Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 9th Annual Conference on Stem Cell and Regenerative Medicine Berlin, Germany.

Day 2 :

Keynote Forum

Y James Kang

Sichuan 3D Bio-printing Institue, China

Keynote: Vascular tissue regeneration from implantation of 3D bio-printed stem cell grafts in rhesus monkeys

Time : 09:00-09:40

Conference Series Stem Cell Congress 2017 International Conference Keynote Speaker Y James Kang photo
Biography:

Y James Kang received his PhD in Toxicology and Zoology at Iowa State University in 1989 and completed Postdoctoral training in Biochemistry and Molecular Biology at Cornell University Medical Center in 1990. He was an Assistant Professor of Pharmacology and Toxicology at the University of North Dakota School of Medicine during 1991-1996, Associate Professor of Medicine of Pharmacology and Toxicology and University Scholar at the University of Louisville School of Medicine during 1996-2001, and Professor and Distinguished University Scholar from 2001. He served several NIH study sections from 1997 to 2006 and other federal agencies including USDA, US-EPA and US-Veterans Administrations from 1996 to 2005. He was elected as Fellow of the Academy of Toxicological Sciences in 2001. He is currently a China National One-Thousand-Talents Professor and Director of the Regenerative Medicine Research Center at Sichuan University West China Medical College. He is also the President and CEO of Sichuan 3D Bio-printing Research and Technology Institute, the Director of Sichuan Regenerative Medicine Research and Technology Center, the Editor-in-Chief of the journals, Cardiovascular Toxicology and Regenerative Medicine Research and the Editor of the book series, “Methods in Pharmacology and Toxicology”.

Abstract:

Dr Kang’s research team has been engaged in 3D bio-printing and they invented living cell-based bio-ink called biosynsphere and developed a series of 3D bio-printers. Among these bio-printers is a vascular graft printer. Using this printer and the bio-ink, they have created a novel stem cell-based vascular graft producing technology, the product was successfully implanted as an abdominal artery interposition graft into rhesus monkeys. The bio-printed vascular grafts were composed of adipose stem cells and once implanted into monkeys, they were fully differentiated into the naturally-identical blood vessel structure and performed the expected biological function in monkeys. This DEVELOP (Destination-Engaged Vessel Evolving Lineage Organ Production) approach is ready to be applied to humans.

  • Tissue Regenaration | Cell and Gene Therapy | Regenaration and Therapeutics | Regenarative Medicine and Market | Tissue Science and Engineering | Biomaterial Engineering
Location: Muggelsee

Chair

Tamara Lah Turnšek

Mednarodna podiplomska šola Jožefa Stefana, Slovenia

Co-Chair

Viacheslav M Mikhailov

Institute of Cytology - Russian Academy of Sciences, Russia

Session Introduction

Viacheslav M Mikhailov

Institute of Cytology - Russian Academy of Sciences, Russia

Title: Cell transplantation as tool for foetuses growth reg
Biography:

Viacheslav M Mikhailov received education in Medicine from Mechnikoff’s Medical University during 1958-1964 years. He pursued Postgraduation from Embryological Department of Institute of Experimental Medicine (1958-64) and PhD degree (1970). His PhD thesis entitled: “The analyses of mechanisms of pathogenic action of immune antirenal antisera for mammalian embryogenesis”. He was awarded DS degree for thesis: “Life Cycle of Decidual Cells”, by title Leader Research Fellow and by Professor title in 1998 and 2003 respectively. He is the Head of Cell Populations Genetic Group of Institute of Cytology RAS.

Abstract:

Cell interactions between uterus and foetuses influence for success of pregnancy and of fetal development in mammals. In case of human and rodents the embryos and foetuses are surrounded by decidua cells (DC) that form decidua membrane. Without DC blastocyst development of experimental animals stops at the initial stages of gastrulation and doesn’t go beyond somites formation. DC participate in the trophic relationships between mother and foetus, their specific functions include the prevention of the development of inflammation in the endometrium and the regulation of immunological conflict between mother and foetus. There are also results that single intravenous transplantation of pregnant rats Percoll derived mononuclear bone marrow cells (BMC) suspension of 4-5, 7-9 or 11-12 pregnant days to rats with the same date of pregnancy influences for survival and weight of foetuses of 18th of pregnancy. After BMC, intravenous transplantation during 4-5 days of pregnancy the weight was preserved but survival of foetuses was decreased. The weight of 18th day foetuses after transplantation during pregnant 7-9 days (835 ± 15 mg) was significant increase in comparison with weight of normal and control foetuses (745 ± 11). The survival of foetuses was also preserved. In case of intravenous transplantation during placentation at 11-12 days of pregnancy the weight of foetuses was decreased, the weight of placentas was increased and survival of foetuses was disturbed. The retardation of foetuses growth after BMC transplantation during placenta formation (587 ± 5 mg) at pregnant 11-12 days may be explained by cytotoxic action of uNK cells for embryo. At the same time, the sub-dermal BMC transplantation at 11 and 13 pregnant days stopped the embryotoxic action of uNK cells, increased the weight of foetuses and preserved the survival of embryos.

Biography:

Helen McGettrick completed her PhD in 2006, followed by 3 Postdoctoral Research fellowships at the University of Birmingham (UK). She was appointed as a University Fellow in Inflammation Biology in 2011, and a year later successfully won a five-year Arthritis Research UK Career Development Fellowship. She was recently awarded the prestigious Garrod Prize by the British Society for Rheumatology in 2016. She is an Honorary Lecturer at the University of Glasgow and Newcastle University (UK). She has more than 30 published articles with about 760 citations, and has filed 3 patents and received funding from Wellcome Trust, Pfizer and British Heart Foundation.

Abstract:

Chronic inflammation is associated with formation of ectopic fat deposits that might represent damage-induced aberrant mesenchymal stem cell (MSC) differentiation. Such deposits are associated with increased levels of inflammatory infiltrate and poor prognosis. Here we tested the hypothesis that differentiation from MSC to adipocytes in inflamed tissue might contribute to chronicity through loss of immunomodulatory function. We assessed the effects of adipogenic differentiation of MSC from bone marrow or adipose tissue on their capacity to regulate neutrophil recruitment by endothelial cells and compared the differentiated cells to primary adipocytes from adipose tissue. Bone marrow derived MSC were immunosuppressive, inhibiting neutrophil recruitment to TNFα-treated EC, but MSC derived adipocytes were no longer able to suppress neutrophil adhesion. Changes in IL-6 and TGFβ1 signaling appeared critical for the loss of the immunosuppressive phenotype. In contrast, native stromal cells, adipocytes derived from them and mature adipocytes from adipose tissue were all immuno-protective. Thus, disruption of normal tissue stroma homeostasis, as occurs in chronic inflammatory diseases, might drive abnormal adipogenesis which adversely influences the behavior of MSC and contributes to pathogenic recruitment of leukocytes. Interestingly, stromal cells programmed in native fat tissue retain an immuno-protective phenotype.

Biography:

Jasbir Rattu is a Consultant Industrial Pharmacist EU qualified person focussed on innovative regenerative medicines and advanced therapy medicinal products for unmet medical need.

Abstract:

IQ= QRM x QbD x QMS. Quality risk management is an enabling process that supports the product life cycle and is an integral part of an organisation’s pharmaceutical quality system. The ultimate goal of the QRM process is to bring focus and effort to the issues that impart the highest risk to product quality and/or patient safety. Therefore, as QRM is ultimately linked to the protection of the patient, a modern PQS cannot truly function properly in the absence of an effective, integrated QRM system. QRM is a systematic process for the assessment, control, communication and review of risks to the quality of the medicinal product. It can be applied both proactively and retrospectively. Therefore, the objective is not just to identify risk, but to provide information to help make better and informed decisions to mitigate and reduce risk, thus improving the process. Hence, it is critical to understand and effectively and optimally use the most appropriate risk management tools and approaches. The consistent and effective analysis of risks associated with manufacturing processes and quality systems typically leads to more robust decisions reduces uncertainty and leads to greater confidence in outcomes. These QRM elements must be embedded throughout the organisation through policies, procedures and reinforced via effective training. Quality by design requires a deep understanding of the product and process science which impacts quality and hence performance. Quality Management System is the set of systems which ensure high quality is continuously improved e.g. audit systems, change control systems, deviation management systems etc. Continuous improvement of QRM multiplied by excellent QbD understanding and again by reliable Quality Management Systems provides a powerful optimised integrated quality system. This presentation discusses best practice including my experience of successfully implementing integrated quality systems in high risk areas such as regenerative medicine and other areas of the biopharmaceutical industry including applications to cutting edge advanced therapy medicinal products e.g. cell and gene therapies such as CAR-T cells in immuno-oncology to the tissue engineering of organs such as the eye and stem cell-based therapies for treating optic neuropathies such as glaucoma.

Speaker
Biography:

Luca Gentile graduated in Developmental Biology in 2000 and received his PhD in Bioengineering and Bioinformatics at the University of Pavia (ITA). In 2003, he was invited to the Center for Animal Transgenesis and Germ Cell Research (Kenneth Square, PA, USA) for studying reprogramming in single reconstructed mouse embryos. In 2004, he joined the Max Planck Institute for Molecular Biomedicine (Münster, GER), working on the onset of cellular reprogramming. In 2010, he was appointed Head of the Planarian Stem Cell Laboratory (Münster, GER), working on the conserved mechanisms of pluripotency. In the same year, he organized the first International Meeting on Planarian Biology, which in 2018 reaches its 4th edition (Madison, WI, USA). Since March 2015, he leads the Pluripotency & Regeneration Group at the Fraunhofer Institute for Biomedical Engineering (Sulzbach, GER). His subjects of investigation are the derivation of human cardiac organoids and the functional characterization of stem cell subpopulations in planarian S. mediterranea.

Abstract:

Over the last 60 years, 14% of all drugs were withdrawn post-marketing owing to their cardiotoxic effects. Cardiovascular diseases are responsible for nearly half of all deaths in Europe. This is largely due to the poor regeneration capability of the human heart, which culminates in scar tissue formation and reduced function. Although advanced medical procedures increased the survival rate, cell-based therapies fall short in delivering a functional engraftment, and many patients progress towards end-stage heart failure. We used 3D printing and ultra-high viscosity alginate to produce a hydrogel scaffold called the Heart-patch, which could sustain cardiac muscle identity as a functional unit over long time in culture. Human induced pluripotent stem cell-derived cardiomyocytes cultured on the Heartpatch are closer to adult CMs that those cultured on traditional plastic, making the Heartpatch a physiologically relevant model of the heart. Biocompatible UHV alginate is 3D printed using a novel, viscosity-independent, printing method. It allows both patterning the scaffold’s surface and tuning its stiffness. Additionally, surface modifications can be applied with click-it chemistry, allowing a specific ECM protein composition that mimics the cardiac niche. Cardiomyocytes on the Heartpatch can be cultured for longer periods (3-4 months) than their counterparts on cell culture plastic, exhibiting self-sustained macro contractions visible to the naked eye. Gene expression profiling and immunocytochemistry analysis indicate increased cardiomyocyte maturation, enhanced cell-cell interactions and a high degree of both electrophysiological coupling and cytoskeletal maturation. As a result, force transduction resembles more closely the in vivo situation, as also supported by a novel optical-based analysis of the self-sustained depolarization. On the Heart-patch, hiPSC-CMs acquire an additional degree of freedom. As an individual working unit, the Heartpatch is an enhanced cardiac muscle model that enables disease modeling and high-throughput compound screening, with potential future applications in regenerative medicine.

Speaker
Biography:

Jiamin Teng is a Professor in the Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center in Shreveport (LSUHSC), USA. She completed Master’s Degree from Tongji Medical University in Wuhan, China and her PhD in Nihon University School of Medicine in Tokyo, Japan. Her research focuses on glomerulosclerosis of kidney damage and the stem cell regeneration, which can be translated to diabetic nephropathy due to similarities in pathogentic mechanisms involved. Her laboratory has developed unique experimental models such as in vitro live cell micromanipulation model, in vitro 6 dimensional live cell observation models, ex vivo kidney perfusion model, and in vivo model for the kidney glomeruli damage and reparation. These models have well served for the purpose of kidney pathogenic and regeneration.

Abstract:

Many investigators have supported the idea that mesenchymal stem cells (MSCs) participate in the process of repair / regeneration exclusively by providing paracrine factors that enabled the process.  Using a model of mesangial damage induced by glomerulopathic immunoglobulin light chains (G-LCs) and then repaired or regenerated by MSCs in AL-amyloidosis, the role of MSCs in the process was investigated. In-vitro and ex-vivo experimental platforms were used to address the issue. The in vitro 6 dimensional (6D) live cell imaging system was used to observe the damage of mesangial cells (MCs) and the alteration of the mesangial matrix incubated with G-LCs. In the ex vivo model, G-LCs were perfused through the renal artery. The respective lesions were reproduced in both platforms. Then, tagged MSCs were introduced. Immunofluorescence, immunohistochemistry and electron microscopy were used to evaluate samples obtained at different time frames. Stains for smoothelin, muscle specific actin, smooth muscle actin, CD29, and 68 were used to monitor phenotypic transformation of MSCs in the process of regeneration. Our results showed that MSCs initially transformed from an undifferentiated to a macrophage phenotype to clear the damaged mesangial areas where transformed MSCs phagocytosing cellular debris resulting from apoptotic mesangial cells and damaged matrix elements of amyloid fibrils. Following the cleaning process, MSCs acquired morphologic and immunophenotypic characteristics of MCs as they proceeded to lay down new mesangial matrix. MSCs manifest great plasticity as they proceed to repair the damaged mesangium in both our models. The fact that they transform to a macrophage phenotype followed by transformation to MCs allows them to perform different crucial functions during the process of regeneration. The restored mesangium is possible as new MCs derived from MSCs are able to reproduce the normal mesangium.

Speaker
Biography:

Sharmila Fagoonee is a Researcher at the Institute of Biostructure and Bioimaging of the Italian National Research Council at Molecular Biotechnology Center in Turin, Italy. After her BSc (hons) in Biology at Mauritius and MSc in Cellular Biology at Bordeaux, France, she got her PhD in Cell and Molecular Biology at the University of Turin. She also holds a MSc in Molecular Biotechnology. During her PhD, she worked on heme and iron metabolism in mouse models of liver diseases. After a period at BIDMC, Harvard, Boston, she shifted to study the differentiative capacity of mouse pluripotent spermatogonial stem cells in vitro and in vivo. Using bioinformatics analysis, she identified new pluripotency- and stemness-related genes in pluripotent stem cells. She is currently interested in human stem cells-based therapy for liver diseases and in studying the mechanisms leading to and in developing new biomarkers for liver fibrosis.

Abstract:

Adult stem cells have gained in attractiveness over embryonic stem cells for liver cell therapy due to their origin, multipotentiality, and the possibility of autologous transplantation. In the last two decades, great strides have been made in spermatogonial stem cell research showing the potentiality of these stem cells in organ regeneration and in the field of regenerative medicine. Spermatogonial stem cells derived from post-natal or adult mice are capable of differentiating into spermatozoa in vivo. A spontaneous embryonic stem cell-like conversion is also observed in vitro in long-term cultures. These pluripotent spermatogonial stem cells can be induced to differentiate into cells of the three germ layers without ethical constraints. We have recently shown that functional hepatocytes can be derived from mouse pluripotent spermatogonial stem cells for in vitro and in vivo use. These cells can thus provide a very promising source of hepatocyte-like cells for the regeneration of damaged tissue and for restoring liver functionality.

Speaker
Biography:

Christopher A Bradley completed his PhD in Biochemistry & Molecular Biology at Louisiana State University Health Sciences Center in Shreveport. His Postdoctoral research was in the fields of eukaryotic protein synthesis, proteomics, cancer, osteoarthritis, and virology. Industry experience in detergent chemistry and nutraceuticals preceded his current position in regenerative medicine as Product Development Director for Lattice Biologics, a manufacturer of stem cell and tissue allografts.

Abstract:

The extracellular matrix consists of structural and functional molecules secreted by the resident cells. The 3-dimensional organization and composition of an ECM is distinctive for each tissue type. Solubilized gel-forming ECM could potentially be used to develop products such as 3D culture substrates, growth factor delivery vehicles, or scaffolds that promote tissue regeneration. One goal of ours is to design a 100% allograft custom-fabricated human ECM-derived 3D microenvironment that mimics the biological properties of native ECM. Toward this goal, we evaluated the biochemical and functional characteristics of ECM isolated from human placenta, adipose, and cadaveric muscle tissues. In addition, we compared the properties of muscle-derived ECM isolated from young versus old donors. Proteomic analysis showed the presence of proteins involved in matrix remodeling, angiogenesis, cell growth and differentiation. ECM protein variability largely depended on tissue type and donor age. Regardless of tissue origin, ECM-based 3D gels affected cell morphology by reducing cell spreading. Differentiation of adipose-derived stem cells subjected to ECM sourced from various tissues showed no effect on adipogenic ADSC differentiation. Contrastingly, we found that the presence of young muscle ECM augmented chondrogenic ADSC differentiation, while old muscle ECM favored osteogenic ADSC differentiation. In summary, tissue- and age-related properties of ECM should be considered in developing 3D culture models for basic and translational research.

Biography:

Andriana Margariti has developed significant expertise in stem cell biology, with particular emphasis on cell reprogramming, chromatin remodelling, cell signalling and endothelial cell biology. Her research program is based on the remarkable idea of direct reprogramming which will allow her to realize fundamental principles of cell reprogramming and establish homogeneous populations of endothelial cells. Development of fast and robust new methodologies that produce well-characterized, homogenous, clinical-grade cells suitable for tissue repair/re-modelling would have great utility. Her research team has generated patient-specific iPS cells and PiPS cells and they now have exciting novel data and they are working towards to develop a remarkable new highly efficient strategy of cell reprogramming. Importantly, the potential of the reprogrammed endothelial cells to enhance angiogenesis and neovascularisation and to promote perfusion of ischemic tissue will be established. Her research is demonstrating the therapeutic potential of “re-born” reprogrammed endothelial cells which would have transforming consequences for regenerative and personalized medicine.

Abstract:

The capability to derive endothelial cell (ECs) from induced Pluripotent Stem (iPS) cells holds huge therapeutic potential for diabetes. This study elucidates the precise role of the RNA-binding protein Quaking isoform 5 (QKI-5) during EC differentiation from both mouse and human iPS cells and dissects how RNA-binding proteins can improve differentiation efficiency towards cell therapy for important vascular diseases such as diabetes. iPS cells represent an attractive cellular approach for regenerative medicine today since they can be used to generate patient-specific therapeutic cells towards autologous cell therapy. In this study, using the model of iPS cells differentiation towards ECs, the QKI-5 was found to be an important regulator of STAT3 stabilisation and VEGFR2 activation during the EC differentiation process. QKI-5 was induced during EC differentiation, resulting in stabilisation of STAT3 expression and modulation of VEGFR2 transcriptional activation as well as VEGF secretion through direct binding to the 3’ UTR of STAT3. Importantly, iPS-ECs overexpressing QKI-5 significantly improved angiogenesis and neovascularization and blood flow recovery in experimental hind limb ischemia. Notably, human iPS cells overexpressing QKI-5, induced angiogenesis on Matrigel plug assays in vivo only seven days after subcutaneous injection in SCID mice. These results highlight a clear functional benefit of QKI-5 in neovascularization, blood flow recovery and angiogenesis. They, thus, provide support to the growing consensus that elucidation of the molecular mechanisms underlying EC differentiation will ultimately advance stem cell regenerative therapy and eventually make the treatment of vascular diseases such as diabetes a reality.

Francesco Neri

Institute on Aging – Fritz Lipmann Institute, Germany

Title: Intragenic DNA methylation prevents spurious transcription initiation
Speaker
Biography:

Francesco Neri obtained PhD in Biotechnology in Siena (Italy) working on embryonic stem cells epigenetics. Then he worked as Postdoc between Human Genetics Foundation (HuGeF) in Torino (Italy) and the Radboud University Medical Centre, Nijmegen (Netherlands) where specialized his research on the DNA methylation specific epigenetic mark on stem cell, differentiation and colon cancer. During this period, he characterized new mechanisms of gene transcriptional regulation, developed new genome-wide methods and identified the role of the intragenic methylation. Since 2016, he is the Group Leader of the “Epigenetics research group” at the Fritz Lipmann Leibniz Institute on Aging (FLI) in Jena (Germany). His current research is supported by the Sofja Kovalevskaja starting grant of the von Humboldt foundation, and it is focused on the aging-dependent epigenetic aberrations occurring in adult stem cells promoting colon cancer.

Abstract:

DNA methylation is a heritable epigenetic modification required for embryonic development, which causes transcriptional repression when established on gene promoters. Recent studies have reported that Dnmt3b binds preferentially to the gene bodies by interacting with the histone modification H3K36me3. While the molecular and biological functions of intragenic DNA methylation are still unknown, the deregulation of this epigenetic feature has been associated with several diseases. Here, we show that the Dnmt3b-dependent intragenic DNA methylation protects the gene body from RNA Polymerase II (RNA Pol II) spurious entry and cryptic transcription initiations. Using different genome-wide approaches, we demonstrate that loss of Dnmt3b leads to an increase of the RNA Pol II engagement within gene bodies resulting in the onset of spurious intragenic transcription initiations. Finally, we demonstrate that inhibition of RNA Pol II spurious entry depends on the enzymatic activity of the Dnmt3b recruited by H3K36me3. Our results elucidate the functional role of the Dnmt3b-dependent intragenic DNA methylation, and the existence of a RNA Pol II-triggered epigenetic crosstalk involving SetD2, H3K36me3, Dnmt3b and DNA methylation, to ensure gene transcription initiation fidelity. This security feedback is probably lost during cancer development, where a global intragenic hypomethylation frequently occur, which increases transcription instability and promotes (stochastic) tumor cell heterogeneity.

Speaker
Biography:

Raphael Gorodetsky is the head of the Laboratory of Biotechnology and Radiobiology at the Sharett Institute of Oncology, Hadassah - Hebrew University Medical Center in Jerusalem, where he is employed as a faculty member since 1989 (affiliated to the Hebrew University Medical School in Jerusalem). He received his M.Sc. and PH.D from the Hebrew University in 1985 and had his Postdoc at UCLA Medical Center (1985-1988). Among other subjects he was involved in cancer research and radiobiology studies, as well as in projects in regenerative medicine. His earlier studies were associated with trace elements physiology in health and disease. Later directions of his research focused mainly on the invention of fibrin based biomaterials used for tissue regeneration and cell therapies and on different aspects of cancer research and radiation biology. Among the new ventures he co-founded was Hapto Biotech in 2000, where he served as the chief scientist (later merged with Ortec to form Forticel International, NY). In this area he designed fibrin based matrices for tissue regeneration, specifically of bone and cartilage. In recent years he was deeply involved in the development of placental stromal cell based treatment for regenerative and tumour control purposes, including the mitigation of radiation effects. These findings were applied clinically by Pluristem Therapeutics. In parallel he is involved in the development of new anticancer immunotherapies. Besides authoring more than 100 peer reviewed scientific publications and chapters, he edited and authored the book “Stem cells and Tissue Repair” in 2010 (by the Royal Society of Chemistry RSC, Cambridge, UK). He served as secretary of Israel Stem Cells Society (2014-2017) and has been a member in a number of related national and international societies.https://stemcell-regenerativemedicine.conferenceseries.com

Abstract:

We developed a technique for direct isolation of human placenta stromal cells from tissues of fetal origin (fPSC) by a unique process which allows the direct migration of the desired cells from tissue fragments to culture dishes. These selected placental stromal cell population was found to be highly potent indirect enhancers the regeneration of failing bone marrow and mitigation of acute radiation syndrome (ARS) following total body irradiation. The study was based on our previous record with similar cells of mixed maternal and fetal origin produced by a corporate in bioreactors to fit for clinical applications. The advantage of IM treatment by fPSC was that these stromal cells were found to be more immunocompetent and could reside longer in the injected muscle with no apparent adverse effects that were described following IV delivery of MSC of different origins, where most of the injected cells were trapped in the lungs. The cell treatments induced rapid indirect therapeutic effects. In studies on mitigation of radiation effects these cells enabled to the fast repopulation of the bone marrow lineages with subsequent regeneration of the peripheral blood cells. This saved the animals from the lethal effects of ARS with dramatic significant raise in ~8Gy irradiated mice from less than a third to almost 100%, with fast recovery of the bone marrow and peripheral blood cells. Cytokines analyses showed that the injected xenogeneic cells respond to the stress of the heavily irradiated mice by secretion of a wide range of related pro-regenerative cytokines. The IM based fPSC treatments have also been investigated as cell therapy for treating bone marrow failure due to different other causes. Other indications tested for optional PSC treatment include regeneration of the salivary glands after heavy dose head and neck irradiation, regeneration of tissues affected by autoimmune diseases. These included inflammatory bowel disease (IBD) and autoimmune inflammatory processes in the brain, such as multiple sclerosis. Further detailed studies are performed to better understand the indirect mechanism of action of the PSC by stress induced activation of relevant family of genes in these cells.

Biography:

Yuval Rinkevich is the young Principle Investigator with supervising and mentoring position at the Institute for Lung Biology and Disease, Comprehensive Pneumology Center (CPC), Helmholtz Zentrum, Munich, Germany. He obtained a PhD degree in Biology from Technion of Israel Institute of Technology, Haifa in 2008. From 2008 until 2014, he was postdoctoral fellow of Prof. Irving L. Weissman, Stanford Institute for Stem Cell Biology and Regenerative Medicine at Stanford University, USA. In 2014 he became Basic Life Science Research Associate at Stanford University. He was on the Team of inventors of two Patents regarding ‘Isolation and Characterization of Progenitor Cells from Mesothelium and Methods’ and ‘Compositions for the Prevention and Treatment of Surgical adhedions’. His lab is exploring the stem cells, embryonic lineages and mechanisms by which tissues/organs regenerate following injury, at multiple levels of biological organization. His research is currently funded by grants from the Else-Kröner-Fresenius Stiftung (EKFS), Human Frontier Science Program Organisation (HFSPO), German Research Foundation (DFG) and the Fritz Thyssen Stiftung (FTS). He is member of several Scientific Societies and publishes in peer-reviewed journals, gives lectures at numerous, distinguished Conferences and Institutes as COST, GRC, ICB, TUM, EMBO, TERMIS, BMF and ATS and was recently elected to the ETRS Board.

Abstract:

All mammals and humans undergo metamorphosis in response to injury, from Regeneration To Scarring (RTS). Here we follow two functionally diverged fibroblastic lineages (ENFs & EPFs) and document their lineage successions during backskin development that coincides with RTS. We show that ENFs are dermal sculptures that develop and regenerate native architectures during fetal life, and that their lineage decline over time imposes a dermal tissue absent of such events. We show that EPFs are scar producers even at fetal stages, wherein their numbers are below a threshold needed to generate macroscopic scars, but that their dynamics predicts scar emergence. We show that clonal advantages to EPFs rather than programed cell lineage death, most likely are primary succession mechanisms, and that RTS can be partly circumvented by transplantations of fetal ENFs or decellularised fetal dermis. Our findings provide a mechanism for regenerative decline in mammals, carry clinical implications by suggesting that human dermal regeneration could be reached by coxing or transplanting ENFs alone, and provide a model for comparative regeneration studies between taxon groups.

Biography:

Yibing Li the Founder and President of Midwest Joint Pain Institute and Centre for Pain Management & Rehab, is an interventional Physiatrist. She is American board certified in physical medicine and rehabilitation, pain medicine and small fiber nerve conduction studies. She graduated from Shenyang Medical College in China and completed residency at BJH/Washington University School of Medicine at St. Louis. She has over 12 years of clinical experience in spine and joint pain management. She has special interest and extensive training in regenerative medicine and is one of frontiers in performing prolotherapy, PRP, amniotic allograft tissue and bone marrow stem cell therapies in the United States.

Abstract:

Overview: To monitor the clinical outcomes from comprehensive regenerative therapies with prolotherapy, platelet rich plasma (PRP) and human amniotic allograft matrix injections to treat patients with chronic joint pain and all degrees of osteoarthritis in an outpatient clinic setting.

 

Introduction: Ligament laxity, tendinopathy, cartilage degeneration and previous joint injuries or surgeries are the major root causes of persistent joint pain and osteoarthritis. Dextrose prolotherapy, PRP and amniotic allograft injection into joints and surrounding weakened ligaments and tendons have been recommended to reduce chronic joint pain, improve functional status, and correct underlying causes of osteoarthritis. The current report is to assess the clinical outcomes of these comprehensive therapies among patients who had persistent joint pain and all degrees of degenerative joint disease/OA, ligament laxity or tendinopathy from imaging studies and failed with most of other conservative treatments.

 

Methods:  From September 2015 to August 2017, 118 patients  who had chronic joint pain and various degrees of osteoarthritis were treated with a series of injection of prolotherapy, PRP and human amniotic allograft tissue matrix to different parts of the body (N=94, 11, 9, and 4 to knees, hips, shoulders, and ankle/foot joints, respectively) under ultrasound needle guidance. The age range of the patient population is from 38-99 years, 62 males and 56 females. 31.4% of the patients had mild to moderate degenerative joint disease (DJD) and 64.4% had severe DJD. 19.6% of the patients had history of joint surgeries. Outcomes are the difference or % of change in VAS pain scale, upper extremity DASH and lower extremity 80-point functional scales, measures obtained from physical exams, X-ray and musculoskeletal ultrasound between pre- and 2-3 months, 4-6 months, 6 months and above post-treatments.

 

Results: After 2-6 months post-initial treatments, on VAS pain scales, 13.5% of the patients reported 0/10 or no pain and overall 74.6% reported at least 25%-50% pain reduction. 76.2% reported improvements in LEFS scale. Almost all patients showed varied degrees of improvements in both from soft tissue regenerations on ultrasound comparison and the physical exams between pre- and post- treatments. On the X-ray comparison, 66.7% showed improvement while 33.5% showed no change. None had experienced any adverse reactions from these injections.

 

Conclusion: Comprehensive regenerative therapies are safe, effective, non–surgical treatment options for patients with persistent joint pain and functional limitations from soft tissue degeneration/tears and osteoarthritis. Further treatments are recommended to some patients and continuous clinical studies are warranted to evaluate the long term benefits.

Biography:

Yibing Li the Founder and President of Midwest Joint Pain Institute and Centre for Pain Management & Rehab, is an interventional Physiatrist. She is American board certified in physical medicine and rehabilitation, pain medicine and small fiber nerve conduction studies. She graduated from Shenyang Medical College in China and completed residency at BJH/Washington University School of Medicine at St. Louis. She has over 12 years of clinical experience in spine and joint pain management. She has special interest and extensive training in regenerative medicine and is one of frontiers in performing prolotherapy, PRP, amniotic allograft tissue and bone marrow stem cell therapies in the United States.

Abstract:

Overview: To evaluate the clinical outcomes of comprehensive regenerative therapies with mesenchymal stem cells (MSCs) from bone marrow, platelet rich plasma (PRP) and dextrose prolo-solution injections to treat patients with persistent joint pain and severe osteoarthritis at an outpatient regenerative medicine clinic.

 

Introduction: Ligament laxity and cartilage degeneration are known to be root causes of osteoarthritis. Comprehensive regenerative therapies with the combination of dextrose prolotherapy, PRP and autologous stem cell injections into the pathologic joint and surrounding weakened tendon and ligaments are effective in improving pain and overall function in all degrees of osteoarthritis. In this study from an outpatient clinic, MSCs from bone marrow aspirate with PRP and dextrose prolotherapy were used for 19 patients (14 male, 5 female, age from 30-70 years old) from Sept. 2016 to August 2017. Among them, 18 patients had severe degree of degenerative joint disease/osteoarthritis; one had severe shoulder pain from rotator cuff tendon tears. All of them failed with traditional treatments and then selected for alternative treatments to surgeries. Treatments were made with one time PRPs and MSC bone marrow stem cells injections to 20 knees, 1 shoulder, 1 ankle and 2 hip joints, respectively under imaging needle guidance. Outcomes included the changes in VAS pain scale, upper or lower extremity functional scale, X-rays and measurements from musculoskeletal ultrasound pre- and post-treatments.

 

Results:  After 2-6 months post initial treatments, data was collected in 18/19 patients. There were an average of 58.2% pain reduction at VAS pain scale and 46.7 % improvement in functional scales. X-ray improvements seen in 2 out of 3 patients. Over 90% patients showed varies degree of new soft tissue growths from musculoskeletal ultrasound exam comparisons. No adverse reactions or complications were observed in all patients.

 

Discussion and Conclusions: Comprehensive regenerative therapies, as above, are new cutting-edge treatment compared to the alternate invasive orthopaedic surgeries after failure of other traditional standard-of-care treatments. Bone marrow stem cell aspiration and subsequent joint injections with added PRP and dextrose prolotherapy are safe, cost-effective and relatively low-risk for patients with persistent joint pain and arthritis caused by torn tendons, ligaments ligament laxity and degenerative cartilages. For moderate to severe degree of joint pain and arthritis, we would recommend more rounds of regenerative therapies to improve the results. Further clinical studies are warranted to evaluate the long-term benefits.

  • Young Research Forum
Location: Muggelsee

Chair

Walter Birchmeirm

Max Delbrueck Center for Molecular Medicine (MDC) in the Helmholtz Association, Germany

Session Introduction

Jeffrey Xue

CPMR - Midwest Joint Pain Institute, USA

Title: Bone marrow stem cell therapy for shoulder rotator cuff tear: a case report
Biography:

Jeffrey Xue is an avid learner and diligent pre-med student. He currently attends Knox College in IL, USA with major in Chemistry and minor in Chinese Study. He works at Midwest Joint Pain Institute during his summer time as a Research Assistant. He has strong interest in researching and studying stem cell therapy and regenerative medicine. He serves as a Student Athletic Advisory Committee Member at Knox and was the Founder and President of the science club at Lake Forest Academy. He has earned over 450 hours volunteer experience in the healthcare field.

Abstract:

Case Description: A 50-year-old male complained of right shoulder pain, popping and clicking with shoulder movements for one month after boxing. He couldn’t raise his right arm above his shoulder. He had difficulty sleeping at night because of the shoulder pain. He had previous right shoulder pain from rotate calf tendons and labrum tear, the pain was resolved from a series of prolotherapy and PRP injection over a year ago after failure of rest, NSAIDs, PT, etc. On the physical exam, he had reduced right shoulder range of motion to 50% normal. He had focal tenderness anterior right shoulder with (+) Hawkin’s test and (-) drop arm test. MSK ultrasound exam reviewed two large incomplete tears in the right subscapularis tendon, measured at 0.33 cm x 0.31 cm and 0.54 cm x 0.31 cm, respectively. He also had chronic and diffuse rotator cuff tendinopathy. Due to the recurrent injury and tendon tears, he decided to pursue bone marrow stem cell therapy. He received 14 cc of mesenchymal stem cells and growth factors injection from bone marrow aspiration to the torn subscapularis and other rotator cuff tendons with sterile techniques and imaging guidance at an outpatient regenerative medicine and PM&R clinic. The whole procedure lasts a few hours without IV sedation and he was discharged home without any complications.

 

Result: On the one month follow-up visit, he reported his right shoulder pain markedly reduced, he can sleep well at night. He can raise his right arm over the shoulder without pain. His shoulder ROM is nearly 90% normal range. On the repeated ultrasound exam, his previous torn subscapularis tendon had totally healed. In three months after the stem cell therapy, he was ready to go back to boxing.

 

Discussion: The adult bone marrow is a rich source of mesenchymal stem cells and growth factors. The bone marrow stem cell injection therapy allows our own body to restore, repair and regenerate damaged tissues and organs. Many research studies show that MSCs are primitive cells with capacities to self-replicate, fight apoptosis, reduce inflammation and differentiate into multiple tissues, including bone, muscles, tendons, ligaments, cartilage and fat. Due to the poor healing process from injuries or aging changes to the avascular structures, such as tendon, ligaments, cartilage, meniscus, labrums, etc., the patients usually suffer from the chronic joint pain and disability. Traditional treatments with NSAIDS, pain medications and arthroscopic surgeries are usually either not effective, lack of long term success or carrying significate risks and requiring prolonged healing time. The regenerative therapies with direct injections with MSCs and growth factors injection to those damaged tissues have clinically proven to be very simple procedures and highly effective treatments by targeting to resolve the root of chronic painful conditions with no downtime and very low risk or complications.

 

Conclusion: Bone marrow stem cell therapy is potentially very effective and alternative treatment to shoulder pain from incomplete rotator cuff tears and tendinopathy with very minimum risks, complications and side effects compared to current most traditional treatment options.

Biography:

Karthika Mayan is currently employed as a Research Assistant, in the study titled “An Exploratory Study on Environmental, Medicinal, Genetic and Dietary Determinants of Chronic Kidney Disease of Uncertain Aetiology Based on Postmortem Tissue Analysis” at Faculty of Forensic medicine, University of Colombo, Sri Lanka. Having completed Master degree in Bioinformatics with a merit pass and Bachelor’s degree in Biochemistry and Molecular biology at University of Colombo. During the MSc degree she investigated that 11 natural compounds can be used as cancer stem cells targeted potential anti-cancer compounds. In between she got the opportunity to work on a research project titled “In silico characterization of a RNA binding protein of cattle filarial parasite Setaria digitata” and published. Last year she has worked as a Research assistant at Bioinformatics and Computational Biology research group in UCSC, University of Colombo, and research topic on Co-evolution of Dengue virus and human. Since her long term goal was to be actively involved research. In the future she hopes that her research experience gained so far will be helpful that she can carry out valuable outcomes from her novel findings.

Abstract:

Cancer stem cells (CSCs) play significant roles in tumor initiation, relapse, angiogenesis, metastasis and therapy. Collectively Wnt, notch and hedgehog are major pathways that have been linked to the resistance of CSCs to conventional therapy. Most conventional therapies are directed at the rapidly growing tumor mass but not at the slow dividing CSCs. Eliminating CSCs has been suggested as a promising approach to cure cancer. In silico, molecular docking simulations were carried out for the binding of 35 selected natural compounds with receptor proteins which are involved in the main signalling pathways of CSCs, such as β-catenin chain A and Smo receptor from the Wnt and hedgehog pathways respectively, using Hex 8.0.0, DOCK6 and AutoDock Vina software. Additionally docking interaction residue analysis, score functions such as Drug score, X score, Per Contact Score (PCS) and Average affinity PCS and drug-likeness study were carried out for the selected compounds. Overall, 11 compounds were identified with good binding energy, interaction, binding affinity and better drug likeness for β-catenin chain A involved in Wnt inhibition. There was no considerable overall binding ability for Smo inhibition. Energy values for Wnt inhibition obtained using AutoDock Vina were as follows. Gedunin (-7.3 kcal/mol), Kaempferol (-6.1 kcal/mol), Methylripariochromene A (-5.3 kcal/mol), Myrigalone G (-5.1 kcal/mol), Catechin (-6.5 kcal/mol), Myricetin (-6.5 kcal/mol), Discretine (-5.6 kcal/mol), Laurolitsine (-5.9 kcal/mol), Myricitrin (-6.3 kcal/mol), Nordicentrine (-6.0 kcal/mol) and Phloretin (-5.4 kcal/mol). These novel Wnt inhibitors need further attention to assess their potential application in CSC targeted therapy.

Speaker
Biography:

Monika Glemžaitė is a second year PhD student in Biochemistry at Vilnius University. Her research is focused on epigenetic mechanisms distinguishing amniotic fluid-derived stem cells functioning. She is a co-author in the international patent application related to CRISPR/Cas9 and has been awarded a Young Inventor Medal by World Intellectual Property Organization (WIPO) for CRISPR/Cas research. She has two publications as a first author and has presented her work in six international scientific conferences. She is also actively spreading science among students and public in Lithuania.

Abstract:

Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) are fetal mesenchymal stem cells having multilineage differentiation potential and found in amniotic fluid. The aim of the present study is to assess the potency of AF-MSCs to differentiate into cardiomyogenic lineage in vitro using DNMT inhibitors, Decitabine, Zebularine, RG108 and p53 inhibitor Pifithrin-α and determine epigenetic changes during this differentiation. Cells were obtained from amniocentesis samples from second trimester women who needed prenatal diagnostics (protocols approved by the Ethics Committee of Biomedical Research of Vilnius District, No 158200-123-428-122). Isolated AF-MSCs were characterized by cell surface markers (CD44, CD90, CD105 positive and CD34 negative) and stem cells pluripotency genes (OCT4, SOX2, NANOG, REX1). Cardiomyogenic differentiation was confirmed by cell staining as well as by expression of cardio myogenesis-related genes NKX2-5, TNNT2, MYH6 and DES using RT-qPCR. MTT assay showed that all differentiation inducers and especially Pifithrin-α suppressed cell proliferation at the beginning of differentiation. Western blot analysis of epigenetic changes revealed the reduction in levels of Polycomb repressive complex 2 (PRC2) proteins EZH2 and SUZ12 and chromatin remodeling enzymes DNMT1, HDAC1/2 and HP1α after AF-MSCs induction with all agents. Also, alterations in marks both keeping active (H3K4me3, H3K9Ac and H4hyperAc) and repressed (H3K27me3 and H3K9me3) state of chromatin were observed. Our results demonstrate various changes in genetic and epigenetic profiles of AF-MSCs differentiated into cardiomyocytes progenitors resulting in global chromatin remodeling. Therefore AF-MSCs can be proposed as a potential alternative of stem cells for regenerative medicine.

Biography:

Collin Looi graduated from International Medical University in 2008. He is currently enrolled in a Master in Orthopaedic programme in Universiti Putra Malaysia. He has two published papers and is a keen researcher, with interest in Stem cells and non-unions of bone.

Abstract:

Granulocyte-Colony Stimulating Factor (G-CSF) is a native hematopoietic cytokine produced by fibroblasts, monocytes and endothelial cells, which primarily regulate neutrophil production and upregulates mesenchymal stem cell proliferation within the bone marrow and peripheral circulation. These mesenchymal stem cells have been described as key components involved in bone fracture healing. We hypothesized that by increasing the concentration of these mesenchymal stem cells by systemic administration of G-CSF, the rate of fracture healing could be expedited, potentially opening doors for further clinical applications. A caprine based animal study was done to assess the effect of systemically administered G-CSF on the rate of bone fracture healing involving 2 groups (control group and G-CSF group) with 5 animals in each group. Rate of fracture healing was assessed via radiographic methods
using a validated score measuring the parameters of bridging and union. Results revealed a median time to bridging of 7 weeks in the control group and 4 weeks in the G-CSF group (interquartile range of 0.5 weeks in both groups), while a median time to union of 13 weeks in the control group and 8 weeks in the G-CSF group (interquartile range of 1.5 weeks in both groups). A Mann-Whitney U test demonstrated a significant difference between the control group and G-CSF group in relation to bridging (p-value 0.008) and
union (p-value 0.008) times. This confirmed our research hypothesis that systemic administration of G-CSF produces an optimized fracture-healing rate.