Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 12th International Conference & Exhibition on Tissue Preservation, Life care and Biobanking (B2B & Networking) Atlanta, Georgia, USA.

Day 1 :

Keynote Forum

J Koudy Williams

Wake Forest Baptist Medical Center, USA

Keynote: Optimizing regenerative medicine therapies for differing patient populations

Time : 09:00-09:30

Conference Series Biobanking 2018 International Conference Keynote Speaker J Koudy Williams  photo
Biography:

J Koudy Williams is a DVM with over 35 years’ experience performing translational research using different animal models of human disease. He has published over 120 full-length manuscripts and 20 chapters and reviews. His focus is on women’s health and recently on regenerative medicine approaches to restoration of the urinary sphincter for women with urinary incontinence. This presentation is a review of determinates of cell therapy effi cacy on urinary incontinence using nonhuman primates as an animal model of aging, chronicity of disease, estrogen defi ciency, and obesity. The presentation concludes with a discussion of regenerative pharmacologic approaches for these different populations. 

Abstract:

Regenerative medicine off ers the promise of an unlimited amount of tissue and organ repair and replacement. Great progress has been made in preclinical studies and many applications are now in the clinical stage. Regenerative medicine is beginning to explore the potential eff ects of effi  cacy in diff erent patient populations. Th e risk urinary incontinence in women is age and obesity-related and is a chronic disease infl uenced by the sex hormone milieu. It is well known that aging and diabetes reduce the ability of the tissue to regenerate. It also stands to reason that these changes may also infl uence the effi  cacy of regenerative medicine approaches to urinary incontinence. In fact, this may explain, in part, why cell therapies for urinary incontinence are so successful in preclinical studies (which historically use younger health animals with acute UI). In contrast, the results of clinical studies in older women with varying body weights, sex hormone status and chronicity of disease. Th is presentation will fi rst review select studies identifying the eff ects of age, gender and hormone status on the ability of cells to stimulate regeneration of tissues. The majority of this presentation will introduce a female non-human primate (NHP) model of induce intrinsic urinary sphincter deficiency (ISD) and then present results of several studies describing the effects of skeletal muscle precursor cell (skMPC) treatment in acute vs. chronic fibrotic ISD; older and younger NHPs and in NHPs with stress-induced dysmenorrhea. Th e presentation will close with the results of recent studies identifying the use of chemokines on sphincter regeneration in this animal model.
 

Conference Series Biobanking 2018 International Conference Keynote Speaker Martin J D’Souza  photo
Biography:

Martin J D’Souza has obtained his PhD degree from the University of Pittsburgh, PA, USA. He is a Professor and Director of Graduate Programs in the College of Pharmacy at Mercer University in Atlanta, Georgia. He also serves as the Director of the Clinical Laboratory and Co-Director of the Center for Drug Delivery Research. He has graduated over 50 PhD students and has published over 100 manuscripts. He has been the recipient of several research grants from the National Institutes of Health (NIH), the American Diabetes Association, the Georgia Cancer Coalition, and Georgia Research Alliance. He serves on several Editorial Boards and is a journal reviewer for over 10 scientifi c journals and has several patents issued in the area of Nanotechnology. 

Abstract:

Regenerative medicine off ers an opportunity to restore or establish normal “pancreatic function”. Type 1 diabetes mellitus (T1DM) is a disease, characterized by lack of pancreatic islet function. Whole tissue transplantation appears to be a viable alternative in the management of T1DM due to limitation of exogenous insulin therapy. Th is study aims at fabrication and evaluation of smart biomaterials such as alginate-chitosan in microcapsule encapsulating insulin-secreting pancreatic islet cells using an automated novel specialized spraying nozzle. Microcapsules were characterized for permeability, stability, and cell viability. Microencapsulated β TC-6 cells were transplanted intraperitoneally into streptozotocin (STZ) induced diabetic mice and monitored for a decrease in blood glucose level and cell implant tolerance. Spherical microcapsules with diameter in the range of 250-350μm were prepared at an air fl ow rate of 250L/hr. Since the process is automated, this allows for small or largescale production with minimal batch-to-batch variation. Microencapsulated β TC-6 cells in alginate capsules demonstrated prolonged viability. Mice that received microencapsulated β TC-6 cells maintained normoglycaemia for the study period of around 35 days. However, the mice that received naked pancreatic islet cells rejected the graft  within 1 or 2 days. In conclusion, microcapsules produced by the specialized nozzle were reproducible with narrow size distribution. Our fi ndings using mice in the in vivo studies revealed that transplantation of microencapsulated β cells may be a viable alternative in the management of T1DM with greater immune acceptance.
 

Keynote Forum

Beverly Torok Storb

Fred Hutchinson Cancer Research Center, USA

Keynote: Marrow fi broblasts induce monocytes to promote endogenous regeneration

Time : 10:00-10:30

Conference Series Biobanking 2018 International Conference Keynote Speaker Beverly Torok Storb  photo
Biography:

Circulating monocytes can transmigrate the vascular wall, enter a specifi c tissue, and diff erentiate into macrophages to perform functions relevant to that microenvironment (ME). Using the bone marrow as a model ME we focused on the ability of marrow fi broblasts to provide signals to monocytes. Th ese marrow fi broblasts appear to remain constant in their support of continuous “regeneration“, otherwise known as blood cell production. To study marrow fi broblasts we isolated and immortalized distinct fi broblast lines from normal human marrow: one designated HS27a expresses CD146, IGA3, and maintains early progenitors in culture. Monocytes co-cultured with HS27a assume a macrophage phenotype and express genes that function in tissue and organismal development. We also cloned a canine equivalent of HS27a, designated DS1, to be used in vivo in canine models of ischemia-induced myocardial infarction (MI) and radiation-induced pulmonary fi brosis (PF). 107 DS1 cells were infused intravenously aft er establishing the existence of an injury: 2 weeks post-infarction in the MI model and 5 weeks aft er irradiation in the PF model. In both, recovery of function was signifi cantly improved in dogs given DS1 cells compared to controls. Histological examination of tissue at necropsy also showed signifi cant diff erences between DS1 treated dogs and controls. Th e more complete tissue regeneration was observed in the lung model in which dogs were euthanized 21 weeks aft er treatment, whereas MI dogs were euthanized only 2 weeks aft er treatment. Importantly 48 hours aft er infusion there was no evidence by PCR of DS1 cells in the regenerating tissues or in the blood. Th is was in agreement with previous studies using 111In-DS1 cells which showed the DS1 cells are trapped in the lung where they are destroyed. However within 6 hours of DS1 infusion circulating monocytes expressed activation markers, and between 2 and7 days post DS1 infusion, large colonies of endothelial-like cells were grown from blood mononuclear cells. We propose that circulating monocytes come in contact with and receive signals from the DS1 cells sequestered in the lung. Th ese signals prepare the monocytes for recruitment to sites of injury where they scavenge debris and promote and regulate a multi-step process of endogenous regeneration.
 

Abstract:

Beverly Torok Storb is a full member of the Faculty at the Fred Hutchinson Cancer Research Center and Director of the Core Center of Excellence in Hematology. Her research focuses on dissecting the marrow microenvironment to understand the regulation of blood cell production and endogenous regeneration. 

Conference Series Biobanking 2018 International Conference Keynote Speaker Vincent S Gallicchio  photo
Biography:

Vincent S Gallicchio earned his PhD in Experimental Hematology at New York University Medical Center. He completed his hematology fellowship at Memorial Sloan Kettering Cancer Center and did post-graduate training at the University of Connecticut Health Science Center. He received his diploma in medicine from the “Vasile Goldis” University of Arad (Romania). He has served on the faculty of Yale Medical School and the University of Kentucky Medical Center before his current tenure as professor of biology, microbiology, and public health sciences at Clemson University, where he also serves as the Director of the Education & Research Center for Trace Elements that operates under the auspices of UNESCO. He has published more than 200 scientifi c articles many dealing with the biology of stem cells, regenerative & personalized medicine and the alternative medical uses of lithium, many book chapters, and full texts. He is a Fellow of the Association of Clinical Scientists, the Association of Schools of Allied Health Professions, and the Royal Society of the Arts. In 2003, at the 200th anniversary of RSA, because of his long-standing effort to educate and train biomedical laboratory scientists from England, he was recognized for his efforts by being presented to Her Majesty, Queen Elizabeth II at Buckingham Palace. 

Abstract:

Lithium (Li) salts have been widely used in psychiatry as mood stabilizing agents for 66 years. Li is found in variable amounts in foods, especially grains, vegetables, and in various geographical areas. Additionally drinking water provides a signifi cant source of the element. Dietary intake in humans depends on location, type of foods consumed, and fl uid intake. Traces of Li have been detected in human organs and tissues, leading speculation that the element is responsible for specifi c functions in the human body. It was not until the 20th century that studies performed in the 1970's and 1990's, primarily in chickens, cows, goats, and rats, maintained on Li defi cient diets demonstrated higher mortality, and altered reproductive and behavioral abnormalities. Such defi ciencies have not been detected in humans; however, studies performed on populations living in areas with lower Li levels in water supplies have been associated with higher rates of suicides, homicides, and the arrest rates for drug abuse and other crimes. Th us, Li appears to play a signifi cant role in early fetal development evidenced by high Li levels during the early gestational period and perhaps social behavior. Biochemically, the mechanism of Li action involves multifactor and interconnected pathways with enzymes, hormones, vitamins, transcription, and growth and transforming factors involved. Th is body of evidence now appears suffi  cient to label Li as an essential element with the recommended RDA for a 70kg adult of 1mg/ day. Of extreme importance for the future is the increasing clinical data indicating Li can be used eff ectively for the treatment of acute brain injuries, e.g., ischemia and chronic neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Tauopathies, and Huntington's disease. Th is conclusion is based on evidence showing Li as important in neurogenesis, neuronal repair, as well as protecting neurons from neurotoxicity.
 

Conference Series Biobanking 2018 International Conference Keynote Speaker Timothy Lyden  photo
Biography:

Timothy Lyden graduated from the University of Maine-Orono in 1992 with his Ph.D. in reproductive cell biology. In 2001 he relocated to UW-River Falls from The Ohio State University Medical School. During the previous 11 years, he had worked as a biomedical researcher focused on the normal human placenta at both Ohio State and Wright State University Medical Schools. During that time he held positions as a Senior Post-Doctoral Fellow, Research Scientist and Research Assistant Professor serving as a co-investigator for nearly $4 million in NIH research grant projects. In 2001, he relocated to UW-River Falls in order to balance his scholarship with more teaching as well as to develop an ongoing independent research program. Following several successful smaller projects focused on various aspects of placental cell biology during 2001-03, he shifted his research focus in 2004 to modeling cellular and tissue aspects of developmental and tumor biology using tissue engineering methods. He is now a Full Professor of Anatomy and Physiology in the Bio-Medical track at UW-River Falls. In addition, he participates in teaching within both the Biotechnology and Neuroscience programs and maintains an active research center involving undergraduates in mentored projects. Throughout his career, he has authored or co-authored more than 22 chapters and papers while also presenting more than 200 posters around the world. 

Abstract:

Tissue engineering and regenerative medicine have developed in various different directions while collectively growing into the burgeoning new fields that we see today. One area of interest to our laboratory seeks to apply engineered or “artificial” tissues as alternatives to cell cultures and live animals in modeling cancer. Our laboratory at the TCIC has been applying engineered tissue approaches over the past 14 years to explore the use of these technology solutions to study normal and cancerous tissues. From initialization/colonization to long-term progression, metastasis and target attachment/invasion we are exploring artificial tumor tissues using a variety of different natural and synthetic substrates as well as several different culture conditions. Her work focused on two specific types of tumors which will be presented, melanoma and breast cancer. In each of these cases, we have successfully developed in vitro tumor models which present with many features observed in patient samples while providing a platform to directly study cellular and tissue interactions and mechanisms involved at various stages of these diseases. In the case of melanoma, models of tumor generation and progression using a hydrogel-based matrix combined with B16F1 and B16F10 mouse melanoma cell-lines will be the focus. These studies demonstrate the capacity of in vitro 3D models to very closely replicate clinical observations of cutaneous melanoma. In addition, since these in vitro models are maintained intact for extended growth periods, out to 6 months in these studies, the resultant artificial tumors effectively demonstrate the process of tumor progression as well. A hallmark of this progression is the generation of two-three cellular populations that display clearly distinct morphological and behavioral characteristics. In the second case, human breast adenocarcinoma cell line MCF-7 was employed with several different natural matrix materials in addition to our standard hydrogels, to generate a library of studies with artificial tumors tissues extending over very long-term culture periods, up to 4 years in some cases. Results from several types of scaffold and their implications as evidence of the value of this modeling system will be presented. In these studies collectively, tumor progression leads to the staged development of single cell release followed by cancer cell cluster release and definitive spheroid formation followed by distant colonization of the culture wells. Our working hypothesis is that these stages and the released products, particularly the clusters/spheroids, represent a direct correlate to clinical metastasis. We are also studying the nature and behaviors of these released cells under various conditions and using a variety of methods include immunolabeling, flow cytometry, and western blotting. Taken together, these presented studies demonstrate the power and effi cacy of in vitro 3D artificial tissues as models of clinical disease in cancer and support our assertion that these essentially represent designer “Lab-Animals-in-a-Dish”.
 

Keynote Forum

Kelvin Brockbank

Clemson University and Tissue Testing Technologies LLC, USA

Keynote: Ice-free cryopreservation of natural and bioengineered tissues

Time : 11:45-12:15

Conference Series Biobanking 2018 International Conference Keynote Speaker Kelvin Brockbank  photo
Biography:

Kelvin Brockbank, CEO, and Founder of Tissue Testing Technologies LLC is a Research Professor of Bioengineering at Clemson University and Adjunct Professor of Regenerative Medicine and Cell Biology at the Medical University of South Carolina. His research interests include cell, tissue and organ cryopreservation for test systems and transplantation and manufacturing methods for cell-based bioengineered therapy products. His work has led to the establishment of two successful publicly traded low-temperature technology platform companies, CryoLife Inc., and Lifeline Scientifi c. He has over 500 publications and presentations at national and international conferences including more than 30 patents related to hypothermic, frozen and vitrifi ed biomaterial preservation. 

Abstract:

Effective improved tissue banking methods for natural and engineered tissues, complex vascularized allotransplants and organs are desperately needed for transplantation. Banking of living cellular tissues using current tissue banking practices employing conventional cryopreservation by freezing is not feasible due to the well-documented damage caused by ice formation. An alternative ice-free cryopreservation approach is vitrification. Formation of ice is prevented by the presence of high concentrations of cryoprotectants with preservation of extracellular matrix components and optional preservation of cells. Ice-free vitrifi cation works for a variety of natural and engineered tissues, using a formulation consisting of DMSO, formamide and propylene glycol, known as VS55, but have been unsuccessful at sample volumes over a few mLs. The major constraints for scale-up of cryopreservation by ice-free vitrification have been avoidance of ice nucleation during warming and mechanical forces generated by glasses at low temperatures. In this presentation, I will focus on strategies for avoidance of ice nucleation. Our fi rst successful strategy for large tissue samples was an 83% formulation based upon the same cryoprotectants, known as VS83. This formulation can be used to retain viable chondrocytes in large osteochondral graft s or for non-viable cardiovascular graft s with retention of extracellular matrix integrity, depending upon the way in which the formulation is added and removed before and after vitrifi cation. Non-viable cardiovascular graft s with intact matrix have been a major research focus for the last 10 years and both in vitro and in vivo results demonstrated signifi cantly reduced immunogenicity in heart valves, including reduced memory T-cell proliferation and most recently modulation of TGF-β1 from latent to active form among other statistically signifi cant eff ects. We have been successful in scaling up the viable preservation of large tissue samples using either nano warming, inductive heating of iron nanoparticles, or convection warming using improved ice-free vitrification formulations.
 

Keynote Forum

Svetlana Gramatiuk

Ukraine Associated of Biobank, Ukraine

Keynote: Biobank sustainable plan in developing countries

Time : 12:15-12:45

Conference Series Biobanking 2018 International Conference Keynote Speaker Svetlana Gramatiuk  photo
Biography:

Svetlana Gramatiuk (MD, Ph.D.) serves as President of Ukraine Associated of Biobank that she co-founded in 2017. She was also the Medical Director Research Biobank ASK-Health (2015-2016) and the Ukraine Editor of the journal Advanced Research Biobank and Pathophysiology from 2017. Previously, she also established and/or managed several biobanks in Ukraine. In addition to his unique expertise in Biobanking, she also did Master of Science Biobanking in Medical University Graz and has an in-depth knowledge of oncology biomarker research in the position holding a from Head Department Medical and Research Laboratory in the Grigoriev Radiology and Oncology Institute and having completed a Post-Doctoral Fellowship at the Kharkiv National Medical University (Department Pathophysiology from Kharkiv, Ukraine

Abstract:

Quality biological samples stored in Biobanks are essential for the provision of appropriate health services and also act as a reservoir for the development of precision medicine and biotechnological innovation. The Ukrainian Association of Biobanks (UAB) coordinates the network procedures, based on ESBB, ISBER, BBMRI, and NCI guidelines, which are standardized across the entire network. Policies and documents including Patient Consent Policy, Patient Information Sheet, Biobank Informed Consent Form, and Sample Application Form after extensive and constructive discussion in UAB have been agreed for use in each hospital and medical institute. An optimum sequence for document preparation and submission for review have been outlined. At the outset, UAB employed ESBB standard operating procedures (SOPs) for tissue collection, storage, and processing. Harmonization of policies and SOPs is an ongoing activity and standardization is achieved via workshops and training in data protection, database management, data sharing, tissue collection and storage, ethical considerations, sample access, and quality control. It was important at the initiation of UAB to develop a sequence for policy development, review, and approval. It was essential that policies were compliant with both national and European guidelines and directives. UAB introduced a standardized Patient and Volunteers Information Letter, Informed Consent Form of Biobank and Sample Access Policy in Ukraine for the first time. It was difficult, in the beginning, to distinguish the order in which to develop, to agree and finally to submit documents of UAB for review. The Ireland pathway was identified as the most effective for Ukrainian local circumstances and it was decided to adopt a similar sequence of document development and submission. Fist, the consensus was reached with each hospital within the network. Second, a review of documents by the Risks and Legal Department in UAB was performed. Parallelly a formal legal statement was obtained from an independent lawyer specialized in medical research.
 
Conclusion: The UAB short- to medium-term objectives include expanding the current network and to include each of the major medical research centers in Ukraine. Enlargement of the network could be undertaken with modest capital investment and UAB medium size will facilitate a swift and dynamic approach to network expansion. Ideally, the Ukrainian biobank network should be integrated into working groups within larger European networks in the future as it is affiliated with the Science and Innovation Working Group for international biobanking ESBB. By now, the UAB is reaching its goal: to develop infrastructure and facilitate the biomedical research nationally and internationally.

 

  • Stem cell and Stem cell biobanking | Vitrification | Fertility preservation | Biobank sustainability: current status and future prospects | Tissue engineering | Next generation Biobanking | Cryopreservation methods| Bio preservation and its Advances | Biobanking & expertise networks |

Session Introduction

Kejin Hu

University of Alabama at Birmingham, USA

Title: Role of bromodomain extra terminal proteins in cellular reprogramming

Time : 13:45-14:05

Speaker
Biography:

Kejin Hu received his PhD degree in animal molecular biology in 2003 from Hong Kong University. He received postdoctoral training in human pluripotent stem cell biology at University of Wisconsin – Madison. He has more than 12 years of experience in pluripotent stem cells and cellular reprogramming. He has established his own laboratory at the Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham (UAB). He is currently an Assistant Professor in stem cell biology and cellular reprogramming at UAB. He has been involved in the improvement of iPSC technology. His method of iPSC reprogramming is widely used, and human iPSC lines he generated are also widely used. He has authored more than 20 scientifi c articles. His lab is deciphering the molecular regulation of pluripotency, and dissecting the molecular mechanisms of cellular reprogramming. The works from his laboratory enhance our understanding of iPSC reprogramming process. His research is supported by Alabama Institute of Medicine, American Heart Association, and NIH.
 

Abstract:

Pluripotent stem cells (PSCs) have differentiation potentials into any type of cells in our bodies and therefore hold great promise for regenerative medicine. The conventional PSCs are embryonic stem cells (ESCs) derived from the inner cell mass (ICM) of a pre-implantation embryo. The clinical application of ESCs is hampered by issues of ethical concern, technical limitation, a limited supply of human embryos, and non-autologous nature. PSCs can be induced from fibroblasts or other somatic cells by ectopic expression of a few transgenes, e.g. OCT4, SOX2, KLF4, and C-MYC. Induced pluripotent stem cells (iPSCs) have removed many problems associated with the use of ESCs. However, iPSC reprogramming still has problems of low efficiency, incomplete reprogramming, epigenetic memory of the starting cells, stochastic nature, immunogenicity, and reprogramming-associated mutagenesis. The molecular mechanisms of iPSC reprogramming remain poorly understood. My lab found that proteins of bromodomain extra-terminal (BET) family play different roles in iPSC reprogramming process. By screening a human cDNA library, we discovered that a short isoform of human BRD3, BRD3R, displayed reprogramming activity while other BET proteins lack reprogramming activity. BRD3R also gains mitotic activities, which may be partially responsible for its gained reprogramming activity. Interestingly, our further detailed study showed that chemical inhibition of BET proteins at low concentration enhanced iPSC reprogramming by three distinct BET inhibitors, while a high concentration of these BET inhibitors impairs iPSC reprogramming. At the same time, our RNA-seq data showed that such mild chemical inhibition of BET proteins erased fibroblast transcriptional program. Our published and unpublished data indicate sophisticated roles of BET protein in cellular reprogramming.
 

Eric B Kmiec

Christiana Care Health System and University of Delaware, USA

Title: CRISPR-directed gene editing creates genetic heterogeneity surrounding the sickle cell disease point mutation

Time : 14:05-14:25

Speaker
Biography:

Eric B. Kmiec is well-known for his pioneering work in the fi elds of molecular medicine and gene editing. Throughout his professional career, He has led research teams in developing gene editing technologies and genetic therapies for inherited disorders such as Sickle Cell Disease. He is the recipient of multiple research awards from the National Institutes of Health (RO1s, R21s), the American Cancer Society, and private foundations including the 2012 Proudford Foundation Unsung Hero Award in Sickle Cell Disease. He has been a member of numerous editorial boards, NIH study sections and review boards and is the (primary/senior) author of more than 145 scientifi c publications (mostly in genetic recombination and gene editing).

Abstract:

CRISPR-directed human gene editing in somatic cells and stem cells is rapidly transitioning from bench to bedside with clinical trials beginning to appear throughout the world. Our laboratory and others have demonstrated that the repair of a point mutation can be facilitated by the combined activity of a single-stranded oligonucleotide and the CRISPR/Cas9 system. Using K562 cells, we found that the presence of the appropriate CRISPR/Cas9 complex leads to an enhancement in the frequency of gene editing. While gene editing activity does in fact increase in a dose-dependent fashion, we fi nd a heterogeneity of modifi ed genetic sequences that are created and maintained even when repair at the sickle cell disease mutation in the human beta-globin gene is successful. Our data suggest that CRISPR complexes leave a genetic footprint at the target site by creating a DNA junction heterogeneity as a byproduct of the reaction. Th is heterogeneity could be called on-site mutagenesis and one of the most interesting is a curious pattern of DNA nucleotides adjacent to the double-stranded break site; footprints of the human delta-globin gene. A targeted population of cells contains one allele of apo appears to be a chimera of the HBB and the hemoglobin delta gene (HBD). Our results suggest the genes with similar sequences (related family members) such as HBB and HBD could be involved in template repair of double-strand DNA breaks independent of exogenously added donor DNA. We further evaluated the relationship among cellular delivery, nuclear uptake in CD34+ progenitor cells, oft en viewed as the benchmark metric of successful gene editing, and single base repair. We took a similar combinatorial approach using single-stranded oligonucleotide and a CRISPR/Cas9 ribonucleoprotein to convert wild-type HBB into the sickle cell genotype. Confocal microscopy data show that the CRISPR/Cas9 ribonucleoprotein tends to accumulate at the outer membrane of the CD34+ cell nucleus when NEON Transfection System is employed, while the ribonucleoproteins do pass into the cell nucleus when nucleofection is used. When the correct delivery system is maximized, we once again a similar genetic pattern of on-site heterogeneity created by the activity of the CRISPR Gene editing complex. Our results indicate that more stringent criteria must be established to facilitate the clinical translation and scientifi c robustness of gene editing for sickle cell disease, most critical in stem cell therapy.
 

Speaker
Biography:

Naresh Kumar Rajendran has completed his Ph.D. (Medical Biochemistry) in 2015 from University of Madras, India and currently working as a Postdoctoral Reseach Fellow in Laser Research Centre, Faculty of Health Sciences and University of Johannesburg, South Africa. He has two years of research experience at the University of Tokyo, Japan. He has published more than 15 papers in peer-reviewed journals and has been serving as a reviewer for 5 international journals. He has presented his research outcomes in various international and national conferences. He is engaged in national and international collaborations and co-supervising postgraduate students.
 

Abstract:

Delayed wound healing is considered one of the most common and serious complications of diabetes mellitus. High glucose levels induce oxidative stress and it leads to the central pathogenesis of chronic complications associated with diabetes. Controlling the factors leading to oxidative stress outbursts may emerge as an effective step to treat diabetic chronic wounds. Photobiomodulation (PBM) is a technique based on the use of low-powered light to regulate various pathophysiological cellular effects, including inflammation and chronic wounds. However, the exact mechanism of action behind PMB remains unclear. The present study aimed to determine if PBM at 660nm attenuates oxidative stress by inhibiting the FOXO1 signaling pathway. Four experimental group’s namely normal, wounded, diabetic and diabetic wounded WS1 fibroblast cells were exposed to laser irradiation at a wavelength of 660nm and a fluence of 5J/cm2; non-irritated cells served as experimental controls. Migration of cells was determined by inverted microscopy at 0, 24 and 48h. Cells were harvested at 0, 12, 24 and 48h. Cell viability was determined by the trypan blue exclusion assay and adenosine triphosphate (ATP) luminescence assay. Indirect ELISA was used to determine the levels of HMOX1, SOD, and CAT in cultured cells. Western blotting and immunofluorescence were performed to analyze AKT and FOXO1. RT-PCR was used to determine gene expression of TGF-β, PDGF, and VEGF. Irradiation of cells resulted in a significant decrease in FOXO1 and increase in AKT protein. The treatment also upregulated the expression of TGF-β1, PDGF and VEGF genes. The level of antioxidants such as HMOX1, SOD, and CAT was also increased in irradiated groups. The present study confirmed that PBM at 660nm enhances wound healing through attenuation of oxidative stress by regulating the FOXO1 signaling pathway.
 

Speaker
Biography:

Yoo-Hun Suh is now Chaired Professor and President of the Neuroscience Research Institute of Gachon University. He was the founding president of Korea Brain Research Institute. Furthermore, he is a Professor Emeritus of College of Medicine at Seoul National University. He won Korea’s Most Distinguished Scientist Award, the National Government Medals, and many other prizes. He was selected one of 20 outstanding Korean Medical Scientists and one of 21 outstanding Korean Scholars of the 21st Century. He is an Editor and Editorial Board Member for 6 SCIs. He fi first cloned the gene for epinephrine synthesizing enzyme, PNMT and has greatly contributed to the discovery of a new potential gene and factors for the AD, the development of potential stem cell and drugs for AD and PD. He has published more than 200 papers and over 50 books. Presently, he is a Member of Board of Trustee, HFSP, a Member of Korean National Academy of Science and Technology, a Member of Korean National Academy of Medicine and a Member of Korean National Science and Technology Council. In addition, he is a Member of International Scientific Advisory Board of AD & PD and a Member of International Scientific Advisory Board of International Conference of Alzheimer’s Disease and related disorders.
 

Abstract:

Alzheimer’s disease (AD) is characterized by the accumulation of amyloid plaques and neurofibrillary tangles accompanied by cognitive dysfunction. We examined whether intracerebrally or intravenously transplanted human adipose-derived stem cells (hASCs) could have therapeutic or preventive effects in AD/PD mice model. We demonstrated that intracerebral or intravenous injection of hASCs rescued memory deficit and gave benefits of blocking the pathogenesis in the brain of AD Tg mice by reducing the number of plaques and neuropathology. Among stem cells, autologous human adipose-derived stem cells (hASCs) elicit no immune rejection responses, tumorigenesis, or ethical problems. We found that intravenously transplanted hASCs passed through the BBB and migrated into the brain. The e-learning, memory, and pathology in an AD mouse model (Tg2576) mice greatly improved for at least 4 months after intravenous injection of hASC. Th e number of amyloid plaques and Aβ and APP-CTs levels decreased signifi cantly in the brains of hASC-injected Tg mice compared to those of the Tg-sham mice. Intravenously or intracerebrally transplanted hASCs significantly rescued memory defi cit and neuropathology in the brains of Tg mice by upregulating IL-10 and VEGF and be a possible use for the prevention and treatment of AD. We are currently conducting clinical phase I & II study of hASCs for an AD in the USA. Here, we demonstrated that transplantation of neural stem cells into 12-month-old Tg2576 brains markedly improved both cognitive impairment and neuropathological features by reducing β-amyloid, secretion of anti-inflammatory cytokines, endogenous neurogenesis, as well as synapse formation. In contrast, the stem cell transplantation did not cover cognitive dysfunction and β-amyloid neuropathology in Tg2576 mice aged 15 months when the memory loss is manifest. However, the optimal stage of the disease for stem cell transplantation to have a therapeutic effect has yet to be determined. Overall this study underscores that stem cell therapy at an optimal time frame is crucial to obtain maximal therapeutic effects that can restore functional deficits or stop the progression of the AD. The treatment of Parkinson’s disease (PD) using stem cells has long been the focus of many researchers, but the ideal therapeutic strategy has not yet been developed. The hASC were intravenously injected into the tail vein of a PD mouse model induced by 6-hydroxydopamine. The behavioral performances were significantly improved at 3 weeks after the injection of hASC. Additionally, dopaminergic neurons were rescued, the number of structure-modified mitochondria was decreased, and mitochondrial complex I activity was restored in the brains of the hASC-injected PD mouse model. Overall, this study strongly indicated that intravenously transplanted hASC may have therapeutic potential for PD by recovering mitochondrial functions.
 

Speaker
Biography:

Matthew Fischer is a Former Government Attorney with a passion for the emerging stem cell industry. Prior to joining the fi rm, he served as a Senior Attorney Advisor in the US Department of Health and Human Services, Office of Medicare Hearings and Appeals. He advised Federal Administrative Law Judges (ALJs) on reimbursement appeals, including Medicare contractor overpayment audits (i.e., RAC, ZPIC, and PSC), statistical sampling extrapolation issues, Medicare Part D exceptions, and secondary payer appeals. His current practice is concentrated in healthcare with a strong focus on stem cell regulatory development and enforcement.
 

Abstract:

There are no off the shelf solutions when it comes to starting a new business or adding a new component to a practice. Between navigating regulations, receiving training, and marketing the service, there's a lot to address in a short time. Trying to do it all yourself? You may be a highly trained clinician, but given healthcare’s ever-changing regulatory environment, it is highly encouraged for providers to seek experienced counsel first. To get started, here is a short summary of what to expect. The first issue is always protection when starting a business or adding a new service. Take the case of an orthopedic physician that wants to add stem cell treatments (e.g. PRP) to his or her practice. The initial inclination is usually to create a new entity separate from the medical practice. What the physician is unaware of is that this may create exposure to state self-referral laws. Typically, under these types of laws, the intent is not a requirement. Therefore, is it important to determine if your state has this type of law and if so, how to structure the new venture before moving forward? The FDA has the authority to regulate stem cell products. Most products are regulated by the FDA as an HCT/P while others are not. Many direct-to-consumer providers are unsure about what this means and need to know what they are able to do and say. Thus, it is imperative to be up to date on the issues and know what boundaries have been set by the FDA. With the rise of stem cell science and treatments, many state boards have warned physicians that they could face potential disciplinary action for failing to meet the prevailing professional standard of care or for performing an experimental procedure without first obtaining full, informed consent. Many physicians would like to have a mid-level practitioner (i.e. NP or PA) assist or provide injections. Once again, state regulations need to be reviewed to determine who can provide treatments and what level of supervision is required.
 

Biography:

Stanca A Birlea MD has completed her Ph.D. at the age of 34 from the University of Medicine Cluj-Napoca Romania and postdoctoral studies on the Genetics of Vitiligo and Other Autoimmune Diseases in Professor Richard Spritz lab from University of Colorado School of Medicine. She is an Associate Professor in the department of dermatology at the University of Colorado. She has published more than 25 papers in reputed journals and has been serving as an editorial board member of repute Pigment Cell & Melanoma Research Journal. Her projects focus is on melanocyte regeneration in vitiligo, a collaboration between Department of Dermatology–Chair Professor David Norris- and Gates Center for Regenerative Medicine-Director Dr Dennis Roop.
 

Abstract:

Vitiligo is an autoimmune depigmentation disorder characterized by white spots on the skin that cause profound social and psychologic stigma in patients. Vitiligo is caused by CD8+ T cell-mediated destruction of epidermal melanocytes (MCs). Vitiligo repigmentation requires proliferation and migration of MC precursors from the hair follicle (HF) bulge to repopulate the interfollicular epidermis, and the strongest stimulus for this process is Narrow Band UVB (NBUVB). To better understand this process, we developed a research platform that used skin biopsies collected from 6 vitiligo patients treated with NBUVB and 6 untreated vitiligo patients, using rapid fluorescent immunostaining combined with laser capture microdissection to collect RNA from bulge MC precursors and mature MCs from the epidermis of the regenerated vitiligo skin. The total RNA captured from MCs was subjected to whole transcriptome RNA sequencing, followed by gene expression analysis. We found upregulation of TNC, GJB6 and THBS1 transcripts in the bulge MC precursors of NBUVB-treated vitiligo skin as compared with epidermal MCs of regenerated NBUVB-treated vitiligo skin, and of β-catenin as being the top upstream transcription regulator of this process. We also identified that GLI1, a candidate stem cell-associated gene, was significantly modulated by NBUVB in the bulge MCs. The above pathway and signals are potentially key-players in the activation of bulge MC precursors during vitiligo repigmentation.
 

Biography:

Renee Cottle is an Assistant Professor of Bioengineering at Clemson University. She earned her Ph.D. in Biomedical Engineering from the Georgia Institute of Technology and Emory University in 2015. She completed a T32 Postdoctoral Fellowship in the Cardiovascular Research at MUSC in 2016 and started her faculty position at Clemson in August 2016. Her expertise is in gene editing, gene therapy, and non-viral delivery strategies. Her research group is focused on cell-based gene therapies for inherited metabolic diseases of the liver, including familial hypercholesterolemia. Her research addresses technical barriers for advancing novel gene therapies for genetic disorders.

Abstract:

CRISPR and associated protein (Cas9) system have proven to be the most promising gene editing tool available for therapeutic applications due to its facile design and robust targeting activity in mammalian cells. When coupled with a donor template, CRISPR-Cas9 nucleases trigger the homologous directed repair pathway to precisely incorporate new gene sequences into the genome, which can be leveraged for the correction of genetic diseases in the patient’s cells. Despite the advantages of this approach, there are several barriers to its application in humans. One major hurdle is delivering a sufficient amount of CRISPRCas9 complexes and donor templates into target cell types. In previous research, we investigate microinjection, traditionally applied for in vitro fertilization, for direct, controlled delivery of nucleases and donor template into human hematopoietic cells as a novel therapeutic strategy for sickle cell disease. We characterized a microinjection system, investigated the effects of microinjection on cell functionality, and demonstrated proof-of-principle of gene editing in human hematopoietic K562 cells microinjected with TALENs and CRISPR-Cas9 along with donor template. We found that injection negligibly affects the cell proliferation potential, provides high cell viability, and can be used to control the exposure of nucleases in injected cells. A major drawback of microinjection is the low throughput. In contrast, nucleofection is amenable for therapeutic applications. Currently, we are optimizing nucleofection of CRISPR-Cas9 nucleases and donor templates into hepatocytes as a therapeutic strategy for familial hypercholesterolemia. We show >90% on-target CRISPR-Cas9 activity in primary hepatocytes, spurring further development into a novel cell-based gene therapy for metabolic liver diseases.
 

Speaker
Biography:

Alessandra Giuliani is Permanent Researcher and Aggregate Professor in Physics Applied to Cultural Heritage, Environment, Biology and Medicine at the Polytechnic University of Marche, Clinical Science Department. Within the Physics Group, she coordinates the research in Physics Applied to Biomaterials, Tissue Engineering and Regenerative Medicine. The purpose of her research is to study, using advanced physical techniques (such as microdiffraction, computed microtomography, holotomography), based on synchrotron radiation, all the structural changes of various biological tissues (mice bone under conditions of micro and macro gravity and / or of transgenic type, dental implants of various origins, tendons treated with collagen membranes, infarcted rat hearts treated with cardiac progenitor cells, mice dystrophic muscle injected with human AC133+ cells). A particular attention is paid to the vascularization issue of the regenerated tissue using an innovative imaging technique - the computed holotomography. She is author of around 54 peer-reviewed journal papers, chapters on 7 books internationally distributed and numerous works and abstracts related to National and International Congress presentations. Her researches in Physics applied to Tissue Engineering and Regenerative Medicine have been the subject of >45 presentations to Congresses and Schools, the most of them as invited speaker.

Abstract:

The evaluation of engineered tissues is usually performed by light microscopy on one or more histological sections. Th is conventional analysis provides only bi-dimensional (2D) information with the consequent risk that the selected sections do not properly represent the entire biopsies. In recent years there has been an increasing interest in a novel approach to evaluate diff erent engineered tissues by means of synchrotron micro-tomography (SCT). Using SCT, tissue regeneration subsequent to graft ing hosting sites with diff erent types of biomaterials (with or without stem cells seeding) was recently explored. SCT was shown to be fundamental to explore the dynamic and spatial distribution of regenerative phenomena, also in complex anatomic structures. Traditionally, absorption imaging with SCT is conducted with almost no distance between sample and detector. Homogeneous materials with a low attenuation coeffi  cient (like collagen, unmineralized extracellular matrix, vessels, nerves, etc.) or heterogeneous materials with a narrow range of attenuation coeffi  cients (like the case of heterologous bone scaff olds or graded mineralized bone) produce insuffi  cient contrast for absorption imaging. For such materials, the imaging quality can be enhanced through the use of phase contrast tomography (PCT), oft en achieved with an increased distance between sample and detector (propagation-based imaging). In the present lecture, the most recent breakthroughs in regenerative medicine will be shown, demonstrating the unique capabilities of the SCT in off ering not only an advanced characterization of diff erent biomaterials (to understand the mechanism of their biological behavior as tissue substitute) but also to investigate the growth kinetics of regenerated tissues in diff erent environments.
 

 

Bonginkosi Duma

National Health Laboratory Service, National Institute for Occupational Health, South Africa

Title: Quality indicators as a measure of good practice at the National Biobank in South Africa

Time : 16:40-17:00

Speaker
Biography:

Bonginkosi Duma has expertise in Quality and Biobanking and is passionate about innovation and getting solutions to cure diseases. Concerned about health and wishing to improve the health and wellbeing of South Africans and make a lasting impact on people around the world. As a Biobank Manager, he manages one of the largest biobanks in South Africa and is also a Quality Assurance Manager and auditor for South African National Accreditation Systems (SANAS) body for South Africa. He is a member of ISBER and sits on the committee for standards and also the ESBB working group on training. He also serves as a vice chairperson of the WHO BCNET committee based in IARC in France.
 

 

Abstract:

The National Health Laboratory Service (NHLS) Biobank is within the National Institute for Occupational Health (NIOH) which has been accredited for ISO: 15189; 17020; 17025 by the South African National Accreditation System (SANAS). Furthermore, there is adherence to ISBER’s Best Practices for Biorepositories as well as compliance with the National Health Act 61 of 2003 which provides a framework for a structured uniform health system within the Republic of South Africa. Th e model of the NHLS Biobank is designed to manage and secure biomaterial collections and storage as well as the associated data for research purposes both in the short-term as well as long-term. Th e Biobank encompasses multiple components of biospecimen for cancer, cell culture, genetics, molecular biology and for non-communicable diseases. Th e storage capacity is large scale for internal NHLS and external clients, can store over a million specimens with additional infrastructure recently created with an additional capacity of four million samples. All stages of the quality cycle, as part of the Quality System Essentials (QSEs), are continually being assessed, namely; equipment, facilities, and safety, organization and personnel, purchasing and inventory, documents and records, information management, corrective action, occurrence management. Th ese QSEs grouped into 3 categories; resource management, process management as well as improved management form quality indicators and ensure maintenance of QMS. Th e biomaterial data tracking system is through specialized soft ware and Biobank Information Management System (BIMS). To date, a substantial amount of resources and time has been invested in continued personal development in ensuring that quality standards are maintained. Future plans of the NHLS Biobank include implementation of the ISO 9001:2015 standard, ISO/TC 276, enhanced networks and stakeholder involvement as well as continued technical improvements on pre-analytical specimen factors which ultimately impact on specimen quality. Th ese future plans tie- in with maintaining quality within the NHLS Biobank.

 

Biography:

Dalia A Elgamal has completed her PhD at the age of 33 years from Faculty of medicine, Assiut University, Egypt and postdoctoral studies from the same university. She has published 15 papers in reputed journals and has been serving as an editorial board member of repute. She is paying a major concern to basic researches that may lead to possible/defi nite improvement in health services and decreasing disabilities, morbidity and mortality. eg. Studying male and female infertility at cellular and molecular basis using experimental trials. In addition to stem cell isolation, differentiation as a new trend in regenerative medicine.
 

 

Abstract:

Type-1 diabetes mellitus is a chronic autoimmune disorder in which genetic and epigenetic factors contributed equally to its pathogenesis. Histone deacetylase (HDAC) inhibitors such as sodium butyrate (NaB) had been reported to protect beta-cell damages and improve the glucose homeostasis by the modulation of p38/ ERK MAPK pathway. Th e aim of this work is to evaluate the role of NaB on ultrastructure of pancreatic beta- cells and PI3/Akt pathway. 30 juvenile male albino rats (5-6 weeks) were divided into 6 groups: Group I: Untreated control. GroupII: NaB control, received 500mg/kg/day NaB i.p. for 3 weeks. Group III: 3 days diabetic control received STZ (60mg/kg) i.p. Group IV: 3 weeks diabetic control received STZ (60mg/ kg) i.p .Group V: pre-treatment with NaB for 3 weeks prior to diabetes induction. Group VI: post-treatment with NaB for 3 weeks aft er diabetes induction. Plasma glucose, insulin levels, glucose tolerance were evaluated. Light, electron microscopy and immunohistochemistry was performed using ki67, caspase3, insulin and acetylated histone H3. NaB treatment resulted in a signifi cant improvement in plasma glucose level, plasma insulin level / expression and ameliorated the diabetes-induced histological alternations. Decrease in number of apoptotic cells had been demonstrated. Additionally, it inhibited the HDAC activity and increased the acetylation of histone H3 and expression of phosphorylated Akt.
Conclusion: Th ese fi ndings provide evidence that NaB might be useful for the treatment of juvenile diabetes.
 

 

Biography:

Devyani Joshi obtained her bachelor’s degree from the University of Mumbai, India. She is a graduate student in Mercer University College of Pharmacy, Atlanta, GA. As a graduate student, her area of research is particulate vaccines against infectious diseases and novel treatments for neurodegenerative diseases.
 

Abstract:

Parkinson’s disease affects millions of people the world over and the incidence and severity of this disease continue to increase. This disease is characterized by decreased levels of catecholamines including dopamine in the brain. PC-12 cell, a pheochromocytoma cell line from Rattus norvegicus, is a cell line that is capable of producing catecholamines. These cells can synthesize, store and be stimulated to release dopamine. Additionally, a PC-12 cell has the capacity to undergo neuronal differentiation in response to a nerve growth factor and can be a useful and important feature of PC-12 cells for Parkinson’s Disease (PD) studies. The neuronally differentiated cells directly model sympathetic neurons which are one of the neuron types affected by Parkinson’s disease. PC-12 cells were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% Horse Serum, 5% Fetal Bovine Serum and 1% Penstrip at 37°Ð¡ and 5% CO2. For fabrication of the microcapsules, 1% w/v solution of sodium alginate was prepared, and PC-12 cells were added to it and allowed to stir for fifteen minutes. After stirring, the alginate–cells suspension was sprayed through a 1.40mm nozzle using a Buchi spray dryer B-190 into the calcium chloride solution (1.5% w/v). The microcapsules in calcium chloride solution were allowed to stir for fifteen minutes and washed with phosphate buffered saline (PBS) and centrifuged twice at ×285g (1200 RPM) to remove excess of calcium ions. The suspension of microcapsules was then transferred to the chitosan glutamate solution (0.5% w/v), stirred further for fifteen minutes and washed with PBS, centrifuged twice at ×285g (1200 RPM) to remove the excess chitosan glutamate. The suspension of the microcapsules was then transferred to the media and kept in the incubator at 37°Ð¡ and 5% CO2. The parameters affecting the size of the microcapsule were air flow rate, pump speed controlling the flow rate of the alginate cell suspension, the distance between the nozzle and calcium chloride solution, nozzle diameter, and spray rate. In this study, we emphasized the air flow rate through the nozzle to alter the size of the microcapsules and kept all the other parameters constant. We found that maintaining the higher flow rate leads to the reduction in the size of the microcapsules due to high shear at the tip of the nozzle. The size of the microcapsules was in the range of 250–350μm. The alginate microcapsules were spherical in shape and no deformities were observed. The short-term stability studies showed that the cells were viable in the capsule for up to 7 days. The microcapsules containing these cells can be delivered into the brain through the intra-cranial injection for the treatment of the Parkinson’s Disease (PD). These encouraging primary results will lead the way for further research in the area and testing in murine models of Parkinson’s disease.