breast cancer MDA‐MB‐231 cells migration, proliferation, and invasion. The results revealed that 3‐O‐[N‐(3′‐chlorobenzenesulfonyl)‐carbamoyl]‐oleanolic acid (11) and 3‐O‐[N‐(5′‐fluorobenzenesulfonyl)‐carbamoyl]‐oleanolic acid (12) were the most active hits at low μm concentration. Western blot analysis indicated the activity of 1, 11, and 12 might be related, at least in part, to the suppression of Brk/Paxillin/Rac1 signaling pathway. Pharmacophore modeling study was conducted to better understand the common structural binding epitopes important for the antimigratory activity. The sulfonyl carbamoyl moiety with an optimal bulkiness electron‐deficient phenyl ring is associated with improved activity. This study is the first to discover the antimigratory and anti‐invasive activities of oleanolic acid and analogues through targeting the Brk/Paxillin/Rac1 axis.


Marine Drugs | 2015

Araguspongine C induces autophagic death in breast cancer cells through suppression of c-Met and HER2 receptor tyrosine kinase signaling.

Mohamed R. Akl; Nehad M. Ayoub; Hassan Y. Ebrahim; Mohamed M. Mohyeldin; Khaled Y. Orabi; Ahmed I. Foudah; Khalid A. El Sayed

Receptor tyrosine kinases are key regulators of cellular growth and proliferation. Dysregulations of receptor tyrosine kinases in cancer cells may promote tumorigenesis by multiple mechanisms including enhanced cell survival and inhibition of cell death. Araguspongines represent a group of macrocyclic oxaquinolizidine alkaloids isolated from the marine sponge Xestospongia species. This study evaluated the anticancer activity of the known oxaquinolizidine alkaloids araguspongines A, C, K and L, and xestospongin B against breast cancer cells. Araguspongine C inhibited the proliferation of multiple breast cancer cell lines in vitro in a dose-dependent manner. Interestingly, araguspongine C-induced autophagic cell death in HER2-overexpressing BT-474 breast cancer cells was characterized by vacuole formation and upregulation of autophagy markers including LC3A/B, Atg3, Atg7, and Atg16L. Araguspongine C-induced autophagy was associated with suppression of c-Met and HER2 receptor tyrosine kinase activation. Further in-silico docking studies and cell-free Z-LYTE assays indicated the potential of direct interaction between araguspongine C and the receptor tyrosine kinases c-Met and HER2 at their kinase domains. Remarkably, araguspongine C treatment resulted in the suppression of PI3K/Akt/mTOR signaling cascade in breast cancer cells undergoing autophagy. Induction of autophagic death in BT-474 cells was also associated with decreased levels of inositol 1,4,5-trisphosphate receptor upon treatment with effective concentration of araguspongine C. In conclusion, results of this study are the first to reveal the potential of araguspongine C as an inhibitor to receptor tyrosine kinases resulting in the induction of autophagic cell death in breast cancer cells.


European Journal of Pharmacology | 2014

3-O-[N-(p-fluorobenzenesulfonyl)-carbamoyl]-oleanolic acid, a semisynthetic analog of oleanolic acid, induces apoptosis in breast cancer cells.

Mohamed R. Akl; Heba E. Elsayed; Hassan Y. Ebrahim; Eman G. Haggag; Amel M. Kamal; Khalid A. El Sayed

Oleanolic acid (OA), a pentacyclic triterpene acid widely distributed in food and traditional herbal remedies, exhibits diverse therapeutic effects. OA has been subjected to various chemical modifications to optimize its anticancer effect. Among other analogs, 3-O-[N-(p-fluorobenzenesulfonyl)-carbamoyl]-oleanolic acid (PFOA) was semisynthesized from OA. This study evaluates the cytotoxic effects of PFOA on MDA-MB-231, MCF-7, BT-474, and T-47D human breast cancer cells. Acute treatment of PFOA inhibited breast cancer cell viability in a dose-dependent manner. Treatment of PFOA at cytotoxic doses significantly induced apoptosis in cancer cells as shown by flow cytometry analysis. Activation of apoptosis in MCF-7 and BT-474 cells seemed to be initiated through induction of Fas ligand, which resulted in activation of caspase-8 and PARP-1, whereas apoptosis in MDA-MB-231 cells was initiated by the activation of caspase-9, caspase-3 and PARP-1. The mechanism of apoptosis induction in T-47D involves activation of PARP-1. PFOA decreased the expression of EGFR, HER-2, MET and ERα in human breast cancer cell lines. These findings suggest that PFOA inhibits cell growth, activates apoptosis, and decreases the expression of key proteins involved in progression of breast cancer.


Marine Drugs | 2016

Discovery of Novel Antiangiogenic Marine Natural Product Scaffolds

Hassan Y. Ebrahim; Khalid A. El Sayed

Marine natural products (MNPs) are recognized for their structural complexity, diversity, and novelty. The vast majority of MNPs are pharmacologically relevant through their ability to modulate macromolecular targets underlying human diseases. Angiogenesis is a fundamental process in cancer progression and metastasis. Targeting angiogenesis through selective modulation of linked protein kinases is a valid strategy to discover novel effective tumor growth and metastasis inhibitors. An in-house marine natural products mini-library, which comprises diverse MNP entities, was submitted to the Lilly’s Open Innovation Drug Discovery platform. Accepted structures were subjected to in vitro screening to discover mechanistically novel angiogenesis inhibitors. Active hits were subjected to additional angiogenesis-targeted kinase profiling. Some natural and semisynthetic MNPs, including multiple members of the macrolide latrunculins, the macrocyclic oxaquinolizidine alkaloid araguspongine C, and the sesquiterpene quinone puupehenone, showed promising results in primary and secondary angiogenesis screening modules. These hits inhibited vascular endothelial growth factor (VEGF)-mediated endothelial tube-like formation, with minimal cytotoxicity at relevant doses. Secondary kinase profiling identified six target protein kinases, all involved in angiogenesis signaling pathways. Molecular modeling and docking experiments aided the understanding of molecular binding interactions, identification of pharmacophoric epitopes, and deriving structure-activity relationships of active hits. Marine natural products are prolific resources for the discovery of chemically and mechanistically unique selective antiangiogenic scaffolds.


European Journal of Pharmaceutics and Biopharmaceutics | 2015

PEGylated γ-tocotrienol isomer of vitamin E: Synthesis, characterization, in vitro cytotoxicity, and oral bioavailability.

Ahmed Abu-Fayyad; Fathy A. Behery; Asmaa A. Sallam; Saeed Alqahtani; Hassan Y. Ebrahim; Khalid A. El Sayed; Amal Kaddoumi; Paul W. Sylvester; Jennifer L. Carroll; James A. Cardelli; Sami Nazzal

Vitamin E refers to a family of eight isomers divided into two subgroups, tocopherols and the therapeutically active tocotrienols (T3). The PEGylated α-tocopherol isomer of vitamin E (vitamin E TPGS) has been extensively investigated for its solubilizing capacity as a nonionic surfactant in various drug delivery systems. Limited information, however, is available about the PEG conjugates of the tocotrienol isomers of vitamin E. In this study two PEGylated γ-T3 variants with mPEG molecular weights of 350 (γ-T3PGS 350) and 1000 (γ-T3PGS 1000) were synthesized by a two-step reaction procedure and characterized by (1)H NMR, HPLC, and mass spectroscopy. The physical properties of their self-assemblies in water were characterized by zeta, CMC, and size analysis. Similar physical properties were found between the PEGylated T3 and vitamin E TPGS. PEGylated T3 were also found to retain the in vitro cytotoxic activity of the free T3 against the MCF-7 and the triple-negative MDA-MB-231 breast cancer cells. PEGylated γ-T3 also increased the oral bioavailability of γ-T3 by threefolds when compared to the bioavailability of γ-T3 formulated into a self-emulsified drug delivery system. No significant differences in biological activity were found between the PEG 350 and 100 conjugates. Results from this study suggest that PEGylation of γ-T3 represents a viable platform for the oral and parenteral delivery of γ-T3 for potential use in the prevention of breast cancer.


Marine Drugs | 2014

The Marine-Derived Sipholenol A-4-O-3',4'-Dichlorobenzoate Inhibits Breast Cancer Growth and Motility in Vitro and in Vivo through the Suppression of Brk and FAK Signaling

Mohamed R. Akl; Ahmed I. Foudah; Hassan Y. Ebrahim; Sharon A. Meyer; Khalid A. El Sayed

Sipholenol A is a natural sipholane triterpenoid isolated from the Red Sea sponge, Callyspongia siphonella. Previous studies showed the antimigratory and antiproliferative activities of the semisynthetic sipholenol A esters against breast cancer cell lines. This study investigated the effects of sipholenol A-4-O-3′,4′-dichlorobenzoate (SPA) on the growth, migration and invasion of diverse human breast cancer cells. Results showed that SPA inhibited the growth of the human breast cancer cells, MDA-MB-231, MCF-7, BT-474 and T-47D, in a dose-dependent manner. Immunofluorescent analysis showed that SPA significantly reduced Ki-67-positive cells in MDA-MB-231 cells. Flow cytometry and Western blot analyses revealed that SPA treatment suppressed MDA-MB-231 cell growth by inducing cell cycle arrest at the G1 phase. In addition, SPA suppressed breast cancer cell migration, invasion and decreased Brk and FAK activation in a dose-dependent manner. Molecular docking study suggested a perfect fitting at the FAK’s FERM domain, inhibiting the main autophosphorylation site, Y397, which was further confirmed by Western blot analysis. Most known small molecule FAK inhibitors target the kinase domain, creating several off-target side effects. The in vivo studies showed that SPA treatment suppressed breast tumor growth and Ki-67, CD31, p-Brk and p-FAK expression in orthotopic breast cancer in nude mice. In conclusion, SPA inhibited the growth, invasion and migration of breast cancer cells possibly via deactivating Brk and FAK signaling, suggesting good potential for therapeutic use to control invasive breast cancer.


European Journal of Pharmacology | 2017

The olive oil phenolic (-)-oleocanthal modulates estrogen receptor expression in luminal breast cancer in vitro and in vivo and synergizes with tamoxifen treatment

Nehad M. Ayoub; Abu Bakar Siddique; Hassan Y. Ebrahim; Mohamed M. Mohyeldin; Khalid A. El Sayed

ABSTRACT Luminal breast cancer represents a therapeutic challenge in terms of aggressive disease and emerging resistance to targeted therapy. (‐)‐Oleocanthal has demonstrated anticancer activity in multiple human cancers. The goal of this study was to explore the effect of (‐)‐oleocanthal treatment on growth of luminal breast cancer cells and to examine the effect of combination of (‐)‐oleocanthal with tamoxifen. Results showed that (‐)‐oleocanthal inhibited growth of BT‐474, MCF‐7, and T‐47D human breast cancer cells in mitogen‐free media with IC50 values of 32.7, 24.07, and 80.93 &mgr;M, respectively. Similarly, (‐)‐oleocanthal suppressed growth of BT‐474, MCF‐7, and T‐47D cells in 17&bgr;‐estradiol‐supplemented media with IC50 values of 22.28, 20.77, and 83.91 &mgr;M, respectively. Combined (‐)‐oleocanthal and tamoxifen treatments resulted in a synergistic growth inhibition of BT‐474, MCF‐7, and T‐47D cells with combination index values of 0.65, 0.61, and 0.53 for each cell line, respectively. In‐silico docking studies indicated high degree of overlapping for the binding of (‐)‐oleocanthal and 17&bgr;‐estradiol to estrogen receptors, while (‐)‐oleocanthal and tamoxifen have distinguished binding modes. Treatment with 5 mg/kg or 10 mg/kg (‐)‐oleocanthal resulted in 97% inhibition of tumor growth in orthotopic athymic mice bearing BT‐474 tumor xenografts compared to vehicle‐treated animals. (‐)‐Oleocanthal treatment reduced total levels of estrogen receptors in BT‐474 cells both in vitro and in vivo. Collectively, (‐)‐oleocanthal showed a potential beneficial effect in suppressing growth of hormone‐dependent breast cancer and improving sensitivity to tamoxifen treatment. These findings provide rational for evaluating the effect of (‐)‐oleocanthal in combination with endocrine treatments in luminal breast cancer. Graphical abstract Figure. No caption available.


Oncotarget | 2016

The oleocanthal-based homovanillyl sinapate as a novel c-Met inhibitor

Mohamed M. Mohyeldin; Mohamed R. Akl; Hassan Y. Ebrahim; Ana Maria Dragoi; Samantha S. Dykes; James A. Cardelli; Khalid A. El Sayed

The hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-Met) signaling axis has gained considerable attention as an attractive molecular target for therapeutic blockade of cancer. Inspired by the chemical structure of S (−)-oleocanthal, a natural secoiridoid from extra-virgin olive oil with documented anticancer activity against c-Met-dependent malignancies, the research presented herein reports on the discovery of the novel olive-derived homovanillyl sinapate (HVS) as a promising c-Met inhibitor. HVS was distinguished for its remarkable potency against wild-type c-Met and its oncogenic variant in cell-free assays and confirmed by in silico docking studies. Furthermore, HVS substantially impaired the c-Met-mediated growth across a broad spectrum of breast cancer cells, while similar treatment doses had no effect on the non-tumorigenic mammary epithelial cell growth. In addition, HVS caused a dose-dependent inhibition of HGF-induced, but not epidermal growth factor (EGF)-induced, cell scattering in addition to HGF-mediated migration, invasion, and 3-dimensional (3D) proliferation of tumor cell spheroids. HVS treatment effects were mediated via inhibition of ligand-mediated c-Met activation and its downstream mitogenic signaling and blocking molecular mediators involved in cellular motility across different cellular contexts. An interesting feature of HVS is its good selectivity for c-Met and Abelson murine leukemia viral oncogene homolog 1 (ABL1) when profiled against a panel of kinases. Docking studies revealed interactions likely to impart high dual affinity for both ABL1 and c-Met kinases. HVS markedly reduced tumor growth, showed excellent pharmacodynamics, and suppressed cell proliferation and microvessel density in an orthotopic model of triple negative breast cancer. Collectively, the present findings suggested that the oleocanthal-based HVS is a promising c-Met inhibitor lead entity with excellent therapeutic potential to control malignancies with aberrant c-Met activity.


Bioorganic & Medicinal Chemistry | 2016

The indole alkaloid meleagrin, from the olive tree endophytic fungus Penicillium chrysogenum, as a novel lead for the control of c-Met-dependent breast cancer proliferation, migration and invasion

Mohamed S. Mady; Mohamed M. Mohyeldin; Hassan Y. Ebrahim; Heba E. Elsayed; Wael E. Houssen; Eman G. Haggag; Randa F. Soliman; Khalid A. El Sayed

Fungi of the genus Penicillium produce unique and chemically diverse biologically active secondary metabolites, including indole alkaloids. The role of dysregulated hepatocyte growth factor (HGF) and its receptor, c-Met, in the development and progression of breast carcinoma is documented. The goal of this work is to explore the chemistry and bioactivity of the secondary metabolites of the endophytic Penicillium chrysogenum cultured from the leaf of the olive tree Olea europea, collected in its natural habitat in Egypt. This fungal extract showed good inhibitory activities against the proliferation and migration of several human breast cancer lines. The CH2Cl2 extract of P. chrysogenum mycelia was subjected to bioguided chromatographic separation to afford three known indole alkaloids; meleagrin (1), roquefortine C (2) and DHTD (3). Meleagrin inhibited the growth of the human breast cancer cell lines MDA-MB-231, MDA-468, BT-474, SK BR-3, MCF7 and MCF7-dox, while similar treatment doses were found to have no effect on the growth and viability of the non-tumorigenic human mammary epithelial cells MCF10A. Meleagrin also showed excellent ATP competitive c-Met inhibitory activity in Z-Lyte assay, which was further confirmed via molecular docking studies and Western blot analysis. In addition, meleagrin treatment caused a dose-dependent inhibition of HGF-induced cell migration, and invasion of breast cancer cell lines. Meleagrin treatment potently suppressed the invasive triple negative breast tumor cell growth in an orthotopic athymic nude mice model, promoting this unique natural product from hit to a lead rank. The indole alkaloid meleagrin is a novel lead c-Met inhibitory entity useful for the control of c-Met-dependent metastatic and invasive breast malignancies.


Nutrition and Cancer | 2017

Rutin as A Novel c-Met Inhibitory Lead for The Control of Triple Negative Breast Malignancies

Heba E. Elsayed; Hassan Y. Ebrahim; Mohamed M. Mohyeldin; Abu Bakar Siddique; Amel M. Kamal; Eman G. Haggag; Khalid A. El Sayed

ABSTRACT Triple negative breast cancer (TNBC) has high metastatic and mortality potential and lacks effective and selective therapeutic options. Aberrant dysregulation of the receptor tyrosine kinase c-Met promotes TNBC progression, motility and survival and therefore considered a valid therapeutic target. Among various identified anticancer agents, plant polyphenols (PPs) including flavonoids, have been shown to be safe and proven for their antitumor activity through modulating diverse macromolecular targets. This study reports the bioassay-guided identification of the common flavonol glycoside rutin as breast cancer cell proliferation, migration and invasion inhibitor. The cell free Z′-LYTE kinase assay, Western blot and in silico docking experiments uncovered, for the first time, c-Met kinase as a potential mechanistic target for rutin-mediated anticancer effects on TNBC cell lines. Likewise, the intraperitoneal injection of rutin at 30 mg/kg, 3X/week, significantly reduced the growth of the TNBC MDA-MB-231/GFP orthotopic xenograft in nude mouse model. These results clearly designate the functional dietary flavonoid rutin as a potential lead for the prevention and control of c-Met-dependent breast malignancies.

Collaboration


Dive into the Hassan Y. Ebrahim's collaboration.

Top Co-Authors

Avatar

Khalid A. El Sayed

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Mohamed M. Mohyeldin

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Heba E. Elsayed

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Mohamed R. Akl

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Abu Bakar Siddique

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmed I. Foudah

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Asmaa A. Sallam

University of Louisiana at Monroe

View shared research outputs
Top Co-Authors

Avatar

Mohammad M. Hailat

University of Louisiana at Monroe

View shared research outputs
Researchain Logo
Decentralizing Knowledge