Network


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

Hotspot


Dive into the research topics where Fadee Mondalek is active.

Publication


Featured researches published by Fadee Mondalek.


Journal of Nanobiotechnology | 2006

The permeability of SPION over an artificial three-layer membrane is enhanced by external magnetic field

Fadee Mondalek; Yuan Yuan Zhang; Bradley P. Kropp; Richard D. Kopke; Xianxi Ge; Ronald L. Jackson; Kenneth J. Dormer

BackgroundSensorineural hearing loss, a subset of all clinical hearing loss, may be correctable through the use of gene therapy. We are testing a delivery system of therapeutics through a 3 cell-layer round window membrane model (RWM model) that may provide an entry of drugs or genes to the inner ear. We designed an in vitro RWM model similar to the RWM (will be referred to throughout the paper as RWM model) to determine the feasibility of using superparamagnetic iron oxide (Fe3O4) nanoparticles (SPION) for targeted delivery of therapeutics to the inner ear.The RWM model is a 3 cell-layer model with epithelial cells cultured on both sides of a small intestinal submucosal (SIS) matrix and fibroblasts seeded in between. Dextran encapsulated nanoparticle clusters 130 nm in diameter were pulled through the RWM model using permanent magnets with flux density 0.410 Tesla at the pole face. The SIS membranes were harvested at day 7 and then fixed in 4% paraformaldehyde. Transmission electron microscopy and fluorescence spectrophotometry were used to verify transepithelial transport of the SPION across the cell-culture model. Histological sections were examined for evidence of SPION toxicity, as well to generate a timeline of the position of the SPION at different times. SPION also were added to cells in culture to assess in vitro toxicity.ResultsTransepithelial electrical resistance measurements confirmed epithelial confluence, as SPION crossed a membrane consisting of three co-cultured layers of cells, under the influence of a magnetic field. Micrographs showed SPION distributed throughout the membrane model, in between cell layers, and sometimes on the surface of cells. TEM verified that the SPION were pulled through the membrane into the culture well below. Fluorescence spectrophotometry quantified the number of SPION that went through the SIS membrane. SPION showed no toxicity to cells in culture.ConclusionA three-cell layer model of the human round window membrane has been constructed. SPION have been magnetically transported through this model, allowing quantitative evaluation of prospective targeted drug or gene delivery through the RWM. Putative in vivo carrier superparamagnetic nanoparticles may be evaluated using this model.


Audiology and Neuro-otology | 2006

Magnetic nanoparticles: inner ear targeted molecule delivery and middle ear implant.

Richard D. Kopke; Ronald A. Wassel; Fadee Mondalek; Brian P. Grady; Kejian Chen; Jianzhong Liu; Donald D. Gibson; Kenneth J. Dormer

Superparamagnetic iron oxide nanoparticles (SNP) composed of magnetite (Fe3O4) were studied preliminarily as vehicles for therapeutic molecule delivery to the inner ear and as a middle ear implant capable of producing biomechanically relevant forces for auditory function. Magnetite SNP were synthesized, then encapsulated in either silica or poly (D,L,-Lactide-co-glycolide) or obtained commercially with coatings of oleic acid or dextran. Permanent magnetic fields generated forces sufficient to pull them across tissue in several round window membrane models (in vitrocell culture, in vivo rat and guinea pig, and human temporal bone) or to embed them in middle ear epithelia. Biocompatibility was investigated by light and electron microscopy, cell culture kinetics, and hair cell survival in organotypic cell culture and no measurable toxicity was found. A sinusoidal magnetic field applied to guinea pigs with SNP implanted in the middle ear resulted in displacements of the middle ear comparable to 90 dB SPL.


Biomagnetic Research and Technology | 2007

Magnetic characterization of superparamagnetic nanoparticles pulled through model membranes

Allison L Barnes; Ronald A. Wassel; Fadee Mondalek; Kejian Chen; Kenneth J. Dormer; Richard D. Kopke

BackgroundTo quantitatively compare in-vitro and in vivo membrane transport studies of targeted delivery, one needs characterization of the magnetically-induced mobility of superparamagnetic iron oxide nanoparticles (SPION). Flux densities, gradients, and nanoparticle properties were measured in order to quantify the magnetic force on the SPION in both an artificial cochlear round window membrane (RWM) model and the guinea pig RWM.MethodsThree-dimensional maps were created for flux density and magnetic gradient produced by a 24-well casing of 4.1 kilo-Gauss neodymium-iron-boron (NdFeB) disc magnets. The casing was used to pull SPION through a three-layer cell culture RWM model. Similar maps were created for a 4 inch (10.16 cm) cube 48 MGOe NdFeB magnet used to pull polymeric-nanoparticles through the RWM of anesthetized guinea pigs. Other parameters needed to compute magnetic force were nanoparticle and polymer properties, including average radius, density, magnetic susceptibility, and volume fraction of magnetite.ResultsA minimum force of 5.04 × 10-16 N was determined to adequately pull nanoparticles through the in-vitro model. For the guinea pig RWM, the magnetic force on the polymeric nanoparticles was 9.69 × 10-20 N. Electron microscopy confirmed the movement of the particles through both RWM models.ConclusionAs prospective carriers of therapeutic substances, polymers containing superparamagnetic iron oxide nanoparticles were succesfully pulled through the live RWM. The force required to achieve in vivo transport was significantly lower than that required to pull nanoparticles through the in-vitro RWM model. Indeed very little force was required to accomplish measurable delivery of polymeric-SPION composite nanoparticles across the RWM, suggesting that therapeutic delivery to the inner ear by SPION is feasible.


Journal of Biomedical Materials Research Part A | 2010

Enhanced angiogenesis of modified porcine small intestinal submucosa with hyaluronic acid-poly(lactide-co-glycolide) nanoparticles: from fabrication to preclinical validation.

Fadee Mondalek; Richard A. Ashley; Christopher C. Roth; Yusuf Kibar; Nabeel Shakir; Michael A. Ihnat; Kar Ming Fung; Brian P. Grady; Bradley P. Kropp; Hsueh Kung Lin

Hyaluronic acid-poly(de-co-glycolide) nanoparticles (HA-PLGA NPs) were synthesized to stabilize the porous structure of porcine small intestinal submucosa (SIS), to improve surface biocompatibility and to enhance performance in tissue regeneration. HA-PLGA NPs were characterized for size, zeta potential, surface morphology, and HA loading. Human microvascular endothelial cells responded to HA-PLGA NPs and HA-PLGA modified SIS (HA-PLGA-SIS) with elevated cell proliferation. HA-PLGA-SIS significantly enhanced neo-vascularization in an in ovo chorioallantoic membrane angiogenesis model. The angiogenic capability of the newly fabricated HA-PLGA-SIS was tested in a canine bladder augmentation model. Urinary bladder augmentation was performed in beagle dogs following hemi-cystectomy using HA-PLGA-SIS. The regenerated bladder was harvested at 10 weeks post augmentation and vascularization was evaluated using CD31 immunohistochemical staining. Bladder regenerated with HA-PLGA-SIS had significantly higher vascular ingrowth compared to unmodified SIS. This study shows that HA-PLGA NPs may represent a new approach for modifying naturally derived SIS biomaterials in regenerative medicine.


Journal of Nanobiotechnology | 2010

Inhibition of angiogenesis- and inflammation-inducing factors in human colon cancer cells in vitro and in ovo by free and nanoparticle-encapsulated redox dye, DCPIP

Fadee Mondalek; Sivapriya Ponnurangam; Janita Govind; Courtney W. Houchen; Shrikant Anant; Panayotis Pantazis; Rama P. Ramanujam

BackgroundThe redox dye, DCPIP, has recently shown to exhibit anti-melanoma activity in vitro and in vivo. On the other hand, there is increasing evidence that synthetic nanoparticles can serve as highly efficient carriers of drugs and vaccines for treatment of various diseases. These nanoparticles have shown to serve as potent tools that can increase the bioavailability of the drug/vaccine by facilitating absorption or conferring sustained and improved release. Here, we describe results on the effects of free- and nanoparticle-enclosed DCPIP as anti-angiogenesis and anti-inflammation agents in a human colon cancer HCT116 cell line in vitro, and in induced angiogenesis in ovo.ResultsThe studies described in this report indicate that (a) DCPIP inhibits proliferation of HCT116 cells in vitro; (b) DCPIP can selectively downregulate expression of the pro-angiogenesis growth factor, VEGF; (c) DCPIP inhibits activation of the transcriptional nuclear factor, NF-κB; (d) DCPIP can attenuate or completely inhibit VEGF-induced angiogenesis in the chick chorioallantoic membrane; (e) DCPIP at concentrations higher than 6 μg/ml induces apoptosis in HCT116 cells as confirmed by detection of caspase-3 and PARP degradation; and (f) DCPIP encapsulated in nanoparticles is equally or more effective than free DCPIP in exhibiting the aforementioned properties (a-e) in addition to reducing the expression of COX-2, and pro-inflammatory proteins IL-6 and IL-8.ConclusionsWe propose that, DCPIP may serve as a potent tool to prevent or disrupt the processes of cell proliferation, tissue angiogenesis and inflammation by directly or indirectly targeting expression of specific cellular factors. We also propose that the activities of DCPIP may be long-lasting and/or enhanced if it is delivered enclosed in specific nanoparticles.


World Journal of Urology | 2015

Temporal expression of hyaluronic acid and hyaluronic acid receptors in a porcine small intestinal submucosa-augmented rat bladder regeneration model.

Fadee Mondalek; Kar Ming Fung; Qing Yang; Weijuan Wu; Wenli Lu; Blake W. Palmer; Dominic Frimberger; Beverley Greenwood-Van Meerveld; Robert E. Hurst; Bradley P. Kropp

IntroductionHyaluronic acid (HA), a non-sulfated glycosaminoglycan, is an essential component of the extracellular matrix (ECM). Since HA is involved in many phases of wound healing and may play a key role in tissue repair and regeneration, this study was intended to understand temporal and spatial expression of HA and HA receptors (HARs) during the course of bladder regeneration in rats.Materials and methodsSprague–Dawley rats were subjected to partial cystectomy followed by augmentation with porcine small intestinal submucosal (SIS) prepared from distal sections of the small intestine. SIS-augmented bladders were harvested between postoperative days 2 and 56.ResultsBladder regeneration proceeded without complications. All augmented bladders had complete urothelial lining and smooth muscle bundles by day 56 post-augmentation. Temporal and spatial distributions of HA and HARs were studied by immunohistochemistry in regenerating bladders. The strongest HA immunoreactivity was observed in the ECM on postoperative days 28 and 56. Cluster of differentiation 44 (CD44) immunoreactivity was detected in the cytoplasm of urothelial cells on day 56; and LYVE-1 immunoreactivity was exclusively limited to lymphatic vessels on days 28 and 56.ConclusionsWe demonstrated that HA was synthesized throughout the course of bladder wound healing and regeneration; and HA deposition coincided with urothelial differentiation. Expression of CD44 and LYVE-1 followed the same temporal pattern as HA deposition. Therapeutic modalities through local delivery of exogenous HA to improve the outcome of SIS-mediated bladder regeneration might need to be coordinated with HAR expression in order to achieve maximal regenerative responses as opposed to fibrosis.


Gastroenterology | 2011

Nanoparticle-Based Delivery of DCAMKL-1 SiRNA and DAPT Increases MicroRNA-144 and Inhibits Colorectal Cancer Tumor Growth via a Notch-1 Dependent Mechanism

Sripathi M. Sureban; Randal May; Dongfeng Qu; Fadee Mondalek; Sima Asfa; Rama P. Ramanujam; Shrikant Anant; Courtney W. Houchen

G A A b st ra ct s downregulated in gastric cancer tissues relative to non-tumor gastric mucosae. Downregulation of miR-29c was associated with progression of gastric cancer, and was more prominent in advanced gastric cancers than in gastric adenomas and early gastric cancers (p<0.05). In addition, expression of the oncogene Mcl-1, a target of miR-29c, was significantly increased in gastric cancer tissues relative to non-tumor gastric mucosae. Activation of miR-29c by celecoxib induced suppression of Mcl-1 and apoptosis in gastric cancer cells. Celecoxib activated the expression of miR-29c, but not that of miR-29b, suggesting that celecoxib may modulate the binding of transcriptional factors between miR-29b and miR-29c. The database (www.gene-regulation.com/pub/programs.html), indicated that C/EBPα binding sites were located in the promoter region of miR-29c. The chromatin immunoprecipitation (ChIP) assay showed that immunoprecipitation with the C/EBPα antibody was significantly increased in AGS cells treated with celecoxib, indicating that celecoxib activates the expression of miR29c by enhancing the binding of C/EBPα to the promoter. Conclusions : Downregulation of the tumor suppressor miR-29c plays critical roles in the progression of gastric cancer, and that miR-29c is a novel therapeutic target for gastric cancer. Selective COX-2 inhibitors may have clinical promise for the chemoprevention of gastric cancer via activation ofmiR-29c.


The Journal of Urology | 2009

BLADDER REGENERATION IN A CANINE MODEL USING HYALURONIC ACID/POLY(LACTIC-CO-GLYCOLIC) ACID NANOPARTICLE-TREATED PORCINE SMALL INTESTINAL SUBMUCOSA

Christopher C. Roth; Yusuf Kibar; Fadee Mondalek; Richard A. Ashley; Hsueh-Kung Lin; Dominic Frimberger; Bradley P. Kropp

Christopher C. Roth*, Fadee G. Mondalek*, Yusuf Kibar † , Richard A. Ashley*, Cardin H. Bell*, John A. Califano*, Sundar V. Madihally , Dominic Frimberger*, Hsueh-Kung Lin* and Bradley P. Kropp* *Department of Urology, University of Oklahoma Health Sciences Center, Oklahoma City, ‡ School of Chemical Engineering, Oklahoma State University, Stillwater, OK, USA, and † Department of Urology, Gulhane Military Medical Academy, Ankara, Turkey Accepted for publication 7 May 2010


Journal of Nanobiotechnology | 2011

Nanoparticle-based delivery of siDCAMKL-1 increases microRNA-144 and inhibits colorectal cancer tumor growth via a Notch-1 dependent mechanism

Sripathi M. Sureban; Randal May; Fadee Mondalek; Dongfeng Qu; Sivapriya Ponnurangam; Panayotis Pantazis; Shrikant Anant; Rama P. Ramanujam; Courtney W. Houchen


Biomaterials | 2008

The incorporation of poly(lactic-co-glycolic) acid nanoparticles into porcine small intestinal submucosa biomaterials.

Fadee Mondalek; Benjamin J. Lawrence; Bradley P. Kropp; Brian P. Grady; Kar Ming Fung; Sundar V. Madihally; Hsueh Kung Lin

Collaboration


Dive into the Fadee Mondalek's collaboration.

Top Co-Authors

Avatar

Rafal Farjo

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Alexander B. Quiambao

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Bradley P. Kropp

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Courtney W. Houchen

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ronald A. Wassel

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kar Ming Fung

University of Oklahoma Health Sciences Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge