Network


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

Hotspot


Dive into the research topics where Irini Gerges is active.

Publication


Featured researches published by Irini Gerges.


Biomaterials | 2015

Versatile fabrication of vascularizable scaffolds for large tissue engineering in bioreactor

Alessandro Tocchio; Margherita Tamplenizza; Federico Martello; Irini Gerges; Eleonora Rossi; Simona Argentiere; Simona Rodighiero; Weiwei Zhao; Paolo Milani; Cristina Lenardi

Despite significant progresses were achieved in tissue engineering over the last 20 years, a number of unsolved problems still remain. One of the most relevant issues is the lack of a proper vascularization that is limiting the size of the engineered tissues to smaller than clinically relevant dimensions. Sacrificial molding holds great promise to engineered construct with perfusable vascular architectures, but there is still the need to develop more versatile approaches able to be independent of the nature and dimensions of the construct. In this work we developed a versatile sacrificial molding technique for fabricating bulk, cell-laden and porous scaffolds with embedded vascular fluidic networks. These branched fluidic architectures are created by highly resistant thermoplastic sacrificial templates, made of poly(vinyl alcohol), representing a remarkable progress in manufacturability and scalability. The obtained architecture, when perfused in bioreactor, has shown to prevent the formation of a necrotic core in thick cell-laden constructs and enabled the rapid fabrication of hierarchically branched endothelium. In conclusion we demonstrate a novel strategy towards the engineering of vascularized thick tissues through the integration of the PVA-based microfabrication sacrificial approach and perfusion bioreactors. This approach may be able to scale current engineered tissues to clinically relevant dimensions, opening the way to their widespread clinical applications.


Acta Biomaterialia | 2014

Three-dimensional hypoxic culture of human mesenchymal stem cells encapsulated in a photocurable, biodegradable polymer hydrogel: a potential injectable cellular product for nucleus pulposus regeneration

Deepak Kumar; Irini Gerges; Margherita Tamplenizza; Cristina Lenardi; Nicholas R. Forsyth; Yang Liu

Nucleus pulposus (NP) tissue damage can induce detrimental mechanical stresses and strains on the intervertebral disc, leading to disc degeneration. This study demonstrates the potential of a novel, photo-curable, injectable, synthetic polymer hydrogel (pHEMA-co-APMA grafted with polyamidoamine (PAA)) to encapsulate and differentiate human mesenchymal stem cells (hMSC) towards a NP phenotype under hypoxic conditions which could be used to restore NP tissue function and mechanical properties. Encapsulated hMSC cultured in media (hMSC and chondrogenic) displayed good cell viability up to day 14. The genotoxicity effects of ultraviolet (UV) on hMSC activity confirmed the acceptability of 2.5min of UV light exposure to cells. Cytotoxicity investigations revealed that hMSC cultured in media containing p(HEMA-co-APMA) grafted with PAA degradation product (10% and 20%v/v concentration) for 14days significantly decreased the initial hMSC adhesion ability and proliferation rate from 24hrs to day 14. Successful differentiation of encapsulated hMSC within hydrogels towards chondrogenesis was observed with elevated expression levels of aggrecan and collagen II when cultured in chondrogenic media under hypoxic conditions, in comparison with culture in hMSC media for 14days. Characterization of the mechanical properties revealed a significant decrease in stiffness and modulus values of cellular hydrogels in comparison with acellular hydrogels at both day 7 and day 14. These results demonstrate the potential use of an in vivo photo-curable injectable, synthetic hydrogel with encapsulated hMSC for application in the repair and regeneration of NP tissue.


Acta Biomaterialia | 2014

Poly(amido-amine)-based hydrogels with tailored mechanical properties and degradation rates for tissue engineering

Federico Martello; Alessandro Tocchio; Margherita Tamplenizza; Irini Gerges; Valentina Pistis; Rossella Recenti; Monica Bortolin; Massimo Del Fabbro; Simona Argentiere; Paolo Milani; Cristina Lenardi

Poly(amido-amine) (PAA) hydrogels containing the 2,2-bisacrylamidoacetic acid-4-amminobutyl guanidine monomeric unit have a known ability to enhance cellular adhesion by interacting with the arginin-glycin-aspartic acid (RGD)-binding αVβ3 integrin, expressed by a wide number of cell types. Scientific interest in this class of materials has traditionally been hampered by their poor mechanical properties and restricted range of degradation rate. Here we present the design of novel biocompatible, RGD-mimic PAA-based hydrogels with wide and tunable degradation rates as well as improved mechanical and biological properties for biomedical applications. This is achieved by radical polymerization of acrylamide-terminated PAA oligomers in both the presence and absence of 2-hydroxyethylmethacrylate. The degradation rate is found to be precisely tunable by adjusting the PAA oligomer molecular weight and acrylic co-monomer concentration in the starting reaction mixture. Cell adhesion and proliferation tests on Madin-Darby canine kidney epithelial cells show that PAA-based hydrogels have the capacity to promote cell adhesion up to 200% compared to the control. Mechanical tests show higher compressive strength of acrylic chain containing hydrogels compared to traditional PAA hydrogels.


Acta Biomaterialia | 2015

RGD-mimetic poly(amidoamine) hydrogel for the fabrication of complex cell-laden micro constructs

Alessandro Tocchio; Federico Martello; Margherita Tamplenizza; Eleonora Rossi; Irini Gerges; Paolo Milani; Cristina Lenardi

The potential of the 3D cell culture approach for creating in vitro models for drug screening and cellular studies, has led to the development of hydrogels that are able to mimic the in vivo 3D cellular milieu. To this aim, synthetic polymer-based hydrogels, with which it is possible to fine-tune the chemical and biophysical properties of the cell microenvironment, are becoming more and more acclaimed. Of all synthetic materials, poly(amidoamine)s (PAAs) hydrogels are known to have promising properties. In particular, PAAs hydrogels containing the 2,2-bisacrylamidoacetic acid-agmatine monomeric unit are capable of enhancing cellular adhesion by interacting with the RGD-binding αVβ3 integrin. The synthesis of a new photocrosslinkable, biomimetic PAA-Jeffamine®-PAA triblock copolymer (PJP) hydrogel is reported in this paper with the aim of improving the optical, biocompatibility and cell-adhesion properties of previously studied PAA hydrogels and providing an inexpensive alternative to the RGD peptide based hydrogels. The physicochemical properties of PJP hydrogels are extensively discussed and the behavior of 2D and 3D cell cultures was analyzed in depth with different cell types. Moreover, cell-laden PJP hydrogels were patterned with perfusable microchannels and seeded with endothelial cells, in order to investigate the possibility of using PJP hydrogels for fabricating cell laden tissue-like micro constructs and microfluidic devices. Overall the data obtained suggest that PJP could ultimately become a useful tool for fabricating improved in vitro models in order to potentially enhance the effectiveness of drug screening and clinical treatments.


Langmuir | 2009

Effect of pH on Water Proton NMR Relaxation in Agmatine-Containing Poly(amidoamine) Hydrogels

Lucia Calucci; Claudia Forte; Irini Gerges; Elisabetta Ranucci

Transverse and longitudinal relaxation times of water protons were measured and magnetization transfer experiments were performed on cross-linked amphoteric poly(amidoamine)s containing different proportions of agmatine side groups swollen in buffer media with pH values in the 1.4-8.5 range in order to obtain information on water/polymer interactions as a function of pH and polymer composition. The experimental results indicated that water interacts with polymer chains mainly via hydrogen bonding to carboxylate groups and via proton exchange with amino groups. The introduction of agmatine in the polymer disfavors water/polymer interactions possibly because of polymer structural changes and competing electrostatic interactions with carboxylate groups.


Macromolecular Bioscience | 2016

A Tailor-Made Synthetic Polymer for Cell Encapsulation: Design Rationale, Synthesis, Chemical-Physics and Biological Characterizations.

Irini Gerges; Margherita Tamplenizza; Eleonora Rossi; Alessandro Tocchio; Federico Martello; Camilla Recordati; Deepak Kumar; Nicholas R. Forsyth; Yang Liu; Cristina Lenardi

This study presents a custom-made in situ gelling polymeric precursor for cell encapsulation. Composed of poly((2-hydroxyethyl)methacrylate-co-(3-aminopropyl)methacrylamide) (P(HEMA-co-APM) mother backbone and RGD-mimicking poly(amidoamine) (PAA) moiteis, the comb-like structured polymeric precursor is tailored to gather the advantages of the two families of synthetic polymers, i.e., the good mechanical integrity of PHEMA-based polymers and the biocompatibility and biodegradability of PAAs. The role of P(HEMA-co-APM) in the regulation of the chemico-physical properties of P(HEMA-co-APM)/PAA hydrogels is thoroughly investigated. On the basis of obtained results, namely the capability of maintaining vital NIH3T3 cell line in vitro for 2 d in a 3D cell culture, the in vivo biocompatibility in murine model for 16 d, and the ability of finely tuning mechanical properties and degradation kinetics, it can be assessed that P(HEMA-co-APM)/PAAs offer a cost-effective valid alternative to the so far studied natural polymer-based systems for cell encapsulation.


International Journal of Polymeric Materials | 2016

Creep-resistant dextran-based polyurethane foam as a candidate scaffold for bone tissue engineering: Synthesis, chemico-physical characterization, and in vitro and in vivo biocompatibility

Irini Gerges; M. Tamplenizza; S. Lopa; C. Recordati; F. Martello; A. Tocchio; Leonardo Ricotti; Chiara Arrigoni; P. Milani; Matteo Moretti; Cristina Lenardi

ABSTRACT A highly crosslinked composite dextran-based scaffold (named DexFoam) was tailored to overcome specific deficiencies of polymeric and ceramic bone scaffolds and to guarantee a bone-mimicking microenvironment for the proliferation of human mesenchymal stem cells in vitro. The creep resistance for up to 90% compressive stain, the capability to regain the original shape after deformation, and the good thermal stability in both physiological and “body limit” conditions make DexFoam a valid alternative to the currently available bone scaffolds. Histopathological evaluation for host reaction and tissue colonization of DexFoam scaffold, implanted subcutaneously in mice, demonstrated its in vivo biocompatibility and biodegradability. GRAPHICAL ABSTRACT


Cell Transplantation | 2016

Stem Cell Delivery with Polymer Hydrogel for Treatment of Intervertebral Disc Degeneration: From 3D Culture to Design of the Delivery Device for Minimally Invasive Therapy.

Deepak Kumar; Alexander M. Lyness; Irini Gerges; Cristina Lenardi; Nicholas R. Forsyth; Yang Liu

Nucleus pulposus (NP) tissue damage can induce detrimental mechanical strain on the biomechanical performance of intervertebral discs (IVDs), causing subsequent disc degeneration. A novel, photocurable, injectable, synthetic polymer hydrogel (pHEMA-co-APMA grafted with PAA) has already demonstrated success in encapsulating and differentiating human mesenchymal stem cells (hMSCs) toward an NP phenotype during hypoxic conditions. After demonstration of promising results in our previous work, in this study we have further investigated the inclusion of mechanical stimulation and its impact on hMSC differentiation toward an NP phenotype through the characterization of matrix markers such as SOX-9, aggrecan, and collagen II. Furthermore, investigations were undertaken in order to approximate delivery parameters for an injection delivery device, which could be used to transport hMSCs suspended in hydrogel into the IVD. hMSC-laden hydrogel solutions were injected through various needle gauge sizes in order to determine its impact on postinjection cell viability and IVD tissue penetration. Interpretation of these data informed the design of a potential minimally invasive injection device, which could successfully inject hMSCs encapsulated in a UV-curable polymer into NP, prior to photo-cross-linking in situ.


Journal of The Mechanical Behavior of Biomedical Materials | 2017

3D porous polyurethanes featured by different mechanical properties: Characterization and interaction with skeletal muscle cells

Lorenzo Vannozzi; Leonardo Ricotti; Tommaso Santaniello; Tercio Terencio; Reinier Oropesa-Nuñez; Claudio Canale; Francesca Borghi; Arianna Menciassi; Cristina Lenardi; Irini Gerges

The fabrication of biomaterials for interaction with muscle cells has attracted significant interest in the last decades. However, 3D porous scaffolds featured by a relatively low stiffness (almost matching the natural muscle one) and highly stable in response to cyclic loadings are not available at present, in this context. This work describes 3D polyurethane-based porous scaffolds featured by different mechanical properties. Biomaterial stiffness was finely tuned by varying the cross-linking degree of the starting foam. Compression tests revealed, for the softest material formulation, stiffness values close to the ones possessed by natural skeletal muscles. The materials were also characterized in terms of local nanoindenting, rheometric properties and long-term stability through cyclic compressions, in a strain range reflecting the contraction extent of natural muscles. Preliminary in vitro tests revealed a preferential adhesion of C2C12 skeletal muscle cells over the softer, rougher and more porous structures. All the material formulations showed low cytotoxicity.


Molecular Imaging | 2015

In vivo imaging study of angiogenesis in a channelized porous scaffold.

Margherita Tamplenizza; Alessandro Tocchio; Irini Gerges; Federico Martello; Cristina Martelli; Luisa Ottobrini; Giovanni Lucignani; Paolo Milani; Cristina Lenardi

The main scientific issue hindering the development of tissue engineering technologies is the lack of proper vascularization. Among the various approaches developed for boosting vascularization, scaffold design has attracted increasing interest over the last few years. The aim of this article is to illustrate a scaffold design strategy for enhancing vascularization based on sacrificial microfabrication of embedded microchannels. This approach was combined with an innovative poly(ether urethane urea) (PEUtU) porous scaffold to provide an alternative graft substitute material for the treatment of tissue defects. Fluorescent and chemiluminescent imaging combined with computed tomography were used to study the behavior of the scaffold composition within living subjects by analyzing angiogenesis and inflammation processes and observing the variation in x-ray absorption, respectively. For this purpose, an IntegriSense 680 probe was used in vivo for the localization and quantification of integrin αvβ3, due to its critical involvement in angiogenesis, and a XenoLight RediJect Inflammation Probe for the study of the decline in inflammation progression during healing. Overall, the collected data suggest the advantages of embedding a synthetic vascular network into a PEUtU porous matrix to enhance in vivo tissue integration, maturation, and regeneration. Moreover, our imaging approach proved to be an efficient and versatile tool for scaffold in vivo testing.The main scientific issue hindering the development of tissue engineering technologies is the lack of proper vascularization. Among the various approaches developed for boosting vascularization, scaffold design has attracted increasing interest over the last few years. The aim of this article is to illustrate a scaffold design strategy for enhancing vascularization based on sacrificial microfabrication of embedded microchannels. This approach was combined with an innovative poly(ether urethane urea) (PEUtU) porous scaffold to provide an alternative graft substitute material for the treatment of tissue defects. Fluorescent and chemiluminescent imaging combined with computed tomography were used to study the behavior of the scaffold composition within living subjects by analyzing angiogenesis and inflammation processes and observing the variation in x-ray absorption, respectively. For this purpose, an IntegriSense 680 probe was used in vivo for the localization and quantification of integrin αvβ3, due to its critical involvement in angiogenesis, and a XenoLight RediJect Inflammation Probe for the study of the decline in inflammation progression during healing. Overall, the collected data suggest the advantages of embedding a synthetic vascular network into a PEUtU porous matrix to enhance in vivo tissue integration, maturation, and regeneration. Moreover, our imaging approach proved to be an efficient and versatile tool for scaffold in vivo testing.

Collaboration


Dive into the Irini Gerges's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eleonora Rossi

University Hospital of Basel

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leonardo Ricotti

Sant'Anna School of Advanced Studies

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge