Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 5th International Conference on Tissue Engineering and Regenerative Medicine Berlin, Germany.

Day :

  • Tissue Engineering | Tissue Repair & Regeneration
Location: Kiel 1-3

Chair

Barritault Denis

OTR3, France

Co-Chair

Pedro Morouço

Polytechnic Institute of Leiria, Portugal

Biography:

Abstract:

PIF Premise: Endogenously secreted only by viable embryos, PIF has determining immune-modulatory and transplant acceptance functions throughout pregnancy–a “perfect” regulatory scenario. Synthetic PIF replicates native peptide functions in representative preclinical/clinical non-pregnant setting. Specifically, PIF comprehensively addresses neuro-repair locally and systemically.PIF Effects: PIF exerts autotrophic effects on native stem cells. In newborn/early pregnancy PIF has neurotrophic/protective effects; the notochord initiates embryogenesis. In prematurity and multi-injury-induced brain damage (HIE), PIF reverses corpus striatum and cortex damage. In adult multiple sclerosis, (EAE) PIF promotes re-myelination while reversing chronic paralysis up-to-total resolution.PIF Mechanism of Action: Mechanistically, PIF crosses intact BBB targeting brain and spinal cord microglia, neurons and blood vessels. In HIE PIF reduces let-7 microRNA, PKC/PKA phosphorylation while increasing IL10. In adult EAE, PIF regulates brain phosphoproteins and spinal cord proteome to reduce oxidative stress and protein misfolding while promoting Na+/K+/Ca++, glucose and amino acid transport. Thus PIF promotes neuron assembly and synaptic transmission while reducing systemic inflammation. PIF promotes endogenous brain stem cells proliferation and differentiation. The effect of PIF is superior to and potentiates intra-cranially injected stem cells.Conclusion: PIF is in Fast-Track FDA Phase Ib clinical trial (NCT#02239562). Collectively, PIF high-safety and comprehensive effect on CNS and peripheral nervous system coupled with intimate interaction with endogenous stem cells support PIF’s planned phase II clinical trial for neuro-repair.

Biography:

Anne des Rieux has completed his PhD in 2006 from Université Catholique de Louvain (UCL), Belgium and preformed her Post-doctoral study at the Northwestern University, Chicago. She is an Associate Professor at UCL and is developing a team within the Louvain Drug Research Institute (LDRI). She has published more than 30 papers in reputed journals.

Abstract:

Traumatic spinal cord injuries (SCI) cause devastating neurological deficits and disabilities. Symptoms vary greatly,ranging from pain and paralysis. We focus on drug and cell delivery for spinal cord repair following traumatic SCI. We incorporated FGF-2 and VEGF in scaffolds, and GDNF and VEGF in hydrogels and studied their impact on spinal cord injury and functional recovery. We observed a functional recovery of rats treated with GDNF. In order to further stimulate spinal cord repair, our approach is to combine drug and stem cells with hydrogels, to ensure local delivery of drugs and survival of cells at the injury site. Human dental stem cells from the apical papilla (SCAP) have been selected based on their origin (neural crest) and ease of access. We first assessed the impact of SCAP encapsulation in different hydrogels. We then compared in vivo the influence of SCAP delivery with the implantation of the whole tissue they originate from (apical papilla) in a rat spinal cord hemisection model. Functional recovery was observed in the group treated with the papilla. Genomic and proteomic analysis are being performed to identify the markers involved. Second, SCAP were grown on growth factor-loaded microcarriers and embedded in hydrogel. Impact on cell survival and gene expression is being studied, after which the system will be implanted in a SCI model. In conclusion, we are developing a multidisciplinary approach based on the combination of drug and cell delivery to stimulate spinal cord repair.

Biography:

Rekha Samuel is a Pathologist working at the Centre for Stem Cell Research (CSCR), Christian Medical College, India. She did her Post-doctoral Training in Dr. Rakesh K Jain’s Laboratory at Massachusetts General Hospital, Boston, on an overseas fellowship that was funded by the Department of Biotechnology,Government of India. She is the Principal Investigator of the Vascular Biology Laboratory at the CSCR. Her current research focuses on examining the Microvascular Defects of Type 2 Diabetes using the Gestational Diabetes Mellitus Placenta. She has recently been awarded the European Foundation of Study for Diabetes/Sanofi 2015 grant.

Abstract:

Human iPSC derived vasculogenic cells may offer a long sought solution for obtaining large numbers of autologous cells sufficient for tissue engineering and opportunities to model vascular disease in a dish. We report a novel approach for the derivation of endothelial precursor cells from hips cells using triple combination of selection markers: CD34, neuropilin-1(NP-1) and KDR, and an efficient 2-D culture system for hiPS cell-derived endothelial precursor cell expansion. Functionality of blood vessels in cranial window models of severe combined immunodeficient (SCID) mice was determined by non-invasive longitudinal in vivo multiphoton laser scanning microscopy for parameters that included red blood cell (RBC) velocity, blood flow and permeability to bovine serum albumin. We successfully generated endothelial cells from hiPS cells obtained from healthy donors and formed stable functional blood vessels in vivo - lasting for 280 days in SCID mice. The RBC velocities of engineered blood vessels were comparable to normal endogenous host vessels (1.36±0.3 mm/s), and demonstrated a higher permeability as compared to endogenous vessels. We also generated mesenchymal precursor cells (MPCs) from hiPS cells in parallel. Moreover, we successfully generated functional blood vessels in vivo using these endothelial cells and mesenchymal precursor cells derived from the same hiPS cell line. In parallel, we have isolated hiPSC-derived ECs and PVCs from type-1 diabetes and maturity-onset diabetes of the young (MODY) cases. The T1D-iPS engineered blood vessels were functional for 4 months in vivo while MODY-iPS-ECs failed to form functional blood vessels. Human iPSC-derived vasculogenic cells may be an abundant source to examine vascular defects of diabetes in a dish.

Biography:

Scott L Nyberg is a trained Liver Transplant Surgeon and a Biomedical Engineer. His engineering training includes a BS degree in Chemical Engineering from MIT and PhD in Biomedical Engineering from University of Minnesota. His medical training includes an MD degree from Johns Hopkins and Transplant Surgical training from the University of Minnesota. He directs the Artificial Liver Program and Liver Regeneration Program at Mayo Clinic Rochester, where he has worked as a Transplant Surgeon since 1996. He has published over 175 papers in reputed journals, and serves on the Editorial Board of Journal of Hepatology and Liver Transplantation.

Abstract:

Acute liver failure is a serious, potentially life-ending, medical problem. Spontaneous recovery occurs in less than 50% of cases. Recovery would be more frequent if a supportive therapy were available to correct the toxic milieu of acute liver failure to prevent its extra hepatic manifestations and to assist in liver regeneration. Therefore, we have developed a novel supportive therapy, the spheroid reservoir bio-artificial liver (SRBAL), composed of 20-40% of the hepatocyte mass of a normal human liver. The greater cell dose is accomplished with anchorage-independent aggregates of primary hepatocytes (spheroids) engineered by a novel rocked mixing technique. Results of this pivotal preclinical study demonstrate that the SRBAL improved survival in an allogeneic model of acute liver failure. Survival benefit correlated with the rate of ammonia detoxification and lowering of intracranial pressure indicating a neuroprotective effect of this cell-based therapy. Plans for clinical evaluation of the SRBAL are underway. In addition, we have bioengineered a novel animal, the FAH deficient pig, to serve as an in vivo“incubator” for large scale production of primary human hepatocytes. The presentation will include progress towards a next generation “humanized” SRBAL employing human hepatocytes produced in the FAH deficient pig.

Biography:

Liudmila Leppik has completed her PhD in Molecular Biology at the Laboratory of Human Gene Structure and Function at the Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry in Moscow, Russia and Post-doctoral studies at the German Cancer Research Center in Heidelberg, Germany. She is currently a Postdoctoral Research Fellow and Assistant Director at Frankfurt Initiative for Regenerative Medicine at the Goethe-University in Frankfurt/Main.

Abstract:

Electrical stimulation has been successfully used to treat bone defects clinically for many years. Recent in vitro studies have demonstrated that ES can change cell behavior such as migration, proliferation and differentiation. The goal of this project was to combine bone tissue engineering (TE) and electrical stimulation (ES) approaches in in vivo and in vitro experiments to determine its effectiveness and characterize the underlying mechanisms, respectively. In vitro experiments were performed in a customized six well ES culture chamber, in which we exposed adipose- (AT-MSC) and bone marrow- (BM-MSC) derived MSC to ES in 2D and 3D culture. Results showed that ES changed osteogenic gene expression patterns in both AT- and BM-MSC,and that these changes differed between the two groups. We found that ES increased BMP2 and TGF-ß1 expression in MSC suggesting calcium signaling as a potential mechanism. In in vivo experiments, a rat femur critical size defect (CSD) model was used and treated with AT-MSC seeded on calcium phosphate scaffold (β-TCP) material. Femurs from three groups of rats(40 per group; control=ß-TCP alone; sham=ß-TCP+AT-MSC; experimental=ES+ß-TCP+AT-MSC) were assessed at one and eight weeks post treatment by mean of histology, gene-expression and mechanical testing. The results of this ongoing in vivo experiment will be presented at the upcoming meeting. These initial findings may support the use of ES in TE applications which could potentially improve and expand their use in the clinical setting.

Eva Sykova

Institute of Experimental Medicine ASCR, Czech Republic

Title: Stem cells and biomaterials for the treatment of spinal cord injury, stroke and ALS
Biography:

Eva Sykov is a Physician, Scientist, an internationally recognized expert in Neuroscience and Cell Therapy and was the Director of the Institute of Experimental Medicine from 2001-2016. Her research focuses on neurologic diseases and stem cells. She has worked at Charles University, Prague, Founding and Directing the Department of Neuroscience, 2nd Medical Faculty (1996-2012) and leading the Center for Cell Therapy and Tissue Repair (2000-2011). In 2008 she built the Innovative Biomedical Center, which houses a business incubator that assists in the transfer of results in regenerative medicine research into practice.

Abstract:

Stem cells alone or in combination with biomaterials have been investigated for their therapeutic potential in animal models of spinal cord injury (SCI), stroke and ALS. We compared human mesenchymal stem cells (MSCs) from bone marrow, a conditionally immortalized human stem cell line from fetal spinal cord (SPC-01) and human induced pluripotent stem cellderived neural precursors (iPS-NPs) for their capacity to migrate towards lesion sites, differentiate and induce regeneration.Transplanted cells were labeled with iron-oxide nanoparticles for MRI tracking. Animals were tested using motor and sensory tests. Animals with chronic SCI were implanted with HPMA-RGD hydrogels seeded with MSCs. In SCI, the best functional improvement was found after iPS-NP transplantation, followed by MSCs and SPC-01. MRI proved that the cells migrated into the lesion, survived for several months and differentiated into motoneurons and glia. Improved motor and sensory scores in chronic SCI were found after the implantation of biomaterials seeded with MSCs. Natural ECM hydrogels bridged the SCI lesion cavity, modulated the immune response and provided the substrate for in vivo neural tissue regeneration. Grafting of  iPS-NPs reversed stroke-induced somatosensory and motor deficits and protected the host substantia nigra from atrophy. MSC were also intrathecally applied to rat ALS-model (SOD1). Overall survival in the cell-treated group compared with the shamtreated group was prolonged, the cell-treated rats showed better motility, grip strength and greater numbers of perineuronal nets in the spinal cord. Our results demonstrate that the intrathecal application of MSCs can slow down progression of the disease.

Biography:

Enrique Brandan is a Professor and Chairman of Department of Cell and Molecular Biology, Faculty of Biological Sciences at the Pontificia Universidad Católica de Chile, Santiago, Chile. He has earned his BSc in Biology at Universidad de Chile and PhD in Cell Biology at Universidad Catolica de Chile. He was an International Scholar of the Howard Hughes Medical Institute, USA for 10 years. He is a Member of the National Academy of Sciences, Chile. He has been interested in the signaling pathways that control skeletal muscle differentiation and in the mechanisms associated to fibrosis present in several skeletal muscular dystrophies. He has been concentrating in to develop new strategies to fight fibrosis improving skeletal muscle strength and cell therapeutic approaches.

Abstract:

Duchenne muscular dystrophy (DMD) is characterized by absence of the protein dystrophin, muscle wasting and fibrosis.We have evaluated the role of the profibrotic connective tissue growth factor (CTGF/CCN2) in a genetically model (mdx-CTGF+/- mice) and in a decreasing CTGF activity model, by blocking CTGF antibodies in mdx mice. In both models, we observed a decrease of fibrosis together with a significant less muscle damage compared to mdx mice. The decrease or blocking of CTGF caused an improvement of muscle strength and improvement of wild type cell graft. Thus, CTGF could be novel therapeutic agents to treat muscle fibrosis associated to DMD and improve cell therapeutic approaches.

Biography:

W Bonani has received his PhD in Materials Science from the University of Trento in 2011 and in Mechanical Engineering from the University of Colorado in 2012. He is currently holding a Post-doctoral position at the University of Trento (INSTM research unit). During his career, he has developed and studied scaffolds for tissue engineering applications (vascular, bone, cartilage, neural), systems for drug encapsulation and delivery, extraction, manipulation and processing of biopolymers (silk, collagen, alginate) and cellularized 3D building blocks for tissue modeling. He has published more than 15 papers in peer-reviewed journals.

Abstract:

Mammary acini organization plays a fundamental role in tissue development and physiology, but also could offervaluable insights in cancer onset and progression. 3D in vitro models represent a new paradigm in biological sciences.Reconstituted Matrigel® Matrix was successfully used to model 3D tissues; however, Matrigel® encompasses a wide range of different factors, presents large batch-to-batch variation, is expensive and difficult to isolate. Here we designed tunable alginatebased substrates and assessed the assembly of MCF10A human breast-epithelial cells with a focus on the role of laminin-1 in acini maturation and cavitation. Collagen and laminin-1 were used to increase cell adhesive properties of the alginate matrix and to modulate cell activity. We exploited an overlay model consisting of an alginate-based hydrogel substrate and the addition of laminin-1 in the cell culture medium and followed mammary acini morphogenesis. The evolution of cell spheroids was followed for 21 days; immunofluorescence was performed to investigate acini maturation, cavitation and cell polarization were also analyzed in terms of area and roundness. Image analysis was performed to analyze spheroids/acini for area and roundness. We developed an efficient method for the development of 3D cell spheroids/acini in vitro and studied the effect of laminin-1 on spheroids formation (added to medium). This work established tunable, instructive and medium-throughput in vitro culture systems consisting of alginate-based hydrogel substrates and enriched culture media for the development of mammary acini. The modulation of culture substrates and signaling molecules could represent a platform to study the effects of cellular microenvironment on acinar morphogenesis.

Biography:

Andrzej Lange has graduated with a Medical degree with distinction from the Medical School in Wrocław, Poland. Currently, he is a Professor in the Institute and Founder and Head of the Lower Silesia Center of Cellular Transplantation in Wrocław. His international experience started as a Leverhulme Fellow in the Middlesex Hospital Medical School, London. During 8 year period, he used to spend three months yearly as a Visiting Scientist in the Institute of Experimental Medicine and Biology in Borstel, Germany. He has been a Short Term-Visitor and lectured in a number of European and North American scientific institutions. He is author of 239 scientific papers in peer-reviewed journals.

Abstract:

Graft vs. Leukemia effect preventing relapse in patients with hematologic malignancies receiving allogeneic hematopoietic stem cell transplantation (alloHSCT) can be augmented by infusion of donor lymphocytes (DLI) in patients at the risk of relapse. However, this procedure can ignite the graft vs. host disease in about 30% of patients post DLI. The lower rate of leukemia relapse in patients receiving alloHSCT as opposed to patients post autoHSCT prompted us to develop a new approach in DLI delivery to facilitate anti-leukemic allo-reactivity not increasing the risk of GvHD. The aim of our study is to regenerate the immune system originated from transplanted (HSCT) material especially within the lymphocyte compartmentexerting cytotoxic potential but not facilitating GvHD. The elaborated and clinically implemented approach consisted of the following steps: (1) Identification of relapse post alloHSCT with the presence of leukemic cells in the marrow or in the bones in extramedullary relapse; (2) Obtaining from the donor of mononuclear cell population containing 61%±2.34 (sem) of CD3+ cells in the lymphocyte gate with the use of cell separator; (3) The cells are divided into portions securing CD3+ cells infusion in a dose of 10E6 kg-1 BW for the first infusion, 10E7 for the second approach and 10E8 for the third infusion, frozen in the liquid nitrogen and stored in our tissue bank and (4) Donor cells are injected directly to the posterior iliac crest in patients with marrow or, to the affected bones in patients with extramedullary relapse. The 17 procedures were performed in 5 patients. To have an access to the marrow cavity or to the bones affected by leukemia, the routine trocar for bone harvesting or the bone injection gun, were employed, respectively. The procedure was safe and well tolerated and no symptoms of GvHD were noted after the procedures. In a patient with extramedullary relapse, the healing of the bone lesions was seen clinically and in the evaluated nuclear resonance images. The positive response lasted 7 months until the fatal marrow relapse emerged. In 4 patients with medullary relapse, the reduction of leukemic cells or stable disease was seen in all patients except one, after 1st and 2nd DLI procedures. The patients were under the observation from 3 to 16 months. The positive response was consolidated either by the second transplant (2 patients) or the maintenance approach with the use of Sorafenib (1 case) or anti-CD20 MoAb (one  CLL case). Notably in all marrow evaluations performed along the treatment, there was an excess of CD8+ cells in the marrow as compared to the blood especially seen within CD8+ and CD279+ cells compartment. In conclusion, an accumulation of CD8+ donor lymphocytes harboring CD279+ cells in the marrow cavity of patients relapsing post alloHSCT receiving IB-DLI associates with a positive anti-leukemic response. This observation strongly suggests the presence of T cells cytotoxic response in the marrow infiltrated by leukemia restored by IB-DLI.

Biography:

Luminita Simion Labusca is a Medical Doctor, Consultant in Orthopedics and Traumatology. She has completed her PhD in Regenerative Medicine. She has worked as Medical Practitioner, Clinical Researcher and Scientist involved in basic science in various national and international institutes in Belgium, Ireland,Denmark and as well as in South Korea. Scientifically, she has had international collaboration across multitude of EU and US. She has both EU wide and international recognition as a Scientist in Orthopedic and Traumatology, Sports Medicine, Regenerative Medicine, Cell Biology. She is a Reviewer in several international scientific journal boards. She has edited special issue of international peer reviewed journals. She is a Founding Member of Regenero, Romanian Regenerative Medicine Association.

Abstract:

The use of nano-scale materials, particularly of magnetic nanoparticles (MNP) has evolved as an increasing field of research in life sciences. In the past two decades, various iron oxide based MNPs are being explored for applications in magnetic separation, magnetic resonance imaging (MRI), drug delivery, as tracking agents within the emerging field of cell therapy, for guided tissue engineering as well as for hyperthermia (HT) applications. Our research focus on the development of proprietary Fe-Cr-Nb-B MNPs for local delivery of HT aiming the treatment of solid malignancies, for magnetic enhanced cell delivery as well as for magnetic guided tissue engineering. Medium and long term interaction of MNPs with human mesenchymal and stem cells of various origins (bone marrow, adipose tissue) was investigated in regard with their proliferative, differentiation and immune modulatory potential.Cell viabilility, proliferative as well as mesenchymal lineage differentiation potential was unaffected by the presence of bare as well as chitosan coated MNPs of clinically relevant amount (1-2 mg/mL). Particle retention inside the cell was demonstrated up to 28 days with several particularities related to cell type. We are currently investigating MNP loaded cell behavior after exposure to magnetic field.

Biography:

Marta Pokrywczynska is the Head of the Department of Regenerative Medicine at Nicolaus Copernicus University (NCU) (Bydgoszcz, Poland). She has completed her MSc and PhD in Medical Biotechnology from the NCU. Her research area focuses on “development of new tissue engineering and regenerative medicine
technologies”.

Abstract:

Background: Development of an effective method of urinary bladder regeneration is associated with identifying the pathways that play a key role in the regeneration process. Until now, the regeneration pathways in tissue engineered urinary bladder have not been determined.
Aim: The aim of this study was to analyze regeneration pathways in the tissue engineered urinary bladder.
Method: The study was performed on 40 Wistar rats. Adipose tissue was harvested from 20 rats and adipose derived stem cells(ADSCs) were isolated. After hemicystectomy, bladders were augmented with bladder acellular matrix (BAM) (n=20) or BAM seeded with ADSCs (n=20). 10 rats were sacrificed in each group after three and six months. The total RNA was isolated from reconstructed bladder wall and then quantity, purity and integrity of RNA were evaluated. Gene expression was evaluated using microarray and GeneSpring software.
Results: Isolated RNA revealed good purity, concentration and RNA integrity number above seven in all samples. Gene expression analysis indicated 711 differentially expressed transcripts in bladders reconstructed with BAM seeded with ADSCs compared to bladders reconstructed with unseeded BAM, six months after the reconstruction and 8 241 differentially expressed genes, three months after the reconstruction. A large number of differentially expressed genes were involved in a lot of pathways, including B and T cell receptor signaling pathway, IL-3, IL-6, IL-2 and IL-5 signaling pathways, inflammatory response pathway, GPCR signaling pathway and Wnt signaling pathway.
Conclusion: Microarray gene expression analysis allows to create a regeneration patterns in tissue engineered urinary bladder.

Biography:

J R Burgess runs a very successful Non-Surgical Orthopedics practice that utilizes medical fitness as part of their care for achieving better outcomes. Currently,they assembled a 28000 sq. ft. facility with well over 8000 patients who are seeing life changing results. He along with Dr. Baumgartner formed MedFit and has licensed their model to over 30 clinics around the world.

Abstract:

With the uncertainty regarding the future of the health care system, administrators, medical professionals and private practices are looking for solutions to a broken system. Today’s crisis leads to a new opportunity that will combat rising health care costs and improve patient outcomes. The obesity epidemic and the incidence of disease and injury have forced the need for a preventable, medically integrated, outcome based model of medical fitness. Exercise and proper nutrition is the foundation to all functional medicine and anti-aging treatments as well as a necessity for healing and preventing injuries in patients. As we optimize body composition through increasing lean muscle mass while decreasing the fat mass, many of the hormone imbalances that are causing chronic disease can be eliminated. As specialists in our field, we need to have the tools to educate and train our patients on proper exercise and lifelong nutrition to maximize the healthy changes in the lives of our patients. As physicians, we have the ability to influence our patients to make positive changes in their fitness and nutrition that will impact there clinical outcomes. We just need to implement those into our current practice models. Five learner objectives are: Review the current literature on to use medical fitness to change the course of history; understand the need for physicians to prescribe medical fitness and the power we have to make change; maximizing orthopedic outcomes through integrated fitness and nutritional services; compliment any medical practice model to increase cash based revenue and obtain new patients at the same time and; easy ways to implement medical fitness into your practice.

Biography:

Heba Al-Zer is a Dentist who has completed her PhD in Stem Cells Biology from the University Medical Center Hamburger-Eppendorf in Germany, 2015. She is an Assistant Professor of the Conservative Department at the School of Dentistry, University of Jordan. Her work focuses on “The properties and the potentials of the human dental pulp stem cells (DPSCs)”.

Abstract:

The peripheral nervous system (PNS) has an intrinsic ability for repair and regeneration even after severe injury. Schwann cells play a major role in PNS axon regeneration. They are able to dedifferentiate, re-enter the cell cycle and promote axonal regrowth by phagocytosis of the axonal and myelin debris, recruiting macrophages to the injury site, secreting several neurotrophic and key transcription factors and forming a unique column of cells called bands of Bungner within their endoneurial tubes for guiding the axonal sprouts from the proximal stumps. In clinical practice, nerve autografting is considered to be the “clinical golden standard” for promoting repair of segmental peripheral nerve injury and bridging a critical gap defect,but the results are still unsatisfactory. In addition, the reported outcomes of synthetic nerve conduits and allogenic nerve grafts have been voluminous and often conflicting. Therefore, cell-based therapies by transplantation of Schwann cells within appropriate scaffolds have been introduced as a promising treatment modality for peripheral nerve regeneration. The dental pulp of adult human teeth contains different stem cells populations, which show broad diversity and potentials. Here, the NC derived DPSC population is discussed in term of their culturing method and induction of differentiation into SCs. The NC derived DPSC population is to be recruited in the future for peripheral nerve injury regeneration after their induction into SCs in vitro. We recommend that these DPSC-derived SCs can be considered as a superior alternative source of SCs compared with a nerve donor source. DPSCs are feasible, cost and time efficient and are harvested without complicated surgical procedures.

  • Stem Cell Transplant | Ethical & Legal Issues
Location: Kiel 1-3

Chair

Scott L. Nyberg

Mayo Clinic School of Medicine, USA

Co-Chair

Roberta Di Pietro

Gabriele d’Annunzio University, Italy

Biography:

Claire Henchcliffe has received her Doctorate at Oxford University UK. After completing Medical Training at the College of Physicians and Surgeons of ColumbiaUniversity, New York and the New York Presbyterian Hospital, she established the Weill Cornell Parkinson’s Disease and Movement Disorders Institute, New York,where she serves as a Director. She is a Vice Chair for Clinical Research and Associate Professor of Neurology at the Weill Cornell Medical College, New Yorkand combines clinical practice and research in Parkinson’s disease. Her research interests focus on developing new therapies for Parkinson’s disease, in particularcell-based approaches and optimizing clinical trial designs.

Abstract:

Neurorestorative approaches in Parkinson’s disease (PD) based upon new stemcell technologies are imminent. As clinicaltrials are starting, it is critical to consider how to best ensure a genuinely informed consent.We therefore focused upon current barriers to such a goal and identified the following themes:
 
(1)Barriers to adequate disclosure of risks and benefits(investigator-dependent): In first-in-human trials, risks and benefits are incompletely defined. A substantial literature in fetal tissue transplant in PD is only partially applicable, and the possibility of serious risks remains, including tumor formation, graft rejection, and risks of immunosuppressive agents if administered.
 
(2)Barriers to understanding clinical research methodology(participant-dependent):Research participants may have difficulties understanding concepts integral to clinical research, such as equipoise. There is also strong evidence that the therapeutic misconception influenced participant’s consent in previous surgical trials in PD. Cognitive dysfunction, common in PD, may or may not impair understanding of informed consent information, thus complicating evaluation of capacity to consent.
 
(3) Need for extended consent process (investigator and participant-dependent): The traditional model of a single limited visit to address informed consent appears to be insufficient in upcoming clinical trials that involve complex scientific underpinnings, uncertainty in risks and benefits, and recruitment of participants with a neurodegenerative disorder that commonly affects cognition. Extended informed consent processes to consider include the use of multimedia educational materials, assignation of a “research partner”, and multiple disclosure sessions.

Biography:

Jan Kriz has completed his PhD from the Charles University in Prague, Czech Republic and Post-doctoral studies from the University of Western Ontario in London, Ontario, Canada. He is the Principal Investigator of projects focused on “improvement of pancreatic islet transplantation efficiency in the IKEM”, at leading transplant center in the Czech Republic and one of the four most active centers in transplantation of insulin producing tissue. He has published more than 30 papers in reputed journals.

Abstract:

The pancreatic islet transplantation represents a real treatment for type-1 diabetic patients suffering from hypoglycemia unawareness syndrome. The wider application of this method is limited among others by a suboptimal efficiency of islet engraftment. In clinical practice, islets are commonly infused into the hepatic portal vein, where they spontaneously settle in terminal branches. Immediately after infusion, they induce the Instant Blood Mediated Inflammatory Reaction (IBMIR),which results in considerable destruction of graft cells and subsequent non-optimal long term function. The better site for islet transplantation should predominantly provide the adequate oxygen supply without a direct contact to the recipient blood, and should be created using a minimally invasive surgery without any signs of inflammation in time of transplantation. Therefore,the most groups prefer a long term (at least four weeks) creation of subcutaneous cavities with completely finished healing process in time of islet administration. Practically, they transplant islet into the almost avascular fibrous tissue. The main goal of this study is the identification of optimal timing of islet transplantation after the insertion of polymeric macroporous scaffold under skin. We intend to detect the vascular density, blood perfusion and intensity of inflammation during individual phases of tissue healing in the close vicinity of inserted scaffolds. Using a marginal mass of islets transplanted in optimal time,we demonstrate the superior metabolic control in diabetic Lewis rats.

Thomas Skutella

University of Heidelberg, Germany

Title: Germ stem cell reprogramming
Biography:

Thomas Skutella has completed his MD from Frankfurt, Germany and Post-doctoral studies from Max Planck Institute for Psychiatry, Munich and the Institute of Anatomy, Charite, Berlin. He was the Director of the Center for Regenerative Medicine, Tübingen and is currently the Head of a Department in the Institute for Anatomy and Cell Biology, Heidelberg.

Abstract:

It has been suggested that the generation of human germline stem cells from primordial germ cells (PGCs) or spermatogonial stem cells (SSCs) may provide simple and non-controversial access to an individual cell-based therapy. These human studies are based on mouse models of germ stem cell reprogramming, in which the molecular mechanisms underlying the shift to pluripotency could be investigated in more detail. However, recent studies revealed the impossibility of a conversion of adult mouse SSCs and human PGCs into a truly pluripotent state. For the conversion of PGCs it appeared to be necessary to conduct a two-factor induction of pluripotency with Oct4 and Sox2. From the CD49f MACS- and matrix-selected fractional part of enriched human adult spermatogonia, adult human germ stem cell cultures were grown. When compared to authentic pluripotent stem cells, these human adult germ stem cell (haGSC) cultures were not fully reprogrammed. For this reason, an investigation of the molecular blockages of pluripotency in germ cells is necessary. It would be deeply interesting to improve culture conditions, to apply bolting factors, such as small molecules deterring from the molecular block that hinders a full germ stem cells (GSCs) or haGSC conversion to molecular pluripotent stem cells, and to enlarge the restricted time window of SSC to pluripotent stem cell (PSC) conversion.

Biography:

Ivana Haunerova is a Quality Assessor for biotech and advanced therapy medicinal products in the State Institute for Drug Control in the Czech Republic. She has completed her graduation in Biochemistry at the Institute of Chemical Technology, Prague. Since 2009, she is a Member of the Committee for Advanced Therapies at EMA.

Abstract:

Development of an advanced therapy medicinal products and their shift from the basic research to the clinical application is very difficult task. The legislation and regulatory requirements are in force but the research at this area is so rapid that we are facing new and new challenges. On one hand, it is necessary to define the rules so that the regulatory field is transparent,consistent and the decisions are predictable. As a consequence, there are some situations when the requirements seem to be too restrictive. On the other hand, there is a strong wish to have effective treatment on the market as soon as possible.Regulators need to balance both point of views. The national regulatory authorities and especially European Medicine Agency(EMA) offer different procedures that can be used by the product developers. These procedures such as classification of the borderline products, certification and scientific advices can be very helpful for the planning of development and saving time and resources, if used appropriately. At the beginning of the planning of cell-based product development, it is important to know what the regulatory framework for the specific product is, what are the procedures that can or should be used and what is the manufacturer goal (national hospital production or European market). The risk-based approach should be used to define risk and other important aspects but also the requirements that are not relevant for this particular case. Those findings must be taken into consideration during the whole development phase.

Biography:

Toshio Miki is an Associate Professor of Surgery at the Keck School of Medicine of USC. He has received his MD and PhD degrees from Nihon University in Tokyo, Japan. With the goal of overcoming the organ shortage faced by patients awaiting liver transplants, he started studying Xenotransplantation and Hepatocyte Transplantation. In his search for alternative cell sources for hepatocyte transplantation, he discovered stem cell-like populations in the human amnion in the year 2005. Since then, he has been best-known for his studies of placental stem cells and their clinical applications.

Abstract:

Placental stem cells are a readily available cell source for regenerative medicine, which can be procured in a non-invasive manner, and there are few ethical concerns regarding their use. The hepatic differentiation potential of human amniotic epithelial cell (hAEC) suggests usefulness in cell therapies for congenital liver metabolic disorders including ornithine transcarbamylase (OTC) deficiency. OTC deficiency is the most common urea cycle disorder, which can cause severe brain damage and death. To determine if hAEC transplantation has a therapeutic effect against OTC deficiency, we transplanted hAECs into the liver pulp of wild type (WT) and spf/ash mice and measured the following parameters: Engraftment potential,OTC enzymatic activity, levels of urine metabolite markers and behavioral response to excess ammonia. We present our findings that hAECs can engraft in immunocompetent mouse livers and provide a therapeutic effect. While OTC enzyme activity from untreated spf/ash controls produced 12.75±0.91 μM citrulline/mg protein/h, spf/ash mice treated with hAECs (spf/ash-hAEC)produced increased activity at 19.08±1.33 μM citrulline/mg protein/h (p<0.001). In addition, spf/ash-hAEC mice possessed levels of urine metabolites more closely to levels found in WT controls (p<0.05), confirming the improvement in OTC enzyme activity. Lastly, spf/ash-hAEC mice better tolerate ammonia challenge, exhibiting improved behavioral phenotypes compared to untreated controls (p<0.01). This growing body of evidence better suggests that hAECs can be used for cell replacement therapy to treat OTC deficiency.

Biography:

Željka Večerić-Haler is a Medical Doctor and Specialist of Nephrology. As a Nephrologist, she faces numerous challenges from the field of Regenerative Medicine on daily basis. She has completed her PhD from University of Ljubljana, Faculty of Medicine in 2016. Her research interests include studies on stem cell transplantation.

Abstract:

Mesenchymal stem cells (MSCs) are one of the promising tools in regenerative medicine. Non-clinical studies have shown that MSC transplantation markedly improved animal survival as well as functional and morphological parameters of various injured organs. Beneficial effects of MSCs can be attributed to different mechanisms such as their ability to secrete various immunomodulatory molecules as well as other soluble factors that have antiapoptotic, antifibrotic and antioxidative effects. Despite their beneficial effects, the outcome of the MSC transplantation is not always successful. It is suggested that one of the major problems influencing the efficacy of stem cell therapy is the poor MSCs survival following transplantation.Placing MSCs into a foreign tissue or harmful microenvironment may defeat their beneficial effects due to overload of proinflammatory cytokines or insufficient resistance of transplanted stem cells to oxidative and inflammatory stresses at the injured sites. It is interesting that most of the studies that evaluated beneficial effects of human MSCs in cisplatin-induced acute kidney injury have been so far performed on immunocompromised animals; although it is known that both innate and adaptive immune system is important contributor to the pathogenesis of acute kidney injury and can significantly affects the extent of nephrotoxicity. For instance, severly immunocompromised animals are susceptible to cisplatin nephrotoxicity, while mice without T lymphocytes, CD4+ or CD8+ T cells are protected against cisplatin nephrotoxicity. We evaluated the efficacy of MSC transplantation on mice with normal and compromised immune system and found out that difference in immunological environment affected not only the level of inflammation and oxidative stress at the site of injury but beneficial effects of MSC as well.

Biography:

Ingrid Lang has completed her Masters and her PhD degree from Karl-Franzens University, Graz. After additional studies and experience as High School Teacher in Biology, she was appointed as Assistant Professor and subsequently as Associate Professor of Histology and Embryology at the Institute of Cell Biology, Histology and Embryology, Medical University Graz. She has established a method for the isolation and characterization of fetal and adult endothelial cells from the macro and the microvasculature. She has experience in the field of “placenta research, stem cells and vascular biology”. She has published more than 60 articles in reputed journals.

Abstract:

We recently showed that placental-derived mesenchymal stem/stromal cells (PMSC) enhance endothelial cell viability and network formation of endothelial cells in vitro and induce angiogenesis in a mouse model in vivo by the secretion of paracrine factors or direct cell-cell contact. Thus, beside potential therapeutic aspects in disorders caused by insufficient angiogenesis such as chronic wounds, PMSC are promising tools for supporting vessel formation in engineered tissue constructs. Here, we assessed the impact of PMSC on the integrity and stability of endothelial cells in vascular grafts exposed to shear stress. ePTFE grafts were used to culture endothelial cells derived from the human placenta (hPEC) and PMSC in a 3D tissue culture model. hPEC were attached to the inner graft surface and a constant low (0.015 dyne/cm2) or physiological flow (0.92 dyne/cm2) was applied by a perfusion system. hPEC were supported by PMSC attached to the outer graft surface.PMSC improved the viability of hPEC exposed to 0.015 and 0.92 dyne/cm2 as shown by a decreased LDH release of 18% and 47%, respectively. Angiogenesis array analysis revealed that hPEC exposed to 0.92 dyne/cm2 secreted decreased levels of GRO,MCP-1, TIMP1, TIMP2 and angiogenin under co-culture with hAMSC. hPEC exposed to flow stimulated the migration of hAMSC. Our data demonstrate that PMSC improve adhesion, viability, and stability of endothelial cells in perfused vascular prostheses. Increased angiogenic properties and enhanced migration of cells favor endothelialization in order to provide a non-thrombogenic surface of small diameter vascular grafts in clinical use.

Biography:

Heyong Yin graduated in July 2016 with a master’s degree in surgery at Nankai University and Chinese PLA General Hospital. His researches are focus on Cartilage tissue engineering and stem cell. Since September 2016 he joins department of surgery, University of Munich (LMU) as a PhD candidates and continues his research into regenerative treatment of orthopaedics injuries to ligaments, tendon and hyaline cartilage.

Abstract:

We propose a method of preparing a novel cell carrier derived from natural cartilage extracellular matrix (ECM),designated cartilage ECM -derived particles (CEDPs). Through a series of processes involving pulverization, sieving,and decellularization, fresh cartilage was made into CEDPs with a median diameter of 263±48 μm. Under microgravity culture conditions in a rotary cell culture system (RCCS), bone marrow stromal cells (BMSCs) can proliferate rapidly on the surface of CEDPs with high viability. Histological evaluation and gene expression analysis indicated that BMSCs were differentiated into mature chondrocytes after 21 days of culture without the use of exogenous growth factors. Functional cartilage microtissue aggregates of BMSC-laden CEDPs formed as time in culture increased. Further, the microtissue aggregates were directly implanted into trochlear cartilage defects in a rat model (CEDP+MSC group). Gait analysis and histological results indicated that the CEDP+MSC group obtained better and more rapid joint function recovery and superior cartilage repair compared to the control groups, in which defects were treated with CEDPs alone or only fibrin glue, at both 6 and 12 weeks after surgery.In conclusion, the innovative cell carrier derived from cartilage ECM could promote chondrogenic differentiation of BMSCs,and the direct use of functional cartilage microtissue facilitated cartilage regeneration. This strategy for cell culture, stem cell differentiation and one-step surgery using cartilage microtissue for cartilage repair provides novel prospects for cartilage tissue engineering and may have further broad clinical applications.

  • Regenerative Medicine | Models of Regeneration | Cell and Organ Regeneration | Regenerative Medicine Market
Location: Kiel 1-3

Chair

Hans-Georg Simon

Northwestern University, USA

Co-Chair

Jaroslaw Fabis

Medical University of Lodz, Poland

Biography:

Hazem Barmada is a Cardiovascular & Thoracic Surgeon. He has completed his Graduation from St. Andrews and Manchester Universities in 1971 and 1974, respectively. He was active in Martial Arts in Britain from 1968 to 1984 and Medical Officer to the Martial Arts Commission and other major karate organizations in GB from 1974 to 1984. He is a Fellow of the Royal College of Surgeons of Edinburgh in General and Cardiothoracic Surgery and a Member of the Society of Thoracic Surgeons and American College of Phlebology. He has two patents and published a number of papers. His interests include “regenerative medicine and stem cell research, sports injuries and venous and lymphatic disease”.

Abstract:

Stromal vascular fraction (SVF) was first isolated from adipose tissue around 1970. SVF has proved very effective to manage many hitherto difficulties or untreatable conditions, including severe degenerative joint disease (DJD), medically. Nonetheless, it has not been effective in every case, especially in the more severe cases of DJD, possibly reflecting the diversity of the pathology within each category. Extracorporeal Pulse Activation Technology (EPAT), alias dictus shock or Pressure Wave Therapy (ESWT), is a non-invasive technology platform of Storz Medical that employs acoustic pressure waves to affect a mechanic-transduction response at the cellular level, which leads to cell activation, improved metabolism, angiogenesis, neovascularization and improved healing. This has been used as a stand-alone modality for treating various musculoskeletal conditions in Germany over the past two decades. In a small series of patients with severe DJD who have either refused surgery or had undergone multiple unsuccessful surgeries, we found the addition of EPAT to the deployment of SVF intravenously as well as locally, were perceived to be highly effective in decreasing pain, reducing stiffness and increasing functionality. It was also perceived that giving 2-43 treatments of EPAT to the region in question, days apart and one treatment immediately before the SVF deployment, enhanced the benefit. This possibly recruited more local repair processes, attracting more regenerative cells, and releasing growth factors and cytokines for repair.

Romaldas Maciulaitis

Lithuanian University of Health Sciences, Lithuania

Title: Translational challenge in renal regeneration: Establishment of posology
Biography:

Romaldas Maciulaitis has completed his MD and PharmD studies more than 20 years ago and after PhD studies in Clinical Pharmacology he was involved into Drug Regulatory and Scientific Appraisals at the national (Lithuania) and international (European) levels. In 2009, he initiated new experimental Pharmacology research direction in his University in Kaunas on Regenerative Pharmacology applying cell preparations with research program focusing on PK/PD of cell therapies in renal and cartilage injuries. He has published more than 20 papers in reputed journals.

Abstract:

The scientific and regulatory communities are confronted during more than 10 last years with rather numerous controversial views in what is important for successful clinical translation for what is seen as promising during non-clinical studies. Not the last reason why is this still valid today is the fact that there are numerous gaps in knowledge obtained in the fundamental understanding of the biological rationale of regenerative capacities in case of various cell and gene based technologies. On the other hand, regulatory environment of this technology is heavily driven by the principles of conventional drug development and it is not necessarily easily transferable to regenerative drugs, starting from defining the effective dose. The renal regeneration could serve as one of the typical examples of this controversy. One of the biggest remaining uncertainties in cell therapy for kidney regeneration is the understanding of the mechanisms involved in the therapeutic effect. Among the possible mechanisms of action of mesenchymal stem cells (MSCs) for the treatment of acute kidney injury are the reduction of cell apoptosis and the  anti-inflammatory effects via paracrine/endocrine mechanisms. The results of our studies with one of novel MSC type cells: Skeletal muscle-derived stem/progenitor cells does support alternative mode of action, that MSCs improve the functional and morphological renal recovery directly, by migrating and populating the renal cortex. This debate is not only the academic issue. The answer facilitates translation from the very beginning, enabling developer specify the potency of the product, later the dose definition, magnitude and dose response.

Biography:

Bin Xue has completed his PhD from Nanjing Normal University and was a Visiting Scholar at Harvard School of Medicine. He is the Director of Nanjing University School of Translational Medicine Lab. He has published more than 25 papers in reputed journals.

Abstract:

Background & Aims: Liver injury triggers a highly organized and ordered liver regeneration (LR) process. Once regeneration is complete, a stop signal ensures that the regenerated liver is at appropriate functional size. The inhibitors and stop signals that regulate LR are unknown, and only limited information is available about these mechanisms.
Methods: A 70% partial hepatectomy (PH) was performed in hepatocyte-specific (PP2Ac_-), deleted (PP2Ac_-/-) and control (PP2Ac_+/+) mice. LR was estimated by liver weight, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and cell proliferation; and the related cellular signals were analyzed.
Results: We found that the catalytic subunit of PP2A was markedly up-regulated during the late stage of LR. PP2Ac_-/- mice showed prolonged LR termination, an increased liver size compared to the original mass and lower levels of serum ALT and
AST compared with control mice. In these mice, cyclin D1 protein levels, but not mRNA levels, were increased. Mechanistically, AKT activated by the loss of PP2Ac_ inhibited glycogen synthase kinase 3 (GSK3beta) activities, which led to the accumulation of cyclin D1 protein and accelerated hepatocyte proliferation at the termination stage. Treatment with the PI3K inhibitor wortmannin at the termination stage was sufficient to inhibit cyclin D1 accumulation and hepatocyte proliferation. Conclusions: PP2Ac_ plays an essential role in the proper termination of LR via the AKT/GSK3/Cyclin D1 pathway. Our findings enrich the understanding of the molecular mechanism of LR termination control and provide a potential therapeutic target for treating liver injury.

Biography:

Jaroslaw Fabis is the Head of the Department of Arthroscopy, Minimally Invasive Surgery and Sport Traumatology of Medical University of Lodz, Poland. He is a Member of many scientific societies and has got a distinction to be the corresponding Member of the ASES. His latest scientific activities and publications are focused on “molecular and structural aspects of tendon, muscle, menisci and cartilage degenerative process and their regenerative capabilities”.

Abstract:

From epidemiological point of view, rotator cuff injury is the third orthopaedic entity after spine and arthritic changes. It is connected with loss of functional independence of hand due to shoulder dysfunction. The aim of rotator cuff reconstruction is to restore the muscle function which is the key target of the operation. The proper understanding of mechanism of muscle atrophy and degeneration is connected with extensive investigation of two phenomena’s. The first is physiological sarcopenia and the second is acceleration by tear of tendon. However, in spite of current knowledge and improvement of surgical technique, the rate of recurrence of tear after reconstruction is extremely high. Therefore, it is nothing strange that surgeons and scientists are looking for help coming from rapidly growing medical field-regenerative medicine. There are three key structures related to application possibilities of regenerative medicine-tendon, muscle and bony bed. In all, three cases are of key importance-proliferation, apoptosis and autophagy of tenocytes, muscle cells and osteoblasts as well as production and degradation of extracellular matrix. All these processes influence each other on the way multi-molecular feedback modified by inflammation and oxidative stress. The current approach to this issue are stimulation of healing process by platelet rich plasma and selected growth factors, blocking of pro-inflammatory cytokines and selected molecules as well as receptors, antiinflammatory cytokines, implementation of mesenchymal stem cells and different scaffolds. All these methods are separate research areas of regenerative medicine and their skillful combination of connection opens up new horizons.

Biography:

Shan Ping Yu has received his PhD from Stony Brook University and Post-doctoral training in Neuroscience from the Howard Hughes Medical Institute. He is currently the O Wayne Rollins Endowed Chair Professor at Emory University. He has published more than 120 peer-reviewed papers in reputed journals such as Science, PNAS, Journal of Neuroscience, FASEB Journal, Journal of Cerebral Blood Flow and Metabolism, Cell Death and Differentiation and Neurobiology of Diseases and Stroke. He has been serving as an Editorial Board Member of more than 10 scientific journals.

Abstract:

Neurogenesis in the sub-ventricular zone (SVZ) of the adult brain may contribute to tissue repair after brain injuries. Although the regulation of neurogenesis by molecular genes and signal pathways has been extensively investigated, especially in the dentate gyrus (DG) of the hippocampus, whether SVZ neurogenesis can be regulated by neuronal activities is not well defined. Using the spatial and cell type specific optogenetic technique combined with multiple approaches of in vitro, ex vivo and in vivo examinations, we showed morphological and functional evidence of a novel neuronal link between the striatum and SVZ. Transgenic mice expressing the light-gated channelrhodopsin-2 (ChR2) channel in glutamatergic neurons, optogenetic stimulation of the glutamatergic activity in the striatum triggered glutamate release that consequently evoked membrane currents, Ca2+ influx and increased proliferation of SVZ neural stem cells, mediated by AMPA receptor activation. In ChR2 transgenic mice subjected to focal ischemic stroke, optogenetic stimuli to the striatum not only promoted cell proliferation but also the migration of neuroblasts into the peri-infarct cortex with increased neuronal differentiation and improved long-term functional recovery. These data provide the first morphological and functional evidence showing a unique striatum-SVZ connection that up-regulates SVZ neurogenesis and may be a therapeutic target for promoting adult neurogenesis and brain tissue repair.

Biography:

Abstract:

Introduction: People tend to get injured, leading to wounds with losses of multiple tissues including skin, subcutaneous tissue, btendons and bones. There effective management is a complex process, but the outcomes many a time remain unpredictable. The complications involve further tissue loss, infections, and non-healing ulcers leading to huge morbidity. Throughout the world large resources have been spent, consuming billions of dollars, towards manage these wounds and lessen the morbidity. A simple therapy for effective wound management if evolved will transform this health problem and save lot of sufferings.
Aim: To study an ideal solution for wound management led by regenerative medicine product.
Method: The “Sandeep’s Technique for Assisted Regeneration of Skin” (STARS) is a therapy for complete healing of wounds with autologous self activated platelet rich plasma (PRP), imparted as local subcutaneous infiltrate in the wound margin or underneath the necrotic flap, on every 4th day till complete regeneration takes place and wound heals. The wounds were dressed daily/alternate day with only moist saline. It is a prospective case series study of 75 wounds associated with problems such as compound fracture, amputations, infections, post surgical necrotizing flaps treated with STARS, at Acharya Vinobha Bhave Rural Hospital, J N Medical College, Wardha, India.
Findings: The final outcome was complete/near complete healing of all the wounds without requiring any surgical or intense medical intervention, with a near normal skin including sole, with minimum of scar just like primary healing.
Interpretation: The STARS technique for PRP led wound healing in acute wounds is a safe, easily reproducible and widely accessible way of treating even complex wounds. It can be imparted in a very standard manner across the entire health care centre from territory to primary, without any risks. It needs just a little training towards PRP preparation and a low cost centrifuge machine. The results are predictable and good quality wound healing is ensured with control of infection and Pain. It obviates the need for surgical, medical interventions and intense local care. STARS may be the turning point in the history of wound management. A regenerative product (autologous PRP), is being used widely and easily, to overcome one of the largest problem of the humans; hoping to bring relief to millions who continue to suffer because of affordability and accessibility of expert health care. It is single, simple, affordable and effective therapy for wound management, just as an ideal treatment should be.

Biography:

Hans-Georg Simon has completed his PhD at the University of Freiburg, Germany and after Post-doctoral training at Harvard Medical School, USA, he has research experiences in “academic and industrial settings”. Being a Developmental Biologist for over two decades, he is investigating the reactivation of developmental programs as a mechanism to restore damaged or missing tissues and/or organs. He has won Marcus Singer Award for excellence in Regeneration Research.

Abstract:

The ability to functionally repair tissues damaged by disease or injury remains a significant challenge for regenerative medicine. While tissue repair is limited in higher vertebrates, particular in humans, regeneration of complex tissues occurs widely throughout the animal kingdom. Employing multiple regeneration-competent species in a comparative and complementing manner is currently providing a blueprint for regeneration at the cellular and molecular levels. Using new t and zebrafish model organisms, we are investigating the underlying principles that control the differentiated state of the cell and regulate regenerative processes. Performing multi-tissue microarray analyses, we have recently discovered concerted gene activities in different tissue types indicative of a molecular signature of regeneration. We have demonstrated a key role for the extracellular matrix (ECM) at the wound site, revealing that the transition from a stiff structural ECM environment to a softer, regulatory matrix and then back to the default stiff matrix of differentiated tissues is a critical driving force for regeneration. Employing novel 3D-imaging techniques to regenerating skeletal and cardiac muscle and explanting muscle cells into defined environments, we generated experimental evidence for distinct ECM components providing biochemical and mechanical signals that cooperatively regulate muscle regeneration. Using a cross-disciplinary approach incorporating developmental biology, cell signaling, and material sciences, we are now leveraging the gained knowledge towards engineering artificial matrices as a mimetic of the natural regeneration-specific ECM. We will discuss our progress towards deciphering the matrix code to unlock dormant regenerative abilities in mammals and develop new strategic opportunities in regenerative medicine.

Biography:

Yaegaki K is a Doctor of Dental Science. He has completed his PhD in Medicine from Kureme University Medical Biochemistry and Post-doctoral studies from University of British Columbia. He was trained as Oral Maxillofacial Surgeon at School of Medicine Kurume University. His specialty was facial injuries and cancer. He is the Director and Head of Oral Health at Nippon Dental University, which is the oldest dental school in Asia, also a Dean of PhD program. He has published more than 100 papers and 20 books. He has been serving as an Editorial Board Member of repute.

Abstract:

One of the reasons to limit the liver transplantation is fewer available organs than number of the patients on the waiting list. The liver regeneration might be one of the alternatives of the transplantation. Several clinical studies with employing the transplantation of mesenchymal stem cells from blood, adipose tissue or others were reported. Transplantation of those cells might show decline of hepatic reserve function rather than treating conditions of the liver. The objective of adult stem cell transplantations might be to launch “bridge to transplant” strategy rather than treating liver condition. We have shown that human dental pulp stem cell involves potential to treat lethal liver disease. Hence, we have previously treated the biliary liver cirrhosis and acute liver injury in nude rats with transplanting the regenerated liver tissues which were originated from human dental pulp. Nonalcoholic fatty liver disease (NAFLD) has been ascertained as one of the most prevailing liver condition. Hence, the objectives of the research was to evaluate the clinical potential of our transplantation protocols using swine model of progressive liver failure developed from NAFLD. After four weeks of transplantation into six swine with the condition, secondary liver in the spleen and the regenerated liver were produced. Epithelial cells of biliary ducts were partially replaced with human cells. Serum albumin level recovered from 1.5 g/dL to over 3.0 g/dL. Other factors, i.e. HPT, choline esterase, collagen type IV, ALT etc were also improved, although any of them in the positive control has not been changed.

Biography:

Roberta Di Pietro has obtained her degree in Medicine cum Laude in 1985 and the Specialization in Sports Medicine cum Laude in 1988 from University of Chieti, Italy. She has worked as a Visiting Scientist at the Biochemistry Department, AFRC, UK, at the Pathology Department, USUHS, USA and at the Institute of Human Virology, University of Maryland, USA. In 2005, she became Full Professor of Histology and Embryology at the University of Chieti. She has joined the Editorial Board of Current Pharmaceutical Design as an Executive Guest Editor and recently, the Editorial Academy of the International Journal of Oncology as an Honorary Member. She is an author of 176 scientific publications plus international e-book chapters, editorials, Italian textbooks and 1 Italian patent.

Abstract:

The amniotic membrane (AM) is the innermost part of the placenta, in direct contact with the amniotic fluid. In recent years, the interest toward placenta stem cells has been increasingly growing. At present, two main stem cells populations have been identified in AM: Amniotic epithelial cells (AECs) and amniotic mesenchymal stromal cells (AMSCs). Although AM is an excellent source of cells for regenerative medicine, also due to its immune-modulatory properties and low immunogenicity. Thus, the aim of our study was to map the human AM under physiological conditions to identify possible differences in structural features and regenerative capacity of its components. Human term placentas were collected from healthy women after vaginal delivery or caesarean section at Fondazione Poliambulanza-Istituto Ospedaliero of Brescia or at the SS. Annunziata Hospital of Chieti. Samples of AM were isolated from four different regions according to their position relative to umbilical cord (central, intermediate, peripheral and reflected). By means of immunohistochemistry, morphometry, flow cytometry, electron microscopy, CFU assays and AECs in vitro differentiation we demonstrated the existence of different morphofunctional features in the different regions of AM, highlighting that AECs are a heterogeneous stem cell population. This should be taken into account to increase efficiency of amniotic membrane application within a therapeutic context.

Biography:

Abstract:

The collaboration network of English publications on animal-derived regenerative implantable medical devices based on tissue engineering technology and its evolving processes and current states were mapped in this paper. 10159 English papers published before 1st January 2015 were obtained in eight databases. Social network analysis was conducted on these papers by utilizing UCINET software and Statistical Analysis Software for Informetrics (SASI) researched and developed by Peking University. The collaboration network has evolved from scattered formation to single-core dominated, and then to a core-edge one. Collaboration has become more frequent and wider. Network density and centrality have decreased. USA, UK and China are the top three countries with Wake Forest University, Harvard University and Tufts University being the top three contributing institutions cooperated mostly during the period during 2010 to 2014. In conclusion, more collaboration among different institutions and countries is needed. Edge institutions and developing countries should expand their scope of collaboration.

Biography:

Olivia Candini is a Post-doctoral Associate in the Laboratory of Cell Therapies of the Department of Medical and Surgical Sciences at the University of Modena, Italy. Since last 5 years she has been working with mesenchymal stem cells (MSC) to better understanding the impact of aging on progenitor properties. Based on her combined expertise in cell and molecular biology, she is therefore deeply engaged in innovative approaches to enhance MSC performance and skeletal regeneration.

Abstract:

It is known that progenitor cells undergo an age-dependent decline in their number and function resulting in tissue aging and disease. Since their ex vivo isolation has been introduced in regenerative medicine, these impairments may limit stem cells performance ex vivo and possible after transplantation. Therefore, we considered that the identification of possible molecular fingerprints driving aging can contribute in better age-related disease understanding and in generating tools to favor ex vivo progenitor performance for improved therapeutic benefits. Starting from an age-related comparison of a know stem cell type, we identified aged progenitor- specific miRNA signature that involves only 7 microRNA. Focusing on their targets, we selected HOXB7 as age related gene and whose expression was inversely correlated with senescence. Forced HOXB7 expression was associated with an improved cell growth, a reduction of senescence and an enhanced osteogenesis, linked to a dramatic increase of autocrine bFGF secretion. Based on these original observations we proposed an ex vivo HOX-based reprogramming strategy aimed to empowering key features of mesenchymal adult progenitors for innovative approaches of tissue regeneration and repair.

Biography:

Zhong Wang has completed his PhD from Oregon Health and Science University and Post-doctoral studies from University of California, Berkeley. He was a Principal Investigator at Massachusetts General Hospital, Harvard Medical School and Harvard Stem Cell Institute before he joined the Department of Cardiac Surgery, University of Michigan, USA.

Abstract:

Cell-based therapy represents a highly promising approach for the treatment of heart diseases but its validation requires extensive preclinical studies. Major challenges include lack of clinically relevant large animal reporter models, the identification of ideal cell source, and the extremely low retention and survival rates of transplanted cells. To address these critical challenges in heart regeneration, we have generated stable and high-level expression reporter swine and rabbit animals. For the first time in the field, we characterized and targeted the porcine ROSA26 (pROSA26) locus and generated ROSA26-EGFP swine reporters readily inducible by Cre expression. These swine reporters will enable precise quantification of transplanted cells versus host cells. We also generated knock-in pigs containing Cre-T2A-tdTomato at endogenous ISL1 locus, which allow us to trace the specification and reactivation of ISL1+ CPCs in pig hearts. Meanwhile, we have established cutting-edge injectable cell microcarriers for tissue regeneration. In particular, we have developed new nanofibrous hollow microspheres (NF-HMS) that mimic the extracellular matrix architecture at the nanometer scale. Building on pioneering works in cardiac stem cell field, we are able to robustly generate embryonic cardiac progenitor cells (CPCs) from pluripotent stem cells for heart regeneration. Our results show that NF-HMS greatly enhances the CPC retention, survival, and integration in infarcted hearts of large animals. Our progress using these combined approaches in heart regeneration will be presented. Our integration of advanced cell source, biomimetic carrier, and large animal models for heart regeneration should provide general principles in developing an informative model system for regenerative medicine.

Biography:

Abstract:

The Interventional Orthopedics Foundation Registry was first founded in 2005 to track patients treated with culture expanded, bone marrow mesenchymal stem cells, as well as bone marrow concentrate (BMC). It is the largest and oldest clinical registry in existence in the arena of orthopedic stem cell therapy. 15 clinical studies have been published from this prospective dataset including large scale safety (n=2372) of all tracked treatments, the efficacy of BMC and fat graft for knee OA (n=681), BMC for hip OA (n=196) and BMC for shoulder OA and rotator cuff tears (n=102). Several other small case series have been published in the areas of stem cell treated knee anterior cruciate ligament (ACL) tears, hand osteoarthritis, and fracture delayed and non-union. In addition, a dosing paper for knee arthritis has also been published showing a lower limit of cellular content needed to provide functional relief. This lecture will review this published data as well as additional non-published data culled from the registry. The preliminary results of three new randomized controlled trials on the use of a specific protocol of BMC to treat knee osteoarthritis, shoulder rotator cuff tears, and knee ACL tears will also be reviewed.

  • 3rd workshop on Stem Cells Applications in Regenerative Medicine, Flow Cytometry and Medical Tourism
Location: Kiel 1-3
Biography:

Vasiliki E Kalodimou is the Director of the Flow Cytometry-Research and Regenerative Medicine Department at IASO Hospital in Athens, Greece. She has studied and worked with progenitor cells from placenta, umbilical cord and adipose tissue along with their applications in regenerative medicine and flow cytometry. She has publications in these fields, including Research Fellowships. She has published 2 books about flow cytometry, the Greek edition was published in 2010 and in 2013 the book was published from AABB Press USA. In 2015 she published her 3rd book, “A Handbook to Mesenchymal Stem Cells in Regenerative Medicine”. She is a Flow Cytometry/Stem Cell Specialist, Leader at AABB CT Subsection: CT Product Manufacturing and Testing-USA, Scientific Consultant in Regenerative Plastic Surgery Institute/Pure Aesthetics, Brazil, AABB Assessor-USA and Editor in 12 scientific journals, Editor-in-Chief in 4 scientific journals and her biography was included in Who’s Who in The World 2014, 2015 & 2016 edition.

Abstract:

Background: Recent reports have shown that mesenchymal stem cells (MSC’s) could be used for transplantation in various animal models. Mesenchymal stem cells exhibit remarkable plasticity and harbor potential for use in therapeutic applications, such as lung fibrosis.
Aim & Method: The aim of this study was to isolate mesenchymal stem cells from rat bone marrow, count the actual MSC’s numbers and their viability using flow cytometry and infuse them on an experimental injured animal model. We used magnetic beads (MACS) as a delivery system for novel mesenchymal stem cells (MSC’s) isolated from rat bone marrow and the total number was counted using a Beckman Coulter FC 500 flow cytometer.
Results: Bone marrow samples from 60 Wistar rats, >250 g and six weeks old, were used for the isolation of MSC’s. All samples were stored at room temperature and processed immediately. The MSC’s were isolated and the absolute numbers of MSC’s as well as the viability of each sample was determined using flow cytometry. We used trypan blue to distinguish the viable cells and the mesenchymal cells for microscopic analysis as well.
Conclusion: We succeeded to isolate MSC’s from all 60 samples with a mean value of 1.14x106, while the number of unsorted bone marrow cells required for a transplant is 1.05x106 by bibliography. We found that the mean viability value of the samples was 77.6% suggesting good sample collection and processing conditions.

Biography:

Efi Roboti has over 28 years of experience in healthcare sector. Her clinical practice in Athens currently provides Gynecological-Obstetrics (with over 3200 successful child deliveries) and anti-ageing services to over 15000 patients from Greece and neighboring countries.

Abstract:

Women today live almost half their lives in menopause. Post-menopausal women, those who had breast cancer, cervical and uterine cancer and ovarian cancer have intense symptoms of atrophy after chemotherapy, radiotherapy or pharmaceutically induced early menstruation cessation i.e. dryness, a burning feeling, irritation, painful intercourse and frequent urinary tract infections. The rejuvenation of their genitalia and the vagina will help to dramatically improve the quality of their lives. Our aim is to prove that the infusion of mesenchymal stem cells contained in adipose tissue has an important contribution and long-term results: In appearance and function of the skin and mucosa of the external female genitals and the vagina; to reduce pain in women with dyspareunia; at the entrance of the vagina showing thinning of the epithelium and
adhesions of granulation tissue; to reduce urinary incontinence (from stress incontinence) and the improvement or treatment of lichen sclerosus of the vulva to restore the quality of life and sexual function in the affected women.

Biography:

Nikolaos G Tsamopoulos has completed his Medical studies at the University of Rome, Universita Degli Studi di Roma, La Sapienza, where he was awarded his MD degree. He has completed his Fellowship in Interventional Neuroradiology at Ospedale Niguarda Granda di Milano at the Interventional and Therapeutic Neuroradiology Department. In 1999, he was graduated from the Hellenic Naval Reserve Officers Academy. In 2005, he has accomplished stage in Neurosciences at the Centre Hospitalier Sainte Anne in Paris. In 2008, he has completed the full cycle of the International Diploma in Neurovascular Diseases, sponsored by the Paris Sud University. In 2011, he was awarded Doctor of Philosophy (PhD) at the National and Kapodistrian University of Athens Medical School. Since 2010, he is the Director of the Interventional Neuroradiology Department. In 2014, he became the Project Coordinator of Health Care Research and Technical Development Projects at The National Technical University of Athens (NTU), Greece.

Abstract:

We proposed a new model of optic nerve damage in glaucoma, the model of venous neurovascular pathways to neuroinflammation in glaucoma, based on the theory that glaucoma is a neurodegenerative disorder and optic nerve is amenable to damage imposed by factors affecting the central nervous system. Percutaneous venous transluminal angioplasty appears to be effective and feasible with a high degree of procedural success. PVTA seems to positively influence the clinical status by improving the mean deviation (MD).The results are encouraging and bring promise of a safe and effective approach to adjustment therapeutic approach for patients with chronic open angle glaucoma and moderate to severe visual field defects, with intraocular pressure controlled in the low teens. However, randomized controlled trials will be needed to provide true efficacy and safety data, to convince the clinicians and to become a more high level, evidence-based guideline recommendation.

Isavella Mitrou

University of Birmingham, UK

Title: Stem tourism and economic potentials
Biography:

Isavella Mitrou has studied International & European Politics at the University of Piraeus and continued with an MA in International Studies-Diplomacy at the University of Birmingham and an Executive MSc in European Studies at the Economics and Business University of Athens. She has attended a summer school on Human Rights at the University of Birmingham. After an Internship at the Greek MFA in the Directorate for International Organizations, she started working at the Marketing and Sales Department of a leading medical group in Athens. After deciding a change on her career, she focused on the field of medical tourism and she has been certified on the field.

Abstract:

Research on stem cells has greatly progressed over the past few years. In the beginning, it seemed as something unreachable but the breakthroughs achieved, such as the first transplant of stem cells origin have proven us wrong. Although this innovative field expands throughout the globe, there are still difficulties on its further expansion. On the other hand, medical tourism has lately been considered as a field where a substantial profit can be achieved. Patients from all over the world, seek treatment in other countries taking different aspects into consideration such as expenses and the quality of the services provided. Traditionally advanced countries in medicine, such as the US, have received enormous numbers of international patients for consecutive years, but lately these figures have spread out across many smaller countries due to the changes in legislation and the advances they have achieved. Combining the innovations in stem cells’ research and the promising future they hold, extremely successful companies focusing on research have been created, raising the expectations into a higher level.

Georgia Tsakona

Expert in Hospital Economics/Research, Greece

Title: Hospital research & blue economy
Biography:

Georgia Tsakona studied Economics Accounting at the Greece business administration, University of Patras. She continued attending educational training programs: 410 hours H/Y; 175 hours Business Administration; 50 hours development of personal skills and 50 hours working time management and stress. From 2004 to 2008 she was a Teacher in Charge of Administrative Economic Department of the Technical Institution of Professional Training (IEK). Simultaneously, she worked in civil service center (CSC) at Nafplio-Greece. Since 2008 she works as an Office Responsible Personal Presidential Secretary, in the fields of social partners' Federation of Professionals, “Craftsmen and Merchants Argolidas ". For the last few years, she is focusing in the field of hospital economics and research and in the field of blue economy and hospital research.

Abstract:

The field of regenerative medicine is comprised of an estimated 120 public and 600 private companies as well as a plethora of universities. This vital community of major research institutions and companies are succeeding in advancing both the basic science and its translation into clinical testing and commercial products. In a Blue Economy, the current unsustainable models of production and consumption will be outdated and eliminated. A vicious cycle of over-exploitation of labor and the Earth, with an ever-increasing burden of carbon emissions, will cease. It will become a virtuous cycle of using what is available, increasing social capital incrementally and bringing innovations to the marketplace that help meet the basic needs of all. The blue economy can enhance our lifestyles, benefit our planet and use and conserve materials and energy in remarkable ways.

Biography:

Eleni Triantafyllou graduated from the Department of Biological Applications and Technologies, University of Ioannina, Greece. She performed on thesis with title: "Investigation of post-transcriptional regulation factors of gene expression in brain tumors". She is now a Post-graduate student in Master’s Program in Reproductive and Regenerating Medicine. She always had a strong interest in Regenerative Medicine as well as Genetics and Biotechnology.

Abstract:

A cord blood bank is a facility which stores umbilical cord blood for future use. Cord blood stem cells are blood cell progenitors which can form red blood cells, white blood cells, and platelets. This is why they are currently used to treat blood and immune system related genetic diseases, cancers and blood disorders. Furthermore, recent studies have shown that cord blood has unique advantages over traditional bone marrow transplantation, particularly in children, and can be lifesaving in rare cases where a suitable bone-marrow donor cannot be found. Public cord blood banks function like public blood banks, in that they accept donations from anyone, discard donations that fail to meet various quality control standards and use national registries to find recipients for their samples. Matches are most likely to be better in a public than a private bank. One disadvantage is that they do not charge storage fees and so medical centers do not always have the funds required to establish and maintain them. Traditionally, public cord blood banking has been more widely accepted by the medical community. Private cord blood banks store cord blood solely for potential use by the donor or donor’s family. However, because of its cost, limited likelihood of use and inaccessibility to others, private banking is not usually recommended to low-risk families. Moreover, cord blood from private banks is more likely to suffer from various problems such as potential lower quality control and lower medical usefulness of using a patient’s own potentially diseased cord blood. Nearly, all cord blood transfusions come from public banks, rather than private banks, partly because most treatable conditions can’t use one’s own cord blood.

Biography:

Alexandra Baranowski is currently studying Biological Natural Sciences at the University of Cambridge, UK. She has spent a month in Madagascar over her penultimate summer of school researching on the Behavioral Patterns of the Coquerel’s sifaka lemur, to contribute to the conservation efforts of local and international scientists stationed there. She is presently in the Committee of the Hellenic Society of Cambridge University.

Abstract:

Adipose-derived stem cells (ASCs) are most commonly obtained from white adipose tissue, and exhibit multi-potency, displaying osteogenic, myogenic, adipogenic and chondrogenic properties. As a result, ASCs are becoming progressively more promising for the treatment of debilitating diseases such as cardiovascular conditions and diabetes mellitus, and starting to play a fundamental in the field of regenerative medicine and tissue engineering. Perhaps one of the main areas of interest concerns gynecological applications whether this is for the regeneration of tissue following a mastectomy or vaginal rejuvenation, the hope is for long-term effects that will replace current limited techniques, and dramatically improve the lives of post-menopausal women in particular.

  • Biomaterials and Bioengineering
Location: Kiel 1-3

Chair

Andrzej Lange

Polish Academy of Sciences, Poland

Co-Chair

Hazem Barmada

Mississippi Stem Cell Treatment Center, USA

Session Introduction

Narendra Saxena

Datta Meghe Institute of Medical Sciences, India

Title: Re-engineering the sole defects with “STARS” technique
Biography:

Abstract:

Introduction: Normal sole is a very specialized skin, customized to bear weight. It is limited to be only present on flexor/volar aspect of feet. A partial loss of sole is not uncommon and can happen as a result of trauma, diabetes or tropical ulcer, etc. Such loss is irreparable as it is not possible to do surgically; or otherwise, reconstruct it by transferring cross sole flap will lead to huge donor site morbidity. In selected cases, a partial thickness grafts or free/rotational skin flaps from other sites was done to overcome such losses. But these are susceptible to failures leading to recurrences, infections, dehiscence, etc., leading to morbidity and losses. In many other cases, such as diabetes/tropical ulcer amputation may be resorted for cost-effective rehabilitation.
Aim: The aim of the study is to re-engineer the sole defects with “STARS” technique.
Method: The authors have developed a technique using the autologous platelet cells in PRP form for regenerating and engineering the growth of normal sole in such condition. It is “Sandeep’s Technique for Assisted Regeneration of Skin” (STARS) for complete healing of wounds with autologous self activated Platelet Rich Plasma (PRP). This technique involves the autologous PRP local infiltrate in the wound margin on every 4th day, till complete regeneration takes place and wound heals. The wounds were dressed daily/alternate day with only moist saline. This is an early disclosure of 12 prospective case associated with ulcers/ wounds of sole.
Findings: The observations in terms of diagnosis, age, sex, defect sizes, number of therapeutic STARS sessions and final clinical outcomes were disclosed. All defects of sole, healed to normal/near normal clinical features on count of thickness, color and sensitivity. In addition, randomized histopathological biopsies from such engineered sole was done and studied for micro logical characteristics, which also shows the normal HPE regeneration of sole, in contrast to natural healing which demonstrates a cicatrized tissue base.
Interpretation: With the STARS technique led by PRP gradual normal sole reconstruction is possible despite co-morbidities. It is a safe and effective regeneration modality, without in-tense surgical and medical intervention. This is perhaps revolutionary and was not possible earlier. A complete healing to normalcy of a specialized skin such as sole by an autologous regenerative medicine is a giant step forward and such clinical reengineering will shape the future of therapeutic handling of regenerative medicine products towards previously thought irreparable damages/losses, including sole.

Biography:

Samar Hassab-Allah Kassem has completed his PhD in Biochemistry from Helwan University. She has got 3 diplomas in Nutrition, Laboratory Analysis and Education. She is a Lecturer of Biology, Biochemistry and Ecology at the 6 October University, Egypt. She has worked as an Associate Professor of Health Science, Biochemistry and Nutrition at Dammam University, KSA (2012-2013). She was responsible for designing the Strategic plan for Faculty of Physical Therapy and was Ex-director of Community Service at the Faculty of Physical Therapy. She has published 8 papers in local and international journals and has been invited for Board Membership in Clinics on Oncology, USA.

Abstract:

Regenerative therapy is very promising approach, but should not be limited to stem cell transplantation as its applications are restricted by number of limitations; and should instead seek for a strategy that retrieves the initial healing capacity of a tissue,. In this regard, endogenous activation of stem cell recruitment and their homing into the injury sites is a promising approach for therapeutic purposes. We have previously shown that Avemar and Echinacea effectively induce mobilization and homing of CD34+ stem cells to myocardial tissue and modulate the pro-inflammatory cytokines which may help in the repair of cardiomyocytes. They might be taken as prophylactic or as adjuvant therapy in those individuals at high risk to develop MI, as well as in MI patients. Our study offers novel insights into the extraordinary regenerative capacity of natural products. This study can be extended to encompass other natural products and their effects on degenerative diseases. So, evaluation of their implications in health and diseases is an urgent need as the number of patients with preventable degenerative diseases is growing. This review will discuss how can we select and use them efficiently. It will also update our present knowledge about the potential benefit of specific dietary interventions combined with specific natural products as non-pharmacological tool for preventing and combating such degenerative disorders. Some terms relevant to stem cell biology like cell tolerance, resistance, adaptation and behavior need to be redefined.

Biography:

Mazl Chanova E has completed her PhD in early 2011 from the Institute of Macromolecular Chemistry AS CR (IMC), Prague under the auspices of the University of Chemistry and Technology, Prague, Czech Republic. She is a Member of the Department of Bioactive Polymers at IMC, a leading polymer institute in the central Europe. She is interested in the biomimetic surface modification, cell-surface interactions and surface analytic techniques of biomaterials. She has published more than 12 papers in reputed journals.

Abstract:

A specific distribution of cell-adhesion structures, such as RGDS-peptide, over the biomaterial surface and the synergy with other signaling bio-motifs play a crucial role in regulating cell-biomaterial interactions in tissue engineering. In this study, we investigate the creation of surfaces with single peptide nano-clusters with different spatial distribution and multi-functional surfaces with at least two different peptides nano-clusters, which were prepared by deposition of functionalized PLA-b-PEO. The model surfaces were prepared by spin-casting of PLA-b-PEO copolymer colloidal solutions with selected composition of non-functionalized, maleimide-functionalized and alkyne-functionalized copolymers on a PLA film. The surfaces were grafted with RGDS, KRSR and KRTGQYKLGSKTGPGQK peptides via Michael addition and azide-alkyne cycloaddition. The second end of the peptide chains was flanked with biotin which was used for determination of peptide surface distribution by visualization of streptavidin-labelled gold nanospheres (A-Au NS, 40 nm) selectively bound to biotin using AFM. The concentration of peptides was determined by radioactivity measurements of analogous peptides with iodine label. The surfaces were tested in cell tissue cultures with bone, endothelial and stem cells. The selected surfaces cultured with osteosarcoma MG63 cells were tested under dynamic condition. PLA-b-PEO copolymers in selective solvents form self-assemblies, surface deposition of which results in nanoscale-organized surfaces. Using the deposition of various ratios of functionalized and non-functionalized nano-colloids, the surfaces with overall RGDS-peptide concentration in the range of 8.5 to 15.5 pmol/cm2 and peptide cluster distance from 320 up to 1250 nm were obtained. The effect of RGDS clusters distance, synergic effect of additional peptide sequence as well as the influence of mechanical loading on cell response will be demonstrated.

Biography:

Abstract:

Introduction: Limited regeneration occur spontaneously following spinal cord injury (SCI). Biomaterials hold great promising for the regeneration of many tissues including spinal cord (SC). The neurograft collaborative consortium proposed a novel micro-porous collagen conduit to restore SC functions. The conduit was designed to create a bridge across the lesion and provide a trophic support to the survival of neurons and axons outgrowth. The conduit was tested in a new paraplegic rat model that mimicked irreversible acquired paraplegia and in a rat transection model.
Methods: Paraplegia was induced in a rat model by a contusion at thoracic vertebra T9. Four weeks after contusion the injured portion of the SC was removed and replaced by the conduit (conduit), transected without implantation (transection control) or left untreated (contusion control). The motor functions were evaluated for 8 weeks after implantation using the Basso, Beattie and Bresnahan (BBB) rating scale. The inflammation and regeneration of the SC with/without conduits were investigated using histopathologic evaluation. The conduit was also combined with mesenchymal stem cells (MSC) and tested in single transection model. The transection was performed at T9 and the SC was implanted with the conduit (conduit), the conduit combined with neural/glial-differentiated MSC (conduit+MSC) or left empty (control transection). SCI regeneration was evaluated similarly over 12 weeks. The conduit was tested for its biocompatibility following ISO 10993 standard for irritation, cytotoxicity, acute systemic toxicity, degradation kinetic and genotoxicity.
Results: The conduit demonstrated its biocompatibility in all testing performed according to ISO 10993 standards. Its degradation kinetic was compatible with in vitro culture of MSC, allowing conduit functionalization before in vivo implantation. Its degradation kinetic was also compatible with the SC regeneration process, given that, after treatment period, the conduit was adherent to the surrounding spinal cord and restored the physical continuity of the spinal cord. Independent of the paraplegia model tested, BBB evaluation demonstrated no significant improvement of motor functions following implantation of the conduit, with or without MSC. The histopathologic evaluation is under process.
Discussion & Conclusion: To achieve cellular regeneration and functional recovery upon SCI has been a demanding challenge leading to the development of highly complex therapeutic systems including a biomaterial device with specific characteristics and bioactive agents (cells/ molecules). Such systems should ensure suitable mechanical properties, cell-adhesion, electrical activity and biodegradability. While these parameters have been seized within neurograft development, no motor function was restored after transection+conduit implantation; demonstrated by the BBB rating evaluation. Independent of the in vivo model used, outcomes were equivalent. Regeneration of the spinal cord is still being investigated by extensive histopathologic analysis for further understanding of the ongoing repair mechanisms and future fine-tuning of the therapeutic system. Nevertheless, the neurograft conduit has demonstrated its biocompatibility, becoming valuable as a scaffold to test other combination of active molecules and/or stem cells.

Biography:

Elisa Maillard has completed her PhD from Strasbourg and Post-doctoral studies from Oxford University. She has worked on both basic research and clinical islet
isolation and culture. She is the Lab Manager of the European Center of Diabetes Study. She has published 13 articles in reputed journal such as Biomaterials.

Abstract:

Islet transplantation is a promising and minimally invasive therapy to restore normoglycaemia in brittle type 1 diabetic patients. However, the procedure is pancreas consuming since 2 to 3 pancreas are needed for a single patient. Therefore,efforts in research are focusing on improvement of islet survival during the process to decrease pancreas requirement. The stressful event of islet/exocrine separation deprives cells from extracellular matrix contact and oxygen supply; which are two of the major reasons of the loss of approximately 60% of islets. Interest had risen over the last couple of years in biomaterials in islet transplantation regarding transplantation, but also for the culture steps. The uses of biomaterials to create an artificial environment for islets post isolation increase their survival and improve transplantation outcome. In the same way, oxygen provider arouses enthusiasm of the community, and numerous teams tested the beneficial effect of oxygen supply from the pancreas retrieval to islet implantation step. The combination of both agents showed a real benefit for islet viability and function in vitro, providing more robust islet for sustaining the transplantation event. In vivo study highlighted several problems with transplantation sites, the liver. Indeed depending upon the material used, the inflammatory reaction is triggered. Therefore,alternative sites are investigated today; with, in particular, the omental pouch which gives the opportunity to keep matrices post implantation.

Biography:

Mohamed Hamdy Doweidar is an Associate Professor at the Mechanical Engineering Department, University of Zaragoza, Spain. Besides, he is a Member of the Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) at the Aragon Institute of Engineering Research (I3A) and the European Society of Biomechanics (ESB). He has participated in many national and international investigation projects. He has authored and co-authored many books and numerous articles in international journals. His investigation interests include computational biomechanics, cell simulation, finite element method, natural element method and computational fluid dynamics.

Abstract:

The mechanical stimulus is one of the essential stimuli for cell differentiation and/or proliferation. One of the recent challenges is how to apply this stimulus in a local and remote regimen. In this study, we proposed a pioneer approach to apply a mechanical local cell stimulus in a remote manner via encapsulated magnetic nanoparticles exposed to a magnetic field. This technique can help in an effective way to control the tissue regeneration process within the human body. The proposed strategy is formulated within a previously developed three-dimensional (3D) computational model, which is here extended, to study the cell behavior when it senses local changes in the mechanical properties of its extracellular matrix. We assumed that the cells are cultured within 3D nonlinear hydrogels, which is modeled as Neo-Hookean hyperelastic material. A comparison between the cell behavior within a force-free and force-induced substrates has been done for neurogenic (0.1-1 kPa), chondrogenic (20-25 kPa) and osteogenic (30-45 kPa) extracellular matrix. Our findings show that mesenchymal stem cells differentiation and proliferation can be triggered by inducing its substrate with an internal force. They need a longer time to grow and maturate within force-free substrates than within force-induced substrates. Besides, at the instance of mesenchymal stem cells differentiation into a compatible phenotype, the magnitude of the net traction force increases within chondrogenic and osteogenic substrates while it reduces within neurogenic substrates. This is consistent with experimental studies and numerical works recently published by the same authors. However, in all cases the magnitude of the net traction force considerably increases at the instant of cell proliferation because of cell-cell interaction.

David Salamon

Brno University of Technology, Czech Republic

Title: Direct building of 3D microchannel structure for nutrients delivery
Biography:

David Salamon has completed his PhD from Slovak Academy of Sciences and Post-doctoral studies from Stockholm University. He has worked 4 years at University of Twente, Netherlands and joined newly opened Central European Institute of Technology at Brno University of Technology in year 2012 as a Researcher. He has published more than 23 papers in journals, one book chapter and his research activity is mainly in area of processing of advance ceramic materials.

Abstract:

Next generation of the hard tissue scaffolds will be made to accommodate nutrient channels to support cells live inside of the 3D scaffold. Nowadays, diffusion through scaffold and a tissue usually limits transport, and forms potentially hypoxic regions. Hydroxyapatite is biocompatible material that supports cell adhesion and proliferation, therefore ideal for the preparation of bone grafts. Furthermore, chemical stability of this bioceramic material allows stable liquid flow through a porous structure. In this study, a 3D hydroxyapatite structure containing microchannels via microtemplating was prepared. Coating: The method uses micro fibers coated by hydroxyapatite suspension, which are sintered to achieve micro-channels and replicate shape of the fiber. High mechanical flexibility in the green stage allows direct shaping into 3D structure. Sintered structures contains asymmetric membrane walls with flux suitable for nutrition delivery, which keeps microchannels for nutrition support and waste removal separated from cell growth area. The metabolic activity of the cells was determined and the cell morphology was visualized. The ability for these structures to support cell adhesion and proliferation was shown to be favorable over a period of 7 days. The presented straightforward concept for building 3D structure containing tailored microchannels should be suitable especially for bioreactors applied as a bone grafts.

Biography:

Elena Afrimzon has completed her PhD from Central Institute of Immunology, Microbiology and Infectious Diseases (Alma-Ata, Kazakhstan). She is Senior Researcher at the Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and the Technology of the Cellome, Department of Physics, Bar-Ilan University. She is the initiator of the development and utilization of a novel hydrogel micro-chamber array for protracted culturing of live cells and 3D multicellular objects, which permits kinetic live cell studies. She has published more than 25 papers in reputed journals.

Abstract:

Biological tissues normally possess varying levels of rigiditiy, which contribute to the performance of their physiological functions. Changes in tissue rigidity may reflect transformation from a normal to a pathological state. Cancer cells within the tumor are influenced by the mechanical conditions of their microenvironment, which can drive cell fate. Multicellular three-dimensional models provide a more suitably organized, in vivo-like structure, and respond better to external stimuli than 2D cultures. In the present study, we propose a efficient approach to mimic the desired surrounding rigidity in vitro for 3D breast cancer object/structure formation and growth. Non-adherent, non-tethered 3D objects were generated from single cells within a hydrogel array, cultured under various mechanical conditions and measured at single-object resolution exploiting the advantageous mechanical and optical properties of agarose. This study demonstrates differences in the in vitro development of 3D breast cancer structures under various rigidity conditions. Study of individual 3D breast cancer structures reveals that significant differences in object growth rate, morphology and vital features are associated with the extent of environmental rigidity, the point in time at which a change occured and the initial number of seeded cells. The 3D objects initiated from less than six cells are significantly different from those initiated by more cells and demonstrate a growth rate independent from surrounding rigidity. Additionally, the control culture of 3D objects grown freely under low-rigidity conditions lacks the specific subset of the preinvasive phenotype which developed in the stiffer surroundings.

  • Mesenchymal Stem Cells
Location: Lubeck

Chair

Susan D Reynolds

Nationwide Children’s Hospital and The Ohio State University, USA

Co-Chair

Jan Kriz

Institute for Clinical and Experimental Medicine (IKEM), Czech Republic

Biography:

Hajime Ohgushi has completed his MD in 1976 and PhD from Nara Med University in 1980. He was a Research Associate in the Department of Biology at Case Western Reserve University USA from 1985 to 1987, Lecturer and Assistant Professor in Department of Orthopedics, Nara Medical University from 1989 to 2000.Currently, he is the Head of the Orthopedics and Deputy Director at Ookuma Hospital. He has more than 200 publications mostly related to tissue engineering/regenerative medicine and total citations are more than 10000.

Abstract:

In our body, cells having self-renewal and multi-differentiation capabilities exist in many tissues and the cells are called adult stem cells. It is well known that one of the stem cells (mesenchymal stem cells;MSCs) reside in bone marrow tissue. Number of the MSCs is very low; however the number increases after in vitro culture expansion. The cultured MSCs are currently used for various tissue regenerations in clinical settings. We have started their clinical applications towards bone tissue regeneration since 2001. We have also treated patients having cartilage damage. The MSCs used are derived from patient’s bone marrow, thus we use autogenic MSCs for the treatment. The autogenic cells do not induce transplantation immunity and can be used without fear of donor derived infection. However, autogenic MSCs cannot be used for the patients having genetic disorder which cause impairment of the targeted tissue regeneration. For example, for the purpose of bone tissue regeneration, if the patients have mutations of bone tissue-related genes, autogenic cells are hard to be used for treatments of his/her bone fragility. One of the bone-related gene is alkaline phosphatase (ALP), which is necessary for bone tissue development. The patients having the gene mutation of the ALP (Hypophosphasia) cause skeletal abnormalities resulted in short limbs and soft skull bones. Due to impairment of osteoblastic differentiation capability of the patient MSCs, we have used allogenic MSCs for treatments of the patients. This presentation summarizes clinical experiences using autogenic/allogenic MSCs for the purpose of various tissue regenerations.

Brian M Mehling

Blue Horizon International, LLC, USA

Title: Clinical applications of mesenchymal stem cells
Biography:

Brian M Mehling is a practicing American Orthopedic Trauma Surgeon, Researcher and Philanthropist. He has started his path in medicine through undergraduate study at Harvard University obtaining Bachelor of Arts and Master of Science degrees in Biochemistry from Ohio State University. Completing his Degree of Medicine at Wright State University School of Medicine, he received his Post-graduate education through Residencies and Fellowships at St. Joseph’s Hospital in Paterson, NJ and the Graduate Hospital in Philadelphia, PA, while pursuing a PhD in Chemistry. He operates his own practice, Mehling Orthopedics, in both West Islip, NY and Hackensack, NJ.

Abstract:

Musculoskeletal conditions are the most common cause of severe long-term pain and physical disability, and they affect hundreds of millions of people around the world. Osteoarthritis (OA) is the most common form of arthritis and the leading cause of chronic disability. OA is estimated to affect 630 million people worldwide. OA is characterized by degeneration of articular cartilage, limited intra-articular inflammation with synovitis and changes in peri-articular and subchondral bone.BHI Therapeutic Sciences offers a novel method of arthritis therapy using a patient’s own stromal vascular fraction (SVF) cells including mesenchymal stem cells. The therapy is available at Malacky Hospital in Slovakia. Malacky Hospital is staffed with expertly-trained medical professionals and is one of the premiere hospitals in Central Europe. Blue Horizon International Slovakia is licensed by the Ministry of Health of Slovak Republic to provide adipose stem cell treatments for orthopedic joint applications - knees, hips, shoulders and ankles. Procedures utilize cutting-edge technology and adult stem cells only. In 2015,73 patients underwent the therapy with own stromal vascular fraction cells. 64 patients have follow-up evaluation results.Affected area includes knees (37 patients), hips (25 patients) and hips and knees (2 patients). Results from patients’ follow-up examinations and MRI scans showed that stem cell therapy was safe for the patients. Follow-up examination results conducted 10 days, 3 and 6 months after treatment have shown significant improvement of clinical condition relating to pain relief, improved mobility, which was shown also on the follow-up MRI scans of the affected joints.

Biography:

Michael Weber completed his Diploma in Chemistry and Biochemistry from the University of Marburg in Germany and Medical studies from University of Göttingen,Germany. From 1974-1976, he was High School Teacher in Chemistry, Biology and Physics. From 1984-1987, he was Assistant Doctor at University clinic of Göttingen. He is authorized Educator for general medicine, natural medicine and acupuncture by the government medical association in Hannover, Lower Saxony.He is a President of the American Association of Biological Laser Therapy and Acupuncture (AALA) and President of the International Society for Medical Laser Applications (ISLA). He is a Board Member of NAALT (North American Association for Laser Therapy) and Co-editor of the Journal "Pain and Acupuncture".

Abstract:

In this study, new methods of laser therapy will be presented for use in regenerative medicine. Laser needles with different wavelengths and penetration depths can be used externally as a highly effective and pain free method for pain management.Laser needles can be applied on all parts of the body. Infrared lasers can penetrate the skull bone and are used today for benefit on brain diseases. However, penetrating laser light also initiates mitochondrial stimulation with tissue regeneration.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. Endogenous stem cells released from the bone marrow will be stimulated in the blood stream and leads to improved organ function. Interstitial and intra-articular laser therapies are new methods using a fiber optic catheter application of different lasers in the depth of the tissue or directly in joints close to the spot of injury. It is successful in treatment of herniated, nerve injuries and advanced osteoarthritis of the knee, shoulder, hip and other joints. Lasers can also invasively stimulate PRP and stem cells in joints or in the blood stream. They have excellent effects on tissue regeneration because they provide light of all spectral colors for stimulation of the mitochondria by enhancing ATP-production. So laser light activates intravenously or intra-articulary applied mesenchymal stem cells with better survival and differentiation.

Biography:

Ryang Hwa Lee is an Assistant Professor of Medicine at the Institute for Regenerative Medicine, Texas A&M University System Health Science Center. She has earned her PhD in Physiology from School of Medicine, Pusan National University in 2003 on the “Characterization of human adipose and bone marrow derived mesenchymal stem cells (MSCs)”. In 2003, she joined the Center for Gene Therapy at Tulane University Health Sciences Center as a Post-doctoral Fellow for a continuous study on MSCs with Dr. Darwin Prockop, a pioneer in the MSC field. She has joined the Institute for Regenerative Medicine in 2008 as a Research Scientist and the Faculty of Texas A&M Health Science Center in the year 2009. Currently, her research group focuses on investigating the underlying cellular and molecular mechanisms of therapeutic effects of MSCs in animal models of human diseases including sterile inflammation-mediated injury, type 1 diabetes and cancer.

Abstract:

Human mesenchymal stem/progenitor cells (hMSCs) from bone marrow and other tissues are currently being administered to large numbers of patients. The trials are proceeding even though cultures of the cells are heterogeneous, and there is large variability among preparations of hMSCs due to differences among donors, culture conditions, and inconsistent tissue sampling. This is also confounded by the lack of definitive biomarkers to predict the efficacy of hMSCs in vivo. Therefore,the value of the data obtained from current clinical trials could be compromised by variations in the quality of the hMSCs employed. To ensure such trials continue generating quality data, there is a critical need for simple in vitro bioassays to predict hMSC efficacy in vivo. This presentation will provide the first biomarker that can predict the efficacy of hMSCs in suppressing sterile inflammation in vivo.

Biography:

Karen Coopman is a Senior Lecturer in Biological Engineering at Loughborough University. Based at the Centre for Biological Engineering, her research team focuses on “the manufacture of cell-based therapies, tackling issues from scale-up to cryopreservation”. She is also the Director of the Loughborough led EPSRC/MRC Centre for Doctoral Training in Regenerative Medicine and a Member of the BBSRC led Bioprocessing Research Industry Club Steering Group.

Abstract:

With the growth of the cell-based therapy industry, one of the key challenges in the field is the successful preservation of these therapies in order to enable centralized manufacture of an 'off-the-shelf ' allogeneic product. Data from our work on human mesenchymal stem cells (hMSCs) is now emerging which shows the impact that changes in downstream processing (e.g. cell harvesting from microcarriers, washing and re-suspending cells in cryopreservation medium) can have on cell recovery post-thaw despite a consist freezing process being utilized. The idea of optimizing each unit operation in isolation will therefore be questioned and the necessity for end-to-end process design discussed.

Biography:

Irina Aizman has completed her PhD from Tel Aviv University and since then worked as a Research Scientist in California at Fibrogen and SanBio. Currently, she is an Associate Director of Research at SanBio.

Abstract:

Beneficial effects of intracerebral implantation of mesenchymal stromal cells (MSC) and their derivatives are believed to be mediated mostly by factors produced by these cells. However, mesenchymal cell engraftment does not occur, and the majority of cells disappear within a week of implantation. Here, we provide in vitro evidence that dying implanted cells can affect surrounding tissues by releasing their intracellular components. We found that FGF2 and FGF1, but not VEGF and MCP1, levels were high in extracts of mechanically damaged (freeze/thaw) cells despite being low in conditioned media (CM). Extracts induced concentration-dependent proliferation of rat cortical neural progenitor cells (NPC) and human umbilical vein endothelial cells (HUVEC); these proliferative responses were specifically blocked by FGF2-neutralizing antibody. In the neuropoiesis assay with rat cortical cells, both MSC extracts and killed cells induced expression of nestin, but not astrocyte differentiation. However, suspensions of killed cells strongly potentiated the astrogenic effects of live MSC. In implantationrelevant MSC injury models (peripheral blood cell-mediated cytotoxicity and high cell density plating), MSC death coincided  with the release of intracellular FGF2. The data showed that MSC contain a major depot of active FGF2 that is released upon cell injury and is capable of acutely stimulating neuropoiesis and angiogenesis. We therefore, propose that both dying and surviving implanted MSC contribute to the tissue regeneration.

Biography:

Lucilla Lecchi has completed her Degree in Biological Sciences at the University of Milano, Italy. She is the Processing Facility Director of the Milano Cord Blood Bank and responsible of the laboratory for cryopreservation of HPC-A/HPC-M at Centro Trasfusionale Fondazione IRCSS Ca’ Granda Ospedale Maggiore Policlinico, Milan,Italy. She has published more than 59 papers in reputed journals.

Abstract:

This presentation will focus on critical issues related to stem cells cryopreservation procedures of minimally manipulated products containing allogeneic or autologous hematopoietc progenitor cells (HPC). The issues include: Regulations and standards, process design, process validation and qualification, volume reduction, cell concentration, cryoprotectants and cryopreservation, storage, warming events, quarantine, cross contamination during storage and thawing. New approaches of processing are developed such as automatic devices for volume reduction and high cell concentration in the frozen product.DMSO at 10% final concentration is still the most used cryoprotectant for HPC cryopreservation. Although controlled rate freezing is the recommended method for HPC cryopreservation, alternative methods may be used. Last generation vapor storage vessels ensure temperature stability better than older tanks. Moreover, advantages and disadvantages of thawing procedures carried out at the patient’s bedside or in the laboratory will be discussed.

Biography:

Noel D is currently a Research Director in the Inserm Institute for Regenerative Medicine and Biotherapies in Montpellier. She has received her PhD from Bordeaux University in 1992. She then completed Post-doctoral studies at the Institute of Molecular Genetics in Montpellier in the field of Recombinant Retrovirology and Gene Therapy. Since 1999, she works on mesenchymal stem cells and their applications in the treatment of osteo-articular diseases. She has published more than 138 papers in reputed journals and has been serving as Editorial Board Member.

Abstract:

Because of immune-modulatory and trophic properties, mesenchymal stem/stromal cells (MSC) appear as a promising treatment for systemic sclerosis (SSc), a rare intractable disease with fibrosis-related mortality. While autologous approaches could be inappropriate because of alterations in SSc-MSCs, we evaluated allo- and xenogeneic approaches in the relevant hypochlorite (HOCl)-induced murine model of diffuse SSc, recapitulating the main features of the disease. Therefore, we investigated the effect of murine and human (h) MSC in the HOCl-induced SSc model. Subcutaneous fat being an accessible source of MSC, we also compared human bone marrow (BM-MSC) with adipose-derived MSC (ASC). We therefore assessed the effect of BM-MSC from syngeneic BALB/c, allogeneic C57BL/6 mice and xenogeneic hBM-MSC or hASC (3 donors each) in HOCl-challenged mice. We demonstrated that allo- and xenogeneic BM-MSC were as effective as syngeneic BM-MSC in decreasing skin thickness, expression of Col1, Col3, a-SMA and collagen content in skin and lungs. Compared with hBM-MSC, hASC induced a similar reduction in Col1 and a-SMA expression, but a stronger reduction of TNFa, IL1b, and enhanced ratio of Mmp1/Timp1 in target tissues. This therapeutic efficacy was mainly associated with paracrine activity since MSC did not migrate to the skin. Our results indicated similar therapeutic effects using allo-/xenogeneic BM-MSC while hASC displayed potent anti-inflammatory and remodeling properties. Considering heterogeneity between MSC sources and samples, potency assays are needed to optimize MSC-based therapy outcomes in SSc.

Biography:

Abstract:

Hematopoietic stem cells (HSCs) are responsible for blood cell production throughout the lifetime of an individual.However, aging or repeated cell division induces the accumulation of DNA damage, which impairs HSC self-renewal and differentiation capacities. Here, we show that protection of telomeres 1a (Pot1a), a component of shelterin, improves HSC activity under stress. We identified that Pot1a was highly expressed in HSCs, yet this expression declines with age. Knockdown of Pot1a in young HSCs increased DNA damage response (DDR) at telomeric region and induced age-related phenotypes,marked by reduced long-term reconstitution activity and myeloid-biased differentiation. In contrast, overexpression of Pot1a or treatment with exogenous Pot1a protein prevented telomeric DDR and maintained HSC self-renewal activity and rejuvenated the activity of aged HSCs. In addition, we found that the transduction of Pot1a into LT-HSCs reduced production of reactive oxygen species compared with control, indicating the possibility that Pot1a implicates in the energy metabolism of HSCs.Furthermore, human POT1 protein also increased the number of cord blood HSCs. These results indicate that Pot1a/POT1 has an important role in the maintenance of stem cell function under stress condition and can be used to expand functional HSC numbers ex vivo.

Biography:

Vsevolod A Tkachuk has completed his PhD from Moscow State University and DSc from Russian Cardiology Research Center, Moscow. He is a Professor of Biochemistry, Director of Institute of Regenerative Medicine and Chair of Biochemistry and Molecular Medicine in Moscow State University. He has published more than 250 papers in reputed journals and has been serving as an Editorial Board Member of repute.

Abstract:

We investigated effects of adipose derived human mesenchymal stromal cells (MSC) on ischemic limbs of nude mice.Data obtained demonstrate formation of new arterioles and nerve growth with cells and proteins originating from mice. Using of the medium obtained after MSC growing had the same effect on ischemic limbs but stability of new formed arterioles and nerves was much less than after using MSC. It means that MSC stimulates angiogenesis and nerve formation via secretory mechanism, but direct interaction of MSC with ischemic tissue stabilizes regenerated tissue. Proteomic analysis demonstrated secretion by MSC number of angiogenic and neurotrophic factors, as well as matrix metalloproteasis and matrix proteins, anti-inflammatory cytokines and chemokines. Using neutralized antibodies, we demonstrated involvement of some of these proteins in angiogenesis and axons growth. At the same time, exclusion of microvesicles and exosomes from this medium blocked both angiogenic and neurotrophic effects, and using of exosome fraction reconstruct action of medium on ischemic limb regeneration. Such phenomenon can be explained by action of microRNA included in exosomes, since some anti-microRNA block physiological action of exosomes. Treatment of MSC with PDGF or bFGF dramatically changes spectrum of proteins secreted by MSC and spectrum of microRNA included in exosomes as well as physiological effects exosome fraction on blood vessel growth and stability. In our report, we will also discuss the role of MSC in stroma formation of tissues, effect of MSC on some adult stem cells and T-lymphocytes.

Biography:

Jiunn-Jye Sheu is a Cardiovascular Surgeon in Kaohsiung Chang Gung Memorial Hospital, Taiwan. He has completed his graduation from Taipei Medical University.Currently, he is pursuing his PhD with major research focused on “plasma generating machine and its wound healing effect”. He has published more than 75 scientific papers in PubMed. His research interest includes heart remodeling and heart muscle regeneration.

Abstract:

Background: We hypothesized that combined therapy with shock wave (SW) and autologous bone marrow-derived mesenchymal stem cells (BMDMSCs) are superior for alleviating left ventricular (LV) dysfunction.
Methods & Results: Male mini-pigs (n=30) equally divided into group one (sham control), group two [Acute myocardial infarction (AMI) by left coronary artery ligation], group three (AMI-SW), group four (AMI-BMDMSC) and group five (AMISW-BMDMSC) were sacrificed by day 60 and the hearts were collected for studies. Baseline LV injection fraction [LVEF (%)] and LV chamber size did not differ among the five groups (p>0.5). By day 60, LVEF was highest in group one and lowest in group two, significantly higher in group five than that in groups three and four, and significantly higher in group four than that in group three (p<0.001). Cellular and protein levels of VEGF, CXCR4, and SDF-1α were significantly increased progressively from groups one to five (all p<0.05). Small vessel number and protein expressions of CD31 and eNOS were highest in groups one and five, lowest in group two, and significantly higher in group four than those in group three (p<0.001). Protein (MMP-9, TNF-1α and NF-κB) and cellular (CD14+, CD40+) levels of inflammatory biomarkers, protein expressions of oxidative stress (oxidized protein, NOX-1, NOX-2), apoptosis (Bax, caspase-3, PARP), infarct size, and LV dimensions showed a pattern opposite to that of LVEF among all groups (all p<0.001).
Conclusions:Combined SW-BMDMSC therapy is superior for improving LVEF, reducing infarct size and inhibiting LV remodeling.

Susan D Reynolds

Nationwide Children’s Hospital and The Ohio State University, USA

Title: Stem cell treatments for lung disease
Biography:

Susan D Reynolds has completed her PhD in 1992 from the University of Rochester with a Specialization in Developmental Biology and Post-doctoral studies in Lung Stem and Progenitor Cell Biology from the University of Rochester, School of Medicine. She is currently a Principal Investigator at Nationwide Children’s Hospital. She has published more than 70 papers in reputed journals and has been serving as a Reviewer for multiple scientific journals and granting agencies.

Abstract:

Cell therapy has the potential to cure disease through replacement of malfunctioning cells. While the tissue stem cell (TSC) is thought to be the optimal therapeutic cell, transplantation of TSC/progenitor cell mixtures has saved lives. We previously purified the mouse tracheobronchial epithelial TSC and reported that in vitro amplification generated numerous TSC. However, these cultures also contained TSC-derived progenitor cells and TSC re-purification by flow cytometry compromised TSC self-renewal. These limitations forced us to determine if a TSC/progenitor cell mixture would repopulate the injured airway epithelium. We developed a clinically-relevant transplantation protocol and demonstrate that transplanted mouse and human tracheobronchial epithelial TSC/progenitor cell mixtures are 20-25% of airway epithelial cells, actively contribute to epithelial repair and persist for at least 43 days. Two weeks after transplantation, TSC/progenitor cells differentiated into the three major epithelial cell types: Basal, secretory, and ciliated. We concluded that adult tracheobronchial TSC/progenitor cell is an effective cell therapy.

Biography:

Valerie Trichet has completed her PhD from Rennes University and Post-doctoral studies from Edmonton University, Canada and Nantes University, France. She is an Associate Professor at the Faculty of Medicine, University of Nantes in France. She has joined the Laboratory of the Pathophysiology of Bone Resorption (INSERMUMR957) in the year 2006. Her current research interests are focused on understanding the role of Mesenchymal Stem/Stromal Cells in Bone Regeneration and in Primary Bone Tumors.

Abstract:

Conventional therapy of primary bone tumors includes surgical excision with wide resection, which leads to physical and aesthetic defects. For reconstruction of bone and joints, allograft can be supplemented with mesenchymal stem cells (MSCs) or adipose-derived stem cells (ADSCs). Additionally MSC-like cells may be used in tumor-targeted cell therapy.However, we wanted to know whether MSC-like cells may have adverse effects on osteosarcoma development. MSCs/ADSCs were co-injected with human MNNG-HOS osteosarcoma cells in immunodeficient mice and have accelerated the local tumor growth. This pro-tumor effect may be due to MSC/ADSC secreted factors as the in vitro proliferation of osteosarcoma cells was increased up to 2 folds in the presence of MSC/ADSC-conditioned medium. Because of the enhancing effect of MSCs on in vivo/in vitro proliferation of osteosarcoma cells, MSCs may not be good candidates for osteosarcoma-targeted cell therapy. In contrast, MSC-conditioned medium did not change the dormant, quiescent state of osteosarcoma cells cultured in oncospheres.This result indicates that MSC-secreted factors may not be involved in the risk of local recurrence and that MSCs may be safe in tissue reconstruction following bone tumor treatment.

  • Bone Tissue Engineering | Scaffolds in Regenerative Medicine
Location: Lubeck

Chair

Danièle Noel

INSERM, France

Co-Chair

Hans-Georg Simon

Northwestern University, USA

Session Introduction

Menemşe Gümüşderelioglu

Hacettepe University, Turkey

Title: Boron-doped bone scaffolds
Biography:

Menemse Gumudderelioglu is a Professor at Hacettepe University, Chemical Engineering Department since 1998. She has completed her BSc, MSc and Doctorate degrees from the same department. Her main research subjects are biomaterials, controlled release technology, animal cell biotechnology and tissue engineering.She has completed her research studies at London University, Birkbeck College, Harvard University, Medical School, Tufts University and Stratchlyde University. She has more than 100 papers in international peer reviewed journals.

Abstract:

Although boron (B) is increasingly identified as an element that has many benefits especially on bone health, limited number of studies that are investigating the effects of B in osteoblasts in cellular/molecular level are available. Therefore, the exact mechanism of B on bone health is still unknown. However, it is known that B has an important role on the metabolism of calcium, magnesium, vitamin D and steroid hormones which have many indirect effects on bone. The aim of this study was to develop boron-containing polymeric scaffolds to promote regeneration of bone tissue. The scaffolds were prepared by using two different approaches: Embedding of boric acid encapsulated nanoparticles into the scaffolds to achieve sustained B release; combining of polymeric scaffolds with B containing hydroxyapatite (HAp) that is produced by microwave-induced biomimetic method. The scaffolds were produced and characterized in terms of structural properties and in vitro B-release patterns. The effect of released B on the osteogenic activities of MC3T3-E1 preosteoblasts and mesenchymal stem cells derived from bone marrow and adipose tissue, seeded into the scaffolds was investigated in vitro. The results were evaluated with respect to cell viability, bone related ECM gene expressions, and cellular morphology. In conclusion, the cell culture studies proved that the encapsulated boron within the scaffolds can be used as an osteoinductive agent by showing its positive effects on the proliferation and differentiation of MC3T3-E1 cells and MSCs.

Biography:

Andrew R Cohen has joined the faculty in the Department of Electrical and Computer Engineering at Drexel University as an Associate Professor in August 2012.Before coming to Drexel, he was an Assistant Professor at the University of Wisconsin-Milwaukee. He has received his PhD from the Rensselaer Polytechnic Institute in May 2008. He was previously employed as a Software Design Engineer in the DirectX group of Microsoft where he designed operating system software for gaming applications. He is an Associate Editor in the Bioimage Informatics section of the journal BMC Bioinformatics and is a Senior Member of the IEEE.

Abstract:

The ability to characterize cell culture function from a combination of visual and bio-chemical assays will be a key advantage for applications in regenerative medicine. For visual analysis, microscopes are routinely able to image living proliferating cells together with vessels and organelles. The resulting images and movies capture behaviors in live tissue, preserving the cellular microenvironment and spatiotemporal dynamics of development. The imaging data can be combined together with non-imaging (*-omics) data, enabling a comprehensive study of the complex dynamics of disease and development. Transmitted light microscopy allows evaluation of the cells intended directly for therapeutic application, while fluorescence microscopy modifies samples from the cell culture but enables a more detailed bio-chemical examination. In order to make best use of this data, computational tools are required to provide a rigorous and objective characterization of the visual information content of both single images and time-lapse image sequences (movies). This talk will describe a key and under-served area of live cell microscopy–the software tools that are required for the objective and quantitative analysis of the imaging data. LEVER, (lineage editing and validation) a free and open-source collection of algorithms and software tools for measuring clonal development in 5-D images will be described and new visualizations for evaluating cell and clone development will be demonstrated. New approaches for characterizing cell culture morphology from single and time-lapse images will also be described. Examples of measuring development from microscopy images in the neural and hematopoietic systems will be shown.

Biography:

Oded Shoeyov is a Faculty Member of the Hebrew University of Jerusalem. His research is in plant molecular biology, protein engineering and nano-biotechnology.He has authored or co-authored more than 170 scientific publications and is the Inventor or Co-Inventor of 46 patents. He has received the 2012 Israel Prime Minister Citation for Entrepreneurship and Innovation. He is the scientific Founder of 10 companies; among them are Collplant Ltd., an agro-biotech/regenerative medicine company producing human recombinant type-I collagen in transgenic plants for medical implants used in tissue repair.

Abstract:

Consequential to its essential role as a mechanical support and affinity regulator in extracellular matrices, collagen constitutes a highly sought after scaffolding material for regeneration and healing applications. However, substantiated concerns have been raised with regard to quality and safety of animal tissue-extracted collagen, particularly in relation to its immunogenicity, risk of disease transmission and overall quality and consistency. In parallel, contamination with undesirable cellular factors can significantly impair its bioactivity, vis-a-vis its impact on cell recruitment, proliferation and differentiation.Large scale production of recombinant human collagen type-I (rhCOL1) in the tobacco plant provides a source of a homogenic, heterotrimeric, thermally stable “virgin” collagen which self assembles to fine homogenous fibrils displaying intact binding sites and has been applied to form numerous functional scaffolds for tissue engineering and regenerative medicine which have already reached the clinic. In addition we have shown that rhCOL1 can form liquid crystal structures, yielding well-organized and mechanically strong fibers 6 times tougher than human achiles tendon, two properties indispensable to extracellular matrix (ECM) mimicry. Combining resiline, an elastic protein derived from insects, further increased collagen fiber toughness and strain at break. Injections of fibrillated rhCOL1 combined with PRP (platlet rich plasma) to patients suffer from tendinopathy (in tennis elbow) forms in situ matrix composed of collagen and fibrin. The rhCOL1/PRP matrix reduces inflammation and promotes cell proliferation and tissue healing, promotes in situ growth factors generation and release while gradually degrading. Both in vivo and in vitro studies showed superior performance of rhCOL1/PRP compared to PRP alone.

Biography:

Antoine Alves is a Senior Pathologist and Medical Research Manager at NAMSA Lyon, France. He has more than 15 years of experience at NAMSA providing interpretation of tissue/implant interaction according to international standards under GLP conditions. Development of new histopathology procedures for adequate evaluation of medical devices and advanced therapies is one of his lines of research. He has published many papers and book chapters.

Abstract:

Cartilage, unlike bone, does not possess autogenous repair capability and has very limited ability to heal. It has been confirmed that cartilage injuries in the knee joint, if left untreated lead to more serious degenerative joint conditionsn (arthritis) and affect the activities of daily living with severe pain and dysfunction (disability). Cartiheal (Israel) recently CE marked the first in class acellular implant, Agili-C™, designed for joint resurfacing with simultaneous regeneration of hyaline cartilage and subchondral bone. The off-the-shelf implant, applied in a single stage procedure is composed of aragonite with hyaluronic acid functionalization at the cartilage phase. A long term (6 and 12 months) evaluation of the safety and efficacy in a 20 adult goats has been conducted following immediate load bearing: In the Agili-C TM group, the defects were healed and filled with a hyaline cartilage as confirmed by the marked presence of collagen type II and proteoglycans and absence of collagen type I. The subchondral plate and tidemark were reconstructed, whereas a poor repair was observed in the shamoperated group. Two clinical trials involving 148 patients showed over 24 months, a significant improvement of the primary endpoint KOOS score (pain, symptoms, quality of life) with a low rate of adverse events. Second look arthroscopies and MRI confirmed a high level of resurfacing.

Dana Kubies

Institute of Macromolecular Chemistry AS CR, Czech Republic

Title: Polymer porous scaffolds for transplantation of Langerhans Islets
Biography:

Dana Kubies has completed her PhD from the Institute of Macromolecular Chemistry AS CR (IMC), Prague, Czech Republic and Post-doctoral studies from University of Liege, Belgium. She is the Head of the Department of Bioactive Polymers at IMC, a leader polymer institute in the central Europe. She is interested in the application of polymer scaffolds in biomedical applications. She has published more than 28 papers in reputed journals.

Abstract:

The pancreatic islets transplantation (ITx) is a therapeutic alternative for patients with type-1 diabetes mellitus suffering from the hypoglycemia unawareness. The current protocol of ITx is limited, among other factors, by a suboptimal engraftment of islets after infusion into the hepatic portal vein. This results immediately in significant reduction of the graft and in unsatisfactory long-term graft viability predominantly due to an insufficient vasculature and supraphysiological nutrients,drugs and toxins concentration within portal vein blood. Therefore, a considerable effort has been spent to bioengineer an extra-hepatic transplant site rich in vasculature network similar to situation in native pancreas. We aimed to create a highlyvascularized bioartificial cavity using a macroporous polylactide-based scaffold with anisotropic channeled pores as a template,and a host body as the bioreactor. The capsular-shaped scaffolds were prepared using the Dip-TIPS method. We evaluated the effect of the scaffold pore architecture, implantation site and introduction of the vascular endothelial growth factor on the scaffold vascularization. In vivo studies (a rat model) showed the ability of the scaffold pore architecture to guide cell infiltration fully in omentum and partially when implanted subcutaneously. The in vivo magnetic resonance imaging data and immuno-histochemical analysis revealed the enhanced vascularization of the VEGF/scaffolds when compared to unmodified ones. The current study is a significant step forward in the field of biomaterial based extra-hepatic site for islets transplantation.

Biography:

Yuan-shan Zeng is a Professor of Histology and Embryology. He has completed his PhD in Histology and Embryology from West China University of Medical Sciencesin 1991. In 1998, he took Post-doctoral fellow training at Department of Anatomy, Indiana University. He has worked as a Visiting Professor at Department of Anatomy,the Hong Kong University in 2003. He is currently appointed as Head of Department of Histology and Embryology and Vice Director at Institute of Spinal Cord Injury in Sun Yat-sen University. His research program is mainly focused on “The stem cell transplantation and mechanism of repairing of spinal cord injury”. So far, he has published more than 50 papers on international academic journals.

Abstract:

Functional deficits following spinal cord injury primarily attribute to neural cell death and loss of connectivity. Therefore,we designed tissue engineered neural network acting as bridge to rebuilt signal connection between the cut ends of the ruptured spinal cord. Adult stem cells with genetically enhanced expression of tropomyosin receptor kinase C (TrkC) were induced into neuron-like cells after co-cultured with Schwann cells over-expressing neurotrophin-3 in a gelatin sponge scaffold for 14 days. The formation of stem cell-derived neural network was confirmed by electron microscope and recording of spontaneous postsynaptic currents by whole-cell patch clamp. Then, this neural network scaffold was grafted acutely into rats with removal of 2 mm spinal cord tissue. Eight weeks later, stem cell-derived neuron-like cells of the grafts maintained their synaptic connection or formed new connection with regenerative axons. Although biotinylated dextran amine labeled fibers of corticospinal tract only formed a few connections with stem cell-derived neuron-like cells in the grafts, 5-HT labeled fibers formed much more connections. Furthermore, part of these connections was confirmed to be synaptic connections by doublelabeled immunoelectron microscope. Animals with neural network scaffold transplantation resulted in higher BBB score and were improved cortical motor evoked potential. These findings indicate that stem cell-derived neuron-like cells resulting from NT-3/TrkC-induced differentiation can partially integrate into host neural network in rat spinal cord transected. The stem cell-derived neural network in the transection site of spinal cord may accept ascending or descending neural information,meanwhile, transduct it to the host neurons located at both sides of end of the transection site.

Biography:

Bipasha Bose holds a PhD degree in Cancer Biology from Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai,India since 2004. She has about 12 years of Post-PhD Research Experience working in Stem Cell Biology, in academia and industry in India, Belgium and Singapore.She holds key skills in the field of clinical, as well as, R&D grade stem cells. She has several international publications and two patents to her credit. Her present focus is basic biology of adult CD34+/45- stem cells and also various aspects of ocular stem cells and cancer stem cells.

Abstract:

From the perspective of cell therapy and understanding the basic biology of adult stem cells residing in tissues of the different germline origin, we have isolated and characterized CD34+/CD45- stem cells from various organs, other than bone marrow. These studies were aimed at establishing a proof of concept for the translational applicability of such cells in preclinical animal model system. Till now, in clinical scenario, bone marrow has been the exclusive source for CD34+/CD45- for all the therapeutic applications. The advantage of using other cell types for obtaining CD34+/CD45- is avoiding the invasive bone marrow collection procedure. The presence of CD34+/CD45- cells in liver (endoderm), skin (ectoderm) and muscle(mesoderm) were established using immunofluorescence staining. The CD34+/CD45- were isolated from liver, muscle and skin using FACS sorting. Also, CD34+/CD45- cells from muscle had proven to ameliorate muscular dystrophy in mouse model of muscular dystrophy.

  • Poster Presentations
Location: Foyar
  • Young Researcher Forum
Location: Lubeck
Biography:

Claudia Tresoldi is currently a PhD student in Bioengineering at Politecnico di Milano, Italy, where she earned her Master’s degree. Since 2013, she has been working on a project concerning Vascular Tissue Engineering. The aim of her investigation is to develop functional small-caliber vessels able to reproduce the fetal development in vitro by coupling two different technological aspects.

Abstract:

The in vitro simulation of physiological mechanical conditioning through bioreactors plays a pivotal role in the development of functional tissue-engineered blood vessels. Thus, we developed a scaffold-specific fluid-structure interaction (FSI) model under pulsatile perfusion provided by a bioreactor in order to estimate the flow rate that ensures physiological circumferential deformations (εcirc) and wall shear stresses (WSS) on cells seeded on the tubular scaffold. The 2D-axial symmetric FSI model,computed by COMSOL Multiphysics 4.4, represents two domains: The former schematizes decellularized swine artery scaffold,defined as a linear viscoelastic material characterized by its elastic modulus and shear relaxation modulus, previously obtainedby uniaxial stretch tests. The latter domain represents the culture medium, defined as an isotropic, homogeneous, incompressible and Newtonian fluid. A pulsatile and parabolic velocity profile is prescribed at the model inlet, while calculated pressure is prescribed at the scaffold outlet. The pressure is estimated at the scaffold end by solving boundary ordinary differential equations, in relation to construct deformations and to the bioreactor downstream lumped-parameter hydraulic circuit. Our results indicate that the FSI-simulated εcirc-max and εcirc-min are statistically comparable to the experimentally estimated values at the considered flow rate, showing the maximum around 10%. The computed WSS values are in the range of 0.175-2.940 dyne/cm2. Both εcirc (≤10%) and WSS (≤20 dyne/cm2) fall within the physiological range for vascular cells. Therefore,the in silico FSI model well describes scaffold mechanical conditioning when subjected to pulsatile perfusion, properly driving its’ in vitro physiological maturation using scaffold mechanical properties obtained experimentally.

Biography:

Malte Bieber has completed his Master of Science in Engineering from RWTH Aachen University and started his PhD in July 2016 at the Institute of Heat and Mass Transfer.

Abstract:

Stem cell therapy at diseased tissue requires high local concentrations of stem cells. State of the art represents the intravenous stem cell transplantation, leading to distribution of cells within the entire body. Increase of cell concentration is exclusively achieved by utilizing tissue homing, i.e. the property of cells to autonomously migrate to afflicted tissue. An innovative approach aims at high local cell concentrations based on a minimal invasive surgery through flexible endoscopy, by spraying the cells directly onto the area of treatment. Thereby, the endoscope's working channel in combination with a commercially available catheter is utilized for injection of the liquid cell suspension and liquid atomization by a coaxial air stream. Within the realms of endoscopy, the device enables local application of cells, but currently no information is available regarding the influence of atomization parameters on cell survival. Therefore, a first-time parametric study on atomization of cell-suspensions has been performed in this study. The mutual interaction of cells and atomization is experimentally analyzed, aiming at correlations between air velocity, droplet sizes and cell survival rates. The influences of catheter flow, atomization and droplet wall interaction on cell survival are quantified and thus the extensional flow during ligament formation is identified as main cell destruction mechanism. On the basis of the present endoscopic configuration, integral cell survival rates above 80% are achieved, offering the opportunity for optimized homogeneous local coating in tubular shaped bodies.

Biography:

Rajan Narayan has completed his MSc in Biochemistry from Bangalore University in 2010. He has worked as a Scientist at Biocon Limited, in Bangalore, India for a year and then joined the Department of Biotechnology, Indian Institute of Technology Kharagpur in 2011 to pursue PhD in Vascular Tissue Engineering. He has published 4 papers in journals of international repute.

Abstract:

Developing a functional three-dimensional large sized vascularized tissue has been a longstanding objective in the field of tissue engineering. Although the current strategies employed to meet this challenge give stable vessels when grown with mural cells, however it takes weeks to months to get anastomosed and perfused vasculature. Micro fabrication techniques;despite showing great potential, are expensive, requires high expertise and have employed microfluidic chip to develop small dimension vasculature. Modular tissue engineering is an emerging field that generates tissue structures from the bottom-up that mimic the intricate architecture and complexity of native organs and tissues. Vascularization by this approach has been attempted by researchers; however, this approach is not well explored. Previously, we had optimized the in vitro parameters for vascularization via this approach and have shown that endothelialization using human umbilical vein endothelial cells (HUVEC) microtissues (MTs) grown in goat tendon collagen type I-fibrin hydrogel were interconnected, branched and were stable for at least 5 days without mural cells, however; it’s in vivo validation was not performed. In the current study, an attempt was made to validate our in vitro developed 3D vasculature in an immunocompromised mouse in presence and absence of growth factors. Results showed that MTs underwent different stages of evolution and formed interconnected, anastomosed and perfused vasculature without any supporting cells within 12 days post implantation and eventually became more mature by 21 days. Immunohistochemical analysis showed that the neovessels were composed of human endothelial cells. H&E staining showed presence of mouse RBCs in the newly formed vessels, clearly indicating that anastomosis has occurred. These HUVEC MTs if grown with desired cell type may result in a functional engineered tissue construct of clinical relevance.

Biography:

Nidal Ghosheh is currently a PhD student in Molecular Biology at the Institute of Bioscience from the University of Skövde and University of Gothenburg. She is investigating the gene expression and the DNA methylation profile of the genome of human pluripotent stem cells during the differentiation into hepatocyte cells.

Abstract:

Over 600 million people worldwide suffer from chronic liver diseases. End stage liver diseases are treated mainly by organ or cell transplantations. However, shortage of transplantable donor livers requires the search for alternative treatments.Human pluripotent stem cells (hPSCs) are characterized by their unique capacities of self-renewal and differentiation into principal mature cell types of the human body. Therefore, hPSCs provide an excellent source of human cells for use in basic research, drug discovery, tissue engineering, and regenerative medicine. Human PSCs have been successfully differentiated into hepatocytes, which are the dominating functional cell type in the liver. However, fully functional hPSC-derived hepatocytes have still not been achieved. By applying a standardized protocol we have shown the synchronicity of the hepatic differentiation across several hPSC-lines. Analyzing gene expression on the global level demonstrated a high reproducibility of the differentiation procedure and revealed very low variation between biological replicates. Moreover, transcriptional comparison between hPSC-derived hepatocytes and liver tissues identified a set of genes involved in the metapathway bio transferase and oxidation by cytochrome P450 such as CYP2B6, CYP2E1, UGT2B4, FMO4, AKR1C4, and GLYAT; however that show low or no expression in hPSC-derived hepatocytes. Interestingly, some of the cytochrome P450 genes have previously shown to be down-regulated due to epigenetic regulation. Hence, further epigenetic investigation is required to understand the abnormal regulation of differentially expressed genes in hPSC-derived hepatocytes in order to develop strategies for rectifying their expression and generate fully functional hepatocytes.

Katarzyna Mazur

Technical University of Gdańsk, Poland

Title: New finite element with variable young's modulus
Biography:

Abstract:

Image of local tissue stiffness variation in one bone area is available through Computer Tomography. There is the relation between Young’s modulus and radiographic density (expressed in Hounsfield’s units) using an intermediate relation of mineral density ρ and radiological HU. Creating FEM bone model we assign one finite element to one pixel on the tomographic image. In FEM models created with this relation occur major stress concentrations areas that have no reflection in reality.Stress concentrations are caused by notches that create between elements with different Young’s Modulus (in fact bone’s density changes smoothly, in digital CT image changes stepwise). We can prevent it by making CT scans with better resolution but it entails higher radiation dose for the patient and provide no additional medical information. Our propose is to modify FEM calculations by adding to them formula describing various material properties so we get a better model with even less image resolution. This method gives very good results both in plane and solid elements.From the left, we can see high resolution (61x61 mesh) calculations results of the sample (varying properties in its volume)loaded by compressive force acting on its upper edge and supported on the opposite edge. The middle figure is FEM calculations without any changes (3x3 mesh) while on the right figure there are results of calculations with proposed changes made with the same low mesh. In a speech, we will present the results of similar calculations for solid FEM models i.e. those that have not only study but also medical use.

Biography:

Shreyas Kuddannaya is currently a PhD student at Nanyang Technological University. He has completed his Master’s degree from the Delft University of Technology in Netherlands. His PhD research is focused on studying the surface-chemical and micro/nano mechanical environments on mammalian cell behavior in the context of tissue regeneration. He has published in several reputed peer reviewed journals and has presented his research in reputed platforms in bioengineering.

Abstract:

The interactive influence of chemical and topographic microenvironments in the immediate vicinity of anchorage dependent cells such as the mesenchymal stem cells (MSCs) has been a central theme of many studies involving stem cell based tissue regeneration. In this regard, the optimal adhesion of mammalian cells is critical in determining the cell viability and proliferation on substrate surfaces to facilitate long term in vitro studies on cell behavior and regenerative mechanisms. PDMS based microsystems have been greatly used for such in-vitro studies at both cellular and tissue level architectures. However, due to the inherent high hydrophobicity of a polydimethylsiloxane (PDMS) surfaces, cell culture on these surfaces is unfavorable, causing cells to eventually dislodge from the surface. Although physically adsorbed matrix proteins can promote initial cell adhesion, this effect is usually short-lived. To address this issue, a surface chemical modification of PDMS was performed for covalent immobilization of ECM proteins through (3-aminopropyl)triethoxy silane (APTES) and cross-linker glutaraldehyde (GA) cross-linking chemistry. Further, PDMS surfaces developed with micro-pillar and hole (H: 30 μm and D: 30 μm) based topographical features with grooved and non-grooved features obtained by photolithography were included in this study to study the combinatorial substratum effects on MSCs. The hydrophobicity of the native PDMS decreased significantly with the mentioned surface functionalization with a higher anchorage of ECM proteins. Both, surface chemical and topographical features resulted in an enhanced adhesion, proliferation and differentiation of mesenchymal stem cells into osteogenic lineage. We believe that these strategies could also be applied to several other substrate materials by appropriate combinations of self-assembled monolayers (SAMs), ECM proteins and substrate topography to obtain a realistic understanding of cellmicroenvironment interactions which influence critical cell behaviors in the realm of cell based regenerative therapies.

Biography:

Atipat Patharagulpong is currently pursuing PhD from the University of Cambridge, UK.

Abstract:

Topography and mechanical stiffness of the substrate has been shown to influence neural stem cell differentiation. Herein, near-field electrospinning (NFES) was employed to provide a topographically aligned polymeric fibrous scaffold for neural stem cell differentiation, which mimics the similar process found in the brain cortex and the peripheral neurons. Unlike conventional 3D aligned fibers, 2D NFES fibers allow progressive imaging of the cells grown on top of the fibers to study various differentiation parameters such as neurite length and the number of neurites per cell continuously. The expression profile of neuronal marker, Map2, could be used to evaluate how much the stem cells have differentiated into neurons in different scenarios. Neuronal differentiation was found to be affected by the NFES fibers’ sizes, spacing and stiffness, where there is an interesting correlation between the neurite length, the number of neurite and Map2 expression. NFES polymeric fibers were co-electrospun with carbon nanotube to build an electrode which stimulates neuronal stem cells differentiation under the application of 80 Hz sinusoid current waveform at 1.5 mA. NFES fibers could also be composited with thermo-responsive polymer which showed the cell detachment below the LCST of 22ËšC. Finally, coaxial polymeric fibers were patterned inside a microfluidics with incrementally increased spacing, to allow sustained release of the protein inside the core in a gradient fashion. With the system, neuronal migration and differentiation along the aligned nanofibers towards the gradient release of neuronal migration factor could be progressively studied.

Biography:

De Iuliis has completed his Medical School at the age of 25 years from Chieti University, Chieti, Italy. On 2013 He was accepted as a Resident in Clinical Pathology, SS Annunziata University Hospital, University of Chieti and as a postdoctoral fellow in the Department of Medical, Oral and Biotechnology Sciences. Currently he is developing bio-medical projects related to the biochemical mechanisms undergoing autophagy and apoptosis on established tumor cells. Please refer to Pubmed for his first paper as first author. In addition he has two papers in press as a first author and has submitted other five manuscripts.

Abstract:

The E3 ubiquitin ligase TRIM8 has been initially identified as a Ring finger protein expressed in glioblastomas capable of binding SOCS-1, a member of the Suppressor of Cytokine Signaling family of molecules. It has been reported by us that TRIM8 appears to be a negative regulator of SOCS-1 stability by binding to its SH-2/SOCS box domains. Moreover, recent reports have documented that nucleocytoplasmic trafficking of TRIM8 is involved in positive regulation of TNF-α induced NF-kB activation, in the stabilization of p53 on glioblastoma and clear cell renal carcinoma, and in the degradation of PIAS the negative regulator of STAT3. Here, we show for the first time that PIM serine/threonine kinases, also bind and phosphorylate TRIM8, resulting in increased stabilization of the ubiquitin ligase activity. Since SOCS-1 is functionally phosphorylated and also stabilized by PIMs, co-expression of a heterotrimeric TRIM8/SOCS-1/PIM complex promotes partial destabilization of the complexes and pursues degradation of the SOCS-1 protein. Additionally, co-expression of SOCS-1/TRIM8/PIM kinase complexes in 293T cells mitigates the repression of an interferon-γ-mediated signaling in responsive cells in vivo. These data add new partners to the complex network of protein-protein interactions that regulate SOCS-1 function and modulate the cytokine biological response. (Up to 250 words)

Biography:

Julia Lerchbacher-Hieslmayr (nee Lerchbacher) has completed her masters degree in zoology (Mag. rer. nat.) at the age of 19 in 2011 from Karl-Franzens University Graz. For 3 ½ years she worked at the Research Centre of Medical Technology and Biotechnology - fzmb GmbH in Bad Langensalza (Thuringia). Currently she is doing her Ph.D. studies at the Technical University of Munich (Klinikum rechts der Isar). At present, her main interests in the field of regenerative medicine are cartilage regeneration and usability of amniotic membrane.

Abstract:

Cartilage tissue has limited healing capacity. Osteoarthritis (OA) – the most common cause of cartilage lesions – has an incidence of approximately 10% in men and 13% in women aged over 60. This is likely to increase due to aging and obesity. Current treatments include matrix-associated autologous chondrocyte transplantation (MACT) as well as in situ-induction of cartilage repair by microfracture. Therapy of cartilage defects with these methods is unsatisfying. MACT is costly and comprises the disadvantages of two separate operations. Microfracture is limited to smaller defects and known to produce fibrous cartilage. Therefore, a clear need exists for new approaches to stimulate cartilage regeneration. In our study, we investigated the usability of amniotic membrane (AM) as transplant fixation of cartilage defects (1.5 X 2.0 cm) in equine knees. Five groups (N = 6) were included, each with AM cover plus: I. Microfracture, II. Microfracture/cartilage fragments/fibrin, III. Adipose-MSCs/cartilage fragments/fibrin, IV. Bone marrow-MSCs/cartilage fragments/fibrin, and V. no filling. 6 weeks post-surgery arthroscopy was performed. 12 months post-surgery tissue was harvested including native cartilage for comparison. Analyses comprised macroscopic imaging, biomechanics, gene expression and histology. Arthroscopy showed smooth integration of the AM into the defect bed. No adverse reaction was observed during the entire study. Macroscopic analysis showed all treated defects were significantly more covered with newly formed cartilage in contrast to no filling, with best results for two defects of the bone marrow-MSCs group. With adipose-MSCs the stiffness was comparable to native cartilage and Aggrecan, SOX9 as well as Versican expression was higher.

  • Embryonic Stem Cell | induced Pluripotent Stem Cells
Location: Kiel 1-3

Chair

Michael Weber

International Society for Medical laser Applications, Germany

Co-Chair

Brian M. Mehling

Blue Horizon International, LLC, USA

Session Introduction

Hanayuki Okura

National Institutes of Biomedical Innovation, Health and Nutrition, Japan

Title: Therapeutic potential of human adipose tissue-derived multi-lineage progenitor cells in non-alcoholic fatty liver disease
Biography:

Hanayuki Okura has completed her PhD from Osaka University Graduate School of Medicine. She was a Research Fellow for Young Scientists of Japan Society for the Promotion of Science. She is the Deputy Director of Platform of Therapeutics for Rare Diseases, National Institute of Biomedical Innovation, Health and Nutrition, Japan.

Abstract:

Non-alcoholic fatty liver disease (NAFLD) is an increasing cause of chronic liver disease and broadly defined by the presence of steatosis with inflammation and progressive fibrosis. Recently, we have reported the therapeutic potential of adipose tissue-derived multi-lineage progenitor cells (ADMPCs) in liver fibrosis using CCl4-induce chronic mice model. These findings led us to plan next study, whose aim was to assess the effectiveness of ADMPCs in improving NAFLD. ADMPCs were isolated from inguinal adipose tissues of C57 BL/6 mice and expanded. NAFLD model was induced by a single subcutaneous injection of 200 μg STZ two days-after birth followed by feeding a high fat diet beginning at four weeks of age. After randomization of animals, the NAFLD mice received ADMPCs or placebo control via tail vein injection at an age of six weeks and were applied for histological and blood examination at an age of nine weeks. NAFLD model mice with ADMPCs injection exhibited a significant reduction in liver fibrosis and inflammation areas, as evidenced by sirius red staining. Moreover, blood examination showed that plasma adiponectin levels in ADMPCs-treated NAFLD model mice were higher than those in placebo controls. In vitro production of anti-inflammatory cytokines, fibrinolytic enzymes and hepato-protective cytokines examined by ELISA were higher than those of BM-MSCs, suggesting the mode of action of ADMPCs. These results showed the mode of action and proof of concept of systemic injection of ADMPCs in NAFLD, which is a promising therapeutic intervention for the treatment
of patients with NAFLD.

Biography:

Abstract:

Induced pluripotent stem (iPS) cells can efficiently differentiate into the three germ layers similar to those formed by differentiated embryonic stem (ES) cells. This provides a new source of cells in which preclinical allogeneic transplantation models can be established. Our iPS cells were generated from mouse embryonic fibroblasts (MEFs) transfected with the Yamanaka factors, the four transcription factors (Oct4, Sox2, Klf4 and c-Myc), without antibiotic selection or MEF feeders. After formation of embryoid bodies (EB), iPS cells spontaneously differentiated into Flk1-positive cardiac progenitors and cardiomyocytes expressing cardiac-specific markers such as alpha sarcomeric actinin (α-actinin), cardiac alpha myosin heavy chain (α-MHC), cardiac troponin T (cTnT), and connexin 43 (CX43), as well as cardiac transcription factors Nk2 homebox 5 (Nkx2.5) and gata binding protein 4 (gata4). The electrophysiological activity of iPS cell-derived cardiomyocytes (iPS-CMs) was detected in beating cell clusters with optical mapping and RH237, a voltage-sensitive dye, and in single contracting cells with patch-clamp technology. Incompletely differentiated iPS cells formed teratomas when transplanted into a severe combined immunodeficient (SCID) mouse model of myocardial infarction. Our results show that somatic cells can be reprogrammed into pluripotent stem cells, which in turn spontaneously differentiate into electrophysiologically functional mature cardiomyocytes expressing cardiac-specific makers, and that these cells can potentially be used to repair myocardial infarction (MI) in the future.

Biography:

Taghrid Gaafar has completed her MD from Cairo University and Post-doctoral studies from Cairo University School of Medicine. She is a Professor of Immunology & Cell Biology at the Cairo University. She is the Director of RAMSES Stem Cell Research Unit, Children Hospital, Cairo University. She has published more than 20 papers in many reputed journals

Abstract:

Introduction & Aim: Lately, cell replacement using human induced pluripotent stem cells (hiPSCs) is considered a promising therapeutic alternative for cardiovascular diseases. Human iPSCs are endowed with the potential to produce a huge number of functional cardiomyocytes from autologous cell sources without the concern of ethical problems or immunological rejection. However, existing protocols for cardiac differentiation of hiPSCs are inefficient limiting their applications. Therefore, the objective of our study was to examine the modulation of signaling pathways to efficiently enhance cardiac differentiation of NP0040 hiPSCs into cardiomyocytes.
Methods: Cardiac differentiation was assessed by temporally modulating the regulatory elements of the signaling pathways: Wnt, BMP-4, FGF-2 and ascorbic acid in a monolayer-based culture system under serum-free, feeder-free conditions with subsequent purification by lactate method.
Results: Here, we showed that sequential treatment with glycogen synthase kinase-3 (GSK-3) inhibitor, bone morphogenetic protein-4 (BMP-4), fibroblast growth factor-2 (FGF-2) and ascorbic acid followed by inhibitor of wnt production-2 (IWP-2) and ascorbic acid produced a high yield of pure (up to 92%) cardiac troponin-T (cTnT)-positive cardiomyocytes that contracted spontaneously as coordinated sheets in multiple (>15) independent experiments. These cardiomyocytes were maintained as spontaneously contracting cells in culture for more than 1 month. The cardiomyocytes exhibited normal cardiac sarcomere organization expressing sarcomeric α-actinin, cTnT, MLC2v and the gap junction protein, Connexin 43. Reverse-transcriptase PCR revealed the expression of Isl-1, NKx2.5, Tbx5, Tbx20, GATA4, Mef2c, Hand1, MYH6, MYL2, MYL7, TNNT2 and TNNI3. Field Potential recording indicated that cardiomyocytes were electrically coupled to one another. The characteristics of the recorded action potential showed mainly ventricular-like waveforms.
Conclusion: Our data indicated that induction of mesoderm and its subsequent specification into cardiac fate from hiPSCs requires well-orchestrated interaction between Wnt, BMP-4, FGF/MEK and MEK/ERK signaling pathways. Controlling these pathways allows efficient production of cardiomyocytes that may provide a possible source for cell-based therapy.

Biography:

Aleksandra Klimczak has completed her PhD and DSc degree from the Institute of Immunology and Experimental Therapy PAS, Wroclaw, Poland. She gained extensive experience in transplant immunology associated with hematopoietic stem cell transplantation and in studies on tolerance inducing strategies in vascularized composite allograft models (Cleveland Clinic, USA). Currently, she is strongly involved in the studies assessing tissue distribution and biological characteristics of MSC originating from different human organs and their potential application in regenerative medicine. She has published 76 papers including 9 book chapters in peerreviewed worldwide journals.

Abstract:

Multipotent mesenchymal stromal/stem cells (MSC) can be isolated from different tissues. Their morphology, immunophenotype and differentiation potential are dependent on their tissue of origin. MSC isolated from adult human tissues are ideal candidate for tissue regeneration and tissue engineering, however, invasive methods of tissue harvesting is making this procedure uncomfortable for the tissue donors. We analyzed biological properties of MSC isolated from human bone marrow (BM), skeletal muscle and skin from post-operative tissues collected from limbs amputated due to critical limb ischemia. Long-term cultures of MSCs were performed in standard medium. Cells were observed up to 12 passages (P) and examined for phenotype and differentiation potential. Adherent cells, isolated from all examined tissues, express phenotype characteristic for naïve MSC: CD73, CD90, CD105, however their expression down-regulated during follow-up period (after P7). A fraction of MSC isolated from skeletal muscle express myogenic markers: CD56, desmin (up to P7) and dystrophin (expression increased after P7). Co-expression of CD73/CD146, CD90/CD146 and CD105/CD146 was detected on the proportion of adherent cells originated from BM, skeletal muscle and skin, and this suggest that they are proangiogenic progenitors. A fraction of cells expressing CD146 strongly co-expressed PDGFR-α. Cells isolated from examined tissues were capable to differentiate into chondrocytes, osteoblasts and adipocytes. Thus, MSC isolated from BM and skin biologically represent multipotent cells capable to differentiate into different type of tissues, whereas progenitor cells isolated from skeletal muscle have tissue-specific character. Post-operative tissues may serve as an alternative source of MSCs for potential clinical applications.

Biography:

Sudheer Shenoy P holds a PhD degree in Biochemistry from Kuvempu University, India in 2005. He has about 20 years of pre and post-PhD research experience in Tissue Engineering and Stem Cell Biology in India and Singapore. He holds key skills in the field of Generating Clinical, as well as, R&D grade Stem Cells. He has several international publications and 3 patents to his credit. His present focus is basic biology of adult perivascular mesenchymal stem cells and also various aspects of its application in cell therapy.

Abstract:

Pericytes are a source of mesenchymal stem cells (MSCs) having multilineage differentiation potential that are found on the wall of blood vessels. We have isolated, purified and characterized pericytes from liver as CD146+CD34-CD45-CD56- from wild-type (WT) and myostatin null (Mstn-/-) mice. CD146+ cells isolated from WT liver expressed myostatin and pericytes from both the genotypes expressed pericyte and adult stem cell markers and did not express αSMA and GFAP. CD146+ cells could be readily differentiated into adipogenic, osteogenic and chondrogenic lineages. When subjected to myogenic differentiation, these CD146+ cells behaved contrastingly as fibrogenic and myogenic precursors when isolated from two different genotypes WT and Mstn-/- respectively. Presence or absence of myostatin in vitro may play a role in determining the fate of pericytes like cells in liver. Furthermore CD146+ cells from the liver can be useful candidates for cell therapy studies.

Biography:

Jane Lebkowski has been actively involved in the development of cell and gene therapies since 1986 and is currently a Chief Scientific Officer and President of R&D at Asterias Biotherapeutics Inc, where she is responsible for all preclinical and product development of Asterias’ products. From 1998 to 2012, she was a Senior Vice President of Regenerative Medicine and Chief Scientific Officer at Geron Corporation. She led Geron’s human embryonic stem cell program, being responsible for all research, preclinical development, product development, manufacturing and clinical development activities. She has completed her PhD in Biochemistry from Princeton University in 1982 and completed a Post-doctoral Fellowship at the Department of Genetics, Stanford University in 1986. She has published over 70 peer reviewed papers and has 13 issued U.S. patents. She is on the Board of Directors of the American Society for Gene and Cell Therapy. She has served as an Industry Representative to FDA’s Office of Cell, Tissue and Gene Therapy Advisory Board.

Abstract:

Human embryonic stem cells (hESCs) can proliferate indefinitely yet, upon appropriate cues, differentiate into all somatic cell lineages. These two properties of hESCs enable the development of hESC-derived therapeutic cell populations which can be batch manufactured in central manufacturing facilities, cryopreserved, and distributed for “on demand” use at healthcare providers. Protocols have been developed to differentiate hESCs into neural, cardiomyocyte, hepatocyte, islet, osteoblast, chondrocyte, and hematopoietic cell populations which have been shown to be functional in either in vitro or in vivo animal models of human disease. Our group has established protocols to produce oligodendrocyte progenitors that upon transplantation into animals with spinal cord injuries can remyelinate denuded axons, induce axonal sprouting, and improve locomotor activity. Extensive preclinical studies have been completed to examine the activity, biodistribution, dosing, delivery, and potential toxicity and tumorigenicity of the oligodendrocyte progenitors. The safety of these cells is now being tested in the clinic in subjects with complete spinal cord injuries. In addition, our team has developed methods to produce dendritic cells from hESCs that have the antigen processing and presentation functionality to stimulate immune responses. In collaboration with Cancer Research UK, Asterias is preparing for a clinical trial using these hESC derived dendritic cells as a cancer immunotherapy in non-small cell lung carcinoma in the neoadjuvant setting.

Biography:

Abstract:

Coronary artery disease (CAD) resulting myocardial infarction is number one killer in the world and prevalence of this disease is also increasing in the developing countries. The treatment in the form of drugs, arterial stenting and bypass surgery is costing billions of dollars. It has been observed that endothelial cells dysfunction plays an important role in causing the disease. Various risk factors like diabetes mellitus, obesity, hypertension and hypercholesterolemia and smoking have been identified in the epidemiological studies. Endothelial dysfunction results initially and finally leads to thrombosis which will result lack of blood supply to the heart. Studies have shown that endothelial progenitor cells (EPCS) are involved in angiogenesis by differentiating into mature endothelial cells and aids in the repair mechanism. There are limited studies reporting the reduced number of endothelial cells and early senescence in coronary artery disease. Our study grouped 25 subjects in the age group of less than 50 years who were suffering with coronary artery diseases. Circulating EPC’S (CD34+/CD133+) by flow cytometry have shown significant lower number in coronary artery disease patients. EPC/TL (Kb-Genoms) was also markedly lower in coronary artery disease patients. Compared to controls the mean EPC/TA (IU/cells) was lower compared to controls. In younger patients, lower EPC’S number and shorter EPC’S telomere length and reduced telomerase activity were observed. Future gene therapy focusing attention on telomere aspects will help, as the disease prevalence is increasing and resulting in increased mortality. New drugs to increase telomerase activity and increasing telomere length will be the future hope for longevity.

Biography:

Agota Apati has completed her PhD in 2003 from Semmelweis University School of Medicine and Post-doctoral studies from National Blood Service. She is the Leader of Human Pluripotent Stem Cell lab in Research Centre for Natural Sciences of Hungarian Academy of Sciences. She is interested in differentiation, signaling and disease modeling of human embryonic and induced pluripotent stem cells. She has published more than 30 papers in reputed journals.

Abstract:

Human pluripotent stem cells (hPSC) provide new approaches to study the development and differentiation of various cell types of the human body and research tools for disease modeling. Meanwhile, the basic signaling events, including Ca2+-signals, have not been comprehensively evaluated in these cells. Therefore, we have started to study intracellular calcium responses to various ligands and the calcium signaling pathways in hPSCs and in their differentiated derivatives. First, we developed a protocol using the small molecule fluorescent Ca2+-indicator Fluo-4 and confocal microscopy. While this method was informative, several technical problems hindered this approach. To overcome these difficulties (i.e. dye-loading and toxicity), we generated hPSC lines which stably express a genetically encoded Ca2+-indicator (GCaMP2) protein by using a transposon-based gene delivery method. In this stable indicator system, we successfully studied the effects of various ligands in undifferentiated hPSCs and in hPSC-derived cardiomyocytes. To extend our knowledge towards Ca2+-signals in hPSC-derived neural cell types, we also generated GCaMP6 fast expressing neural progenitor cells (NPCs) and then differentiated these to PROX1-positive hippocampal neurons. Next, we investigated the calcium signals in NPCs and PROX1-positive hippocampal neurons expressing GCaMP6 fast, or loaded with Fluo-4 in parallel experiments. We found that specific, ligand induced Ca2+ signals were similar in the two assays, suggesting the applicability of GCaMP6f for the investigation of fast Ca2+ signals in neural cultures. We conclude that characteristics of both the spontaneous and ligand-induced Ca2+-signals, as well as their pharmacological modifications can be successfully examined in these model cells by fluorescence imaging.

Biography:

Kumiko Saeki has completed her graduation from the Faculty of Medicine, Tokyo University in 1988. After Residency training, she entered Graduate School of Tokyo University and was conferred a Doctor degree in 1995. After working as a Post-doctoral fellow, she became the Division Chief of Department of Hematology, Research Institute, International Medical Center of Japan in 1999. Since 2010, she has been the Chief of Stem Cell Therapy, Department of Disease Control, Research Institute, National Center for Global Health and Medicine. She has published more than 40 papers and has been serving as an Editorial Board Member of World Journal of Stem Cells.

Abstract:

Human pluripotent stem cells including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have been expected to provide an excellent tool for regenerative medicine. However, there are two major concerns in their clinical application: Immunological rejection and risk of tumorigenicity in the case of ESCs and iPSCs, respectively. Hence, their clinical application has been considered only in those cases where there are no alternative measures for effective treatments. Here, we propose that human ESCs/iPSCs can contribute to therapeutic development for the treatment of common diseases. As we recently reported, transient existence of human iPSC-derived vascular endothelical cells can exert a sustainable therapeutic effect on an injured artery preventing stenosis induction even after they are immunologically rejected. In this case, immunologically rejection creates beneficial outcomes by nullifying the risk of tumorigenesis without decreasing therapeutic effects. Another utility of human pluripotentstem cells is to provide a feasible tool for drug discovery that are otherwise unavailable. For example, sufficient amounts of brown adipocytes, which has been attracting a great deal of attention as a new target of drug discovery for the treatment of metabolic disorders, are unobtainable from living individuals due to technical as well as ethical problems. However, brown adipocytes can easily be produced from human ESCs/iPSCs as we previously reported. Potential contribution of human pluripotent stem cells to therapeutic development for the treatment of common diseases will be further discussed.

  • Workshop
Location: Kiel 1-3
  • Stem Cells | Stem Cell Treatments
Location: Kiel 1-3

Chair

Xiao-Dong Chen

University of Texas Health Science Center, USA

Co-Chair

Claire Henchcliffe

Weill Cornell Medical College, USA

Biography:

Professor Pestell completed his MBBS and subsequently PhD, M.D., (Melbourne University) FRACP, FACP, MBA (NYU) with post-doctoral studies at Harvard University and Massachusetts General Hospital. He was Director of the Lombardi Comprehensive Cancer Center, the Sidney Kimmel Cancer Center and most recently Executive Vice President of Thomas Jefferson University. He is the author of over 620 published works and 36 books and chapters, with over 45,700 citations, H index 115. He served and or serves on 14 scientific journal editorial boards, external advisory boards of 7 NCI cancer centers, several international research institutes, and review boards for research funding agencies of USA, Italy, UK, Switzerland, Ireland, France, Israel, Australia, and Czech. He is the founder of two biotechnology companies, has multiple issued patents and is a member of National Academy of Inventors (Jefferson Chapter).

Abstract:

Tumor initiating cells convey stem-like features and contribute to therapeutic resistance and tumor metastases. Cancer stem cells are protected against chemotherapy- and radiotherapy- induced death, through mechanisms that protect genomic integrity via induction of DNA damage sensing and DNA repair machinery. In mammalian cells, double-stranded breaks (DSB) may be repaired by either homology directed recombination (HDR), or non-homologous end joining (NHEJ) and single-strand annealing (SSA). Defects in these repair mechanisms can result in chromosomal fusions, translocations and breaks. The molecular mechanisms linking cancer stem cells and DNA repair are poorly understood. Herein, the chemokine receptor CCR5, which is known to contribute to breast cancer progression and metastasis, was shown to promote stem cell like properties and enhance DNA repair. Reintroduction of CCR5 into CCR5-negative cells promoted breast tumor stem cell expansion, metastases, and the induction of DNA repair gene expression. CCR5 was shown to enhance the repair of DSBs by inducing HDR and SSA-based DNA repair. The finding that CCR5 on cancer cells augments DNA repair and cancer stem cell expansion suggests CCR5 inhibitors may enhance the tumor specific activities of DDR-based treatments. (Up to 250 words)

Biography:

Maciej Kurpisz is the Head of Department of Reproductive Biology and Stem Cells, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland. He has completed his graduation from Poznan Medical Sciences University and promoted as MD in Immunology. Since 1996, he is a full Professor. He did professional training in UK, USA, Japan and Germany. He is expert in stem cells, anti-aging and male infertility. He invented experimental therapy for post-infarction heart regeneration. He is a Board Member of eight international journals and recipient of 20 professional awards.

Abstract:

From more than a decade intensive clinical trials with variety of stem cells have been pursued. Among the candidates, the most often used stem cells were originated from bone marrow or adult tissue reservoirs. The low turnover organs (heart, pancreas, central nervous system) were mostly in the focus of regenerative medicine. Due to low renewal and high morbidity, post-infarction heart served as a model for cellular therapies. Cells of myogenic origin, namely, myoblasts were accessible rather from skeletal muscles than from the heart itself, although cardiac precursor cells (CPC) were also applied in some experiments. As early as in 2001/2, human autologous myoblasts to post-infarction heart at the opportunity of coronary artery bypass grafting (CABG) were delivered. Next series included endoscopic delivery of myoblasts by using percutaneous approach and coronary vein (known in the literature as Poznan trial). Myoblasts often induced arrhythmic episodes and were suspected of low degree electromechanical coupling with recipient organ cardiomyocytes as well as they have been low paracrine factors secretors. Therefore in further studies, pre-clinical models (mouse, rats) were exercised when using genetic modifications of human myoblasts with variety of genes. Among others, pro-angiogenic genes, cytoprotective genes and/or conductivity genes were tried with a good success in, in vitro and in situ scenarios. The last clinical trial applied human myoblasts modified with GJA1 gene. Previously, we have tried this modification in rats (with induced ventricular arrhythmia) and mice (prevention of heart remodeling). The latest decoded results of clinical trial will be discussed.

Madalina Georgiana ALBU KAYA

INCDTP – Leather and Footwear Research Institute, Romania

Title: Near field electrospun fibres for neural stem cell differentiation
Biography:

Madalina Georgiana Albu Kaya has completed her PhD in Chemistry from Bucharest University, Faculty of Chemistry and Post-doctoral studies from Osaka University, Institute for Research Protein, Osaka, Japan. She is the Head of Collagen Department from INCDTP-Leather and Footwear Research Institute. She has published more than 70 papers in SCI journals, 10 books and book chapters, 17 patents and patent applications (awarded with 10 Gold Medals and Special Awards), being involved in over 70 research projects since 2001 to present.

Abstract:

A promising solution for soft tissue regeneration is tissue engineering, a multidisciplinary field of research which involves the use of biomaterials, growth factors, and stem cells in order to repair, replace, or regenerate tissues and organs damaged by injury or disease. The success of tissue engineering depends on the composition and microstructure of the used scaffolds. Ideally, scaffolds have to be similar to natural tissues. Collagen is the major component of the extracellular matrix of most soft tissues. As a natural molecule, collagen possesses a major advantage in being biodegradable, biocompatible, presenting low antigenicity and being easily available and highly versatile. Extracted as aqueous solution or gel, we processed type I collagen with different other natural polymers like sericin, hyaluronic acid, chondroitine sulfate, sodium alginate in three-dimensional (3D) forms such as hydrogels or matrices (spongious) by cross-linking or freeze-drying respectively, and successfully used as scaffolds. Human adipose derived stem cells (hADSC), 3T3-L1 preadipocytes, human epidermal keratinocytes and dermal fibroblasts are only some type of cells that proved their ability to build biohybrids in combination with the obtained collagen based scaffolds in a culture system. These findings indicate that the integration of scaffold with cells in vitro provides a potential source of living skin or adipose tissue for grafting in vivo.

Biography:

Peter Chapman-Smith is the founder and Director of the NZ Stem Cell Treatment Centre providing Regenerative Medicine with SVF in New Zealand. As an affiliate member of the Los Angeles based Cell Surgical Network, he contributes to their multicentre multinational safety and efficacy trial that has been running since July 2010. After graduating in Otago NZ in 1976, he was a family doctor and delivering obstetrician for 24 years, worked in anaesthesia for 10 years, and a long standing interest in sports medicine. For the last 20 years he has practised as an Appearance Medicine physician performing tumescent liposuction, skin cancer surgery, and extensively in phlebology doing nonsurgical varicose vein procedures. A registered laser practitioner, he performs regular duplex ultrasound assessment for venous procedures. He works in a private medical clinic in Northland New Zealand.

Abstract:

AMSCs have increasingly been used to successfully and safely to treat osteoarthritis in peripheral joints in the last decade worldwide. Veterinary similar experience has been documented for decades in animals. Adipose tissue has proven useful as a source of adult mesenchymal stem cells, readily available with a mini-liposuction procedure under local anaesthesia alone, and readily repeatable in future is required. Low back pain (LBP) affects 80% of adults at some time, a very high morbidity. The public and private cost in pain and loss of quality of life (QOL) and time off work and leisure is extensive, widely accepted for this all too common disorder. With conservative treatment most cases of LBP will resolve in 6 weeks. Surgical management can be indicated for unremitting nerve compression, only required for a small percentage of cases. AMSCs administered as stromal vascular fraction, SVF, is a new option that can be effective for chronic low back pain of multiple causation including disc disease and facet arthropathy. A single SVF treatment alone may be sufficient, deployed intravenously and concurrently into the lumbar epidural space by translaminar or transforaminal approach, or via the sacrococcygeal ligament as a simple direct caudal injection. Methods of managing LBP with SVF therapy will be presented, and clinical case series data that is encouraging from SVF use in New Zealand. The safety of this simple noninvasive technique is revolutionary.

Biography:

Jun Li has completed his PhD from Zhejiang University and Post-doctoral studies from Cedars-Sinai Medical Center in USA. He is a Professor of Hepatology and the Vice Director of State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine. He is a Member of American Association for the Study of Liver Diseases (AASLD) and a Member of Standing Committee of Stem cells Translational Medicine in the Chinese Society for Anatomical Sciences (CSAS). He has published more than 40 papers in reputed journals including the Gut, Hepatology and Liver International. He has been serving as an Editorial Board Member of Scientific Report.

Abstract:

Stem cell transplantation provides a promising alternative for the treatment of fulminant hepatic failure (FHF). However, it lacks fundamental understanding of stem cells’ activities. Our objective was to clarify stem cell-recipient interactions for overcoming barriers to clinical application. Here, we used an in-house large-animal (pig) model of FHF rescue by human bone marrow mesenchymal stem cells (hBMSCs) and profiled the cells’ activities. The control and transplantation groups of pigs (n=15 per group), both received a D-galactosamine (D-Gal) injection (1.5 g/kg). The transplantation group received hBMSCs via intraportal vein infusion (3×106 cells/kg) immediately after D-Gal administration. The stem cell-recipient interactions were quantitatively evaluated by biochemical function, cytokine array, metabolite profiling, gene sequencing and immunohistochemistry. The results indicated that all pigs in the control group died within an average of 3.22 days, whereas 13/15 pigs in the transplantation group lived >14 days. The cytokine array and metabolite profiling analyses revealed that hBMSC transplantation suppressed D-Gal- induced life-threatening cytokine storms and stabilized FHF within seven days, while human-derived hepatocytes constituted only ~4.5% of the pig hepatocytes. The functional synergy analysis of the observed profile changes indicated that the implanted hBMSCs altered the pigs’ cytokine responses to damage through paracrine effects. Delta-like ligand 4 (DLL4) was validated to assist liver restoration in both pig and rat FHF models. Our results delineated an integrated model of the multifaceted interactions between stem cells and recipients, which may open a new avenue to the discovery of single molecule-based therapeutics that simulate stem cell actions.

Dennis M Lox

Sports and Regenerative Medicine Centers, USA

Title: Sports medicine: Stem cells in the OA and AVN patient
Biography:

Dennis M Lox has attended the University of Arizona where he was Phi Beta Kappa Member. He has received his Medical degree at the Texas Tech University Health Sciences Center and his Residency Training in Physical Medicine and Rehabilitation at the University of Texas, Health Sciences Center at San Antonio. He has maintained a Private Practice in the Tampa Bay, Florida area and presently in Beverly Hills, California. He specializes in musculoskeletal disorders, sports medicine and regenerative medicine. He has edited two medical textbooks, written eight textbook chapters and did scientific publications. He has lectured extensively in the areas of regenerative and sports medicine.

Abstract:

The use of regenerative medicine applications such as stem cell therapies may have a common thread in the management of sports medicine patients. Trauma is a precipitating event in sports for the development of early onset osteoarthritis, as well as avascular necrosis (AVN). An understanding of the underlying pathophysiological processes involved in both post traumatic osteoarthritis (OA) and avascular necrosis (AVN) may direct treatment towards the use of regenerative medicine and stem cells. The progression of post traumatic osteoarthritis and AVN after sports injuries may direct early treatment methodologies towards a regenerative model, rather than a time sensitive progressive degenerative cascade. Case representations will demonstrate a parallel tract for the investigation and management of sports medicine patients who are at risk for, or developed post-traumatic osteoarthritis, and those with avascular necrosis (AVN).

Vinoth Kumar Jayaseelan

Duke-NUS Graduate Medical School, Singapore

Title: Use of stem cell as cellular toxicity test models
Biography:

Vinoth received his PhD, Masters in Science and post-doctoral training from the National University of Singapore in the field of stem cell research. His Bachelors in Dental Surgery was awarded by Bangalore University. He is currently a Clinician Scientist with the National Dental Centre where he devotes his time to clinics as well as to pursue his research interests in stem cell biology and regenerative dentistry. He is the author of several peer-reviewed international publications.

Abstract:

Established mammalian cell lines are commonly used to analyze the cytotoxic and genotoxic potential of environmental factors, drugs, biomaterials as well as chemical, physical and biological agents in vitro. However, these cells poorly reflect human physiology. Better models for use in pre-clinical testing of potential pharmaceutical compounds in clinical trials may potentially also reduce the costly and time consuming attrition of such compounds at later stages due to unacceptable levels of toxicity or lack of efficacy. Much research is also currently being undertaken to find viable and ethical alternatives for animal testing. Stem cells, due to their innate capacity to regenerate different cell types, in vitro, have been validated as a reliable source for in vitro developmental toxicology studies. The recent work that demonstrate the potential for the application of various embryonic and adult stem cell types and its derived progenies as plausible cellular models for in vitro toxicity testing will be discussed. We will also be discussing on the limitations and issue with existing toxicity test models.

Triin Vasar

Hospital of Reconstructive Surgery, Estonia

Title: Fat crafting for treatement of complicated wounds: Case report
Biography:

Triin Vasar has graduated from Faculty of Medicine in University of Tartu in the year 2007 and continued studies in Residency of Plastic and Reconstructive Surgery which she completed in 2014. Her special interests are in fat grafting and regenerative medicine. She has participated in animal studies to study angiogenic potential of ASCs. She is a Member of Estonian Society of Plastic and Reconstructive Surgery, as well as Member of ICOPLAST, EPRAS and IFATS.

Abstract:

Our experience includes treating complicated wounds with fat crafting and stromal vascular fracture (SVF). Treatment of diabetic, trophic and posttraumatic lower limb wounds/ulcers can be sometimes difficult due to their location, infection and compromised vascularity of the limb. Usually, wound/ulcer is infected with multiple resistant bacteria and blood supply to the affected area is poor and those patients have been already offered amputation. Fat crafting improves vascularization of wound/ulcer, more efficient infection control, minimal postoperative scaring and sometimes salvage of the limb. Hence, patient can return to their everyday life with no limitations. In this case report: A 30-year old male patient, who was involved in motorcycle accident in the year 2012 was considered. He suffered unstable left knee injury-total rupture of knees ligaments with injury of popliteal artery. Due to delayed diagnosis of arterial injury, patient developed ischemic contracture of foot and ankle. In 2014, the patient underwent a surgery for correcting ankle position, which was complicated with wound dehiscence, tissue loss, infection and exposure of bone. Due to compromised vascularity the traditional treatment methods were insufficient and unusable. Patient was treated with one session of SVF enriched fat grafting, two months later extra fat grafting procedures and skin grafting. It was observed that, 6 months later the defect healed with minimal scaring and patient returned to normal everyday life.

Biography:

Huaiyong Chen has completed his PhD from Institute of Biophysics, Chinese Academy of Science and Post-doctoral studies from Duke University Medical Center. He is the Vice Director of Department of Basic Medicine in Tianjin Haihe Hospital, recognized as one of the national great health care providers for treatment of lung infections including tuberculosis and flu virus in China. He has published more than 25 papers in reputed journals.

Abstract:

Aberrant systemic artery supply results in recurrent infections in the abnormal lung lobe of intralobar pulmonarysequestration (ILS). Because of inefficient control of such symptom in clinic, surgical resection becomes the treatment of choice. In present study, we observed abnormalities associated with epithelial barriers in ILS specimen including hypersecretion of mucin and reduced expression of surfactant protein C (SPC). Compared to healthy control specimen, keratin 5-positive stem cells were less proliferative in distal airways. SPC-positive alveolar type 2 (AT2) stem/progenitor cells were also less proliferative and less in number. Transcriptional profiling analysis suggests that human aortic endothelial cells differed from human pulmonary artery endothelial cells in signal pathways associated with cell division. Array validation indicates that thrombospondin-1 (TSP1) expression was decreased in vascular cells near lesion part but increased in lesion part of ILS lobe. In vitro culture demonstrates that TSP1 exhibited an inhibitory role in mouse AT2 cell differentiation. The inhibitory effect was abolished when CD36 was knocked out in AT2 cells. These data demonstrate that distal lung stem/progenitor cells are impaired in ILS lobe and imply that restoring epithelial integrity can be beneficial for the future treatments of recurrent infections in lung pathologies.

Biography:

Janna Nawroth is a Principal Investigator (R&D Lead) at Emulate Inc., a company in Boston, MA, USA, focusing on the development and commercialization of Organs-on-Chip Technology. She is an expert in biological fluid dynamics. She has completed her PhD in Biology in at the California Institute of Technology, USA and Post-doctoral studies at the Wyss Institute for Biologically Inspired Engineering at Harvard University, USA. She has published more than 10 papers in distinguished journals that collectively have been cited over 400 times.

Abstract:

Organ-on-Chip technology, the pairing of micro-engineered human tissue with micro-engineered environments, enables the in vitro emulation of human physiology. Our goal is in particular to recapitulate individual organ functions and organorgan interactions, with the goal of elucidating their role in homeostasis, metabolism, development, toxicology and disease. Using computational design studies and working closely with collaborators in academia, industry, and the clinic, we develop micromechanical environments and quantitative readouts specific to particular organs and disease states. This study presents designs and applications of microengineered systems, including the Small-Airway-Chip and the Thrombosis-Chip. In the near future, we aim to study inter-organ signaling and cell migration involved in systemic processes such as cancer, immune-system responses and hormone-mediated remodeling.

Biography:

Xiao-Dong Chen is a Professor in the Department of Comprehensive Dentistry and Chief of the Regenerative Medicine Program in the School of Dentistry at the University of Texas, Health Science Center at San Antonio. His group was the first to establish Cell-Free Native ECM made by Bone Marrow Stromal Cells. To closely replicate the tissue specific microenvironment (niche) ex vivo, he and his team have extended their technology by developing a variety of 3D tissue-specific scaffolds for facilitating stem cell-based applications.

Abstract:

MSC-based therapies have great potential for treating many age-related diseases. Due to biosafety concerns and FDA regulations, autologous stem cells are preferable for cell-based therapies. Unfortunately, the quantity and quality of MSCs decrease with aging, inhibiting progress in developing autologous MSC-based applications. In the present study, we propose an innovative approach for rescuing aged MSCs by separating a sub-population of more “youthful” cells, using biomarkers (size and SSEA-4 expression), from a population of cells obtained from the BM of elderly donors (human subjects); and protecting and amplifying this “youthful” sub-population by culture on a “young microenvironment” using our established patented technology (BM-ECM produced by young cells). The results demonstrate that by applying this approach we were able to identify and isolate a subpopulation of BM-MSCs from elderly donors, displaying a less “aged” phenotype, and culture them on a young microenvironment (BM-ECM) to significantly improve their proliferation and differentiation capacity.

Biography:

Malancha Ta completed her PhD from the National Institute of Immunology (NII), New Delhi, India and postdoctoral studies from NIH, Bethesda, USA. Currently she holds the position of Assistant Professor at the IISER-Kolkata, India and has been working in the area of stem cells for the last 15 years. She has published many papers in reputed journals and her research interest includes investigating the molecular differences between MSCs from different tissue sources and understanding their functioning under different degrees of physiological stress.

Abstract:

Mesenchymal stem cells (MSCs), due to their paracrine, anti-inflammatory and wound healing applications, have emerged as a promising treatment option for various human diseases. However, the efficacy of MSC therapy is currently limited by low retention and poor survival of transplanted cells as demonstrated consistently by clinical studies using bone marrow-MSCs. This is due to the harsh microenvironment marked by oxygen and nutrient deprivation and inflammation at the injured sites. Also, the choice of MSC source could be critical in determining fate and cellular function of MSCs. Therefore, our objective was to investigate the influence of ischemic stress on Wharton’s Jelly (WJ)-MSCs from the umbilical cord, to assess their therapeutic relevance in ischemic diseases. We simulated conditions of ischemia in-vitro by culturing WJ-MSCs under 2% oxygen in a serum deprived and low glucose medium for 48 hrs. Under ischemic stress, WJ-MSCs retained a viable population of greater than 80%. Ischemic WJ-MSCs got arrested at G0/G1 phase of cell cycle but did not show signs of senescence. Moreover, the ischemic WJ-MSCs expressed the characteristic MSC surface antigens and in addition, were negative for the expression of costimulatory molecules, thus maintaining their immuneprivilged status under ischemia. To evaluate the wound healing properties of ischemic WJ-MSCs, we compared gene expression profile between control and ischemic WJ-MSCs by PCR array and found an upregulation of many ECM and adhesion molecules, cytokines, trophic factors in the ischemic population. Our findings thus provide evidence that WJ-MSCs might be therapeutically beneficial and potent in treating ischemic diseases.

Biography:

Kowit-Yu Chong has completed his PhD from Southern Illinois University, School of Medicine and Post-doctoral studies from Oregan Primate Research Center. He is an Associate Professor at Chang Gung University, Taoyuan City, Taiwan. He has published more than 40 papers in reputed journals.

Abstract:

Chronic obstructive pulmonary disease (COPD) is a progressive disease that makes patients hard to breathe. Lung emphysema is caused by inflammatory response, oxidative stress or endothelial cell apoptosis and is a smoking-related disease. Mesenchymal stem cell (MSC)-based cell therapy is a novel approach with great therapeutic potentials for the treatment of lung diseases. Despite demonstration of MSC grafting, the populations of engrafted MSCs have been shown to decrease dramatically 24 hours post-transplantation due to exposure to harsh microenvironments. Hypoxia is known to induce expression of cytoprotective genes and also secretion of anti-inflammatory, anti-apoptotic and anti-fibrotic factors. Hence, hypoxic preconditioning is thought to enhance the therapeutic potency and duration of survival of engrafted MSCs. In this study, we aimed to prolong the duration of survival of engrafted MSCs and to enhance the effectiveness of emphysema transplantation therapy by the use of hypoxia-preconditioned MSCs (HP-MSCs). Our results showed that compared to MSCs, the cyto-protective genes, such as Bcl-2, CAT, HO-1, HGF and VEGF in HMSCs have significant increased, the conditioned medium of HMSCs have enhanced the cell viability of elastase treated murine lung type II epithelial cells (MLE-12). Furthermore, intratracheal instillation of HP-MSCs into elastase-induced emphysema mouse model at day 14, the pulmonary respiratory function have shown significant improvement for up to 30 days of HP-MSCs treatment. Expression of inflammatory factors IL-1β, IL-6, protease, and elastin were all down-regulated in the lung tissues of the treated mice. Moreover, the histopathologic examination indicated significant amelioration of lung damages. In conclusion, we demonstrated that HP-MSCs significant improve the therapeutic effect of the emphysema. This study should improve our understanding of the protective effect of engraftment HP-MSCs in lung injury model. Furthermore, this novel approach may lead a new avenue of therapy for COPD.

Biography:

Beka Solomon has earned her PhD in 1976 from the Weizmann Institute of Science, Rehovoth, Israel. She has joined Tel-Aviv University in 1979 following Post-doctoral studies and training periods at Harvard Medical School and Brigham and Women’s Hospital, Boston, USA. She is a Member of the Editorial Board of Drugs of Today, Recent Patents on CNS Drug Discovery, Neurodegenerative Diseases and Journal of Alzheimer's Disease. She was awarded the prestigious Zenith Award of the Alzheimer Association and received the Dana Foundation Award for Neuroimmunology. In 2007 she was included in Scientific American’s List of 50 of the World’s Leading Innovators.

Abstract:

Senile plaques and neurofibrillary tangles cause synaptic dysfunction and loss of neurons in the brain of Alzheimer's disease (AD) patients, accompanied by inflammatory processes. Tumor necrosis factor α (TNFα), a multipotent cytokine, is considered as a “Master Regulator” of the immune response in the central nervous system and modulator of glutamate toxicity. This effect is enhanced when activated microglia cooperates to release the cytokine in response to CXCR4 stimulation which is increased in tissue of AD patients. CXCR4 is a chemokine receptor which belongs to the G-protein-coupled receptors family and is selective for the CXCL12 ligand. The axis of CXCR4/CXCL12 has a prime role in various physiological routes and mechanisms and release TNFα from astrocyte cell surface. Another mechanism which is being mediated by CXC4/CXCL12 is homing and mobilization of hematopoietic stem and progenitor cells (HSPCs). Under basal conditions TNFα signaling negatively regulates HSPCs repopulating activity. AMD3100 is a FDA approved, small bicyclam molecule that reversibly and selectively blocks binding of CXCL12 to its receptor CXCR4. The binding of AMD3100 to CXCR4 mobilize HSPCs from the bone marrow into the blood. We chronically treated the triple transgenic mice model (3xTg-AD) which demonstrates agedependent changes in TNFα mRNA levels with AMD3100. The data presented here suggests the potentiality of AMD3100, to modulate the TNFα, to improve the cognitive functions, to decrease pathology and to offer a chance of a cure for AD.

Biography:

Abstract:

Introduction: Diabetes Mellitus (DM) is a group of metabolic diseases with hyperglycemic characteristic that occurs due to abnormal insulin secretion, insulin resistance or both. Various kinds therapies of DM, whether it is preventive, pharmacological and life-style changes, have been attempted for managing diabetes, but the prevalence is higher and higher with high mortality and morbidity as well. Therefore, it necessitates the provision of alternative therapies in the management of DM by using Mesenchymal Stem Cells (MSCs) was derived from adipose.
Methods: 40 patients with inclusion criteria of type 2 diabetic with tertiary failure of treatment were given autologous MSCs derived from adipose. MSCs were inserted through catheterization of pancreatic artery, and then was measured the levels of fasting blood sugar, 2 hours postprandial blood sugar, insulin, C peptide, HbA 1C, HOMA- R, HOMA- B pre and post stem cell transplantation in three months later after implantation.
Result: Evaluation in patients receiving stem cell therapy three months after implantation were found the characteristic of the patients dominantly in 50-59 y.o about 40%, with T test, Mean of HbA 1c pre were 8.28, post 6.795 (p= 0.000), mean of fasting glucose pre were148.77, post were 102.32 (p= 0.000), mean 2 hours post prandial glucose were 252.77, post were 129.12 (p=0.000). with Wilcoxon test was found, mean of c peptide pre were 2.44, post were 2.98 (p=0.000), mean of fasting insulin were 8.79, post 7.19 (p=0.447), mean of HOMA IR pre were 4.85 and post were 2.69 (0.000), and the last mean of HOMA B pre were 52.47 and post were 71.04 (p= 0.000).
Discussion: Three months after stem cell implantation plasma glucose was significantly reduce, pancreatic preservation also insulin resistance were significantly improvement.
Conclusion: Adipose derived MSCs can improve pancreatic function and decrease insulin resistance in diabetic patients with tertiary failure treatment.

Biography:

Rahim Mohammadi has completed his DVSc at the age of 39 years from Urmia University. He is Assistant Professor of Surgery in Faculty of Veterinry Medicine, Urmia University, Urmiam Iran. He has published more than 45 papers in reputed journals.

Abstract:

Traumatic injury to peripheral nerves results in considerable motor and sensory disability. Several research groups have tried to improve the regeneration of traumatized nerves by invention of favorable microsurgery. Effect of undifferentiated bone marrow stromal cells (BMSCs) combined with artery graft on peripheral nerve regeneration was studied using a rat sciatic nerve regeneration model. A 10-mm sciatic nerve defect was bridged using an artery graft (IOAG) filled with undifferentiated BMSCs (2 _ 107 cells/mL). In control group, the graft was filled with phosphated buffer saline alone. The regenerated fibers were studied 4, 8 and 12 weeks after surgery. Assessment of nerve regeneration was based on behavioral, functional (Walking Track Analysis), electrophysiological, histomorphometric and immuohistochemical (Schwann cell detection by S-100 expression) criteria. The behavioral, functional and electrophysiological studies confirmed significant recovery of regenerated axons in IOAG/BMSC group (P < 0.05). Quantitative morphometric analyses of regenerated fibers showed the number and diameter of myelinated fibers in IOAG/BMSC group were significantly higher than in the control group (P < 0.05). This demonstrates the potential of using undifferentiated BMSCs combined with artery graft in peripheral nerve regeneration without limitations of donor-site morbidity associated with isolation of Schwann cells. It is also cost saving due to reduction in interval from tissue collection until cell injection, simplicity of laboratory procedures compared to differentiated BMSCs and may have clinical implications for the surgical management of patients after facial nerve transection.