Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 6th World Congress on Cell & Stem Cell Research Philadelphia, Pennsylvania, USA.

Day 2 :

Keynote Forum

Diana Anderson

University of Bradford, United Kingdom

Keynote: General Overview of the Comet Assay

Time : 09:35-10:00

Conference Series Stem Cell Research-2016 International Conference Keynote Speaker Diana Anderson photo
Biography:

Diana Anderson has completed her PhD at the University of Manchester, UK in the Faculty of Medicine. She is the Established Chair in Biomedical Sciences at the University of Bradford. She has published more than 450 papers, 8 books and successfully supervised 30 PhD students, she has Hirsch factor of 51. She is the Editor-in-Chief of a Book Series for the Royal Society of Chemistry and Consultant to many International Organizations such as the World Health Organisation/International Programme of Chemical Safety. She is/has been Member of the Editorial Board of ten international journals.

Abstract:

The comet assay is an important toxicological method for measuring DNA strand breaks in single cells. Cells embedded in agarose on a microscope slide are lysed with detergent and high salt. Electrophoresis at high pH results in structures resembling comets, observed by fluorescence microscopy. The comet tail is formed by DNA fragments moving towards the anode. The assay can be used for genotoxicity testing of novel compounds and exotoxins, human bio-monitoring, molecular epidemiology, basic research into DNA damage and repair and effects of nanoparticles. DNA repair can be measured after treatment of cells with a DNA damaging (challenging) agent and measuring damage remaining after different time intervals or with repair enzymes. This overview will be illustrated with examples from the work of the author’s group and collaborators. These will include studies for genotoxicity testing of various agents for exotoxins the work with DBP halogenated acetic acids, for human biomonitoring and molecular epidemiology the work with mother and babies, diabetes and lead-exposed children and for fundamental research that with drug resistant non-Hodgkin’s lymphoma patients over-expressing p53 mutant protein and lacking DNA repair. Most of this work has been carried out in somatic human lymphocytes but it can also be carried out in germ cells as human sperm. Using sperm, the oestrogens can cause DNA damage which can be diminished with anti-oxidants suggesting an oxygen radical involvement. Also as age increases in men so there is an increase in DNA damage. The comet assay can be modified and used as an innovative blood test to predict cancer. From a regulatory viewpoint, the assay is regarded as an indicator test but is incorporated into guidelines in some countries.

Keynote Forum

Georg F. Weber

University of Cincinnati Academic Health Center, USA

Keynote: Mechanisms of cancer progression: the roles of osteopontin variants

Time : 09:30-10:00

Conference Series Stem Cell Research-2016 International Conference Keynote Speaker Georg F. Weber photo
Biography:

Georg F. Weber attended medical school in Würzburg, Germany. He worked at the Dana-Farber Cancer Institute, Harvard Medical School from 1990 through 1999 and is currently on the faculty at the University of Cincinnati. Georg F. Weber has published about 90 scientific reports, including many in the most respected professional journals, and various monographs, including textbooks on molecular oncology and anti-cancer drugs. He holds several U.S. and international patents. As a component of his mission to combat cancer dissemination, Georg F. Weber is the founder and chief executive officer of MetaMol Theranostics, a company specialized in diagnosis and treatment of cancer metastasis.

Abstract:

Metastasis formation is an essential aspect of cancer. While the organ preference for dissemination is largely governed by tumor–host interactions on the epigenetic level, there is a genetic basis underlying the ability of cancer cells to disseminate. Metastasis genes are comprised of developmentally non-essential stress response genes, which encode homing receptors, their ligands, and extracellular matrix-degrading proteinases. They jointly cause invasion and anchorage-independence. Metastatic potential is conferred to cancer cells by aberrant expression or splicing of these genes, which include variant CD44 and osteopontin. The CD44-dependent spread of tumor cells is mediated by its cytokine ligand osteopontin, which induces directed migration. A C-terminal osteopontin domain ligates the variant exon 6 of CD44 through a protein-protein interaction and is likely to bind to variant exon 3 through a heparin bridge.rnOsteopontin is expressed at high levels by various cancers and contributes importantly to their invasive potential. In contrast, osteopontin derived from host cells induces cellular immunity and could bolster anti-tumor protection by cytotoxic T-lymphocytes. Underlying the functional differences between tumor-derived and host-derived osteopontin are structural characteristics. The osteopontin gene product is subject to alternative splicing, which yields three RNA messages, osteopontin-a (full length), osteopontin-b (lacking exon 5), and osteopontin-c (lacking exon 4). The shorter forms -b and -c are differentially expressed in cancers, but are absent from healthy tissues.rnThe major limiting factor in the process of metastasis formation is the death of the tumor cells before their implantation in target organs. Hence, anchorage-independent survival is essential for metastasis. While untransformed non-hematopoietic cells undergo anoikis consecutive to losing contact with their substratum, cancer cells can survive in the circulation for extended periods of time. The detachment of mammary epithelial cells prompts a loss of glucose transport and ATP deficiency, thus compromising the energy metabolism. Invasive breast tumor cells abundantly express two splice variants of the metastasis gene osteopontin. Osteopontin-a and osteopontin-c synergize in supporting tumor progression via up-regulating the energy production, which leads to deadherent survival. Osteopontin splice variants hold promise as potential drug targets.rn

Break: Networking & Refreshments 11:05-11:25 @ Foyer
  • Stem Cell Therapy, Stem Cell Banking, Bioinformatics and Computational Biology

Session Introduction

Alan Wells

University of Pittsburgh, USA

Title: Enhancing survival of transplanted stem cells in the wound bed
Speaker
Biography:

Alan Wells, M.D., D.M.Sc., is Executive Vice-Chair and Thomas J Gill III Professor of the Departments of Pathology and BioEngineering. Wells holds an A.B. in Biochemistry from Brown University (1979), a D.M.Sc. in Tumor Biology from the Karolinska Institute (1982), and an M.D. from Brown University (1988). Following his studies, Dr. Wells completed a postdoctoral Fellowship in Tumor Biology at the University of California in San Francisco, California, and a Residency in Laboratory Medicine at the University of California in San Diego, California. He has over 230 scholarly publications that have been cited over 15,000 times. These advances have been recognized by numerous grants, patents, and invited keynote talks and professorships, as well as election to the American Society for Clinical Investigation and the American Association of University Pathologists (Pluto Club). He has consulted extensively to industry and has cofounded two companies. None of this would have been possible without the input, aid, and assistance of some 20 graduate students (PhD and MD-PhD) and a similar number of post-doctoral fellows and a half dozen junior faculty colleagues.

Abstract:

Transplantation of stem cells to augment the intrinsic regenerative capacity is a potential therapy for non-healing wounds. However, such approaches have been less than successful, as the majority of introduced cells are lost within days due to cell death from the harsh wound environment that lacks nutrients and presents apoptosis-inducing cytokines. A cellular engineering approach to enhanced survival could overcome this obstacle. Activation of the EGF receptor at the cell surface, but not from internalized receptors in the endosomes, promotes cell survival in the face of cellular starvation, death cytokine signaling, and toxic agents. The signaling cascade involves tonic low-level activation of Erk MAPK and PI3K/Akt pathways. To achieve this mode of signaling the triggering ligand must be retained in or attached to the substratum or matrix. This can be accomplished by either tethering classical EGF receptor ligands to the substratum, or activating the EGF receptor with cryptic ultralow affinity matrikines present in tenascin-C and laminin V. We have creating matrices with (or without) tethered EGF or tenascin-C. These were introduced into acute full thickness wounds in mice, internal soft tissue in mice (subfascial), and critical cortical bone defects in dogs. In all three instances wound healing and vascularization was significantly increased in the presence of EGF receptor ligand. Xenotransplanted stem cells were retained up to one month in immunocompetent mice, compared to less than a week without the tEGF or tenascin-C. The presence of these cells with the tenascin-C limited scarring in a hypertrophic scar model, even 6 months after wounding, when all the transplanted stem cells have been rejected. The extended survival of these transplanted stem cells allows them to educate the local wound environment, and turn the healing towards regeneration and away from non-functional scarring or failure to heal. This represents a new approach to cellular support for dysrepair.

Patricia E. Berg

George Washington University Medical Center, USA

Title: Activation of BP1 is associated with Aggressive Breast Cancer
Speaker
Biography:

Patricia Berg received her bachelor’s degree in mathematics from the University of Chicago, her Ph.D. in microbiology from the Illinois Institute of Technology, then pursued Post Doctoral studies at the University of Chicago. Research at the National Institutes of Health followed. Currently she is a Professor of Biochemistry and Molecular Medicine at George Washington University in Washington, DC, where she is director of a cancer research laboratory. Her work, which centers on BP1, has been published in major journals and has been featured on network television and in major media, including the New York Times and Washington Post.

Abstract:

BP1, a transcription factor (TF) we identified in cancer, is encoded by a homeobox gene. BP1 is overexpressed in breast cancer, prostate cancer, ovarian cancer, acute myeloid leukemia, non-small cell lung cancer, and possibly other malignancies as well. Important characteristics of BP1 in breast cancer, a focus in our laboratory, include findings that: (1) BP1 is expressed in 80% of invasive ductal breast tumors, including 89% of the tumors of African American women compared with 57% of the tumors of Caucasian women. (2) BP1 expression correlates with the progression of breast tumors, from 0% in normal breast tissue to 21% in hyperplasia and 46% in ductal carcinoma in situ. (3) Expression of BP1 is associated with larger tumor size in both women and mice. (4) BP1 appears to be associated with metastasis. Forty-six cases of inflammatory breast cancer were examined and all were positive for BP1 expression, as well as matched lymph nodes in the nine metastatic cases. (5) BP1 overexpression induces oncogene expression. BP1 activates the BCL-2 gene; high BCL-2 protein levels are associated with resistance to drug and radiation therapy. BP1 also activates VEGF and c-MYC, as well as other genes important in angiogenesis, invasion and metastasis. Interestingly, BP1 down-regulates BRCA1. (6) BP1 up-regulates ER alpha, inducing estrogen independence and tamoxifen resistance. In summary, BP1 appears to confer properties on breast cancer cells that lead to a more invasive and aggressive phenotype. Since the functions of homeotic TF are highly conserved, it is likely that BP1 regulates many of the same processes and genes in other malignancies in which it is active.

Speaker
Biography:

Purwati Armand has finished in general practitioner from Airlangga University in 1997, has completed in internal med. Specialist in 2008 from Airlangga University also and taken Doctoral program in Airlangga University 2010-2012. Interest in stem cell field from 2008, be secretary of stem cell laboratory of Airlangga University and also secretary of Surabaya Regenerative Medicine Centre. Have almost 50 publication in journals, papers, and seminar.

Abstract:

Brain in control centre of the body, this organ has a wide range of responsibilities from coordinating our movement to manage our emotion, the brain does it all. For almost hundred years it has been a mantra of biology that brain cell do not regenerate so need to add new neuron when the brain injury but stem cell niche will induce endogenous stem cell in SVZ to regenerate it. Stem cell niche with content of GABA, FGFs, EGF, VEGF, PEDF. Parkinson’s involves the malfunction and death of vital nerve cells in the brain, called neurons. Parkinson's primarily affects neurons in an area of the brain called the substantia nigra. Some of these dying neurons produce dopamine, a chemical that sends messages to the part of the brain that controls movement and coordination. As PD progresses, the amount of dopamine produced in the brain decreases, leaving a person unable to control movement normally. Objective of this research to drive neuron progenitor stem cell from adipose to treat Parkinson. Adipose was isolation from patient and cultur become neuron progenitor stem cell, after second passage the neuron progenitor stem cell was harvesting . Neuron progenitor stem cell was characterized by Noch1 flowcitrometry and LDopa Icc. Neuron progenitor stem cell was produce dopamine ready implant for Parkinson patient by direct implantation. Result: Ten patients with Parkinson diseases with inclusion and exclusion criteria, received neuron progenitor cell implantation, 2 patients no significant improvement and 8 patients have significant improvement, outcome evaluating using mRS (Modified Ranking Scale) and BI (Barthel index ). Conclusion: Neuron progenitor stem cell has significant improvement for Parkinson diseases

Speaker
Biography:

Mennat-allah Elmenyawi is a Assistant lecturer of medical physiology in faculty of medicine-Suez Canal University. She completed her Master degree in 2015, she also attended Workshops such as: bioinformatics and leadership and management in faculty of medicine-Suez Canal University, headache management workshop held in turkey in 2015 (African-turkish association of headache) and Conferences: fourth annual African association of physiology in 2012.

Abstract:

Introduction: Cell transplantation using The Bone Marrow Mesenchymal Stem Cells (BMSCs) and The Schwann Cells (SCs) to alleviate neurological deficits has become the focus of research in regenerative medicine. In attempt to identify the possible mechanisms underlying the regenerative potential of cell transplantation (BMSCs and SCs), this study investigate the most effective therapy of the sole cell transplantation (BMSCs and SCs) by induction of injury in rat’s sciatic nerve, when compared to their co-transplantation. Materials and methods: In this comparative experimental study, adult male albino rats (n=60, 250-300gm) divided into 5 groups: group (I): the control intact sciatic nerve, group (II): the left injured sciatic nerve injected intralesionally with physiological saline, group (III): the left sciatic nerve injected intralesionally with BMSCs, group(IV): the left sciatic nerve injected intralesionally with SCs, group(V): the left sciatic nerve injected intralesionally with BMSCs and SCs. BMSCs and SCs were labeled with Bromodeoxyuridine (Brdu). After 12 weeks, nerve conduction velocity, electromyographic, functional assessments, oxidative and antioxidative effects of cell tranplantation and measurement of BDNF were performed and analyzed by one-way analysis of variance (ANOVA). Results: This treatment led to: (i) improved walking tract as measured by sciatic nerve index in all the treated groups, (ii) increase in nerve conduction velocity and EMG magnitude by using biopack MP150 signifcantly (p<0.01) in SCs and Co-treated groups, (iii) increase in the antioxidant effect and reduction in the oxidative effect of cell transplantation in nerve tissue significantly (p<0.01) in BMSCs and Co-treated groups, (iiii) increase expression of brain derived neurotrophic factor (BDNF) in nerve tissue using real time PCR significantly (p<0.01) in SCs and Co-treated groups. Discussion: The results showed the superiority of the co-transplantation group followed by SCs group in the most of the assessments to BMSCs group which exceptionally succeeded in the increase of the antioxidant and the decrease in the oxidant levels.

Break: Lunch Break:13:05-13:50 @ Benzamin’s Restaurant
Speaker
Biography:

Zahara Mansoor has completed her masters in Regenerative Medcine at the Faculty of Medicine, University of Colombo who is currently drafting her thesis and will be graduating next year in 2016.She is one of the pioneering Stem Cell Scientist in Sri Lanka where stem cell is still at its infancy stage.Prior to this, I have successfully completed a Bachelor of Science, in Biotechnology at University of Bangalore. She also won the 2nd place for the Best Poster Presentation at The Annual Research Symposium, University of Colombo, 2015.Her interest motivated to explore research opportunities in the field of Regenerative Medicine.

Abstract:

Mesenchymal Stem cells (MSCs) are plastic-adherent, fibroblast – like cells with specific surface phenotype, having ability to differentiate into osteoblasts, chondroblasts and adipocytes in-vitro. Umbilical cord (UC) is a readily available without ethical constraints, showing high proliferation rate and osteogenic potential. To derive MSCs from the human UC Wharton’s Jelly (WJ) and osteogenic differentiation was my main objective.Following obtaining ethical approval, five UCs from healthy mothers undergoing elective Caesarian sections were collected, cleaned with phosphate buffered saline, removed blood vessels,digested WJ in 0.5% collagenase 2-3 hours / 0.2% collagenase overnight and cultured in DMEM supplemented with 10% FBS, 1% L-glutamine and 1% penstrep at 37Ëš C in 5% CO2. Cells are passaged at 70%confluency. At fourth passage (P4), osteogenic differentiation medium was added following incubation .Culture maintained for 21 days and cells were stained with 2% Alizarin red and von kossastains.MSCs were determined and characterized using Trypan blue test, Flow cytometry, RT-PCR and karyotypic analysis.Cells were were positive for CD90, CD73 and CD105 and negative for CD34 and CD45 markers expressing Oct-4 and G6PD genes. Karyotypes depicted were normal. Alizarin red stain gave bright orange red and von kossa stain gave black-brown deposits demonstrating the presence of extracellular calcium deposits.UC-MSCS serves as a suitable source for osteogenic regeneration Gene expression demonstrated the embryonic origin of the MSCs which maintained genomic stability upto P4 stage.So my initatiative stem cell research in Sri Lanka improves the therapeutic potential in bone defects and opens up new perspectives for bone tissue engineering.

Speaker
Biography:

Zhiheng Xu was awarded an MD in 1989 from the Second Military Medical University, Shanghai, and a PhD in 1999 from Rutgers University, New Jersey.In 1999 he was a postdoctoral and research associate at Columbia University, New York.He received the Ruth L Kirschstein National Research Service Award in 2003 and the Distinguished Young Investigator Award, National Science Foundation (China), in 2007.

Abstract:

Human autosomal recessive primary microcephaly (MCPH) is a neural developmental disorder hallmarked by significantly reduced brain size and variable intellectual disability. Mutation of WD40-repeat protein 62 (WDR62) is the second major cause of MCPH. We have reported recently that WDR62 regulates the maintenance of neural progenitor cells (NPCs) during cortical development through JNK1 (Xu et al., Cell Reports 2014). However, the detailed biological function of WDR62 and the underlying mechanism by which WDR62 regulates JNK signaling are still not very clear. Here, we demonstrate that MEKK3 forms a complex with WDR62 to promote JNK signaling synergistically and regulate neurogenesis as well as brain size. MEKK3, WDR62 and JNK1 depletion or knockout phenocopy each other in defects including premature NPC differentiation and reduced brain size. These defects can be rescued by the expression of transgenic JNK1, indicating that the complex controls neurogenesis through JNK signaling. We show further that WDR62 protein level is positively regulated by MEKK3 through JNK1-induced WDR62 phosphorylation. Meanwhile, WDR62 is also negatively regulated by specific phosphorylation of WDR62 at T1053, leading to the recruitment of the E3 ligase FBW7 and proteasomal degradation of WDR62. Our findings demonstrate that WDR62 controls the maintenance of NPCs via MEKK3 and JNK1 during cortical development and reveal the molecular mechanisms underlying MCPH pathogenesis.

Simon Berkovich

The George Washington University, USA

Title: Connotation of Life beyond molecular biology
Speaker
Biography:

Professor Simon Berkovich received MS in Applied Physics from Moscow Physical-Technical Institute (1960) and PhD in Computer Science from the Institute of Precision Mechanics and Computer Technology of the USSR Academy of Sciences (1964). He has several hundred publications in various areas of physics, electronics, computer science, and biology. In 2002, Professor Simon Berkovich was elected a member of the European Academy of Sciences "for an outstanding contribution to computer science and the development of fundamental computational algorithms". In 2014, he won the GWU Technology Transfer Innovation Competition. In 2015, he was awarded a status of Emeritus Professor

Abstract:

Modern science resolutely rejects the longstanding belief that understanding of Life needs some ‘vital’ force besides fundamental physics concepts. However, an essential part of biological processes still cannot find a concrete physical explanation. So, physics that does not provide an explanation for biology is not just incomplete, it merely employs an incorrect paradigm. Therefore, right understanding of biology may help to straighten physics, rather than vice versa. The essence of biology lies in information processing. Likewise, it is also supposed “The physical world is made of information with energy and matter as incidentals” (John A. Wheeler). Scientific views often go in parallel with contemporary technologies; correspondingly, at different times living organisms were compared to mechanical apparatuses, hydraulic systems, clock devices, chemical factories, electrical machines, computers etc. Nowadays, the Information Revolution fosters Cloud Computing. This inspires to consider the Universe as an Internet of Things. Such kind of an arrangement is realized in the construction of Holographic Universe arising from our Cellular Automaton model of physics. Workings of interactive holography attain a clear explanation for the strangeness of quantum mechanics, particularly, for the most inconceivable property of nonlocality. Further, in contrast to quantum particles, macromolecules can acquire a content-addressable access to holographic memory, leading to the capabilities of sophisticated behavior. Thus, the Holographic Universe presents an operational framework for biological processes. Instruction sequences for biological objects include signals for information control and impacts for material actuations. Revealing a physical facility that could intervene in this process may provide a new approach to medical treatment.

Speaker
Biography:

Carol M. Jim is currently a Ph.D. candidate working on her dissertation at The George Washington University in the School of Engineering and Applied Science. She is also an adjunct professor of computer science and information technology at Hood College where she received her M.S. in computer science and B.A. in mathematics. She has published in peer-reviewed international conference proceedings and journals on topics including network forensics, data mining and machine learning, game theory, and computational biology.

Abstract:

Although extensively studied, little is still known about the origins of pairs of monozygotic twins and higher order multiples such as triplets and quadruplets. By understanding the mechanism behind the twinning process, further developments in developmental biology can be achieved as well as insights into disease mechanisms and the human aging process. We consider and analyze certain possible cell labeling schemes that model an organism’s development and expose the phenomenon of quadruplet twins during the process. We predict that monozygotic quadruplets are not quadruplets in the traditional sense but rather, are two pairs of monozygotic twins where the pairs slightly differ. The probability of monozygotic twins is discovered to be (1/2)K, and the probability of monozygotic quadruplets, or triplets in the case of the death of an embryo, is found to be (1/8)K, where K is a species-specific integer representing the number of pairs of homologous chromosomes. This investigation into twinning establishes the cell development mechanism. The failure of the internal cellular clock from this mechanism may play an important role in cancerogenesis. The parameter K may determine cancerization with a probability threshold that is approximately inversely proportional to the Hayflick limit, so exposure to small levels of ionizing radiation and chemical pollution may not produce cancer. From the considered model, the mechanism involved in the two opposite circumstances of twinning and cancerogenesis provides a foundation for the understanding of the origins of these two disparate processes.

Speaker
Biography:

Miguel Garber has over 30 years experience in Internal medicine and cardiology, with expertise in regenerative medicine and research. He has more than 10 years experience working with stem cells, including exploring and developing stem cell therapies for cardiomyopathies, osteoarthritis and regenerative medicine at Stem Cell Therapeutics Department of American Medical Information Group and Clinica Quirurgica Quantum. He is currently serving as Medical Director of Revitacell and Clinical Director of Regenerative Medicine department at Humanus. Dr. Garber has made a significant contribution to Stem cell Research and is actually he is involved in Adipose Stem Cell application.

Abstract:

A comprehensive review of the stem cell research literature indicates that bone marrow stem cells (BMSC) constitute the natural repair system of the body and that the number of circulating stem cells appears to be a critical parameter in the effectiveness of stem cell-based tissue repair. On this basis, Endogenous Stem Cell Mobilization (ESCM) emerges as a possible approach to the treatment of a variety of degenerative conditions, including diabetes. BMSC have been shown to have the ability to migrate on their own into the pancreas and to differentiate into functional insulin-producing cells, and mobilization of BMSC has been shown to rescue streptozotocin-induced diabetes in mice. The stem cell mobilizer StemEnhanceTM increased the number of circulating stem cells as well as the number of stem cells that migrated in the pancreas of streptozotocin-treated mice, which significantly increased insulin production and reduced fasting blood glucose. This was confirmed in humans where StemEnhanceTM supplementation for 12 weeks decreased fasting blood glucose and HbA1c levels in type II diabetes patients. A multi-center study is underway to document the effect of plant-based stem cell mobilizers on pre-diabetes. Current evidence suggests that ESCM could be an effective approach to prevent or slow down the development of diabetes, or in certain cases even reverse diabetes.

Break: Networking, Refreshments & Poster Presentations 15:55-16:25 @ Foyer
Speaker
Biography:

I am practicing as an assistant professor in Oral & Maxillofacial surgery department of Isfahan university of medical sciences since 2010 while seeking further training and experience in Craniofacial cleft surgery.My project for getting oral and maxillofacial specialist degree entitled “ A comparison of tissue-engineered bone from adipose-derived stem cell with autogenous bone graft…” has been done in May 2010. This topic with its special harvest of stem cell from adipose tissue for maxillofacial reconstruction has been done for the first time in Iran and the world .It has been published in International journal of oral and maxillofacial surgery 2012.

Abstract:

This study was designed to compare bone regeneration of tissue-engineered bone from adipose-derived stem cell and autogenous bone graft in a canine maxillary alveolar cleft model. In this prospective clinical trial, mesenchymal stem cells (MSCs) were isolated from subcutaneous canine adipose tissue. Undifferentiated cells were incubated with a 3 mm × 3 mm × 3 mm hydroxyapatite/beta-tricalcium phosphate scaffold, in specific osteogenic medium for 21 days. Four mongrel dogs were prepared by removal of two of the three incisors bilaterally and a 15 mm defect in bone was created from crest to nasal floor. After healing, repair was followed by a tissue engineered bone graft from adipose-derived stem cells on one side and corticocancellous tibial auto graft on the other side. Bone regeneration was evaluated by histomorphometry on days 15 and 60 after implantation. The data were analysed with descriptive and t test methods (α = 0.05). Bone formation on the autograft sides was higher than on the stem cell sides at 15 and 60 days, 45% and 96% versus 5% and 70%, respectively. Differences between the two groups at 15 and 60 days were significant (p = 0.004 and 0.001, respectively). Although autograft is still the gold standard for bone regeneration, tissue engineered bone may provide an acceptable alternative.

Speaker
Biography:

Atefeh Roein Tan has completed her Master in Biotechnology at the age of 29 years from Payame Noor University of Tehran. She work as a researcher and lab technician for more than four years in Shefa Neuroscience Research Center in Tehran, Iran. She worked on the project of creating and purifying neural cells and using in the treatment of Amyotrophic lateral Sclerosis as a technician and this project has been patented in September of 2014.

Abstract:

Cell therapy is one of the approaches for the treatment of locomotive deficits in spinal cord injuries and neurodegenerative disorders. Neural stem cells derived from bone marrow stromal cell are considered as a feasible option for cell therapy. Epigenetic experts have reported that cell differentiation during the development process of BMSCs to NSCs is controlled by several factors including growth and environmental factors as well as regulation and induced effects. Several protocols are using different chemicals for inducing neuronal differentiation of BMSCs. In this study, we investigated the feasibility of using of Selegiline as an efficient inducer for neuronal differentiation of rat BMSCs and its effect on gene expression of neurotrophins and their receptors. Based on our results, Selegiline has multiple effects which makes it a good candidate for inducing neuronal phenotype into BMSCs. Also it has ability to induce the expression of some genes like neurotrophic factors. Therefore, it can be considered as an alternative neuroprotective inducer for BMSCs where the induced cell can still be used for cell therapy. Moreover, the local expression of neurotrophin genes suggests a wide range of paracrine and/or autocrine mode of action through their corresponding receptors within the bone marrow. For our experiments, after achieving the optimal concentration of Selegiline the expression of antibodies Nestin , Neurofilament 68,Neurofilament 200, TH, Neu-N and GFAP was evaluated using immunocytochemistry. Furthermore, the expression profile of neurotrophins NGF, BDNF and NT-3 and their receptors (TrkA/B/C, p75NTR) was examined during neural differentiation by RT-PCR.

Speaker
Biography:

Kevin Murray, Sales Manager for BioSpherix, Ltd. BS BioChemistry, MBA Finance & Marketing, 20+ Years experience in the Pharmaceutical/Biotech/Medical Research Industry.

Abstract:

Total quality recognizes that for best cell potency, cells need full-time optimization of all critical cell process parameters (O2, CO2, RH, T). Total quality recognizes that all typical negative side effects of machines on cells (particles, heat, vibration, etc.) must be neutralized to make automation compatible with a cell optimized ecosystem, and those machines must be protected from dust, aerosols, and corrosion. Total quality recognizes that each entire cell production line (all manual and automated steps) must be protected from microbial contamination by full-time, absolutely aseptic conditions. Total quality recognizes that all personnel must be fully protected from cells harboring virus, vectors, prion, and other pathogens. Total quality recognizes that scaling up and out must be efficient. Total quality recognizes that cost efficiency is a fundamental quality attribute, critical for commercial. The Xvivo System is a comprehensive, modular, total quality "platform" for cells.

Mahsa Khayat-Khoei

University of Texas Health Science Center at Houston, USA

Title: Angiogenesis properties of the amniotic membrane stem cells after cryopreservation
Speaker
Biography:

Mahsa Khayat-Khoei has completed her medical education and received her MD degree from Shahid Beheshti University of Medical Sciences in 2012. She will soon receive her MBA degree, and is a research assistant at University of Texas Health Science Center. She has worked as a researcher and clinical trainee at Baylor Saint Luke's Medical Center, as well as a research trainee at MD Anderson Cancer Center in Houston, Texas. She has a patent, several publications in reputed journals and has presented her research in respectable national and international meetings. She has been serving as an editorial board member of several medical journals. Her main research interest is on Amniotic Membrane derived Stem Cells and their different properties.

Abstract:

Human placenta supports the growing fetus and consists of several layers the inner most of them is a membrane with unique capacities. This layer which is called the Amniotic membrane (AM) develops two different types of pluripotent stem cells which have previously shown to express angiogenesis regulatory properties that make them great candidates for cancer and cardiac researches. In order to store and transfer these cells for experimental and possible clinical purposes, cryopreservation is a necessary procedure. However it is debatable whether the cryopreservation negatively influences the Amniotic membrane Stem Cells (AMSCs) characteristics or not. In this study AMSCs were cryopreserved and stored for 6 months at -80 °C. The effect of cryopreservation on these stem cells’ properties was evaluated by comparing the angiogenesis activity of the thawed AMSCs and fresh AMSCs in an animal model. The length and number of branches of formed capillaries were measured via intra-vital microscopy after 5 and 15 days. The amount of angiogenesis promoting factors IL-8 (interleukin-8) and TIMP-2 (Tissue Inhibitor of Matrix Metalloproteinase-2) that are believed to be produced mainly by AMSCs were evaluated using ELISA assay. The effect of cryopreserved AMSCs on angiogenesis was reported to be of similar power to that of fresh cells. These promising results can act as a basis to confirm cryopreservation as a proper and reliable method of storing AMSCs in different clinical and research settings.

Break: Poster Presentations 15:55-16:25 @ Foyer
Speaker
Biography:

Khalid Shah heads the Molecular Neurotherapy and Imaging Laboratory in the departments of Radiology and Neurology at Massachusetts General Hospital. Dr. Shah is also the Director of the Stem Cell Therapeutics and Imaging program in the Center for Translational Research at MGH and also a Principal Faculty at Harvard Stem Cell Institute in Boston. His laboratory focuses on developing therapeutic stem cells for receptor targeted therapies for cancer and testing their efficacy in clinically relevant mouse tumor models. Dr. Shah’s research also explores the development of novel in vivo imaging markers and their potential use in assessing fate of stem cells and their therapeutic efficacy. In recent years, Dr. Shah and his team have pioneered major developments in the stem cell therapy field, successfully developing experimental models to understand basic cancer biology and therapeutic stem cells for cancer, particularly brain tumors. These studies have been published in a number of very high impact journals like Nature Neuroscience, PNAS, Nature Reviews Cancer, JNCI, Stem Cells and Lancet Oncology, validating the use of therapeutic stem cells alone and in combination with clinically approved drugs for cancer therapy. Dr. Shah holds current positions on numerous councils, advisory and editorial boards in the fields of stem cell therapy and oncology. In an effort of to translate the exciting therapies develped in his laboratory into clinics, he has recently founded biotech company, AMASA Technologies Inc. whose main objective is the clinical translation of therapeutic stem cells in cancer patients.

Abstract:

Stem cell-based therapies are emerging as a promising strategy to tackle cancer. Multiple stem cell types have been shown to exhibit inherent tropism towards tumors. Moreover, when engineered to express therapeutic agents, these pathotropic delivery vehicles can effectively target sites of malignancy. Using our recently established invasive, recurrent and resection models of primary brain tumors (GBM) and breast and melanoma metastatic tumors in the brain that mimic clinical settings, we show that that engineered human mesenchymal stem cells and neural stem cells expressing novel bi-functional proteins or loaded with oncolytic viruses target both the primary and the invasive tumor deposits and have profound anti-tumor effects. These studies demonstrate the strength of employing engineered stem cells and real time imaging of multiple events in preclinical-therapeutic tumor models and form the basis for developing novel cell based therapies for cancer. This presentation considers the current status of stem cell-based treatments for tumors in the brain and provides a rationale for translating the most promising preclinical studies into the clinic.

Jaroslaw Blaszczyk

Centre of Molecular and Macromolecular Studies of the Polish Academy of Sciences, Poland

Title: Unexpected opening of the glycosylation site in hexagonal form of CAL-B. Is it functionally related?
Speaker
Biography:

Jaroslaw Blaszczyk has completed his PhD at the age of 30 years from Technical University of Lodz and postdoctoral studies from NIH, National Cancer Institute at Frederick. He became a research assistant professor at Michigan State University. After that he served as the PDB annotator at Rutgers University of New Jersey. Currently he is an assistant professor at the Centre of Molecular and Macromolecular Studies of the Polish Academy of Sciences. He has published more than 67 papers in reputed journals and has been serving at several grant review panels at NCN, the Polish National Science Center.

Abstract:

We discovered the new, hexagonal crystal form of lipase B from Candida antarctica (CAL-B). The NAG (N-acetyl-D-glucosamine) molecules, which were closing the glycosylation site in the orthorhombic form, in our hexagonal structure, unexpectedly, adopt an open conformation. We do not know whether the opening and closing of the glycosylation site by the ‘lid’ NAG moiety, could be related to the opening and closing of the active center of the enzyme upon substrate binding and product release. The packing of molecules in the hexagonal crystal makes the active center of the enzyme very well accessible for the ligand, which, in our opinion, may help in the enzyme-ligand complex formation. Financial support by the Polish National Science Center, grant No. DEC-2012/05/B/ST4/00075, is gratefully acknowledged. The structure is available at the PDB, entry 4ZV7.

Alain Chapel

Institute of Radioprotection and Nuclear Safety, France

Title: Stem cell therapy for the treatment of severe tissue damage after radiation exposure
Speaker
Biography:

For 20 years, he has been developing gene and cell therapy to protect against the side effects of radiation. He collaborates with clinicians to develop new strategies for treatment of patients after radiotherapy overexposures. He has participated in the first establishment of proof of concept of the therapeutic efficacy of Mesenchymal stem cells (MSCs). Currently his work focuses on the development of radio-induced bone marrow aplasia using human hematopoietic stem cells derived from human IPS. He is a member of various learned national and international societies and associate editor of international journals. Hirsch Index 22.

Abstract:

Institute of Radioprotection and Nuclear Safety Radiotherapy may induce irreversible damage on healthy tissues surrounding the tumour. It has been reported that the majority of patients receiving pelvic radiation therapy shows early or late tissue reactions of graded severity as radiotherapy affects not only the targeted tumor cells but also the surrounding healthy tissues. The late adverse effects of pelvic radiotherapy concern 5 to 10% of them, which could be life threatening. However, a clear medical consensus concerning the clinical management of such healthy tissue sequelae does not exist. Although no pharmacologic interventions have yet been proven to efficiently mitigate radiotherapy severe side effects, few preclinical researches show the potential of combined and sequential pharmacological treatments to prevent the onset of tissue damage. Our group has demonstrated in preclinical animal models that systemic MSC injection is a promise approach for the medical management of gastrointestinal disorder after irradiation. We have shown that MSC migrate to damaged tissues and restore gut functions after irradiation. We carefully studies side effects of stem cell injection for further application in patients. The clinical status of four first patients suffering from severe pelvic side effects resulting from an over-dosage was improved following MSC injection in a compassional situation. Bone marrow-derived MSC from the patients´ children were injected to four patients. A quantity of 2 millions to 6 millions of MSC /kg were infused intravenously to the patients. Pain, hemorrhage, frequency of diarrheas and fistulisation as well as the lymphocyte subsets in peripheral blood were evaluated before MSC therapy and during the follow-up. Two patients revealed a substantiated clinical response for pain and hemorrhage after MSC therapy. In one patient pain reappeared after 6 months and again substantially responded on a second MSC infusion. A beginning fistulisation process could be stopped in one patient resulting in a stable remission for more than 3 years of follow-up. The frequency of painful diarrhea diminished from an average of 6/d to 3/d after the first and 2/d after the 2nd MSC injection in one patient. A decline of CD4+ and CD8+ T lymphocytes and an increase of potentially regulatory CD25+ T cells accompanied the clinical response in this patient after the MSC injections. In all patients, prostate cancer remained in stable complete remission. A modulation of the lymphocyte subsets towards a regulatory pattern and diminution of activated T cells accompanies the clinical response in refractory irradiation-induced colitis. No toxicity occurred. MSC therapy was safe and effective on pain, diarrhea, haemorrhage, inflammation, fibrosis and limited fistulisation. For patients with refractory chronic inflammatory and fistulising bowel diseases, systemic MSC injections represent a safe option for salvage therapy. A clinical phase II trial will start in 2016.

Speaker
Biography:

Mohammadhadi fartookzade has Msc degree in MBA and B.s. degree in Micribiologgy. He is the technician in Electron microscope unit. He is working in Iran university of Medical Sciences.

Abstract:

Neural stem cells (NSCs) find in the sub-ventricular zone (SVZ) and the hippocampus of the adult brain. NSCs can give rise to neurons, astrocytes and oligodendrocytes. Locus Ceoruleus (LC) plays an important role in the sleep-wake cycle. The aim of this investigation was study of effect of Neural Stem Cells (NSCs) transplantation on the sleep-wake cycle after bilateral lesion of the locus coeruleus. Fourty-two adult male Wistar rats, were categorized in seven groups [Control, Sham (cannula implantation), lesion, experimental 1 (intravenous transplantation of NSCs), experimental 2 (intravenous transplantation of noradrenergic-like cells (NACs)), experimental 3 (intraventricular transplantation of NSCs), experimental 4 (intraventricular transplantation of NACs)]. Neural stem cells were harvested from SVZ of newborn rat brains. NSCs were differentiated in neurobasal medium, B-27 supplemented with BDNF and GDNF for 5 days. The animals received bilateral 6_hydroxydopamine (6_OHDA) lesion of the LC. For sleep-wake recording 3 EEG and 2 EMG electrodes were implanted. In this study Nestin and Sox2 were expressed in NSCs. NSCs were differentiated into NACs and Tyrosine hydroxylase was detected in these cells. A significant decrease was seen in NREM (Non Rapid Eye Movement) and PS (Paradoxical Sleep) stages and a significant increase was seen in wake and PS-A (Paradoxical Sleep without Atonia) in lesion group in comparison with control and sham groups (P≤0.05). NSCs transplantation in experimental groups prevented of decrease in PS and increase in PS-A (P≤0.05). The results of this study demonstrate NSCs have ablility to differentiate into noradrenergic cells and NSCs transplantation improved disruption of the sleep-wake cycle after bilateral lesion of LC.