Poster Presentation
Biography
Tania Limongi is research scientist in the Physical Science and Engineering Department of the King Abdullah University of Science and Technology (KAUST) in Saudi Arabia. She received in 1999 her Master Degree in Biological Sciences at the University of L’Aquila and her Ph.D. in Microsystem Engineering at the University of Rome Tor Vergata in 2004. She published more than 40 papers in reputed journals. Her work is mainly addressed on the development of new tissue engineering approaches and on the optimization of single molecule localization protocols and techniques.
Abstract
Tissue engineering is a fast growing contest of multiple disciplines that aims to realize tissues and organs for the replacement through congenital defects, cancer, aging and trauma. As an interdisciplinary field, principles of materials science, chemistry, biomechanics, engineering, biology, and medicine are vastly employed in regenerative medicine research. The use of biocompatible polymers for the design and the fast realization of low-cost microfabricated scaffolds has the potential to solve the problem of the urgency of organs donation for patients requiring organ transplantation. The primary goal of biocompatible scaffolds use is to furnish appropriate support for maintain, improve or restore function of damaged cells, tissue and organs. For the preparation of scaffolds, regardless of the tissue type, a great number of natural and synthetic materials were tested and used due to their low toxicity, bioresorbability, and low costs. To provide a satisfying cellular proliferation, optimal diffusion of growing factors, nutrients, gases and an efficient disposal of wasted products, scaffolds should be designed by means of an interconnected porous structure by optimizing mechanical function and mass-transport requirements. In details, micropores provide the optimal background for cells proliferation and, by promoting the physiological interactions between the cells and the extracellular matrix, they result in essential tools for successful tissue regeneration. Microporous structures in 3D biocompatible-engineered devices have been realized by means of printing, stereolithography and thermally induced phase separation. Freeze-lyophilizing method and also salt addition and leaching emerged as useful techniques for biocompatible porous scaffolds fabrication. This work describes the realization of poly (epsilon-caprolactone) porous structures by using salt particles as porogen; the porosity of the material has been created by salt leaching by immersing it in distilled water. The results show that the proposed method is suitable for the manufacture of porous structures and depending on the fabrication process different degrees of porosity can be obtained by varying salt sizes and concentration.
Biography
Eman Alfayez is currently a full time 3rd Year PhD student, Dental Institute King’s College London. She is funded by King Abdulaziz University and the Saudi Ministry of Higher Education. She has worked as a resident Dentist in Prince Sultan and King Fahad Armed Forces Hospitals for two years. In addition, holds a demonstrator position in the Oral Biology Division, Oral Basic and Clinical Sciences Department, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia.
Abstract
Vascularization is critical for tissue engineered constructs, in order to provide nutrients, oxygen and establish an initial integration with the host tissue. Studying vascularization in vitro remains challenging. Defined culturing conditions that mimic the physiological niche are essential for cell differentiation and survival in 3D scaffolds in vitro. The niche comprises both biological and mechanical cues. It has been documented in the literature that mechanical load exerted by bioreactors remarkably enhances culture conditions and cellular metabolism. Since a dynamic environment permits continuous nutrients transport, it prepares scaffolds prior to in vivo implantation. Hence, bioreactor technology is a promising strategy for culturing cells in a dynamic environment. In this study, a bioreactor was used to explore the dynamic effect on endothelial and osteogenic genes expression of co-cultured alveolar human osteoblasts (aHOBs) and human umbilical vein endothelial cells (HUVECs) in 3D scaffolds. Cells were seeded in Smart bone® blocks which are composed of bovine bone matrix, biodegradable polymer and cell nutrients with excellent mechanical properties. Scaffolds were cultured in endothelial conditioned media and subjected to pulsating compressive force of 35 ± 10 N at a frequency of 0.1Hz for approximately 72 hours. The molecular qPCR data showed that mechanical stimulation enhanced gene expressions of vascular and early bone markers when compared to seeded scaffolds incubated under static conditions. The findings from this study demonstrated the importance of tailoring the in vitro environment for seeded scaffolds in order to better understand the physiological conditions and factors that influence blood vessel formation in implanted scaffolds.
Biography
Alina Gabriela Rusu is a Ph.D. student in Chemistry from Technical University “Gheorghe Asachi†of Iasi, Faculty of Chemical Engineering and Environmental Protection and member in the Center for Training and Research in Tissue Engineering and Regenerative Medicine, from University of Medicine and Pharmacy “Grigore T. Popa†of Iasi. In December 2014, she obtained a doctoral scholarship in the POSDRU Programme 159/1.5/S/133652 at “Alexandru Ioan Cuza†University of Iasi. She completed her MSc degree in Prosthetics Bioengineering in 2012. Her research studies are focused on developing and characterization of novel scaffolds based on functionalized chitosan with potential applications in medicine.
Abstract
Introduction. Cell encapsulation in a hydrogel that plays the role of extracellular matrix (ECM) requires a cytocompatible hydrogelation process, without the use of harsh chemical agents. Collagen, the main protein of the ECM, represents an ideal micromedium in which cells can be encapsulated given the natural RGD sequence that mediates cell adhesion and the ability to form hydrogels in physiological conditions. In our study, we have tested the cytocompatibility of a collagen based hydrogel with mechanical properties improved by crosslinking with functionalized polysaccharides. Methods. The biocompatibility of the hydrogels based on collagen crosslinked with partial oxidized gellan was tested by determining the viability of the cells using MTT method. Adipose stem cell proliferation within the hydrogels was evaluated with Alamar Blue test. The morphology of encapsulated cells and hydrogel structure were observed by SEM, after critical point drying. The degradability of the hydrogels was tested in the presence of collagenase, by ninhydrin assay. Results. MTT tests after 24, 48 and 72 hours revealed a viability above 97% compared with control wells. Cells proliferated within the hydrogels as shown by an increase in metabolic activity. Hydrogels present a fibrillar structure with cells integrated and adhered to the fibrils. The presence of gellan as a crosslinker stabilized the hydrogel which degraded slower than the un-crosslinked collagen. Conclusions. We have obtained a cytocompatible collagen-based hydrogel by using functionalized polysaccharides as a structural component and also as a crosslinker. The hydrogels allow cell encapsulation without affecting the viability and cell proliferation and are more stable to enzymatic degradation.
Biography
Hanaa H. Ahmed has completed her Ph.D. at the age of 35 years from Faculty of Science, Cairo University. She is the Head of Hormones Department, Medical Research Division, National Research Centre, one of the biggest research center in Egypt. She has published more than 84 papers in reputed journals and 11 international books and serving as an editorial board member of 9 international journals. She has awarded the Prize of Excellence at the National Research Centre in the Field of Science and Technology, which serves the Advanced Medical Science, (2013).
Abstract
Background: Parkinson’s disease (PD) has been considered a paradigm of degenerative diseases of the nervous system characterized by motor impairment due to malfunction and loss of dopaminergic neurons. Currently there is no therapy clinically available that delays the neurodegenerative process, therefore modification of the disease course is an unmet clinical need. Mesenchymal stem cells (MSCs) have been considered as a promising therapeutic strategy for acute injury and degenerative diseases of the central nervous system. Objective: The current work aimed to elucidate the possible role of single intravenous dose of bone marrow derived MSCs (BM-MSCs) in restraining dopaminergic neurons in experimental model after 2 months. Materials and Methods: Thirty two ovariectomized animals were classified into 4 groups; Group (1) was control, Groups from (2) to (4) were subcutaneously administered with rotenone for 14 days after one month of ovariectomy for induction of PD. Group (2) was left untreated; Groups (3) and (4) were treated with sinemet and BM-MSCs respectively. Y-chromosome gene (sry) was assessed by PCR in brain tissue of the female rats. Serum TGF-β1 and MCP-1 levels were assayed by ELISA technique. Brain dopamine level was assayed fluorometrically while, brain TH and nestin genes expression were detected by sqRT-PCR. Results: The data of the current work revealed that BM-MSCs were able to home at the injured brains and produced significant decrease in serum TGF-β1 and MCP-1 levels associated with significant increase in brain dopamine content, brain TH and nestin genes expression levels. Conclusion: The observed improvements in the studied biomarkers after 2 months from intravenous transplantation of BM-MSCs sheds light on the promising role of BM-MSCs in ameliorating neurodeterioration of dopaminergic neurons through their anti-inflammatory and neurogenic effects. Key words: Dopaminergic neurons degeneration, Bone marrow mesenchymal stem cells, anti-inflammatory action, Neurogenic potential, Rotenone, Rats.
Biography
MeryemSümeyye AKDEMİR has completed her B.Sc. degrees from KaredenizTechnical University, Department of Chemistry. Currently she is M.Sc. student at the bioengineering and biochemistrydepartments. She is a member of Hacettepe University Cell and Tissue Engineering Research Group, being coordinated by Prof. Dr. MenemşeGümüşderelioğlu and Biochromatography and Biodiagnostics Research Group, being coordinated by Prof. Dr. AdilDenizli.
Abstract
Titanium (Ti) and its alloys have been widely used as implant materialsin dental and orthopedic fields owing to their superior mechanical properties and biocompatibility.However, Ti implants have a major problem related to their osseointegration success.One of the effective approaches to overcome this issue is the biomimetric coating of implant surfaces with hydroxyapatite (HA) due to its osteoconductive and osteoinductive properties. Furthermore, many studies have reported that biosignalssuch as melatonin and BMP-6 had positive effects on osseointegration. Thus,integration of growth factors with HA coated Ti based implants is a promising approach, which have not been investigated yet. The searchof ideal carrier systemsto control the rate and the time profile of biosignal releaseis also an important issue.Cyclodextrins (CD) have gained prominence because of their capability of binding biosignals due to their hydrophobic cavity. The aim of this study is to develop HA-CD composite carriers which supports the formation of CD-biosignal inclusion complexes to improve osseointegration in Ti based implant surfaces.For this purpose, polished Ti alloy surfaces were etched with NaOH and then coated with biomimetric HA by 10xSimulated Body Fluid (SBF) treatment. Following this, (2-Hydroxypropyl)-β-cyclodextrin (HPβCD) was impregnated on HA-coated Ti surfaces. Finally, melatonin and BMP-6 were used as biosignal moleculesto make inclusion complexes with cyclodextrin.Fabricated implant materials were characterizedbyTGA,ATR-FTIR, XRD,SEM analysis and water contact angle measurements. Melatonin and BMP-6 release studies were also performed.In vitro cell culture studies were carried out with pre-osteblastic MC3T3-E1 cell line to investigate the effect of HA-HPβCD -composite carriers on osseointegration.
Biography
Alina Gabriela Rusu is a Ph.D. student in Chemistry from Technical University “Gheorghe Asachi†of Iasi, Faculty of Chemical Engineering and Environmental Protection and member in the Center for Training and Research in Tissue Engineering and Regenerative Medicine, from University of Medicine and Pharmacy “Grigore T. Popa†of Iasi. In December 2014, she obtained a doctoral scholarship in the POSDRU Programme 159/1.5/S/133652 at “Alexandru Ioan Cuza†University of Iasi. She completed her MSc degree in Prosthetics Bioengineering in 2012. Her research studies are focused on developing and characterization of novel scaffolds based on functionalized chitosan with potential applications in medicine.
Abstract
Over the last decade, hydrogels have received a considerable attention due to their resemblance to the natural extracellular matrix, providing soft tissue-like environment for cell proliferation and survival, while allowing diffusion of nutrients and other water-soluble metabolites through the hydrogel network. Moreover, they have advantages over other types of polymeric scaffolds, such as biocompatibility, easy control over structural parameters, high water content, and tunable scaffold architecture. A variety of synthetic or natural polymeric hydrogels have been employed as scaffolds in tissue engineering applications, among them chitosan, a biopolymer also frequently used for pharmaceutical and biomedical applications. Moreover, chemical functionalization of the hydrogels with bioactive compounds has been widely used to control the interactions between cells and materials, emerging as an important strategy for developing new hydrogels. Based on these observations, L-arginine, an α-amino acid with remarkable biological properties in wound healing, was chosen to graft onto hydrogels based on N-maleoyl-chitosan and poly(acrylic acid) in order to modulate the physico-chemical properties and specific cellular responses. The morphological structure of these hydrogels was investigated by SEM images and its degradation performance was tested. The swelling behavior in simulated exudate fluid demonstrated an increased capacity of absorbing fluids, while the maximum swelling ratio was achieved in less than 1 minute. Also, the hydrogels cytocompatibility on fibroblast was assessed, revealing cytocompatible characteristics by comparison to control. These results indicated that the grafting of arginine could lead to new type of biologically active biomaterials with potential use as wound dressings.
Biography
Hadeer A. Aglan has completed her Ph.D. at the age of 32 years from Faculty of Science, Ain Shams University, Egypt. She is Researcher at Hormones Department, Medical Research Division, National Research Centre, one of the biggest research center in Egypt. She has published 4 papers in reputed journals and participated in 4 scientific projects.
Abstract
Background: Transplantation of mesenchymal stem cells (MSCs) for treating neurodegenerative disorders has received growing attention recently because these cells are readily available, easily expanded in culture, and when transplanted survive for relatively long periods of time. Objective: The current study was planned to explore the possible role of single intravenous dose of MSCs in mitigation of degenerated cholinergic neurons in experimental model after 2 months. Materials and Methods: MSCs were harvested from bone marrow of femoral bones (BM-MSCs) and excised from adipose tissues of both the omentum and the inguinal fat pad (ADMSCs) of male Sprague-Dawley rats, grown and propagated in culture, then characterized morphologically and by the detection of cell surface markers gene expression. Also, their tripotential mesodermal differentiation capability into osteoblasts, chondrocytes, and adipocytes was confirmed. Regarding the biological experimentation: forty female rats were classified into 5 groups; Group (1) was control, while the groups from (2) to (5) were orally administered with AlCl3 for induction of cholinergic neurons deterioration. Group (2) was left untreated; groups (3), (4) and (5) were treated with cerebrolysin, BM-MSCs and ADMSCs respectively. Y-chromosome gene (SRY) was assessed by PCR in brain tissue of the female rats. Serum TGF-β1, MCP-1 and BDNF levels were assayed by ELISA technique. Brain ChAT expression was determined by immunohistochemical procedure. While, brain nestin gene expression was detected by sqRT-PCR. Also, histopathological investigation of brain tissues was done. Results: The data of the current work implied that BM-MSCs and ADMSCs were able to home at the injured brains and produced insignificant decrease in serum TGF-β1 and MCP-1 levels associated with significant increase in serum BDNF and nestin gene expression levels. Also, they caused significant increase in the number of positive cells for brain ChAT expression and ameliorate the neurodegenerative alterations in the hippocampus. Conclusion: These findings shed light on the promising role of BM-MSCs and ADMSCs in alleviating the retrogression of cholinergic neurons through their anti-inflammatory effect, neurotrophic capacity and neurogenic potential.
Biography
Victor Lopez Davila is a PhD candidate at University College London (UK) in the field of Nanomedicie and Tissue Engineering. He completed his MSc in Nanomedicine in Cranfield University (UK) and his BSc in Biotechnology in Lleida University (Spain) in 2011. He also works part time as a biobank technician for Tissue Access for Patient’s Benefit (TAPB).
Abstract
Background: Colorectal cancer therapy shows very low response rates, leaving room for improvement. In this work we approach the problem from two points of view: loading the MEK inhibitor AZD6244 in organic nanoparticles to improve its delivery to colorectal cancer cells, and testing these nanoformulations in a collagen-based 3D in vitro model of cancer to mimic physiological conditions. Methods: AZD6244-containing GCPQ micelles and DOPE/DC-cholesterol liposomes were fabricated using sonication and extrusion methods, respectively. These formulations were tested, together with the free drug, in HCT116 cell monolayers and a collagen-based 3D cancer model, to determine differences in efficacy between both models. Fluorescent nanoparticles were used to determine their ability to penetrate the scaffold and reach its core. Results: Drug delivered in nanocarriers proved to inhibit proliferation in monolayers much more efficiently than on its own. Conversely, nanocarriers were significantly less effective in 3D compared to monolayers. Penetration studies revealed that this phenomenon was most likely due to the inability of the nanoparticles to overcome physical barriers in the 3D model. Surprisingly, the free drug was more effective in 3D than in 2D, while cells tend to be more sensitive to therapeutic agents in monolayers. Conclusions: These results highlight the need to include 3D in vitro testing during drug development prior to in vivo testing, particularly in the case of nanoformulations.
Biography
Emily is currently working as a student researcher at École Polytechnique Fédérale de Lausanne funded by the Whitaker International Program. In June 2014, she graduated from Drexel University, where she completed both her Bachelors and Masters degrees in Materials Science and Engineering though an accelerated BS/MS program. This fall, she will begin her Ph.D. studies at McGill University where she will work towards the development of graphene hydrogels for orthopedic applications.
Abstract
Since the discovery of growth factors in the 1950’s, safety risks and high cost have presented challenges in the translation of growth factor therapies into clinical applications. While it is known that growth factors under physiological conditions are very effective at low doses, the clinical administration of growth factors requires a much higher dose to promote tissue regeneration. However, these high doses, as the Food and Drug Administration has warned, are associated with risks of cancer in patients. In response to this clinical problem, we developed a platform for increasing the affinity of growth factors for the extracellular matrix (ECM) in order to create a sustained release at the injured site and prolonged receptor activation that mimics the physiological delivery of growth factors. We identified a domain from placental growth factor, (PlGF-2123-144), which has high affinity for many ECM proteins and fused this domain to various growth factors, in particular VEGF-A, PDGF-BB, and BMP-2. We were able to show that the engineered growth factors have a high affinity for ECM proteins and that their administration at low doses exhibits better wound healing in skin and bone defects. We are now expanding this engineering concept to SDF-1α, a potent chemokine involved in the recruitment of progenitor and endothelial cells to ischemic tissues, in the context of cardiac tissue repair after myocardial infarction.
Biography
Min Yao has completed her Ph.D. at the age of 37 years from Kyorin University, School of Medicine, Tokyo, Japan and postdoctoral studies from California University, Riverside. She is the director of institute of traumatic medicine, Shanghai Jiaotong Unversity School of Medicine. She has published more than 10 papers in reputed journals of repute
Abstract
In order to fully restore the structure and function of skin in the both donor and recipient site, a novel grafting with microscopic skin tissue columns (MSTCs) was studied. Methods A new technique of grafting that can harvest and transplant the full-thickness MSTCs with 200~1000um diameter was used. In vitro the viability of MSTCs were evaluated. Three different treatments were applied to the swine full-thickness wounds: open wounds, MSTCs transplantation, and STSG (split-thickness skin grafting) transplantation. After grafting, the wound healing rate and contraction rate were investigated. In addition, the morphological and histological evaluations were applied for comparing the outcomes of the donor sites. Furthermore, the MSTCs from human or black hair mouse were transferred to the nude mice for observing the reconstruction of skin appendages. The sweater glands, sebaceous glands, hair follicles in the recipient site were identified. Results The MSTCs contained epidermis and dermis can be harvested effectively with much less damage to the donor site. After culturing, the MSTCs presented high proliferation viability, especially the cells near by the germinal layer. Full structures of the sweat glands, the sebaceous glands, or the hair follicles were carried by MSTCs of 700um diameter and larger. The donor site was re-epithelialized within 3 days in vitro. The wound healing rate in the MSTCs auto-grafting group was lower than that in the STSG group at one week postoperative. After two weeks, wound healing rates in the three groups were the same. However, the contraction rates in the MSTCs or STSG groups were much lower than that in the open wound group. Compared to the STSG grafting and open wound without grafting, MSTCs grafting gained better outcome of wound healing. In addition, the wounds in the donor site after taking MSTCs healed much faster than that in the STSG donor site with much better morphology. Furthermore, human sweat glands, sebaceous glands or hair follicle in the recipient site of mice were identified after human MSTCs xenografting. The black hair grew very well on the recipient site of nude mouse (no hair) after MSTCs of black hair mouse allografting. Full-thickness microscopic skin tissue columns can be harvested effectively with high activity and less damage in the donor site. Grafting of MSTCs improves the outcomes of full-thickness skin defects efficiently with restoring of skin appendages. Our results demonstrate that the MSTCs grafting will become a very promising technique for skin regeneration in clinical practice.
Biography
Marwa Hamdi El-Khashab completed Bachelor of veterinary medicine, Cairo University at 2008 and worked Demonstrator at surgery, anesthesiology and radiology department at faculty of veterinary medicine, Cairo University at 2009. She completed her master at 2012 and in doctoral preparation stage. Title of doctoral thesis is (Treatment of induced partial hepatic cirrhosis in experimental animal model using differentiated stem cells). She is assistant lecturer now.
Abstract
Liver cirrhosis represents the final and the most common pathway of all chronic hepatic injury and is considered a worldwide health problem as liver slowly deteriorates and malfunctions and scar tissue replaces the healthy hepatocytes. The liver has the amazing potential to regenerate by its own hepatocytes when mild liver damage occurs but due to persistent and severe liver damage, hepatocytes no longer have the capacity to proliferate. Major advances have been made in the prevention, diagnosis, and treatment of liver cirrhosis, including liver transplantation and artificial liver. The ideas of complementing or replacing the damaged liver cells through cell-based therapies (stem cell) have been an extremely promising therapeutic agent for tissue regeneration in recent years. This study was applied on eighteen dogs as a model of cirrhotic liver treated with heterogeneous differentiated stem cells intraportal vein under ultrasonographic guidance. Induction of cirrhosis was carried out via the administration of carbon tetrachloride (CCl4). Dogs were divided into two groups, Group-I: CCl4 with non MSCs treatment and Group-II: CCl4 with MSCs treatment. Liver function was assessed by determination of liver function tests, hepatic ultrasonography with measuring of portal and hepatic veins diameters and finally liver biopsy,. Liver function tests, hepatic ultrasonography, portal and hepatic veins diameter and histopathological properties demonstrated that hepatic cirrhosis was successfully established in dogs. After stem cells transplanted, there was a significant reversal in liver function tests, ultrasound findings, histopathological properties and portal and hepatic veins diameter (P < 0.05).
Biography
Andrew Gallagher is currently doing his PhD at the University of Liverpool in collaboration with Spheritech Ltd. His work is primarily focused on developing a corneal bandage lens capable of delivering drugs and antimicrobials to the cornea in order to aid wound healing. Another project he is currently involved in is focused on the development of a macroporous external wound dressing material.
Abstract
Spheritech Ltd have developed a novel biopolymer which has found many applications, amongst them is the formation of macroporous biodegradable 3D constructs developed specifically to support 3D cell culture (Proliferate®). The porosity of the biopolymer is controlled in such a manner as to accommodate cell and nutrient penetration within. A unique feature of Proliferate® is that it is composed of two naturally occurring components; the poly-amino acid, poly-ε-lysine and a di-carboxylic acid, such as dodecanedioic acid or brassylic acid. Poly-ε-lysine is an edible, non-toxic material currently used as an emulsifier and preservative in foodstuffs whilst dodecanedioic and brassylic acids are processed from plant oils. Due to the monomer composition of the biopolymer it will upon decomposition break down into natural components making it a potential candidate for tissue regeneration and transplantation. Proliferate® is already proven as cytocompatible with many cell types including HEK293T cells, kidney stem cells (mKSC H6), Schwann cells, olfactory ensheathing cells, astrocytes, corneal epithelial cells (HCE-T), conjunctival cells (HCjE-Gi) and keratinocytes (HaCaT). It is also in the process of being developed as a wound dressing material. A less cross-linked variant of the same polymer produces a hydrogel which is being developed as a corneal bandage. It is also being investigated for its potential to transplant corneal endothelial cells to the posterior of the cornea and aid in the regeneration of this region.
Biography
Tao Ni has completed his Ph.D. at the age of 32 years from Shanghai Jiaotong Universtiy , School of Medicine, China and no experience of postdoctoral study. He has published 3 papers in reputed journals of repute.
Abstract
In order to fully restore the structure and function of skin in the both donor and recipient site, a novel grafting with microscopic skin tissue columns (MSTCs) was studied. Methods A new technique of grafting that can harvest and transplant the full-thickness MSTCs with 200~1000um diameter was used. In vitro the viability of MSTCs were evaluated. Three different treatments were applied to the swine full-thickness wounds: open wounds, MSTCs transplantation, and STSG (split-thickness skin grafting) transplantation. After grafting, the wound healing rate and contraction rate were investigated. In addition, the morphological and histological evaluations were applied for comparing the outcomes of the donor sites. Furthermore, the MSTCs from human or black hair mouse were transferred to the nude mice for observing the reconstruction of skin appendages. The sweater glands, sebaceous glands, hair follicles in the recipient site were identified. Results The MSTCs contained epidermis and dermis can be harvested effectively with much less damage to the donor site. After culturing, the MSTCs presented high proliferation viability, especially the cells near by the germinal layer. Full structures of the sweat glands, the sebaceous glands, or the hair follicles were carried by MSTCs of 700um diameter and larger. The donor site was re-epithelialized within 3 days in vitro. The wound healing rate in the MSTCs auto-grafting group was lower than that in the STSG group at one week postoperative. After two weeks, wound healing rates in the three groups were the same. However, the contraction rates in the MSTCs or STSG groups were much lower than that in the open wound group. Compared to the STSG grafting and open wound without grafting, MSTCs grafting gained better outcome of wound healing. In addition, the wounds in the donor site after taking MSTCs healed much faster than that in the STSG donor site with much better morphology. Furthermore, human sweat glands, sebaceous glands or hair follicle in the recipient site of mice were identified after human MSTCs xenografting. The black hair grew very well on the recipient site of nude mouse (no hair) after MSTCs of black hair mouse allografting. Full-thickness microscopic skin tissue columns can be harvested effectively with high activity and less damage in the donor site. Grafting of MSTCs improves the outcomes of full-thickness skin defects efficiently with restoring of skin appendages. Our results demonstrate that the MSTCs grafting will become a very promising technique for skin regeneration in clinical practice.
Biography
Bedriye Topal has received her B.Sc. Degree from Hacettepe University, Department of Chemical Engineering. She is a M.Sc. student at the same department. Also, she is a member of Hacettepe University Cell and Tissue Engineering Research Group, headed by Prof. Dr. Menemşe Gümüşderelioğlu
Abstract
Osteosarcoma is a malignant bone tumor that mostly encountered among pediatric and adolescent age group. As a kind of treatment for osteosarcoma, the section of cancer tissue is surgically removed from bone and a bone defect is occurred. Melatonin (N-acetyl-5-methoxytryptamine), the hormone secreted by the pineal gland, has not only anticancer effect because of its free radical scavenger, but also has an important role on bone regeneration. Despite of these critical functions of melatonin, low water solubility limits its usage with high capacity. In this study, melatonin/hydroxypropyl-β-cyclodextrin (HPβCD) inclusion complex was formed to increase water solubility of melatonin using microwave treatment. Inclusion complex was characterized by Higuchi-Connor’s phase solubility diagram, FTIR, NMR, XRD, DSC and TGA methods. Then, chitosan scaffolds were prepared and inclusion complex was embedded onto scaffolds. This carrier system is considered to affect directly to cancer cells and also provide a mechanical support in damaged area during bone regeneration. The release profile of melatonin from this system was investigated in vitro. Melatonin / HPβCD inclusion complexes were accomplishedly produced in solution state using microwave technology. Phase solubility studies proved that 1:1 (M/M) inclusion complex was obtained as well as other instrumental analysis were verified the occurrence of inclusion complex. In vitro release studies were conducted during 2 hours and approximately 86% of loaded melatonin was released from the scaffolds. In the upcoming future studies, characterization of loaded chitosan scaffolds and cell culture with MG-63 (human osteosarcoma cell line) will be done and specified by using related investigations.
Biography
Inji Shikhaliyeva has graduated from Baku State University, Department of Biology (B.Sc.). She is a M.Sc. student at Hacettepe University, Department of Bioengineering and a member of Hacettepe University Cell and Tissue Engineering Research Group headed by Prof. Dr. Menemse Gumusderelioglu.
Abstract
Boron (B) is a notable trace element in humans which has stimulating potential on osteogenesis. The effect of B on the osteogenic differentiation of hBMSCs, hTGSCs and MC3T3-E1 cell line had been reported. The aim of this study was to evaluate the effects of B-doped hydroxyapatite (HAp) coated chitosan (B-HAp/Ch) scaffolds on osteogenic differentiation of rat mesenchymal stem cells (MSCs) derived from bone marrow (BMSCs) and adipose tissue (AdMSCs), comparatively. AdMSCs were isolated enzymatically from adipose tissue and BMSCs were harvested from the femurs and tibias of the male Sprague-Dawley rats. Freeze-drying method was used to fabricate porous chitosan scaffolds and the scaffolds were coated with HAp and B-HAp in the presence of microwaves. In addition, fibronectin (Fn) was immobilized to the scaffolds to enhance the cell attachment. Cell culture studies were conducted with Ch, HAp/Ch and B-HAp/Ch scaffolds. MSCs at a concentration of 2.5 x 105 cells were seeded into each scaffolds. The cells were cultured with osteogenic medium for 28 days under static conditions. The adhesion behaviour and morphology of cells on the scaffolds were observed by SEM analysis at desired culture periods. Proliferation behaviour of cells were determined using MTT assay. Real-time PCR was used to detect the expression of several osteogenic differentiation-related marker genes such as Col1, β-actin, osteocalcin, osteonektin and RunX2. Our results showed that boron enhances proliferation and osteogenic differentiation of both of the AdMSCs and BMSCs. These findings suggest that B-HAp/Ch can be evaluated as a promising scaffold for bone tissue engineering.
Biography
Noha El Hadary has completed her master degree in the field of molecular physiology in the year 2013 from the German University in Cairo . She is currently employed as an assistant lecturer in the physiology department in the same university. Her field of expertise includes but not limited to exercise immunology, cytokines, muscle physiology and regeneration, immunostaining, measuring physiological parameters especially for muscle performance and use of various computer based software for data acquisition.
Abstract
The aim of this study was to explore the effect of exercise on skeletal muscle performance and muscle regeneration following IRI. Rats were divided into 5 groups: Control group (I), IRI group with 2 hours reperfusion (IIa), IRI group with 2 weeks recovery (IIb), swimming exercise trained group followed by IRI with 2 hours reperfusion (IIIa), swimming exercise trained group followed by IRI with 2 weeks recovery (IIIb). Muscle performance physiological parameters [peak twitch tension (Pt), peak tetanic tension (PTT) and fatigue resistance (FR) time] as well as neonatal MHC and CK were assessed in the gastrocnemius muscle. Additionally, the muscle was histologically examined, morphologically studied to detect percentage area of regenerating muscle fibers and immuno-stained to detect αSMA. Rats that performed regular swimming exercise followed by IRI with 2 hours reperfusion or 2 weeks recovery showed a significant improvement in the muscle contractile parameters compared to the untrained group evidenced by the significant elevation in Pt and PTT and significant prolongation in the FR time. Muscle regeneration was significantly improved in the trained injured group with 2 weeks recovery compared to the untrained group with 2 weeks recovery as evidenced by the significant increase in neonatal MHC gene expression and by the positive areas of regeneration and the significant increase in αSMA. In conclusion, exercise exerted a partial protective role against skeletal muscle dysfunction following IRI through the improvement of skeletal muscle regeneration.
Biography
Jinfeng Fu has completed her Ph.D. at the age of 33 years from Kunming Medical Collage China and no experience of postdoctoral study. She was the director of the Burn Research Institution of Yunnan Province,and the chief of the Department of Burn Surgery of the Second Affiliated Hospital. She is the Standing member of Chinese Burn Association, Chinese Medical Doctor Association and Cosmetic Doctor Association Scar Chapter. She has published more than 5 papers in reputed journals of repute.
Abstract
In order to fully restore the structure and function of skin in the both donor and recipient site, a novel grafting with microscopic skin tissue columns (MSTCs) was studied. Methods A new technique of grafting that can harvest and transplant the full-thickness MSTCs with 200~1000um diameter was used. In vitro the viability of MSTCs were evaluated. Three different treatments were applied to the swine full-thickness wounds: open wounds, MSTCs transplantation, and STSG (split-thickness skin grafting) transplantation. After grafting, the wound healing rate and contraction rate were investigated. In addition, the morphological and histological evaluations were applied for comparing the outcomes of the donor sites. Furthermore, the MSTCs from human or black hair mouse were transferred to the nude mice for observing the reconstruction of skin appendages. The sweater glands, sebaceous glands, hair follicles in the recipient site were identified. Results The MSTCs contained epidermis and dermis can be harvested effectively with much less damage to the donor site. After culturing, the MSTCs presented high proliferation viability, especially the cells near by the germinal layer. Full structures of the sweat glands, the sebaceous glands, or the hair follicles were carried by MSTCs of 700um diameter and larger. The donor site was re-epithelialized within 3 days in vitro. The wound healing rate in the MSTCs auto-grafting group was lower than that in the STSG group at one week postoperative. After two weeks, wound healing rates in the three groups were the same. However, the contraction rates in the MSTCs or STSG groups were much lower than that in the open wound group. Compared to the STSG grafting and open wound without grafting, MSTCs grafting gained better outcome of wound healing. In addition, the wounds in the donor site after taking MSTCs healed much faster than that in the STSG donor site with much better morphology. Furthermore, human sweat glands, sebaceous glands or hair follicle in the recipient site of mice were identified after human MSTCs xenografting. The black hair grew very well on the recipient site of nude mouse (no hair) after MSTCs of black hair mouse allografting. Full-thickness microscopic skin tissue columns can be harvested effectively with high activity and less damage in the donor site. Grafting of MSTCs improves the outcomes of full-thickness skin defects efficiently with restoring of skin appendages. Our results demonstrate that the MSTCs grafting will become a very promising technique for skin regeneration in clinical practice.
Biography
Patricia Rodrigues Orsi is degree in Biological Sciences, having master degree and Ph.D in Biological Sciences (concentration area Pharmacology) by São Paulo State University “Juliode Mesquita Filho†(UNESP), Brazil. Currently is Postdoctoral at Center for the Study of Venoms and Venomous Animals at UNESP, in developing project with bone tissue regeneration, stem cells and scaffolds.
Abstract
New Fibrin Sealant (FS) derived from thrombin-like enzyme purified of snake venom and animal fibrinogen (Center for the Study of Venoms and Venomous Animals– CEVAP, UNESP, Brazil) was usedas scaffoldfor bone marrow Mesenchymal Stem Cells (MSCs) in osteoporosis treatment in rat. Wistar rat MSCs of third passagewere identified to CD90,ICAM1 (positive) and to CD45, MHC-II CD34 and CD11b (negative).Two-months-old ratswere ovariectomized to osteoporosis induction. After 3 months a bonecritical defect(lesion of 5mm) was done in distal ends femur of 56 rats and were filled with FS and FS+MSCs to evaluate effects. Controlgroup (injury and no treatment) and group white(noinjuryand no treatment) also were added. Femur Analyses were performed 14 and 28days after surgery. Bone regenerationwasevaluated by Alkaline Phosphatase (AP) and Calcium (Ca)levels histological analysis, Scanning Electron Microscopy (SEM), X-ray and Tomography. Differencesbetween means weretested byanalysis of variance (ANOVA) followed by significance tests. AP level difference was showed at 28th day. The serum Ca level remained constant. SEM and X-ray revealed a wound healingin28 daysin both treatedgroups. Histological date showed that the wound healing process in treated animals with FS and FS+MSCs was better than control group, during the experimental period.No significant differences were found between castrated and un-castrated groups comparing the density values in Hounsfield scale (HU). The new fibrin sealant showed to be a potential candidate as scaffolds to MSCs in bone regeneration.
Biography
Vera Chernonosova has a master’s degree in Biochemistry from the Novosibirsk State University, Russia (2003). Currently, she is a PhD student in laboratory Dr. P.P. Laktionov at Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences. Her research interests cover the following subjects: electrospinning, biomaterials for regeneration, tissue implants, interactions of cells with nanomaterials
Abstract
Production of fibers by electrospinning from the solutions of polymers as well as their mixtures with low molecular substances or biopolymers provides a wide range of 3D matrixes that mimic natural tissues and provide long term delivery of various compounds included in these 3D matrixes. Obviously the ability to release of the compound from basic polymer enriched fibers, depend from solubility of both basic synthetic polymers and biopolymers as long until the fibers will be formed during polymer drying up. In this work we have investigated release/distribution of human serum albumin (HSA) from/in electrospun produced 3D matrixes (ESM). The ESM were produced by electrospinning from the solutions of PCL with different concentration of HSA (in a range 0,4-30%) in hexafluoroisopropanol (fiber diameter ~1 mkm, thickness ï¾150 mkm). The concentration of the exempt protein was measured by ELISA; other methods like Bradford or NanoOrange kit were not suitable. Protein was modified with N-(2-hydroxyethyl)phenazine to evaluate the protein location at the fiber surface. The data obtained by ELISA demonstrate that the main part of protein remains in ESM (for example, ESM containing 10% HSA releases no more than 1-3% of protein), while no less than 20% of the protein is exposed at the surface of the fibers according to the data received from modification of free amino groups by N-(2-hydroxyethyl)phenazine. Treatment of the ESM by proteinase K releases only half of the protein exposed out of the fibers. It was shown that predominant location of the protein is surface/subsurface area of fibers.
Biography
Anna Andrzejewska has completed her studies in Faculty of Biology at University of Warsaw at the age of 24. She obtained her master degree in biotechnology with specialization in molecular biology in 2012 and then started her PhD studies in Mossakowski Medical Research Centre Polish Academy of Sciences at NeuroRepair Department under supervision of Barbara Åukomska Professor of Immunology and Transplantology. She has already attended 5 conferences including 2 international conferences where she presented short communication of her results. She has published one paper and currently 2 more publications are in preparation.
Abstract
Mesenchymal stem cell (MSCs) therapy is an emerging area of research in regenerative medicine with significant studies being carried out in several clinically important areas. Systemic infusion of MSCs is preferred as a minimally invasive method of transplantation however the success relies upon MSCs migration from the vasculature and their efficient recruitment into appropriate tissues. We hypothesize that the induction of integrin overexpression in MSCs will enhance their adhesion and extravasation through the endothelial wall. The aim of the research was to examine the functionality of modified (mRNA-ITGA4transfected) human bone marrow MSCs (hBM-MSCs ) in vitro (using the microfluidic assay) and in vivo (i.a. transplantation of hBM-MSCs in ouabain-induced brain ischemic rats) studies. The experiments in vitro are based on using special microchannels covered by endothelial cells in which laminar flow of hBM-MSCs was forced and their interaction and adhesion to endothelial cells layer are assessed. After the pressure-driven cell perfusion the naive and ITGA4 overexpressing hBM-MSCs were observed to roll, capture and arrest at the surface of endothelial cell layer. Preliminary studies depicted many more mRNA-ITGA4 transfected hBM-MSCs flowed with the lower speed than naive hBM-MSC. In vivo studies revealed that hBM-MSC injected to the right internal carotid artery migrated into the rat ischemic brain being visible in MRI in the ipsilateral hemisphere. Interestingly, more mRNA-ITGA4 transfected hBM-MSCs than non-modified naive cells were found however a constant loss of both types of hBM-MSCs was noticed during long term observation after their transplantation.
Biography
Patrycja Siedlecka completed her master of science degree in biotechnology at University of Life Sciences in Lublin, Poland. She is a PhD Student at Stem Cell Bioengineering Laboratory at Mossakowski Medical Research Centre Polish Academy of Sciences in Warsaw. She is working on the neuroprotective and anti-inflammatory mechanisms of stem / progenitor cells derived from umbilical cord blood and umbilical cord on nerve tissue. Her current work focuses on the supportive effect of mesenchymal stem cells on vascular network of hippocampal slice culture injured by oxygen-glucose deprivation. She is a member of the Polish Neuroscience Society (PTBUN).
Abstract
Objectives Ischemic stroke results in rapid dysfunction of tissue homeostasis leading to disruption of the proper interactions within structural elements of the neurovascular unit (NVU). Recent publications suggest an important role of endothelial progenitor cells in the reconstruction of damaged microvessels, thus contributing to a faster recovery of the NVU in damaged brain. The aim of this study was to evaluate the ability of mesenchymal stem cells derived from human Wharton jelly (WJ-MSC) to differentiate into endothelial progenitor cells (WJ-EPC) and then support vascular network in rat hippocampal organotypic culture (OHC) model of ischemic injury. Material and methods Mesenchymal stem cells isolated from umbilical cord slices were cultured in classical growth medium (MSCGM) or in endothelial differentiating medium (EGM-2). Cells were characterized by immunocytochemistry, flow cytometry and molecular methods based on expression of mesenchymal and endothelial specific markers (CD90, CD73, CD105, vWF, CD31, VEGF, VEGFR2). Potential angiogenic activity of WJ-EPC was evaluated in vitro by capillary-like structure formation test and DiI-Ac-LDL-uptake assay. After 5 days of incubation OHCs undergo oxygen-glucose deprivation (OGD). Subsequently, WJ-MSC or WJ-EPC were co-cultured with intact or post-ischemic slices. Supporting effect of WJ-MSC and WJ-EPC supplementation on vascular network was verified immunohistochemically with rat EC-specific antibody (anti-RECA-1), which allowed calculation of the percentage area of blood vessels in CA1 and CA3 hippocampal regions. Results and conclusions Our results showed that WJ-MSC after 7 days of incubation in EGM-2 medium acquired typical for EPC cobblestone-like morphology, were able to form capillary-like structures on Matrigel and took up DiI-Ac-LDL. Both cell types were positive for MSC and EC markers CD73, CD90, CD105, VEGFR2, VEGF, but only EPC-differentiated cells expressed vWF and CD31. Moreover, WJ-MSC and WJ-EPC co-cultured with injured OHCs decreased atrophy of the blood vessels and protected CA1 cells against post-ischemic death. These results indicate protective effects of the cells of mesenchymal origin in OGD hippocampal slices, which make them a promising candidate for the treatment of brain ischemia.
Biography
Neveen A. Salem have completed her Ph.D. at the age of 31 years from Faculty of pharmacy Cairo University and postdoctoral studies from National Research Centre. She is now a researcher in National Research Centre. She has published more than 24 papers in reputed journals and she is principle investigator of a project about the therapeutic effect of mesenchymal stem cell derived from bone marrow and adipose tissue on liver fibrosis.
Abstract
Adipose tissue holds a promise for regenerative medicinal approaches. This study investigated its competency in liver fibrotic disease induced in rats by thioacetamide. Adipose tissue derived mesenchymal stem cells (AT-MSCs) were isolated, characterized, and cultured in HGF enriched media. Thirty adult female albino rats were used in the present study divided into three groups (n=10). TA was administered (100 mg/kg, i.p, three times weekly for 6 weeks) to 2 groups, one was treated with a single injection of (3x106/ml) AT-MSCs, while the other group was served as positive control group. After two months of treatment, biochemical parameters were measured and histopathological examination was performed. The engraftment of PKH-labeled differentiated AT-MSCs in injured liver was confirmed. TA administration produced fibrotic changes characterized by increased serum enzymes activity and reduced albumin and fibrinogen. Additionally, hepatic TGF-β and HGF contents were elevated. Treatment with AT-MSCs restored liver functions associated with decreased TGF-β. The expression of HNF4a, FOXa2, and albumin was upregulated, while CK-7, CYP7a1, β-5-Tub were down-regulated. These observations were further manifested in histological and immunohistochemical examination of liver tissue. The present work enlighten the aptitude of adipose tissue in cell therapy and regeneration of fibrotic liver, where functional hepatocytes were reproduced. KEY WORDS: liver fibrosis, adipose tissue, mesenchymal stem cells, TGF- β, CYP7a1, rats.
Biography
Manarbek B. Askarov, PhD., MD. National Research Medical Scientific Center, Department of stem cells transplantation and biotechnology. Director. Kazakhstan (Astana). He has published more than 55 papers in scientific journals. Specialists in regenerative medicine and stem cell investigators.
Abstract
At systemic sclerosis on the background of autoimmune disorder inflammatory and fibroplastic reactions are initiated, proliferative/oblitirating generalized vasculopathy and progressive fibrosis of internal organs are developed. In this regard, use of bone marrow stem cells (BMSC) - the central organ of immunogenesis that are characterized immunomodulatory (natural suppressor activity) and morphogenetic and also angiogenetic and anti-fibrotic effects can be effective pathogenetic justified method of treatment of systemic sclerosis. The aim of the investigation is - to give clinical and morphological estimation of the effectiveness of autologous bone marrow stem cells transplantation at systemic sclerosis. We observed 30 patients aged from 27 till 38 years, with definite diagnosis of systemic sclerosis (SSc) according to the criteria of the American College of Rheumatologists (ACR, 1980). Duration of the disease was more than 3 years. Resistance to immunosuppressive therapy (D-penicillamine, azathioprine and prednisolone) and high index of autoimmune process activity (EScSG), according to the criteria of the European Group for the Study of SSc formed the fasis for carrying out bone marrow stem cells transplantation. Clinical efficacy was estimated by the criteria of the European Group studying SSc. Skin involvement was assessed clinically by changes in modified Rodnan skin score, which is considered as the most obvious clinical sign of scleroderma. Results were also evaluated by serological features (ESR, CRP, γ-globulin, fibrinogen and anti-nuclear antibodies (ANA). Bioptic skin material of top third shin was investigated in stage of induration before and in dynamic of treatment for the morphological assessment of the effectiveness of the treatment. Preparations were stained with hematoxylin and eosine and also Masson-trichrom. Electron- microscopic investigation of the material was performed by the standard method on electron microscope Libra 120 of Carl Zeiss firm. Thus, preculturing autologous hematopoietic stem cells of bone marrow transplantation create conditions for the regulation immune (autoimmune) inflammation on system and local level creating conditions for increasing processes of angiogenesis and create conditions for the restoration of morpho-functional skin condition, improvement in serological markers, stabilisation of internal organ function of patients with systemic scleroderma.
Biography
Camilo Arturo Suárez Ballesteros received a BSc degree in physics from pedagogicaland technological university of Colombia (2010), and MS degree in Sciences (2012) from University of São Paulo (Brazil). In 2013 he began a doctoral studies (PhD) inbiomolecular physics at University of São Paulo (Brazil) under the supervision of Prof. Dr. ValtencirZucolotto. His main research interests include the development of sensor and biosensor in nanomedicine, polymeric nanofibers in regenerative medicineand synthesis and characterization of nanomaterials.
Abstract
There is an urgent need to prevent bacterial infections after surgical wounds and burns.We developed a system of nanofiber scaffold – polymeric nanocapsules of silver nanoparticle how tissue, for placed on the wounds and burns. The method consist in applied a pointer LASER 405 nm on system for controlled delivery of silver nanoparticles (AgNps) innanofiber scaffold.The LASER excites the surface plasmon resonant of Ag Nps which change the polymeric configuration of nanocapsule and allowing its release on nanofiber scaffold.This method avoids the toxic effect of the direct interaction of Ag Nps with human body and increases bacterial inhibition in surgical wounds and burns.The system was characterized by scattering electron microscopy, atomic force microscopy, fourier transform infrared spectroscopy and UV-Vis spectroscopy.We use gram-negative Escherichia coli and gram-positive staphylococcus aureus for antibacterial study.Minimum inhibitory concentration (MIC) shows antibacterial activity of nanocapsule solution [0,9 µg/mL], demonstrating the bactericidal effect only when the LASER is incident on nanocapsule system.AGAR diffusion shows a radius of bacterial inhibition (0.8 mm) when LASER acted over the blanket system.In the two studies, we performed the experiments with broad range of solution concentration of nanocapsule and different time exposition of LASER, shows than around 5 to 7 min it found release Ag Nps for bactericidal effect.Our results suggest that the blanket system when LASER is applied, has an interesting approach for development of novel bactericidal methods in medical applications
Biography
Hamed Alizadeh Sardrood is now studying MSc in Chemical Engineering Faculty of Sahand University of Technology, Tabriz, Iran. He is working on his MSc thesis in tissue engineering and stem cell research lab with the title of: “Study on proliferation and differentiation of hematopoietic stem cells encapsulated in modified alginate microcapsulesâ€.
Abstract
Hematopoietic stem cells (HSCs) need specific growth factors for self-renewing expansion. Mesenchymal stem cells (MSCs) can regulate the growth and differentiation of HSCs through the release of the soluble factors. We used alginate-gelatin based three-dimensional (3D) microcapsules for the co-culture of HSCs with MSCs derived from umbilical cord (UC), to simulate the main components of the bone morrow niche, and the results were compared with non-encapsulated cells. Alginate-gelatin microcapsules with an average size of 600 µm were developed by means of an electrostatic inject device. HSCs were cultured in monoculture and co-culture with UC-MSCs in a 12-well plate both in microcapsules and in free cultures, without adding any growth factors. Cell numbers and activity of HSCs was assessed by hemocytometer cell counting method and MTT assay, respectively. Self- renewing HSCs was identified by flow cytometry method. The maintenance of the primitive CD34+ and CD45+ phenotype of HSCs were analyzed after 7 days of culture. Our results showed that HSCs could proliferate 2 folds in the microencapsulated co-culture condition. The percentage of CD34+ and CD45+ stem cells in the microcapsules with and without MSCs were 80% and 75%, respectively. This research demonstrated that alginate-gelatin microcapsules have a high potential for the co-cultring HSCs and MSCs to enrich HSCs for medical applications without using growth factors, although further investigations is needed in this regard.
Biography
Sorour nemati is now studying MSc in Chemical Engineering Faculty of Sahand University of Technology, Tabriz, Iran. She is working on her MSc thesis in Tissue Engineering and Stem Cell Research Lab with the title of “Study on proliferation and differentiation of hematopoietic stem cells in bubble column bioreactor. She has published one paper in 10th Royan international congress on stem cell biology and technology.
Abstract
One of the most important cell-based therapies is using suspendable stem cells such as hematopoietic stem cells (HSCs). Proper methods are needed for the adequate expansion of HSCs in order to use in clinical applications. In this project expansion of hematopoitic cell line (U937) as a model of the suspendable cell was investigated in alginate–gelatin microcapsules. The microcapsules can provide the cells a three dimensional microenvironment as well as protection from shear stress particularly in bioreactors. U937 cells were encapsulated in alginate-gelatin at a density of 2.5 million cell.ml-1 with an average size of 600 µm which were developed by means of an electrostatic inject device. Cell numbers and activity of U937 was assessed by hemocytometer cell counting method and MTT assay respectively. The results showed U937 cells expanded 23 folds in static culture (T-flask) after 7 days. In free cell culture, however, the cells showed a 14-fold increase in cell number. Indeed the encapsulated cells were cultured in a bubble column bioreactor with an aeration of 0.1 vvm at 21% and 5% O2. The result of this bioreactor showed with decreasing O2 from 21% to 5%, the cells increased 2 folds in the microcapsules, indicating the importance of process parameters such as oxygen concentration and tension on the cell behavior. These results demonstrated that alginate-gelatin microcapsules can be considered as a proper microenvironment for proliferation of suspension cells, although more optimization for dynamic systems is required.
Biography
Vijay Kumar Kuna completed M.Sc in Biotechnology in 2009 from Bangalore University, India and M.Sc in Molecular Biology 2012 from University of Skövde, Sweden and worked as research assistant from March 2013- September 2014 and started with PhD from October 2014 at University of Gothenburg. He published 5 papers in reputed journals while once in Lancet. He supervised 4 Master thesis projects till now. He attended four international conferences on vascular tissue-engineering and transplantation and gave oral presentation in two and poster presentation in two.
Abstract
Vascular diseases affect >25 million individuals in westernized societies. Transplantation using tissue-engineered vascular grafts using autologous cells will benefit them. Previous strategies for repopulating vascular conduits were dependent on successful ex vivo expansion of cells. Here we demonstrate a new approach using peripheral whole blood (PWB) thus eliminating isolation and expansion difficulties associated with cells. We extracted 7-9 cm human allogeneic iliac vein segments and decellularized by agitation with 1% triton, 1% tnbp and DNase for 4h with each until 9 cycles. They were recellularized for 10 days in a bioreactor by perfusing 25 ml PWB from healthy donors for 2 days followed by endothelial and smooth muscle cells media for 4 days with each. Further, two autologous PWB tissue-engineered vein conduits were prepared and used for by-pass procedures in a four-year and 20-months patients. The decellularized vein expressed several important extra cellular matrix proteins, angiogenic growth factors and maintained good biomechanical integrity. Recellularized veins showed a well-formed endothelial cell layer and presence of smooth muscle cells in media. Mainly, VEGFR-2+ /CD45+ and a smaller fraction of VEGFR-2+/CD14+ cells contributed to repopulation of the graft. Electron micrographs showed flat cells on the luminal surface of the grafts consistent with endothelial cells. Clinically, the grafted veins immediately provided the two recipients with a functional blood supply. Both patients have normal laboratory values at 30 and 28 months post-transplantation respectively. The patients have not received immunosuppressive drugs. Thus, a simple autologous blood sample can be used to generate personalized vascular conduits. This represents an important milestone for vascular tissue-engineering.
Biography
Oula El Atat has completed her M.Sc from the Lebanese University, Doctoral School of Science and Technology. Currently,she is a Ph.D student the Regenerative Medicine and Inflammation lab, faculty of medicine, Saint Joseph University, Beirut, Lebanon.
Abstract
The Use of adipose derived mesenchymal stem cells in regenerative medicine is in rise due to their plasticity, their capacity of differentiation, their paracrine and trophic effects. Adipose derived stem cells (AD-MSC) have been proven to have the same characteristics as bone marrow derived stem cells; nonetheless the harvesting of fat is minimally invasive with insignificant donor site morbidity and a low complication rate. The number of stem cells in fat is higher; it has been reported to be 100 to 1000 times more, with higher proliferation rate, and less senescence compared to bone marrow. Despite the large number of cells obtained from adipose tissue, it is usually not enough for therapeutic efficiency in many diseases and even sometimes for plastic surgery uses such as stretch marks, burns or skin lesions, where the need for culturing and expanding the cells in vitro for several weeks. Our objective is to investigate the stemness and the immunomodulatory activity of synovial vascular fraction (SVF) and serial passaged adipose derived stem cells (ADSC) versus their potential tumorigenic activity ADSC were isolated, purified, and cultured in vitro from lipoaspirates using a well-established protocol. The immunophenotypic properties (SVF) and serial passaged ADSC (P0-P4) were observed by flow cytometry. In parallel, we have compared the telomerase activity, the aldehyde dehydrogenase activity, and the relative expression of hTERT, C-MYC, and the nucleotide sequence of p53 through passages. We have also analyzed the cytokines secretion profile of ADSC during passages was a by ELISA kits. The telomerase activity was low in SVF and non-increased during P0 to P4 although it always decreases at day 21 during each passage. Aldehyde dehydrogenase was detected in SVF with no changes with serial passages. The relative expression of hTERT was not detected, and that for C-MYC decrease significantly. A SNP was detected at the nucleotide 417 where C is substituted by G. The level of PGE2, STC1, TIMP1 and TIMP2 was not affected through passages. Our results suggest that the expansion of ADSC does not affect the differentiation capacity of stem cells and do not confer to cells a cancerous state or capacity.
Biography
Manal Kamel, is a Professor of Immunology and head of Immunology and Therapeutic Evaluation Department at Theodor Bilharz Research Institute (TBRI), Giza, Egypt. She graduated from the Faculty of Medicine, Cairo University (M.B.B.Ch.). Her postgraduate studies were in immunology and clinical Pathology (M.Sc.; M.D.) from the same university. She has been carrying out research work on immunology of parasitic diseases of medical importance in Egypt. She supervised (9) M.Sc. and Ph.D thesis, two of them in the field of nanotechnology with stem cells and monoclonal antibodies. She has been awarded the best research article from TBRI in 2009. She has awarded a scholarship from DAAD, W. Germany, for training on the application of different immunochemical techniques for diagnosis of diseases in Institute for Comparative Tropical Medicine and Parasitology, Munich, West Germany. she was awarded a second scholarship at the Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria for advanced techniques of cultures of different parasites. She has been working in Production of different antigens and biological materials for more than 15 years in the Schistosome Biological Supply Program (SBSP), Theodor Bilharz Research Institute (1995 till now).
Abstract
Background Cord blood (CB) derived Unrestricted Somatic Stem Cells (USSCs) with their multipotentiality hold great promise in liver regeneration. This work aims at evaluation of the therapeutic potentiality of USSCs in two experimental models of chronic liver injury induced either by S. mansoni infection in balb/c mice or CCL4 injection in hamsters. Material and methods Isolation, propagation, and characterization of USSCs from CB samples were performed. USSCs were induced to differentiate into osteoblasts, adipocytes and hepatocyte-like cells. Cells of the third passage were transplanted in.two models of liver fibrosis: (1) Twenty hamsters were induced to liver fibrosis by repeated i.p.injection of 100 µl CCl4 /hamster for 8 weeks. This model was designed as; 10 hamsters with liver fibrosis and treated with i. h.injection of 3x106 USSCs (USSCs transplanted group), 10 hamsters with liver fibrosis (pathological control group), and 10 hamsters with healthy livers ( normal control group). (2) murine chronics S.mansoni model : twenty mice were induced to liver fibrosis with S. mansoni ceracariae (60 cercariae/ mouse) using tail immersion method and left for 12 weeks. This model was designed as; 10 mice with liver fibrosis were transplanted with i.v. injection of 1×106 USCCs (USSCs transplanted group). Other 2 groups were designed as in hamsters model. Animals were sacrificed 12 weeks after USSCs transplantation, and their liver sections were examined for detection of human hepatocyte- like cells by immunohistochemistry staining. Moreover, liver sections were examined for fibrosis level, and fibrotic indices were calculated. Sera of sacrificed animals were tested for liver functions. Results CB USSCs, with fibroblast-like morphology, expressed high levels of CD44, CD90, CD73 and CD105 and were negative for CD34, CD45, and HLA-DR. USSCs showed high expression of transcripts for Oct4 and Sox2 and were in vitro differentiated into osteoblasts, adipocytes, and hepatocyte-like cells. In both animal models, in virto induced hepatocyte-like cells were confirmed by cytoplasmic expression of glycogen, alpha fetoprotein, and cytokeratin18. Livers of USSCs transplanted group showed engraftment with human hepatocyte-like cells as proved by cytoplasmic expression of human alpha fetoprotein, cytokeratin18, and OV6. In addition, livers of this group showed less fibrosis than pathological control group. Liver functions in the form of serum AST & ALT level, and serum total bilirubin level were significantly lowered in USSCs transplanted group than pathological control group (p< 0.001). Moreover, the fibrotic index was significantly lower (p< 0.001) in USSCs transplanted group than pathological control group. In addition liver sections ,of i.v. injection of 1×106 USCCs of mice , stained with either H&E or sirius red showed diminished granuloma size and a relative decrease in hepatic fibrosis .Conclusion Our experimental liver fibrosis models transplanted with CB-USSCs showed liver engraftment with human hepatocyte-like cells as well as signs of liver regeneration in the form of improvement in liver function assays and fibrosis level.These data provide hope that human CB- derived USSCs are introduced as multipotent stem cells with great potentiality in regenerative medicine & strengthens the concept of cellular therapy for the treatment of liver fibrosis.
Biography
Joanna Jaworska obtained her Master of Science in Engineering degree in biotechnology with a specialization in molecular biotechnology and biocatalysis from Wroclaw University of Technology in 2011. She started her interdisciplinary PhD studies “Life Sciences for Biomedical Research†in Mossakowski Medical Research Centre Polish Academy of Sciences at the NeuroRepair Department under the supervision of Professor Teresa Zalewska in April 2013. Her research involves the investigation of the potential of endogenous stem cells for brain regeneration after neonatal hypoxic-ischemic brain injury. She has already attended 5 conferences including 2 international conferences where she presented the results of her research. She has two papers published and one accepted for publication.
Abstract
Neonatal hypoxic-ischemic (HI) damage causes severe brain diseases such as cerebral palsy. Treatment of these disorders is a big challange. Mounting evidence indicates that histone deacetylase inhibitors (HDACi) exert neuroprotective effects in brain ischemia associated with stimulation of endogenous neurogenesis. This study investigated whether treatment with HDACi – Trichostatin A, might enhance neurogenesis after neonatal hypoxia-ischemia in the neurogenic zone of dentate gyrus. We utilized a model of HI induced in rats of postnatal day 7. TSA was injected subcutaneously in one single dose for 5 consecutive days starting immediately after hypoxia-ischemia. To estimate the proliferation profile, BrdU (50mg/kg) was injected at various days after HI, whereas to determine the phenotype of newborn cells, BrdU was administered twice daily, at 5 to 7 days after HI. At 3-14 days after hypoxia-ischemia BrdU-positive cells were seen in both hemispheres, with the greatest number of dividing cells in ischemic side and strongest proliferation level occurring at days 3 and 6 after the injury. The labelling pattern revealed structure-dependent differences. Three days after HI the highest density BrdU-positive cells was seen in the hilus, whereas at longer survival time (9-14 days) labelled cells changed their localization towards the subgranular zone (SGZ) of DG. To analyze if BrdU-positive cells represent newly generated neuroblasts and oligodendrocyte progenitors, we used double staining BrdU/ DCX and NG2/BrdU, respectively. We found that administration of TSA stimulates generation of neuroblasts and oligodendrocyte progenitors, restoring their reduced number after HI to control level. Supported by NSC grant no 2012/05/B/NZ3/00436 and POKL.04.03.00-00-060/12
Biography
Sylwia Koniusz has graduated from the Warsaw University of Life Sciences with master's degree in biotechnology. In October 2013 she has started the MMRC-KNOW Interdisciplinary PhD Studies at the NeuroRepair Department at the Mossakowski Medical Research Centre Polish Academy of Sciences in Warsaw with the project "The role of bone marrow mesenchymal stem cells and microvesicles derived from these cells in CNS repair of brain ischemia disorders". Sylwia Koniusz has already attended 5 international conferences during which she presented the results of her study. Moreover, she has published one paper and has two more works in preparation.
Abstract
Mesenchymal stem cells (MSCs) are a potential tool for cell-based therapies in regenerative medicine. Many studies revealed that they produce tropic factors which being responsible for their protective and anti-inflammatory properties. The aim of the study was to evaluate migration of human bone marrow mesenchymal stem cells (hBM-MSCs) transplanted i.a. into focal brain ischemic rats and to check their effecton the host immune response induced by brain injury. The experiments were performed in adult Wistar rats with stroke induced with 1μl/50nmol ouabain (Na+/K+ ATPase pump inhibitor) injected into right striatum. Two days after brain injury, 5x105hBM-MSCs (Lonza) labelled with iron nanoparticles conjugated with rhodamine (Molday, BioPAL) were infused into the right internal carotid artery and the inflow of transplanted cells in the rat brain was monitored using MRI. At day 1,3 and 7 post-transplantation rat brains, spleens and cervical lymph nodes were removed and the immunocytochemical analysis of human markers was performed using anti-CD44 and -STEM121 antibodies.Additionally, immunological host response was accomplished by detection of ED1, CD5, CD45RAand GFAP – positive cells in the brain, spleen and lymph nodes. Results: MRI analysis revealed the positive signal for Molday particles inthe right hemisphere, mostly in cortex, corpus callosum and striatum. Viable hBM-MSCs were observed in the rat brain up to 7 days after their grafting. This was accompanied with the activation of immune cells. Whether hBM-MSCs transplantation contributes to attenuate host immune response induced by focal brain ischemia remainsto be evaluated in the further studies
Biography
Martyna Podobinska has completed her MSc at the age of 24 years from University of Warmia and Mazury in Olsztyn and is currently doing her doctoral research in Stem Cell Bioengineering Unit, Mossakowski Medical Research Centre Polish Academy of Sciences. The subject of her research is to define biomimetic conditions for self-renewal and differentiation of Human Umbilical Cord Blood derived Neural Stem Cells (HUCB-NSC) in vitro, including relationship investigation between spatial arrangement of culture, hypoxia and epigenetic regulation.
Abstract
The aim of the project was to analyse the effect of different scaffolds on the growth, proliferation and differentiation of freshly isolated mesenchymal stem cells from Wharton jelly (WJ-MSC) or neurally-committed progenitor cells derived from umbilical cord blood (HUCB-NSC). Different biological three-dimensional (3D) scaffolds were compared: (1) based on keratin associated proteins, in the shape of small rectangular dots and long hair-like fibers; (2) a new family of scaffold consisting of two components: collagen (Col) and chondroitin sulphate (CS) and (3) electroconductive chitosan scaffold. Results: The analysis performed with scanning electron microscope revealed that both WJ-MSC and HUCB-NSC did not adhere to small rectangular fibers, but remained seated on the long hair-like fibers. Neural stem cells were following the shape of fibers, while mesenchymal stem cells formed clamps filling the space between fibers. Both cell types predominantly adhered to Col-CS scaffolds. HUCB-NSC growing on the surface of the scaffold had branched phenotype and expressed neuronal markers (β-Tubulin III, MAP2), while the cells that penetrated inside the scaffold remained rounded. Under the same conditions WJ -MSC mostly penetrated inside the scaffold and strongly proliferated (Ki67 positive cells), with no signs of differentiation. Both WJ-MSC and HUCB-NSC seeded on chitosan-based scaffolds remained undifferentiated. Conclusion: We demonstrated that geometry, structure and composition of the scaffold have an impact on cells adhesion, proliferation and differentiation, but the response to these parameters varies with the cell type and stage of their development. Non-neurally committed progenitors (WJ-MSC) under the shape influence will mainly proliferate, while neural progenitors (HUCB-NSC) in the same conditions will continue to differentiate.