Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 4th International Conference on Tissue Science and Regenerative Medicine Rome, Italy.

Day 2 :

Keynote Forum

Nagy Habib

Imperial College London
UK

Keynote: Preclinical and clinical results of a subset of CD34+ cells

Time : 09:00-09:25

Conference Series Regenerative Medicine-2015 International Conference Keynote Speaker Nagy Habib  photo
Biography:

Nagy Habib is Lead Clinician for the Liver and Pancreatic Unit at Imperial College Healthcare NHS Trust and Academic Head of the Department of Surgery at the Hammersmith Hospital Campus of Imperial College London. Habib is a translational researcher who pioneered the first clinical trial in the use of plasmid and adenovirus for the treatment of liver cancer, as well as the use of plasmid gene therapy in hydrodynamic gene delivery. He was also the Principal Investigator of the world first clinical trials published on the use of adult bone marrow-derived stem cells for the treatment of patients with liver insufficiency and CD34+ cells in patients with stroke. As a clinical scientist he has conducted and published translational research in liver tumours on oncogene, tumour suppressor gene, epigenetic modification, gene therapy, stem cell therapy, and small activating RNA. He was the inventor and was co-author on the first publication to describe the use of radiofrequency energy in devices for liver surgery (Habib 4X), interventional endoscopy (Habib™ EndoHPB and Habib™ EUS-RFA) and interventional radiology (Habib™ VesOpen). He holds a gold award from the Advisory Committee for Clinical Excellence which is given to recognise and reward the exceptional contribution of NHS consultants, over and above that normally expected in a job, to the values and goals of the NHS and to patient care, and he was named as one of Britain’s top surgeons in December 2011 by the Saturday Times Magazine. He was awarded Honorary Professorships by universities in China, Bulgaria, Greece, and Russia. In November 2012 he was awarded Takreem Laureate for his contribution to technology and science.

Abstract:

The preclinical and clinical results of a subset of CD 34+ cells will be presented. The cells were used in three different clinical trials; (i) patients with liver failure; (ii) patients with acute total ischaemic circulation anterior stroke, and; (iii) patients with diabetes complicated by renal transplant. Safety and efficacy aspects of the studies will be discussed and also a new technique to increase efficacy of stem cells using small activating RNA1.

Keynote Forum

Gordon Blunn

University College London
UK

Keynote: Augmenting the fixation of orthopaedic implants with stem cells

Time : 09:25-09:50

Conference Series Regenerative Medicine-2015 International Conference Keynote Speaker Gordon Blunn  photo
Biography:

Gordon Blunn is a bioengineer at the Institute of Orthopaedics and Musculoskeletal Science at University College London; he is based at the Royal National Orthopaedic Hospital. He has been at this Institution for 25 years and during this period of time he has investigated the use of the fixation of implants to the skeleton. He has translated a number of novel treatments and designs into patients, which has enhanced implant fixation. He is currently the president of the British Orthopaedic Research Society.

Abstract:

Orthopaedic implants are extremely successful however in some instances fixation to the surrounding bone is problematic. In patients with bone cancer a massive prosthesis is used to replace the diseased bone. At the same time patients receive chemotherapy to treat and stop the spread of the cancer. In these cases the loosening rate is high, which leads to prostheses being removed and revised. Bone cancer occurs in young patients and these patients may be faced with numerous revision operations throughout their life, therefore, enhancing fixation of these implants is important. We have shown that augmentation with materials such as hydroxyapatite does reduce loosening but there still remains a number of cases where loosening occurs. The aims of this study were to use mesenchymal stem cells to enhance implant fixation to the bone surface (osteointegration).We carried out an investigation on femoral bone defects in rats, which showed that incorporating mesenchymal stem cells within fibrin enhanced bone formation even in those animals given a chemotherapy regime similar to that received by humans. Fibrin glue was chosen as the carrier as this could be sprayed onto the surface of the implant and formed a stem cell layer. Stem cells survived and proliferated within the fibrin glue and they remained viable after spraying the cells onto the implant surface. In an ovine model autologous stem cells were sprayed onto the surface of the implant at a concentration of 1 million cells per ml. After 6 months increased osteointegration of over 100% was measured. Increasing the number of the cells within the glue up to 10 million per ml had a significant affect. Using cells that had been differentiated into osteoblasts also increase osteointegration.However, using autologous mesenchymal stem cells resulted in little new bone formation and in osteoclastic resorption.

Conference Series Regenerative Medicine-2015 International Conference Keynote Speaker Richard G. Pestell photo
Biography:

Richard Pestell (MD, PhD, MBA, FRACP, FACP, MBBS) is a leader in the global cancer community. He has galvanized international teams to fight cancer, has >600 published works, and multiple patents in cancer diagnostics and treatment including light-activated gene therapy. He founded companies (ProstaGene, AAAPhoenix) and Institutes, led cancer not–for-profits and directed two US Cancer Centers (Lombardi Comprehensive Cancer Center, and the Sidney Kimmel Cancer Center at Thomas Jefferson University. He was most recently Executive Vice President Thomas Jefferson University in Philadelphia, USA. His work in breast and prostate cancer and the cell cycle is highly cited (>41,500 citations, H index >100).

Abstract:

A subpopulation of cells have been identified within tumors referred to as tumor initiating cells (BTIC) (cancer stem cells). These cells have been hypothesized to contribute to therapy resistance and tumor recurrence. Contributions to the understanding of the molecular genetic drivers of TIC using multigenic tissue specific temporally and spatially inducible transgenic mice have shown endogenous mammary tumor stem cells expansion is maintained by endogenous c-Jun, and NFkB. The cell cycle control protein p21CIP1 restrains, and cyclin D1 promotes, TIC populations respectively and Notch requires cyclin D1 for BTIC expansion. \\r\\nRecent studies from this laboratory have shown the RDGN (retinal determination gene network) is a dominant regulator of BTIC. In Drosophila Melanogaster, the Retinal Determination Gene Network (RDGN) determines organismal cell fate. The RDGN human homologues include DACH, EYA, Six, and Toy. We showed the RDGN (DACH1, and Eya) governs BTIC. The Drosophila Eyes Absent Homologue 1 (EYA1) is a component of the retinal determination gene network and serves as an H2AX phosphatase. EYA1 enhanced breast tumor growth in mice in vivo, requiring the phosphatase domain. EYA1-dependent induction of breast cancer cell proliferation and mammosphere formation was cyclin D1-dependent. The induction of cellular proliferation and cyclin D1 abundance, but not apoptosis, was dependent upon the EYA1 phosphatase domain. The relative proportion of cells, with characteristics of breast tumor stem cells were assessed in mice with targeted deletion of Dach1 in the mammary gland. The function of the RDGN and the interface with the cell cycle control genes in BTIC will be discussed\\r\\n

Break: Coffee 10:25-10:40
  • Track 6: Scaffolds
    Track 7: Whole Organ Engineering & Approaches
Location: Rome, Italy
Speaker

Chair

Joseph Purita

Institute of Regenerative and Molecular Orthopedics USA

Speaker

Co-Chair

Andrea Barbetta

Sapienza University of Rome
Italy

Session Introduction

Joseph Purita

Institute of Regenerative and Molecular Orthopedics
USA

Title: Cutting edge concepts in the use of stem cell and PRP injections in an office setting

Time : 10:40-11:00

Speaker
Biography:

Purita is Director of Institute of Regenerative and Molecular Orthopedics (www.stemcellorthopedic.com) in Boca Raton, Florida. The Institute specializes in the use of Stem Cells and Platelet Rich Plasma injections. Dr. Purita is a pioneer in the use of Stem Cells and Platelet Rich Plasma. The Institute has treated some of the most prominent professional athletes from all major sports in both the U.S.A. and abroad. He received a B.S. and MD degree from Georgetown Univ. Dr. Purita is board certified in Orthopedics by ABOS. He is a Fellow American College of Surgeons, Fellow American Academy Orthopedic Surgeons, and a Fellow American Academy of Pain Management. He is also certified in Age Management Medicine. He has lectured and taught extensively throughout the world on the use of Stem Cells and Platelet Rich Plasma. He has been instrumental in helping other countries in the world establish guidelines for the use of Stem Cells in their countries. He has been invited to lecture on these techniques throughout the world as a visiting professor.

Abstract:

The presentation concerns PRP and Stem Cell (both bone marrow and adipose) injections for musculoskeletal conditions in an office setting. Indications are given as to which type of cell and technique to use to accomplish repair. Stem cells, both bone marrow derived (BMAC) and adipose, are used for the more difficult problems. PRP injections are utilized for the less severe problems. Indications are given when to use Stem Cells verses PRP and when to use both. The newest concepts in stem cell science are presented. These concepts include the clinical use of MUSE cells, exosomes, and Blastomere like stem cells. Basic science of both PRP and stem cells are discussed. This presentation defines what constitutes an effective PRP preparation. Myths concerning stem cells are dispelled. One myth is that mesenchymal stem cells are the most important stem cell. This was the initial interpretation of Dr. Arnold Caplan the father of mesenchymal stem cell science. Dr. Caplan now feels that MSCs have an immunomodulation capacity which may have a more profound and immediate effect on joint chemistry and biology. We now learn in the talk that the hematopoietic stem cells are the drivers of tissue regeneration. Also discussed are adjuncts used which enhance the results. These therapies include supplements, LED therapy, lasers, electrical stimulation, and cytokine therapy. The scientific rationale is presented for each of these entities as to how they have a direct on stem cells.

Speaker
Biography:

Andrea Barbetta has completed his Ph.D. in 2001 from University of Durham (UK) and postdoctoral studies from University of Kyoto (Japan) and Sapienza University of Rome. At present, he is lecturer at the Department of Chemistry of Sapienza University of Rome. He has published 40 papers in reputed journals and 1 patent. His research interests are focused mainly in the development of methods for the fabrication of scaffolds for tissue engineering, in particular by means of microfluidics and rapid prototyping.

Abstract:

Microfabrication technologies have been proposed as methods to create vascularized tissues. However, despite significant advances, insufficient aligned cellular organization and limited hierarchical architecture has impeded progress toward mimicking the highly vascularized tissue in 3D. To address these challenges, we introduce a new paradigm of vascularization that uses bioprinting as a robust method for fabricating 3D tissues constructs. This approach is based on a cell-laden fiber deposition technique that uses low-viscous solutions of biocompatible materials and cells and can form 3D, interconnected hydrogel fiber grids with high fidelity and reproducibility. The described method uses calcium-alginate as sacrificial templating polymer during the 3D printing process, and produces methacrylated gelatin cell-laden constructs with features in the order of 100 micrometer.We used this technology to produce 3D pre-vascular networks to be used as scaffold for a second, post-seeded cellular type. Endothelial cells (HUVECs) have been 3D printed in interconnected fiber meshes and spread and matured in tubular structures. Cardiomyocytes have been seeded on top of the endothelial network, giving rise to a pre-vascularized, 3D cellular construct that showed strong spontaneous beating behavior. This methodology, that combines bioprinting and scaffold-based approaches, can represent a new paradigm for the in vitro vascularization of 3D tissues.

Speaker
Biography:

Paola Brun is an Assistant Professor at the Department of Molecular Medicine, University of Padova. She obtained her Ph.D. in 2005. Her scientific research activity covers a range of multidisciplinary topics mainly focused on cell biology, bioengineering and molecular medicine. She actively collaborates with the Departments of Industrial Engineering and Physics to find out innovative materials and techniques able to promote proliferation and differentiation of different cell populations. She has published 55 papers in peer-reviewed journals and she is an editorial board member of seven scientific journals.

Abstract:

Hydrogels are water-swollen insoluble polymeric networks, structurally similar to the extracellular matrix of many tissues. We previously reported that hydrogels tailored using particular physiological fibrils of self-assembling peptides (SAPs, e.g. EAK, RGD-EAK, EAbuK) specifically support adhesion and proliferation of osteoblast cells (Acta Biomaterialia, 2011). However, since SAPs hydrogels are characterized by relatively modest moduli (up to 10 kPa), they are hardly applicable as scaffolds for the treatment of bony defects. In the effort to optimize the extracellular matrix mimetic biocompatibility and stiffness, in this study different SAPs at increasing concentrations were electrospun with poly-É›-caprolactone (PCL). By electron microscopy, FT-IR, XPS, and contact angle measurements we found that the percentages of peptides embedded in the scaffolds directly correlates with the diameters of the electrospun fibers and the hydrophilicity of the matrix surfaces. Moreover the enrichment of PCL with SAPs at different concentrations resulted in different trends in tensile modulus, stress and strain. Biological assays reported that matrices characterized by increased hydrophilicity and proper fiber diameter promoted human osteoblast cells adhesion, supported calcium deposition and increased osteoblast-related genes expression. Thus, 10% RGD-EAK motif induced expression of integrin-binding sialoprotein, osteonectin, and runt-related transcription factor 2 mRNA specific transcript levels as determined by quantitative RT-PCR. In conclusion, increasing concentrations of SAPs modulated the physical properties of PCL scaffolds and in particular rectified the softness of SAPs hydrogels.

Speaker
Biography:

Ignazio Castagliuolo is an Associate Professor at the Department of Molecular Medicine, University of Padova. He graduated in 1987 in Medicine. He spent 7 years at Boston University and Harvard Medical Center in Boston performing scientific research in the field of Gastrointestinal inflammation. Then he moved back to Padua were he has performed research mainly focused on cell biology, bioengineering and molecular medicine. He actively collaborates with the Departments of Industrial Engineering of University of Padua to develop innovative scaffolds to support colonization and differentiation of different cell populations. He has published 150 papers in peer-reviewed journals and he is an active reviewer for several scientific journals.

Abstract:

The enteric nervous system (ENS) is the complex network of neurons and glial cells embedded within the gut wall that autonomously regulates most gastrointestinal functions. In the ENS, distinct neuronal subpopulations encode excitatory and inhibitory neurotransmitters. Indeed, interruptions in the neuronal network and derangement in the architecture of the myenteric ganglia have been linked to gastrointestinal dismotility and visceral sensory disorders. Since the extracellular matrix proteins have been long known to provide cues for migration and regeneration of neuronal cells, in this study we designed novel hydrogel scaffolds for intestinal tissue engineering. Peptides mimicking the extracellular matrix of the ENS were synthesized by Fmoc chemistry and differently conjugated. Enteric neurons were isolated from the longitudinal muscle myenteric plexus of young mice and seeded onto different decorated scaffolds. After 5 days on culture, cell adhesion, neuronal marker expression and neurochemical coding were evaluated by confocal microscopy and quantitative RT-PCR. Hydrogels containing laminin adhesive motif greatly promoted adhesion of neuronal cells relatively to other cells of the myenteric ganglia. Arg-Gly-Asp (RGD) motif supported the adhesion of glial cells and also induced expression of mRNA specific for neurotrophin-3, a soluble factor involved in neurogenesis. Ionic-complementary peptide EAbuK fostered axonal outgrowth. Finally, conjugation of RGD and laminin enhanced the expression of choline acetyltransferase, thus inducing cholinergic neurons. In conclusion, our study demonstrated that neuronal subpopulations are modulated by varying the composition of scaffolds. These observations will be helpful in the designed of engineered scaffolds personalized for the treatment of defects of the ENS.

Speaker
Biography:

Valeria Chiono has a Master Degree in Chemical Engineering summa cum laude (2001) and a Ph.D in Chemical and Materials Engineering from the University of Pisa (2006). She is currently associate professor at the Department of Mechanical and Aerospace Engineering, Politecnico di Torino. She is coordinator of the national FIRB Project “Bioartificial materials and biomimetic scaffolds for a stem cells-based therapy for myocardial regeneration” and of the project “Smart Injectable Drug-Delivery systems for Parkinson’s and Alzheimer’s Disease Treatment”. She has collaborated in several national and international project. She is currently author of 40 manuscripts in international journals and 4 patents.

Abstract:

Scaffolds for myocardial Tissue Engineering (TE) should display biomimetic properties respect to cardiac extracellular matrix (ECM), including elastomeric properties [1]. Cardiac regeneration depends on cardiac progenitor cells (CPCs) as well as the milieu in contact with them. Laminin-1 (LN1), typical of developing heart and over-expressed in pathological heart, promotes CPC proliferation and viability [1]. In this work, a thermoplastic polyurethane (PU) was synthesized from poly(ε-caprolactone) diol (Mn = 2000 Da), 1,4-budandiisocyanate and L-lysine ethyl ester dihydrochloride [2]. Bi-layered scaffolds with 0°/90° lay-down pattern were prepared by additive-manufacturing technique [2]. Functionalisation with LN1 or gelatin (G) was performed in two steps: 1) acrylic acid grafting/polymerization by Argon plasma treatment; 2) carbodiimide-mediated coupling of proteins. Scaffolds with mean fibre diameter of 1525 m and mean spacing of 5055 m were prepared. FITR-ATR analysis of protein-coated scaffolds showed higher intensity of the absorption bands at 3370 cm-1 (-OH and –NH stretching) and 1650 cm-1 (amide I). Contact angle decreased from 90° for PU to 60-65° after G- or LN1-grafting. XPS analysis confirmed acrylic acid grafting/polymerization and protein conjugation. Scaffolds were degraded in vitro by lipase (0.3 mg/ml) in 3 weeks. CPC proliferation on PU-LN1 scaffolds was higher than on PU and PU-G scaffolds, increasing from 8,18% on day 7 to 11,8% on day 14. LN1-functionalization stimulated CPC differentiation into cardiomyocytes and endothelial cells.

Biography:

Marzena Zychowicz, in 2012 in the Mossakowski Medical Research Centre, Polish Academy of Sciences, has accomplished her PhD in the field of neural stem cells and bioengineering surfaces. In the recent project she was interested in the standardization of the biomimetic microenvironment for the regenerative medicine application or toxicity testing. She is also involved in the project concerning mesenchymal stem cells and their application in the neurodegenerative diseases. She is co-author of 13 publications.

Abstract:

Induced pluripotent stem (iPS) cells are promising candidates as patient specific cell sources for transplantation or for modeling of diseases. By combining the human iPS-derived neural cells seeded on 3-D collagen scaffolds with dissociated or organotypic neural tissue we are attempting to create a biomimetic microenvironment for studying mutual interactions. To create the microstructured 3D niche, we have used type-I collagen scaffolds. Since collagen scaffolds can exhibit poor mechanical properties and rapid degradation in vivo, depending of cross linking , we have focused on modification of collagen physicochemical properties with improved mechanical and thermal stability without loss of its bioactivity. The human iPS cells, cultured in feeder free conditions, express markers of pluripotency (Oct, Sox2, Nanog), while after neural commitment and differentiation the cells exhibit nestin, GFAP, PDGFRα, β-tubulin III, Map-2, Dcx, GalC. The differentiated population of cells was seeded onto collagen scaffolds and co-cultured with rat organotypic spinal cord (OSC) slices or dissociated dorsal root ganglion (DRG) neurons. The collagen scaffold alone promoted neurite outgrowth from dissociated DRG neurons and when seeded with iPS-derived cells supported migration of cells from the OSC, however no migration of IPS-derived cells from the scaffold to the OSC could be detected. Such a biomimetic environment could be applied in regenerative medicine for testing directed migration from the nerve tissue into the seeded collagen scaffold.

Break: Lunch 12:40-13:30
  • Workshop on New frontiers in hard tissue regeneration: State of art
Location: Rome, Italy
Speaker

Chair

Roberta Di Pietro

G. d’Annunzio University of Chieti-Pescara
Italy

Speaker

Co-Chair

Ivo Kalajzic

University of Connecticut Health Center
USA

Session Introduction

Gianpaolo Papaccio

Seconda Università degli Studi Napoli
Italy

Title: Dental pulp stem cells: State of the art and suggestions for a true translation of research into therapy

Time : 13:30-13:50

Speaker
Biography:

Gianpaolo Papaccio got the degree in Medicine cum Laude in 1979 and the Master Degree in Legal Medicine cum Laude in 1982, University of Naples Federico II, Italy. He worked as visiting scientist in USA- Washington University, Department of Biostructures, Seattle; in Germany, at the Department of Molecular Medicine, J. Liebig University of Giessen; in Switzerland at the Department of Oral Biology, University of Zurich. He currently works at the Second University of Naples as a Full Professor of Medical Histology and Embryology, School of Medicine. He joined the Editorial Board of Stem Cells, PLOs ONE, among others and he’s a Registered Referee for several Journals of Stem cells, Regenerative Medicine, Cell and cancer biology. He is author of more than 150 impacted scientific publications plus international e-book chapters and Italian textbooks.

Abstract:

Stem cells have the ability to rescue and/or repair injured tissue. In humans, it is possible to isolate different types of stem cells from the body. Among these, Dental Pulp Stem Cells (DPSCs) are relatively easily obtainable and exhibit high plasticity and multipotential capabilities. In particular, they represent a gold standard for neural-crest-derived bone reconstruction in humans and can be used for the repair of body defects in low-risk autologous therapeutic strategies. An electronic search was conducted on PubMed databases and supplemented with a manual study of relevant references. All research described in this review highlights that DPSCs are mesenchymal stem cells that could be used in clinical applications. Unfortunately, very few clinical trials have been reported. Major obstacles imposed on researchers are hindering the translation of potentially effective therapies to the clinic. Both researchers and regulatory institutions need to develop a new approach to this problem, drawing up a new policy for Good Manufacturing Practice (GMP) procedures. We strongly suggest that only general rules be standardized rather than everything. Importantly, this would not have an effect on the safety of patients, but may very well affect the results, which cannot be identical for all patients, due to physiological diversity in the biology of each patient. Alternatively, it would be important to study the role of specific molecules that recruit endogenous stem cells for tissue regeneration. In this way, the clinical use of stem cells could be successfully developed. DPSCs are mesenchymal stem cells that differentiate into different tissues, maintain their characteristics after cryopreservation, differentiate into bone-like tissues when loaded on scaffolds in animal models, and regenerate bone in human grafts. In summary, all data reported up to now should encourage the development of clinical procedures using DPSCs. Key words: adult stem cells; bone regeneration; human grafts, scaffold, tissue engineering, stem cell therapy

Speaker
Biography:

Ivo Kalajzic has completed his Ph.D studies on Identification of stages of mesenchymal lineage differentiation. He is currently Associate professor at Dept. of Reconstructive Sciences at University of Connecticut Health Center. His laboratory focuses on studies on regulation of mesenchymal progenitor cell differentiation during bone fracture healing, and the role of notch signaling during repair. Other research projects within his group include the studies on progenitor cells within the muscle, and other tissues, including developing transplantation approaches to treat osteogenesis imperfecta. He has published more than 50 papers with focus on bone biology and orthopedics.

Abstract:

Heterotopic ossification is formation of bone in atypical locations including muscle. Heterotopic ossification generally occurs as a result of aberrant BMP signaling. However, further studies are required to identify BMP-responsive cells in the muscle and understand how they contribute to heterotopic ossification. Alpha smooth muscle actin (αSMA) is a marker of perivascular cells and mesenchymal progenitors that contribute to bone growth and fracture healing. We aimed to evaluate whether αSMA+ cells can contribute to bone formation during heterotopic ossification. To identify and trace cells we used αSMA promoter-driven inducible Cre (αSMACreERT2) combined with a Cre-activated tdTomato reporter Ai9 to generate SMA9 mice. Mature osteoblast/osteocytes were labeled with the Col2.3GFP reporter. To label muscle satellite cells we utilized Pax7CreERT2/Ai9 mice. Pax7Cre/Ai9 labels cells below the basal lamina consistent with a satellite cell phenotype, while SMA9 labels similarly localized cells, in addition to perivascular cells. Pax7Cre/Ai9+ satellite cells are CD45/CD31-, Sca1- but SM/C2.6+. The SMA9+ cells are CD45/CD31- and ~60% express satellite cell marker SM/C2.6. 5% express MSC markers Sca1 and PDGFRα, and a larger proportion express PDGFRβ. During BMP2-induced heterotopic ossification, SMA9+ cells comprised 28% chondrocytes and 44% of Col2.3GFP+ osteoblasts, while Pax7Cre/Ai9+ satellite cells labeled only muscle fibers. Culture of sorted cells indicated that SMA9+ and Pax7Cre/Ai9+ cells differentiated into myotubes while the negative cells do not. SMA9+ cultured cells showed the highest upregulation of osteogenic gene expression after culture in the presence of BMP2. To clarify which subset of SMA9 cells possess osteogenic potential, we subdivided SMA9+ cells based on expression of SM/C2.6 and evaluated their ability to differentiate into osteoblasts after transplantation into muscle with BMP2. We observed osteogenic differentiation of SMA9/SM/C2.6+ cells suggesting that once the satellite cell population is removed from its niche they can exhibit osteogenic potential. In conclusion, αSMACreERT2 labels a population of mesenchymal progenitor cells in the muscle that contribute to heterotopic ossification induced by BMP2. Our data indicate that the perivascular fraction of cells is responsible for heterotopic ossification, however, muscle satellite cells are capable of osteogenic differentiation after removal from their niche. Further study of these cells may indicate mechanisms by which pathogenic heterotopic ossification occurs.

Gunter Lepperdinger

University Salzburg
Austria

Title: Triggering osteocytogenesis in vitro and in vivo

Time : 14:10-14:30

Speaker
Biography:

Gunter Lepperdinger holds a PhD in Biochemistry, is a developmental biologist by training and has worked in Biogerontology with specialization on Stem Cell Ageing since 2002 at ‘The Inst. for Biomedical Aging Research’ in Innsbruck, Austria. He recently moved to Salzburg University to chair the Stem Cell Biology and Healthy Longevity Research Unit. He is Editor to Karger’s ‘Gerontology’, furthermore serving as an Editorial Board Member for many international scientific journals and engaged in European research projects on Aeging (e.g. MOPACT) and Tissue Engineering (e.g. VASCUBone).

Abstract:

Mesenchymal stromal cells (MSC) can be reliably isolated from human connective tissues. Naïve MSC can be culture-expanded and induced to form osteoblasts in a standardized manner. Further differentiation to gain osteocytes is however hard to achieve; also knowledge concerning the molecular mechanisms which specifically trigger and convey osteocyte differentiation is scarce. Through a serendipitous observation while investigating the role of hyaluronan in MSC biology, we were able to unveil molecular mechanisms of osteocytogenesis. These involve specific stress response mechanisms regarding detoxification by means of glucuronidation, upregulation of O-glycosylation which grossly changes the cellular phospho-proteome, induction of stress granule formation and concomitant mRNA-binding thereby yielding differential regulation of translation of osteogenic transcription factors and the formation of poly-ADP–ribose (PAR) in the cytoplasm otherwise known to coordinate the nuclear DNA repair machinery. We could further show in 3D-mesenspheres that provoking this particular stress response pathway in the presence of osteogenic inducers promotes the calcification of MSC-derived microorganoids resulting in the formation of trabecular-like hard tissue structure bearing osteoblastic linings which engulf marrow-like stroma. These observations could be further corroborated in histological sections of human bone in particular showing the presence of stress granule and PAR deposition in osteocytes residing in compact bone. We could further show accelerated healing of a bone defect model in rat calvaria by provoking this stress response pathway.

Roberta Di Pietro

G. d’Annunzio University of Chieti-Pescara
Italy

Title: Potential of human amniotic fluid stem cells (AFSC) and natural scaffolds in bone regeneration and repair

Time : 14:30-14:50

Speaker
Biography:

Roberta Di Pietro got the degree in Medicine cum Laude in 1985 and the PhD in Sports Medicine cum Laude in 1988, University of Chieti, Italy. She worked as a visiting scientist in UK at the Biochemistry Department, AFRC, Cambridge; in USA at the Pathology Department, USUHS, Bethesda, and at the Institute of Human Virology, University of Maryland, Baltimore. She currently works at the University of Chieti as a Full Professor of Histology. She joined the Editorial Board of Current Pharmaceutical Design as an Executive Guest Editor and she was recognized as a Registered Referee for Archives of Ophthalmological Reviews and Reproductive Biology and Endocrinology. She is now author of 171 scientific publications plus international e-book chapters and Italian textbooks.

Abstract:

Every year over 2 millions bone replacement procedures requiring the use of bone graft materials are performed worldwide. This makes bone second only to blood in the list of transplanted materials. Tissue engineering strategies are a very promising option for the healing of critical-size bone defects. To this aim, various classes of natural scaffolds (including collagen and hyaluronic acid-based hydrogel) have been used in the human clinical setting. However, cell-free scaffolds are inadequate to repair large bone defects. Recent studies have focused on the use of stem cells rather than mature differentiated cell types due to their expansion potential and ease of access. In particular, human AFSCs represent an attractive cell model for transplantation therapy due to the lack of significant immunogenicity, tumorigenicity and ethical issues. Although AFSCs have been investigated for bone repair, the cellular distribution and post-implantation viability remain key issues. Our research group explored whether AFSCs could improve bone healing in a rat model of large bone defect. For in vivo experiments male rats were injured at the femoral diaphysis in order to produce a 5-7 mm diameter full thickness bone gap. Human AFSCs were transplanted in vivo at the lesion sites after being loaded on a HA (natural nanocrystalline carbonated hydroxyapatite-Orthoss®) scaffold. Both gross anatomy and histological observations revealed greater bone tissue remodelling and regenerative response in rat specimens treated with HA scaffold supplemented with AFSCs. Our findings support previous studies showing AFSCs angiogenic effects and indicate the potential of these cells in bone reconstruction.

  • Track 8: Novel Approaches in Guided Tissue Regeneration
Location: Rome, Italy
Speaker

Chair

Aysegul Batioglu-Karaaltin

Istanbul University Cerrahpasa School of Medicine
Turkey

Speaker

Co-Chair

Livia Visai

University of Pavia
Italy

Speaker
Biography:

Massimiliano Colombo has completed Medicine and Surgery at University of Milan and specialization in School in Orthopaedics and Traumatology at University of Milan. He is at present working as orthopaedic surgeon and researcher at unit of Orthopaedic Reparative Surgery and Risk Management, G. Pini Orthopaedic Institute, piazza Cardinal Ferrari 1, University of Milan. He has good knowledge of the problems of bone tissue as regards its regeneration and healing processes. He performed numerous national and international publications on the topic of bone necrosis, delayed consolidation of the fractures, non union, bone defects with particular interest in the field of biotechnology, such as growth factors, bone substitutes and mesenchymal stem cells. He produced: 3 medical scientific monographs; 43 orthopaedic scientific publications in Italian and foreign journals, including many with relevant impact-factor and multiple citations; 17 scientific posters presented at conferences. He performed an economy drug study on the use of growth factors in traumatology entitled "Comparative analysis of costs and cost/effectiveness ratio of the treatment of persistent post-traumatic non-unions of the tibia with autologous transplant of the Iliac Crest or Osigraft® (Eptotermin alfa, rhBMP-7,rhOP-1)”. He is performing a study about the effectiveness of a new system for the insertion of distal screws in the implant of long femoral nails. He recently completed a comparative study on the efficacy of growth factors (rh-BMP-7) with respect to autologous bone graft from the iliac crest in the treatment of 150 nonunions and loss of substance of the long bones.

Abstract:

Objective: to determinate the efficacy of core decompression technique with the use of recombinant morphogenetic proteins, autologous mesenchimal stem cells (MSCs) and xenograft bone substitute into the necrotic lesion of the femoral head on clinical symptoms and on the progression of osteonecrosis of the femoral head. Methods: we studied 38 patients and 40 hips with early stages of osteonecrosis of the femoral head. Results: Core decompression technique with the use of recombinant morphogenetic proteins, autologous MSCs and xenograft bone substitute afforded a significant reduction in pain and in joint symptoms and reduced the incidence of fractural stages. At 34 months, 33 patient reach the clinical and radiographic healing. Conclusion. This long term follow-up study confirmed that core decompression technique with the use of recombinant morphogenetic proteins, autologous MSCs and xenograft bone substitute might be an effective treatment for patients with early stages of osteonecrosis of the femoral head.

Speaker
Biography:

Visai received the Ph.D title in Biochemistry in 1989 and She is presently an Assistant Professor in Biochemistry at the Medicine Faculty of Pavia University. Her scientific experience were performed abroad initially at the Connective Tissue Laboratory, Alabama University in Birmingham (USA) and then to the Center for Infectious and Inflammatory Diseases (IBT), Houston University in Texas (USA).She has been recently a Visiting Professor at the Department of Nanomedicine and Nanotechnology of the Methodist Hospital in Houston, Texas, (USA). She is the vice-director of the Interdepartmental Center for Tissue Engineering in Pavia University and the Head of the Nanotechnology Laboratory at Salvatore Maugeri Foundation in Pavia. She has published more than 129 papers in reputed journals and has been serving as an editorial board member of repute.

Abstract:

Recent advances in tissue engineering and regenerative medicine have shown that controlling cells micro-environment during growth is a key element to the development of successful therapeutic system. Various polymeric scaffolds have been used to support cellular growth and, to a certain extent, favor cell organization and tissue structure. A large pool of stem cell lines such as multipotent and pluripotent stem cells lines are available for these type of studies. Furthermore, the exposure of stem cells to biophysical stimuli has been reported to favor early and rapid activation of the tissue repair process. But all of these approaches has appeared to be rather limited since they do not offer the fine control of the cell micro-environment in space and time. We will be presenting the biological effects exerted by electrospun nanocomposite materials on tuning the cell fate of human multipotent and pluripotent stem cells. Then, we will continue by presenting the effects related to the exposure of human multipotent stem cells to Pulsed Electromagnetic Field (PEMF) or Low Level Laser Intensity (LLLI) on cell proliferation and differentiation towards osteoblasts. At the end, we will show the recent data obtained in the design of 3D scaffolds based on hydrogels as well as polymeric fibers to maintain the self- renewal of human pluripotent stem cells in feeder-free conditions.

Speaker
Biography:

Nelson R. Pinto: University of the Andes, Faculty of Dentistry, Santiago, CHILE. Visiting Professor, Department of Oral Health Sciences / Periodontology , University Hospitals, Catholic University Leuven, BELGIUM. Founder and Chairman of the Research Center of Regenerative Medicine and Tissue Engineering , Concepción, CHILE. Clinical Editor POSEIDO JOURNAL. World Leader on Clinical Applications of L-PRF in Hard and Soft Tissue Regeneration and Wound Healing. Developer of the Technique for the Treatment of Chronic Wounds with L-PRF.(Best Oral Research Presentation : 4th Congress of the World Union of Wound Healing Societies, Yokohama, Japan, 2012) National and International Lecturer on Implant Dentistry and Regenerative Medicine. (+200).

Abstract:

Leukocyte- and Platelet-Rich Fibrin (L-PRF) is one of the four main families of platelet concentrates for surgical use. L-PRF is used to improve healing and promote tissue regeneration. Blood sample is taken without anticoagulant and immediately centrifuged to obtain a L-PRF clot that can be used directly or compressed into a membrane without damaging the cells and growth factors content. The L-PRF clot or membrane contains most of the platelets and leukocytes present in the initial blood harvest plus the platelet growth factors and stem cells that are also trapped within the fibrin network. With this architecture, L-PRF is the source of a strong and slow release of growth factors such as TGF-b1, VEGF, IGF-1, FGF, EGF, PDGF-AB, IL-1ß for more than three weeks in vitro. Through the release of these growth factors trapped within the fibrin gel or through the production of new molecules by the leukocytes, the L-PRF membranes have strong effects on the stimulation of the proliferation of most cell types (fibroblasts, keratinocytes, pre-adipocytes, osteoblasts, bone mesenchymal stem cells) and on the differentiation of the bone cells.The possibility to use L-PRF as a biological scaffold by itself or associate with a biomimetic implant surface as open the opportunity to regenerate soft and hard tissue in such a way that was not possible before. The clinical, immunohistochemestry and histological findings (SEM, Confocal Laser, and Optical Microscopy) of our animals and humans studies over the last 14 years confirm the potential of L-PRF as a biological scaffold for hard and soft tissue regeneration in acute or chronic wounds. What we thought impossible yesterday, could be routine tomorrow… “Natural Guided Regeneration with L-PRF”.

Speaker
Biography:

Aysegul Batioglu-Karaaltin has completed her MD degree at the age of 24 years from Hacettepe University and her residency at the Department of ENT and Head and Neck at Ministry of Health Education and Research Hospital. She is working at Department of Otolaryngology Head and Neck Surgery, Istanbul University Cerrahpasa School of Medicine. She is carrying out more then 10 research about regenerative medicine and tissue engineering in the Head and Neck field. She has published more than 15 papers in reputed journals

Abstract:

Tracheal transplantation appears promising, especially for the patients who have been followed in intensive care units for a long time and who have received recurrent operations after traffic accidents or malignancies and, as a result, have to live with a tracheostomy for the remainder of their lives because of the diagnosis of tracheal stenosis. The treatment of tracheal defects longer than 6 cm (approximately half of the total length of trachea) is still controversial. Autograft materials, cadaveric tracheal allotransplantations, vascularized allotransplantation, cadaveric trachea decellularisation were tried. Cadaveric trachea decellularisation followed by seeding with autologous stem cells of the recipient is the best method with no need to use immunosuppressive medication, no requirement for the second major surgical procedure. Successful human transplantations have also been performed using this technique with bone marrow-derived MSCs. We published first successful in vivo tissue engineered trachea regeneration from a decellularized cadaveric trachea matrix with seeded adult adipose tissue-derived MSCs which was integrated into the recipient tracheal sides. It is required to protect the three dimensional structure of the trachea and its extracellular matrix during decellularisation process. The previous studies mostly employed detergent enzymatic method (DEM) and its modifications for tracheal decellularisation. Although ionic detergents used for trachea decellularisation are very effective in removing cellular remnants, they damage the natural structure of the tissue by affecting the integration of the extracellular matrix. We used combined decellularisation method called protected matrix decellularisation (PMD) which was developed by our group. And we control test of decellularisation both for rabbit and human cadaveric tracheas. As a result new protected matrix decellularisation followed by seeding with autologous adipose tissue-derived MSCs which have same origine with chondrocytes and chondrogenic differansiation is known as higher than other stem cells is promising for trakeal reconstruction.

Speaker
Biography:

Srećko Gajović is Head of Histology and Embryology at University of Zagreb School of Medicine. He is Coordinator of FP7 REGPOT project GlowBrain dedicated to unlock the research potential of Croatian Institute for Brain Research through stem cell and biomaterial applications in the mouse stroke model. He is Editor-in-Chief of Croatian Medical Journal, best general medical scientific journal in this part of Europe, and ex-Chair of European COST Domain Committee for Biomedicine and Molecular Biosciences. He was born in Zagreb, Croatia, where he completed his studies of medicine and get a PhD at University of Zagreb. His postdoctoral positions were at Max Planck Institute for Biophysical Chemistry in Göttingen, Germany, and at Institute for Genetic Engineering and Biotechnology in Trieste, Italy. In his current research, he tries to understand the role of innate immunity in brain repair after stroke, and to develop tissue engineering strategies by combining stem cells and biomaterials for the brain repair.

Abstract:

To achieve the beneficial response and brain regeneration after stroke one of the possibilities is to combine stem cells and biomaterials and to monitor their effects in the living animals. This is the main aim of the FP7 REGPOT project GlowBrain, and for this purpose several in vivo small animal imaging modalities are used, which includes magnetic resonance imaging (MRI) and light based imaging modalities - bioluminescence imaging (BLI), fluorescence imaging (FLI) and Cherenkov luminescence imaging (CLI). To visualise the molecular events related to the activity of TLR2, GAP43, and CASP3 in the brain of the living mouse the maging was performed by IVIS Spectrum Pre-clinical In Vivo Imaging System (Perkin Elmer, US) in genetically modified animals carring luciferase reporter TLR2, GAP43, and CASP3 were upregulated after stroke. To analyze the apoptosis in subset of GAP43 cells the imaging with DEVD-aminoluciferin was performed. The findings suggested that CASP3 activity, not necessarily associated with neuronal apoptosis, increased, and CASP3 and GAP43 might be part of a common molecular pathway involved in early stress response after stroke. This added neuronal stress in addition to inflammation, repair, and apoptosis as important process to be assessed by the bioluminescent imaging as the brain response to stroke. The same strategy of in vivo insight in brain molecular events was applied as well to monitor the biocompatibility of brain applications of the stem cells and biomaterials. The combination of neural stem cells and the polysaccharide biomaterial alginate was designed. The in vivo imaging confirmed that the inflammation after brain transplantation was comparable to the control injection by PBS, proving the biocompatibility of the procedure. The described experimental approach opens the way to design and preclinical testing of innovative therapies for brain diseases.

Break: Coffee 16:30-16:45
  • Special Session on Developments, innovations and breakthroughs in regenerative medicine
Location: Rome, Italy
Speaker

Chair

Jonathan Schwartz

Marvel Medicals
USA

Session Introduction

Joseph Purita

Institute of Regenerative and Molecular Orthopedics
USA

Title: Lipogems a game changer in the field of re-generative stem cell therapy

Time : 16:45-17:05

Speaker
Biography:

Purita is Director of Institute of Regenerative and Molecular Orthopedics (www.stemcellorthopedic.com) in Boca Raton, Florida. The Institute specializes in the use of Stem Cells and Platelet Rich Plasma injections. Dr. Purita is a pioneer in the use of Stem Cells and Platelet Rich Plasma. The Institute has treated some of the most prominent professional athletes from all major sports in both the U.S.A. and abroad. He received a B.S. and MD degree from Georgetown Univ. Dr. Purita is board certified in Orthopedics by ABOS. He is a Fellow American College of Surgeons, Fellow American Academy Orthopedic Surgeons, and a Fellow American Academy of Pain Management. He is also certified in Age Management Medicine. He has lectured and taught extensively throughout the world on the use of Stem Cells and Platelet Rich Plasma. He has been instrumental in helping other countries in the world establish guidelines for the use of Stem Cells in their countries. He has been invited to lecture on these techniques throughout the world as a visiting professor.

Abstract:

The use of adipose tissue has taken on increasing im-portance in the field of regenerative stem cell therapy. This therapy can be divided into two major pathways. One path-way involves plastic and reconstruction surgery. The other major pathway involves orthopedics and its related fields. Other uses are being found in gynecology, urology, and general surgery. Until recently the main uses of adipose tissue centered around the use of enzymatic produced SVF suspensions. Typically these suspensions were made with the use of the enzyme collagenase. Although collagenase has worked well in the past there is now a complaint method of producing a fast, enzyme free lipoaspirate that is turned into a therapeutic product. This new method comes from the Lipogems company. The Lipogems technique has been utilized in over 5,000 patients. It has recently obtained FDA clearance. Lipogems makes use of the high stem cell density of adipose tissue thought to be 1 out of every 100 cells. Lipogems utilizes mild mechanical forces in a closed system to produce micro fractured and purified adipose tissue graft which is non ex-panded and ready to use in a variety of regenerative appli-cations. More importantly, Lipogems adipose product has a very well preserved vascular stroma with slit-like capillaries wedged between adipocytes and and vascular channels. The stem cell niche is preserved enabling the pericytes to have a much higher survivability rate. The intact niche pro-vides a perfect environment for reparative response of acti-vated MSCs. As Dr. A. Caplan has pointed out, pericytes are precursor mesenchymal stem cells. Activated MSCs are crucial for the immune modulation and regeneration. The lipoaspirate of Lipogems shows a higher percentage of ma-ture pericytes and hMSCs than enzymatically digested lipoaspirates. We are now aware that there are novel growth factors secreted by Lipogems lipoaspirate which may explain the therapeutic efficacy of Lipogems. Also im-portant, is the fact that the Lipogems technique may pre-serve the important MUSE cell. Muse cells are unique in that they be pluripotent, have a high survivability rate, and are quite small by stem cell standards. They are considered to be an embryonic like stem cell without any of the risks of regular embryonic stem cells since they are the patient's own. They may represent a quantum leap in regenerative medicine. Since Lipogems preserves these cells it too is a quantum leap.

Speaker
Biography:

Michael Weber has completed his studies in chemistry and biochemistry at University Marburg and at University Goettingen he completed his PhD during 1983. He was researcher at Max-Planck-Institute for Experimentel Medicine in Göttingen. He is the Leader of 3 medical laser centers in Germany and Bangkok in Thailand. He is the President of the International Society for Medical Laser Applications (ISLA) and also Board Member of the North American Association for Laser Therapy (NAALT). He is author of many international publications in the field of Medical Laser Therapy. He established his own company (weber medical GmbH) in 2003 supported by the German government and the EU.

Abstract:

In this lecture new methods of laser therapy will be presented covering nearly the whole range of important diseases in medicine. Laserneedle acupuncture with different wavelengths and penetration depths can be used as a highly effective and pain free method for all indications of acupuncture and pain management. Laserneedles can be applied on the body, on the skull or the ear as well. Infrared, red and yellow lasers can penetrate the skull bone and used today for benefit on brain diseases. Intravenous laser therapy is a systemic application of laser light with infrared, red, green, blue and yellow wavelengths. This method stimulates the immune system, leads to an improved microcirculation and oxygen supply and improves endurance in sports people. Furthermore all wavelenghts stimulate the different complexes of the respiratory chain in the mitochondria leading to an increase of ATP-production and thus are well suited for all fields of regenerative medicine in mitochondrial degeneration. Especially blue laser disscociates nitric oxide from haemoglobin and complex IV in the mitochondria and so leads to improved microcirculation, biogenesis of mitochondria and prevents cell senescense. Nitric oxide also activates telomerase and so protects against degradation of telomers with possibly extended life expectation. We also know today that intravenous laser therapy can activate endogenous stem cells with improvement of organ function.Main therapeutic fields are angiopathies, neuropathies, fibromyalgia and chronic fatigue syndrome, autoimmune and metabolic diseases, depression, all diseases on basis of mitochondrial degeneration and antiaging in general. Interstitial laser therapy is a new method using a fiberoptic catheters for application of different lasers in the depth of the tissue close to the spot of injury. It’s a new method for treatment and regeneration of herniated disks and other nerve injuries. For intraarticular laser therapy a fiberoptic laser catheter will be placed directly in a joint and is a highly effective treatment of advanced osteoarthritis of the knee, shoulder and other joints. All laser colors including non penetrating blue laser can be used successfully. This therapy can be combined with injection of PRP or mesenchymal stem cells with light activation for achieving regenerative effects on the cartilage. New therapeutic approaches for photodynamic cancer therapy are presented as well using the different laser colors externally and interstitially with new photosensitizers for all type of cancers.

Speaker
Biography:

Fabio Valerio Sciarretta attended the Liceo Classico " De Sanctis, " earning a Classic baccalaureate diploma in 1982. In the same year he joined the Faculty ' of Medicine and Surgery of the University of Rome "La Sapienza" . From January 1990 to April 1991, he fulfilled his military obligations in quality ' of Lieutenant physician at the Military Hospital of Cagliari with the post of " Assistant of the Orthopaedics Department " reporting the following final judgment : "Officer with outstanding moral and character accomplishment as well as ' comprehensive and updated professional preparation. He has carried out the task to him entrusted with zeal and high sense of duty and responsibility' providing a valuable contribution to the smooth running of the Department . " During this service , the 7/11/90 he has obtained a written note of satisfaction of the Director of Health ' Military Region of Sardinia for " the results obtained by acting with great professional expertise to a qualified orthopedic surgery ." He is affiliate Member of the American Academy of Orthopaedic Surgeons ( AAOS ), International Society of Arthroscopy , Knee Surgery & Orthopaedic Sports Medicine ( ISAKOS ), European Society of Sports Traumatology Knee Surgery and Arthroscopy ( ESSKA ), International cartilage Repair Society ( ICRS ) also member of the Italian Society of Orthopaedics and Traumatology ( SIOT ) , Society of Surgery of the knee, arthroscopy , sports medicine, cartilage , orthopedic technologies ( SIGASCOT ),member of the Italian Society of Arthroscopy ( SIA). He has been the editor of many Italian edition journals. From 1994 to 2000 he served as director of the first level of health care at the Hospital of St. John the Baptist in Rome of the Association of Italian Knights of the Sovereign Military Order of Malta. Since September 2000 , as a senior health care executive for a specified period from 2000 to 2007 and as a senior health care executive for an indefinite period , he served at the Department of Orthopaedics and Traumatology Hospital of Velletri (ASL RMH ) . Currently practices his profession at the among in the private clinics of Rome

Abstract:

While performing an arthroscopy of the knee, cartilage injuries, of any grade, have a whole reported frequency between 57.3% and 66% of cases. These numbers make it clear that, more and more, young adults under the age of 65 years, present with an advanced disease to joint cartilage. in recent years, have increasingly found their way the regenerative medicine therapies, which allow to correct alterations of the biological basis of the disease's process, rather than addressing only the symptoms. Orthobiologics are based on the use of mesenchymal stem cells that can mainly be obtained from bone marrow or adipose tissue. Since no surgical technique has still been proven to be the gold standard of chondral repair, in order to increase the results of the recently developed single stage chondral repair techniques delivering to defect site a larger number of multipotent cells to improve defect filling and repair, the augmented single stage procedures have been introduced. These techniques represent an evolution of the standard AMIC procedure, where a collagen membrane is used to repair and resurface chondral defects. In the augmented techniques the use of PRP or bone marrow concentrate is added to the collagen membrane. In this paper we present the initial experience with a new system for bone marrow aspiration and concentration, perfectly adapting to the timing of one step cartilage repair AMIC technique. The surgical technique steps are: chondral defect debridement, bone marrow iliac crest aspiration through power driven aspiration cannula, defect's sizing through aluminum template use, microfractures, collagen membrane sizing and soaking with bone marrow concentrate obtained through completely closed centrifugation and, finally, closure of the chondral defect with collagen membrane, bone marrow concentrate and fibrin glue. The surgical technique is safe, lasts 15-20 minutes, is completed all inside the OR and doesn't need cellular expansion. Randomized controlled clinical trials will confirm the results.

Al Sears

Palm Beach Institute for Anti-Aging Medicine
USA

Title: Cracking the telomere code: The first step to reverse aging

Time : 17:45-18:05

Speaker
Biography:

Al Sears, M.D. is the founder of the Center for Health and Wellness, a successful integrative medicine and anti-aging facility in Royal Palm Beach, Florida, with over 25,000 patients. His cutting-edge therapies and reputation for solving some of the most difficult-to-diagnose cases attract patients from around the world. Sears was one of the first to be board-certified in anti-aging medicine. As a pioneer in this new field of medicine, he is an avid researcher, published author, and enthusiastic lecturer.Sears is board-certified as a clinical nutrition specialist and a member of the American College of Sports Medicine (ACSM), the American College for the Advancement in Medicine (ACAM), the American Medical Association (AMA), the Southern Medical Association (SMA), the American Academy of Anti-Aging Medicine (A4M), and the Herb Research Foundation, (HRF). Dr. Sears is also an ACE-certified fitness trainer.Sears currently writes and publishes the monthly e-Newsletter, Health Confidential, and daily email broadcast, Doctor’s House Call, and contributes to a host of other publications in the field. He has appeared on over 50 national radio programs, ABC News, CNN, and ESPN.Since 1999, Dr. Sears has published 15 books and reports on health and wellness with a readership of millions spread over 163 countries.

Abstract:

This lecture explores the discovery of the telomerase enzyme and its role as a crucial indicator of health and longevity. By tracing the evolution of telomere research, including the landmark Harvard study that fully restored youthful function in mice by activating the telomerase enzyme, Dr. Sears unveils the true age-reversing, and health-transforming potential of telomere therapy and the capacity to influence gene expression through novel interventions. As one of the only medical doctors to administer the world’s first telomere therapy, Dr. Sears relates its effects on his own patients, and how his own research uncovered new, more effective ways of supporting telomere length and altering telomere biology. Three learner objectives. By the end of the lecture, participants will understand: • The evidence that supports the telomere’s role as the true cause of aging. • What factors cause the telomere to shorten, and the most reliable interventions to support telomere length. • How the search for telomerase activators uncovered new, more powerful and more affordable means of affecting telomere regulation.

  • Young Research Forum
Location: Rome, Italy
Speaker

Chair

Karin Schütze

CellTool GmbH
Germany

Biography:

Nihal AlMuraikhi is a Ph.D candidate expected to be awarded by the end of this year from Imperial College London. She is a lecturer at the Stem Cell Unit of the Medical School of King Saud University, Riyadh, Saudi Arabia.

Abstract:

Current protocols used in the differentiation of human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) toward erythropoiesis utilize two main approaches; Embryoid Body (EB) formation, which influences heterogeneity of the produced population, and/or co-culture with mouse stromal cells, which makes the xeno-free culture requirement difficult to achieve. In this study, we designed a serum-free experiment on the direct differentiation of hiPSCs toward erythroid cells under hypoxia condition bypassing the EB formation step and no co-culture system. Our protocol involves three phases: phase-I) Hematopoietic induction (7days), where cells were exposed to specific cytokines to trigger the mesodermal formation. Phase-II) Erythroid differentiation (7days) with reduced number of cytokines; EPO, SCF and IL-3; and finally, phase-III) Maturation of erythroid progenitors (14days) by exposing cells to EPO only. All culture phases were performed in hypoxia conditions. Phase-I showed an expression of an early hematopoietic marker, CD34 in parallel to a high expression of CD45, a pan leukocyte marker. Both markers peaked by phase-II then decreased gradually during maturation. Phase-I also showed a high expression of an early erythroid marker, CD71 and a late erythroid marker, CD235a over the culture period. Around 10% of the cells were enucleated erythrocytes by the end of phase-III, which was confirmed by Wright-Giemsa and reticulocytes staining. Functional analysis using CFU showed that the cell population were able to form hematopoietic colonies, specifically, erythroid progenitor. Further studies on maturation and enucleation are required in order to achieve fully mature and functional RBCs phenotype.

Speaker
Biography:

Physical and chemistry graduated teacher, Céline Aubrun is preparing a PhD in mechanical engineering at the Équipe Biomatériaux Artois of the Laboratoire génie civil et géoenvironnement (Université d’Artois). She specialized in biomaterials in the field of bone acrylic cements.

Abstract:

Cements are used in bone surgery to fill gaps issue from pathology or traumatology. We classically distinguish calcium phosphate cements with hydraulic setting and acrylic cements with polymeric setting, mainly formulated with polymethylmethacrylate (PMMA) and possibly include a biocompatible inorganic filler. We studied a cement based on 2-hydroxyethylmethacrylate (HEMA), natural polysaccharides and a bioceramic filler (tricalcium phosphate or hemihydrated calcium sulphate, TCP and plaster, respectively) developed for its advantages: hydrophilicity, biodegradability and mechanical characteristics. It aims filling bone defects with analgesic and support functions. The main interest substituting HEMA to PMMA is due to an in situ quick water-uptake when contact with biological fluids. Induced swelling ensures gap total filling by preventing formation of fibrous tissue. However a deterioration of mechanical properties occurs. Our study so aimed improving formulation of cement in order to limit mechanical properties loss while maintaining its water-uptake ability. Two bioceramic fillers were compared. Studied curing parameters were temperature increase and setting time. Mechanical properties andwater-uptake were measured initially and after 21 days in simulated body fluid (SBF). The highest temperature ranges from 50°C to 80°C and setting time from 1min30s to 4min30s. We found that plaster significantly improved mechanical properties. After 21 days in SBF cement Young modulus was 13.6MPa for plaster and 1.4MPa for TCP (initially 470MPa and 362MPa, respectively), while maintaining equivalent water-uptake (32%). These results open the way to a cement formulation allowing curing parameters and in situbehavior improvement, so that we can consider its use as bone substitute.

Biography:

Aysu Arslan has completed her B.Sc. degree from Hacettepe University, Department of Chemical Engineering. Currently she is M.Sc. student at the same department and she is a member of Hacettepe University Cell and Tissue Engineering Research Group, being coordinated by Prof. Dr. MenemÅŸe GümüÅŸderelioÄŸlu. She has published 1 paper in a related journal.

Abstract:

Polybutyleneadipate-co-terephthalate (PBAT) is an aliphatic-aromatic copolyester based on the monomers 1,4-butanediol, adipic acid and terephthalic acid. Recently, it has been considered as a promising biomaterial for tissue engineering applications due to its non-toxicity, desired physical properties and complete biodegradability under enzymatic conditions. However, existing studies are limited to cytotoxicity and basic cell proliferation assays with non-functional PBAT membranes which is not enough to understand PBAT’s behaviour as scaffolds for tissue regenaration. In this study, 2D and 3D porous PBAT scaffolds were fabricated via solvent casting, electrospinning, solvent casting-particulate leaching and melt molding-particulate leaching methods. Fabricated scaffolds were characterized due to their morphologies, mechanical properties, water absorption capacities, wettabilities and pre-osteoblastic cell promotion in order to estimate its potential for tissue regeneration. PBAT scaffolds were successfully fabricated in various structural forms with up to 90% porosities. Attachment and proliferation of MC3T3-E1 pre-osteoblasts on PBAT surfaces was observed with scanning electron microscope (SEM) on the 4th, 24th hours, 3rd and 11th day of cell culture. Results show that cells favoured the neat PBAT surfaces in all fabricated structural forms. At the 11th day of the cell culture, PBAT surfaces were covered by cells and extracellular matrix secreted by the cells. As a result, its easy processability, good mechanical properties, biocompatibility and well-known biodegradability makes it a powerful candidate for tissue engineering applications. In further studies, tissue engineering potentials of neat and modified 3D electrospun PBAT scaffolds will be investigated.

Alena.O. Stepanova

Meshalkin Novosibirsk State Research Institute of Circulation Pathology
Russia

Title: Influence of electron beam irradiation onto physical and chemical properties of electrospun produced 3D matrices

Time : 18:50-19:05

Speaker
Biography:

Alena Stepanova has graduated from the Novosibirsk State University with master’s degree in molecular biology. She did the research for her master’s degree at the Molecular medicine Laboratory at the Institute of chemical biology and fundamental medicine Russian academy of Science in Novosibirsk. She is working at the same department the PhD Studies with the project “The study fundamentals of the vascular prosthesis fabricated by electrospinning in vitro and in vivo”.

Abstract:

Electron beam irradiation (EBI) is widely used in industrial production for sterilization of medical products. We have evaluated the influence of EBI onto mechanical strength, chemical properties and biocompatibility of electrospun produced 3D matrices (EPM). Sheets of EPM with oriented fibers (fiber diameter 1µm, scaffold thickness 100 µm) or tubes (inner diameter 1.7 mm, wall thickness 100 µm) were prepared from nylon 6, polylactic-co-glycolic acid, polycaprolactone (PCL) and their mixtures with gelatin. Electron accelerator ILU-6 (2.2 MeV, 400 mА, 10 Hz) was used for EBI of the materials in doses 25÷150 kGy with increment 25 kGy. Mechanical strength/structure were tested using Zwick/Roell Z100 testing machine/JSM-6460 LV scanning electron microscopy and differential scanning calorimetry. Human primary umbilical vein endothelial cells (HUVEC) and gingival fibroblasts (HGF) were used to evaluate adhesion and proliferation of cells at the surface of EPM. It was observed, EPM irradiation in a dose of 25 kGy does not affect the mechanic properties of all matrices studied. Irradiation of EPM with a dose higher than 50 kGy leads to embrittlement of all matrixes except from those produced from PCL. A dose of 100 kGy increases the proportional limit of the PCL scaffold, significantly increase adsorption of the protein on the surface of PCL fibers and allowed to introduce durable regions by irradiating EP tubes through the template with open areas. EBI of the matrices does not interfere with the capacity of EPM to support adhesion, viability and proliferation rate of HUVEC and HGF at the surfaces of EPM as it was shown by cells labeling with calcein/propidium iodine and incorporation of ethylenedioxy uridine. Thereby an electron beam irradiation of EPM was show to be a useful instrument to modify mechanic/chemical properties of EPM.

Speaker
Biography:

Shima Salmasi is a PhD student and research associate at the Centre of Nanotechnology and Regenerative Medicine, University College London. She completed her Master of Science in Nanotechnology & Regenerative Medicine and now, as part of her PhD, is researching on an innovative translational project with the aim of improving the current standards of spinal fusion surgery. This is a very interesting project with great potentials to provide a clinically effective solution to overcome the shortcomings of the currently available techniques of spinal fusion surgery using tissue engineering and nanotechnology.

Abstract:

Polyhedral oligomeric silsesquioxane poly(carbonate-urea) urethane (POSS-PCU), a new breed of novel nanocomposite material developed by researchers at the University College London has been extensively tested in terms of its physiochemical, in vitro, and in vivo properties, all of which have shown that its enhanced biocompatibility, superior mechanical engineering properties, and augmented degradative resistance renders POSS-PCU capable of functioning as a scaffold for bioartificial organs, nanoparticles for biomedical applications, and a coating for medical devices. The novelty of POSS-PCU further arises from the fact that it has been used in 3 first-in-human studies as a bypass graft, lacrimal duct, and the world's first synthetic trachea. Bone, as a living tissue, has the ability to constantly remodel, renew and regenerate to repair itself. However, large bone defects are considered as major problem for the clinician and society. Recently, we have investigated incorporation of different ratios of nanohydroxyapatite, the main inorganic component of the natural bone, into POSS-PCU nanocomposite for bone tissue engineering applications. This nanocomposite material could be used to better mimic the mineral component and the micro- & nano-structure of the natural bone and so far we have found promising results in terms of the scaffold supporting and enhancing material-cell interaction; i.e. attachment, proliferation and bone mineralisation of oestrogenic cell lines as well as osteoinductive properties for bone deposition and regeneration.