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 3 :

  • 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.