Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 8th World Congress and Expo on Cell & Stem Cell Research Orlando, Florida, USA.

Day 3 :

Keynote Forum

Diana Anderson

University of Bradford, UK

Keynote: Detection of phase specificity of in vivo germ cell toxicants using an in vitro system

Time : 10:00-10:30

Conference Series Stem Cell Research 2017 International Conference Keynote Speaker Diana Anderson photo
Biography:

Diana Anderson 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, 9 books, successfully supervised 30 PhD students. She has a Hirsch factor of 54. She is Editor -in-Chief of a Book Series for the Royal Society of Chemistry and is Consultant to many International Organisations, such as the World Health Organisation/International Programme of Chemical Safety. She has been the Editorial Board Member of ten international journals.

Abstract:

Studies in the 1960s and ‘70s identified at which stages of the spermatogenic cycle a chemical being examined was most effective in producing genotoxic damage and helped in the understanding of its mechanism of action. The expense of carrying out in vivo assays and the ethical issues involved means that there is a constant need for the development of novel in vitro assays that allow detection of individual germ cell types. Examining germline genotoxicity in the male is generally undertaken in vivo partly because of the difficulty of achieving full spermatogenesis in vitro and partly because mating studies are currently the only reliable way of determining heritable effects. An in vitro assay should have high sensitivity and be suitable
for screening large chemical numbers. Therefore, Staput velocity sedimentation followed by culture of specific rodent cells has been developed for use in such a system. Immunohistochemistry is used to assess the specificity of the separation of the Staput system in identifying the spermatogonia, spermatocytes and spermatids. These three germ cell categories contain the three major events in spermatogenesis: mitotic proliferation, meiosis and spermiogenic differentiation respectively and make suitable groupings for toxicity analysis. It is also possible to consider the early germ stem cells in such a system. DNA damage can be assessed by the Comet assay and apoptosis by the TUNEL assay. We have examined different chemicals in such systems also using gene expression and will discuss the significance of the findings which could significantly reduce animal numbers.

  • Stem Cells | Stem Cell Therapy | Stem Cell Biomarkers | Cellular Therapies | Stem Cells and Cancer | Cell and Organ Regeneration | Cell Differentiation and Disease Modeling | Stem Cell Plasticity and Reprogramming | Tumor Cell Science
Location: Orlando
Speaker

Chair

Paul J Davis

Albany Medical College, USA

Speaker

Co-Chair

Haval Shirwan

University of Louisville, USA

Session Introduction

Brian Mehling

Blue Horizon International LLC, USA

Title: Analysis of outcomes following mesenchymal stem cell therapy in subjects with musculoskeletal conditions

Time : 12:20-12:40

Speaker
Biography:

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

Abstract:

Musculoskeletal conditions are major public health problems and often associated with reduced function and pain. Musculoskeletal related pain is affecting more than one third of the adult population worldwide. Pain reduction is one of the main outcomes to determine the success of therapy of subjects with most common orthopedic conditions. Inflammation plays an important role in the occurrence of acute and chronic musculoskeletal-related pain. Numerous investigations suggest that Mesenchymal Stem Cells (MSCs) represent a valuable tool for therapy of inflammation and regeneration of tissue damage. BHI Therapeutic Sciences offers a novel method of arthritis therapy using a patient’s own stromal vascular fraction (SVF) cells including MSCs. The therapy is available at Malacky Hospital in Slovakia. Blue Horizon International Slovakia is licensed by the Ministry of Health of Slovak Republic to provide adipose stem cell therapies for orthopedic joint applications - knees, hips, shoulders, and ankles. Procedures utilize cuttingedge technology and adult stem cells only. Results from patients’ follow-up examinations and MRI scans showed that stem cell therapy was safe for the patients. Follow-up examination results conducted 10 days, 3 and 6 months after therapy have shown significant improvement of clinical condition relating to pain relief, improved mobility, which was shown also on the follow-up MRI scans of
the affected joints.

 

Speaker
Biography:

Margarita Glazova has received her PhD in 1997 from Sechenov Institute of Evolutionary Physiology and Biochemistry Russian Academy of Science. She has performed her Post-doctoral studies at Turku Centre for Biotechnology, University of Turku/Åbo Akademi, Finland and then at East Carolina University Brody School of Medicine, Department of Physiology, Greenville NC, USA. Currently, she is Head of Lab at the Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia. Her main interests concern the study of adult neurogenesis and neural stem cells differentiation at normal and under neuropathology disease, such as epilepsy. She has published more than 30 articles in peer-reviewed scientific journals

Abstract:

The hippocampal formation is one of the most affected areas, where epileptogenesis is usually associated with dramatic pathological changes in morphology and functions. It is known that epilepsy progression is tightly connected with aberrant neurogenesis in the hippocampus. Animal models of audiogenic epilepsy are useful tools to understand the mechanisms underlying human reflex epilepsies. In our work, we have analysed hippocampal neurogenesis in Krushinsky-Molodkina rats genetically prone to audiogenic seizure (AGS). Our data demonstrated that several AGS stimulated proliferation of neural stem cells. On the other hand, audiogenic kindling led to elimination of the proliferated cells in the hippocampus. Moreover, AGS was accompanied with changing in activity of MAPK cascade, glutamate neurotransmission and expression of exocytosis proteins

 

Speaker
Biography:

Mei Wan is an Associate Professor of the Center for Musculoskeletal Research, Department of Orthopedic Surgery at Johns Hopkins University School of Medicine. She has obtained her PhD in Pathophysiology at Hebei Medical University in 1997. Her research for the past 18 years focuses on characterizing the mechanisms by which bone marrow mesenchymal stem cells (MSCs) are regulated in various physiological and pathological conditions such as bone remodeling, cancer development, vascular disorders and tissue repair/remodeling. In recent years, she found that active TGFβ can be released from tissue in response to perturbations to the local environment such as bone remodeling (Nat. Med. 2009, Cell Stem Cell 2011), arterial injury (Stem Cells 2012, Stem Cell Dev. 2014, Nature Communications 2016) and lung injury (J. Immunol. 2014, J. Immunol. 2016). The released active TGFβ stimulates the migration of MSCs to participate in tissue repair or remodeling. Currently, she is an Editorial Board Member for Journal of Bone and Mineral Research and Bone Research.

Abstract:

Recent studies suggest that a sub-population of MSCs, specifically cells expressing nestin, mobilize from their original niches to the vascular remodeling sites after arterial injury in mice. In the present study, we delineated a molecular mechanism by which the lineage commitment/differentiation of nestin+ MSCs is controlled during vascular repair. Using a genetic nestin+ cell lineage mapping mouse model, we found that nestin+ cells recruited to the injured arteries is a contributor to neointimal formation. Nestin+cells recruited to the remodeling sites represent a mixed population, with MSCs as a predominant component. More importantly, we revealed that TGFβ-activated RhoA/ROCK signaling functions as a molecular switch regarding the fate of MSCs in arterial repair/ remodeling after injury. MSCs differentiate into myofibroblasts when RhoA/ROCK is turned on, endothelial cells when turned off. The former is pathophysiologic resulting in intimal hyperplasia, whereas the latter is physiological leading to endothelial repair. Further analysis revealed that MSC RhoA activation promotes formation of an extracellular matrix (ECM) complex consisting of connective tissue growth factor (CTGF) and vascular endothelial growth factor (VEGF). Inactivation of RhoA/ROCK in MSCs induces matrix metalloproteinase-3-mediated CTGF cleavage, resulting in VEGF release and MSC endothelial differentiation. Our findings uncover a novel mechanism by which cell-ECM interactions determine stem cell lineage specificity and offer additional molecular targets to manipulate MSC-involved tissue repair/regeneration.
 

 

Speaker
Biography:

Abstract:

As innovators move closer to clinical success, a gap in the ability to cost effectively manufacture cell therapies has been identified. To address this gap, we have demonstrated the use of single-use expansion and harvest systems that robustly expand and recover a variety of stem cells. An additional contributor to the system is the inclusion of high quality reagents that are animal origin free, lead to better yields and are supplied with a strong regulatory dossier. We will present data regarding ease of use, yield, viability and characterization for full solution expansion and harvest of manufactured cell therapies. Start to finish solutions for expansion and harvest are key enabling technologies for success in commercializing cell therapies.

Speaker
Biography:

Vincent S Gallicchio received his PhD in Hematology from New York University and his Diploma in Medicine from the University of Arad, Romania. He performed Postdoctoral studies at the University of Connecticut Medical Center and conducted his fellowship in Hematology from the Memorial Sloan Kettering Cancer Center. He has published more than 150 research articles in peer-reviewed journals on topics focused on stem cells and hematology, seven books, many book chapters, and has been awarded eleven United States and one international patent for developmental therapeutics for AIDS and Cancer. He currently serves as a Director of Stem Cell Therapy for out-patient clinical use.

Abstract:

The science driving the current revolution in regenerative medicine continues to focus on advances in research made in tissue biology and engineering along with molecular medicine. The goal is to advance the field such that the application of stem cells can improve clinical outcomes when it addresses the issue of altered or damaged cells, tissues or body organs. Another exciting area
of research in regenerative medicine addresses the need to grow new body organs to replace damaged or dysfunction body parts, thus addressing the current problem, where there is a shortage of organs available from potential donors. If the source of stem cells is derived from the same individual in whom the body tissue or organ needs to be replaced will eliminate the problem of organ transplant rejection. Another area of research will also focus on the expanding sources of stem cells such as the use of induced pluripotential stem cells (iPCs), mesenchymal stem cells (MSC) derived from adipose and aminotic tissue or cord blood beyond blood and bone marrow. Any advance in applied use of stem cells beyond blood and immunological use is limited by the fact that cord cells are hematopoietic stem cells (which can differentiate only into blood cells), and are not pluripotential stem cells, as embryonic stem cells, which can differentiate into any type of tissue, they can be induced to “dedifferentiate” as iPCs. Cord blood has been studied as a treatment for diabetes. However, apart from blood disorders, the use of cord blood for other diseases is not a routine clinical modality and remains a major challenge for the stem cell community. Along with adipose and amniotic tissue, cord blood, chord lining, and Wharton’s jelly, have been explored as sources of MSC in order to treat conditions mediated as a function of aging and inflammation. MSC have been studied in vitro, in animal models, and in early stage clinical trials for cardiovascular diseases, as well as in osteoarthritic conditions, neurological deficits, liver diseases, immune system diseases, diabetes, lung injury, kidney injury and leukemia. This presentation will review the use of MSCs in these clinical indications.

Speaker
Biography:

Meenakshi A Chellaiah is a Tenured Full Professor at the University of Maryland School Of Dentistry, Baltimore, since 2014. Formerly, she was a tenure-track Assistant Professor (2000-2007) and tenured Associate Professor (2008-2013) at the same Institution. She has obtained her PhD from Madurai Kamaraj University,
India and completed her Post-doctoral studies at St. Louis University and Washington University, St. Louis, USA. She has published more than 60 papers in peer reviewed journals and gave oral presentations at over 100 national and international conferences.

Abstract:

Advanced prostate cancer (PCa) is androgen-independent. Studies revealed that tumor–initiating cells negative for androgen receptor (AR-) and p63, express the markers for cancer stem cells (CSCs). Recent studies indicate the involvement of stem cell markers (CD44, α2β1, CD133, NANOG, SOX2 and OCT4) in the maintenance of CSCs and relapse of the disease. A unique prostate stem cell biomarker has not yet been found. Our goal here is to characterize PCa cell lines derived from different metastases and tissue microarray (TMA) sections for CSC markers. We used PCa cell lines derived from lymph node (LNCaP), brain (DU145) and bone (PC3) metastases along with control cell lines (HPR1 and BPH1). Loss of AR activates CD44 expression in PC3 and DU145 cells. CD44 expression is negligible in AR positive (AR+) LNCaP cells. Only a sub population (<5%) of PC3 cells were CD44+/ CD133+. Of all the cell lines tested, the combined expression of CD44 and CD133 was greater in PC3 than DU145 cells. CD133+ PC3 cells were enriched by sequential antibody selection using DYNA beads. As compared with PC3 cells, CD44+/CD133+ enriched PC3 cells are integrin α2β1(+), Integrin αvβ3(-) and highly migratory in nature. Immunohistochemistry analyses of TMA sections demonstrated the expression of CD133 in a few island of cells (~1-2%) within the lumen filled with tumor cells (Stage III and IV) and in basal luminal cells. When comparing the expression levels of other stemness factor such as NANOG, SOX2 and OCT4 in PCa cells indicated above, only SOX2 expression was considerably higher at mRNA level which corresponds with an increased protein levels in PC3 cells. Knockdown of SOX2 not only reduces in vitro migration but also the expression of EMT regulatory proteins (Slug and Snail) in PC3 cells. Existence of sub-populations of CSCs [CD133+, CD44+, and integrin α2β1(+)] in bone metastatic PC3 cells and in the malignant prostatic tissue (CD44+, CD133+) suggest that they may cause PCa relapse and failure of many treatment practices. Furthermore, expression of SOX2 and CD133 in PC3 cells appears unique. These proteins may be involved in the maintenance of CSCs in bone microenvironment and could be used as a prognostic marker to detect bone metastatic spread. Further elucidation of the SOX2 target genes and inhibition of the function of SOX2 and its target gene may be useful in the manipulation of cancer progression in/at-risk of men.

Biography:

Željka Večerić-Haler works as a Medical Doctor, specialist of Nephrology at Nephrology department, University Medical Centre, Ljubljana. As a Nephrologist, she is faced with numerous challenges from the field of Nephrology on daily basis and is specially oriented in the fields of kidney transplantation, regenerative medicine and substitutive therapies of acute and end stage kidney failure. She has completed her PhD from University of Ljubljana Faculty of Medicine in 2016 and her thesis is revealing impact of immunological environment on the behavior and functions of transplanted stem cells. Her interests of research involve studies on stem cell transplantation for regeneration of kidney injury and immune tolerance in kidney transplantation.

Abstract:

In the last decade much effort has been introduced into treating toxic acute kidney injury with novel mesenchymal stem cell (MSC) approaches. Inflammation is known to play a crucial role in cisplatin induced acute kidney injury, were the inflammatory pathways were shown to be predominantly driven by T lymphocytes. However, most preclinical studies of stem cell xenotransplantation are performed on severe complex immunodeficiency animals lacking innate and adaptive immunity responses, therfore markedly
altering the real inflammatory state of the diseased and influencing natural course of illness. MSC were shown to have a great therapeutic potential in many disorders, where, it is not known whether those are equally effective in immunocompetent and immunocompromised microenvironment. Antithymocyte immunoglobulin is a polyclonal antibody preparation with multiple effects on immune system with preferential influence on peripheral T cell depletion. This drug enables dose dependant immunosupression, therefore allowing verification of the hypothesis that therapeutic functions and immunoregulatory properties of MSC are largerly affected by immunologic microenvironment and contribute to regeneration of kidney injury predominantly, when introduced in the non-inflammatory niche.

Biography:

Abstract:

The loss and degradation of articular cartilage tissue matrix play central roles in the process of osteoarthritis (OA). New models for evaluating cartilage repair/regeneration are thus of great value for transferring various culture systems into clinically relevant situations. The repair process can be better monitored in ex vivo systems than in in vitro cell cultures. I have therefore established an ex vivo defect model prepared from bovine femoral condyles for evaluating cartilage repair by the implantation of cells cultured in various ways, e.g., monolayer-cultured cells or suspension or pellet cultures of articular bovine chondrocytes representing different cell compactions with variable densities of chondrocytes. I report that the integrin subunit α10 was significantly upregulated in suspension-cultured bovine chondrocytes at passage P2 compared with monolayer-cultured cells at P1 (p=0.0083) and P2 (p<0.05). Suspension-cultured cells did not promote cartilage repair when compared with implanted monolayer-cultured chondrocytes and pellets: 24.0±0.66% for suspension cells, 46.4±2.9% for monolayer cells and 127.64±0.90% for pellets (p<0.0001) of the original defect volume (percentage of defect). Additional cultivation with chondrogenesis-promoting growth factors TGF-β1 and BMP-2 revealed an enhancing effect on cartilage repair in all settings. The advantage and innovation of this system over in vitro differentiation (e.g. micromass, pellet) assays is the possibility of examining and evaluating cartilage regeneration in an environment in which implanted cells are embedded within native surrounding tissue at the defect site. Such ex vivo explants might serve as a better model system to mimic clinical situations.

Speaker
Biography:

Chrishan S Samuel is an Associate Professor and has completed his PhD from University of Melbourne, Victoria, Australia. He has completed his Post-doctoral studies at the Stanford University School of Medicine and Molecular Medicine Research Institute. He is currently a Senior Research Fellow of the National Health & Medical Research Council of Australia and Head of the Fibrosis Laboratory at the Department of Pharmacology, Monash University (Melbourne, Victoria, Australia). He has over 125 career publications, which have been cited over 5100 times and his research interests are focused on establishing novel therapeutic strategies for organ fibrosis.

Abstract:

While stem cell-based therapies have demonstrated immunomodulatory, anti-inflammatory and tissue-reparative functions in acute disease settings, they are less effective when administered to chronically damaged organs. This is likely attributed to tissue fibrosis which can impair stem cell survival, proliferation, migration and integration with resident tissue cells. Hence, the therapeutic efficacy of human bone marrow-derived mesenchymal stem cells (MSCs) or human amnion epithelial stem cells (AECs) were evaluated in the setting of chronic allergic airways disease (AAD), in the absence or presence of an anti-fibrotic drug (serelaxin; RLX). Female Balb/c mice subjected to the 9-week model of ovalbumin (OVA)-induced chronic AAD, were either vehicle-treated (OVA alone) or treated with MSCs or AECs alone (intranasally (i.n)-administered with 1x106 cells once weekly), RLX alone (i.nadministereddaily) or a combination of MSCs or AECs and RLX from weeks 9-11 (n=6/group). Measures of airway inflammation (AI), airway remodeling (AWR) and airway hyper-responsiveness (AHR) were then assessed. OVA-injured mice exhibited exacerbated AI, epithelial damage/thickness, sub-epithelial and total collagen deposition (fibrosis) and AHR compared to their saline-treated counterparts (all p<0.01 vs. saline-treated controls). RLX or AECs (but not MSCs) alone normalized epithelial thickness and partially diminished the OVA-induced fibrosis and AHR by ~40-50% (all p<0.05 vs. OVA alone). Furthermore, the combination treatments normalized airway epithelial thickness, fibrosis and AHR to that in normal mice and significantly decreased AI. These findings showed that the presence of an anti-fibrotic enhanced MSC- or AEC-induced reversal of the three central components of chronic AAD/asthma, to a greater extent than stem cell-treatment alone.

Speaker
Biography:

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

Abstract:

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

Speaker
Biography:

Yael Porat has obtained PhD in Immunology from Sackler School of Medicine and is the Founder and CEO of BioGenCell, a Biotechnology Company focusing on Stem Cell Therapy and Regenerative Medicine. She previously served as Head of Global Biological Development at Teva Pharmaceuticals and CTO at TheraVitae, where she led research on therapies for patients with cardiovascular diseases.

Abstract:

Background: Vascular diseases including cardiovascular and peripheral vascular are a major cause of morbidity and mortality worldwide. Critical Limb Ischemia (CLI), the most serious form of PVD affects >3 Million people. Within 1 year, 25% of patients die and 30% undergo amputation. We describe a novel technology for generating a therapeutic population (BGC101) of enriched endothelial progenitor cells from a patient’s standard blood sample, using dendritic cells (DCs) to direct stem/progenitor cell (SPC) activity. This one day culture process produces sufficient numbers of potentially therapeutic SPCs for the treatment of patients with CLI who do not have other viable treatment options. The EnEPC-CLI-01 clinical study aims to assess BGC101 feasibility in treating CLI patients. Methods & Results: DCs from healthy and diabetic donors were activated with anti-inflammatory and pro-angiogenic molecules. Co-culture for 12-18 hours of activated DCs with SPCs generated 83.7±7.4×10^6 BGC101 cells with >97% viability and angiogenic/ stemness potential. When administered to nude mice with limb ischemia, BGC101 yielded a high safety profile, improved blood perfusion, capillary density, and limb function within 21 days (p<0.0002). A phase I/IIa Study (open-label in 5 patients followed by randomized placebo-controlled study in 25 patients) is underway. Preliminary results from the first patients demonstrated a high safety profile and promising clinical outcomes. Conclusions: DCs promote the generation of EnEPC within culture after one day. The resulting, BGC101, a potential for treatment of vascular conditions, including arteriosclerotic heart disease, stroke and peripheral ischemia is now undergoing a phase I/IIa study in CLI patients.

Biography:

Arshak R Alexanian is currently the Chief Scientific Officer at Cell Reprogramming & Therapeutics LLC and an Adjunct Associate Professor in the Department of Medicine at the Medical College of Wisconsin (MCW). Previously, he held faculty positions in the Departments of Neurosurgery at MCW (2000-2013) and in the Departments of Anatomy and Neurobiology, as well as in Biochemistry and Molecular Biology, at Colorado State University (1997-2000). He has received training at universities and centers world wide, including the Pasteur Institute and University of Montpelier in France, University of Saarland in Germany, Institute of Biochemistry in China and Russia, and Colorado State University, where he gained extensive experience in the fields of Biochemistry, Molecular Biology, Cell Biology (stem cell biology) and Neurosciences. The areas of interest of his research are the epigenetic regulation of cell fate commitment and differentiation, development of cell reprogramming technologies to produce different neuronal and glial cell types and elucidation of the therapeutic effect of these specialized cell types in several neurological disorders.

Abstract:

Advances in cell reprogramming technologies to generate patient-specific cells of a desired type will revolutionize the field of regenerative medicine. Over the last decade, several cell reprogramming methods such as nuclear transfer, cell fusion and transfection or transduction with pluripotent factors have been developed. However, the majority of these technologies require the exposure of cell nuclei to large reprogramming molecules via. transfection, transduction, cell fusion, or nuclear transfer. These methods raise several technical, safety and ethical issues. Chemical genetics is an alternative approach to cell reprogramming that uses small, cell membrane penetrable substances to regulate multiple cellular processes, including cell plasticity. Recently, using achemical genetics approach (a combination of small molecule modulators of epigenetic target enzymes and neural inducing factors) we have been able to turn human mesenchymal stem cells (hMSCs) directly into neuronal progenitors that have the potential to generate different neuronal subtypes, such as dopaminergic, cholinergic and GABAergic cells when further grown in appropriate neuronal differentiation media. The therapeutic effects of these cells on several neurological disorders have been demonstrated.

Biography:

Abstract:

Measuring gene expression in individual cells is crucial for understanding the gene regulatory network controlling human embryonic development. We applied single-cell RNA -Seq analysis to human preimplantation embryos, primordial germ cells (PGCs),and human embryonic stem cells (hESCs). We also systematically profiled the DNA methylome of human early embryos from the zygotic stage through to post-implantation. We showed that the major wave of genome-wide demethylation is complete at the 2-cell stage, contrary to previous observations in mice. Moreover, the demethylation of the paternal genome was much faster than that of the maternal genome, and by the end of the zygotic stage the genome-wide methylation level in male pronuclei was already lower than that in female pronuclei. Then, we also showed that long interspersed nuclear elements (LINEs) or short interspersed nuclear elements (SINEs) that were evolutionarily young are demethylated to a milder extent compared to older elements in the same family and had higher abundance of transcripts, indicating that early embryos tend to retain higher residual methylation at the evolutionarily younger and more active transposable elements. Furthermore, we analyzed the DNA methylome of human PGCs andfound global demethylation of their genomes. Approximately, 10 to 11 weeks after gestation, the PGCs were nearly devoid of any DNA methylation, whereas the repeat elements still kept high level of residual methylation. Our work provides insights of critical features of the transcriptome and DNA methylome landscapes of human early embryos and primordial germ cells, as well as the functional significance of DNA methylome to regulation of gene expression and repression of transposable elements.

Speaker
Biography:

Paul J Davis is a Professor of Medicine at Albany Medical College, and Chief Scientific Officer at NanoPharmaceuticals LLC, Rensselaer, NY. He is former Chair at the Department of Medicine, Albany Medical College. As an Endocrine Researcher, he has co-authored 250 scientific publications and has co-edited two text books on Angiogenesis (Springer, 2013; Elsevier, 2016). He and Dr. Shaker Mousa described the thyroid hormone-tetrac receptor on integrin αvβ3.

Abstract:

The programmed death-1 (PD-1)/PD-ligand 1 (PD-L1) immune checkpoint modulates activated T cell-cancer cell interactions. The PD-L1 protein generated by tumor cells engages T lymphocyte PD-1 to suppress T cell engagement of tumor cells—protecting tumor cells from immune destruction—and may also induce T cell apoptosis. Overexpression of PD-L1 is seen in various human cancer cells and correlates with decreased patient survival. PD-1 antibodies (Opdivo®; Keytruda®) are effective anticancer agents in subsets of solid tumor and hematologic malignancy patients. Because these antibodies are effective in subsets of patients and also induce important adverse events (AE) in normal tissues that elaborate PD-L1, it is desirable to seek non-immunologic strategies for attacking the PD-1/PD-L1 checkpoint. We have shown that thyroid hormone (L-thyroxine, T4) at physiological concentrations acts non-genomically to stimulate cancer cell proliferation,to block apoptosis and to stimulate tumor-related angiogenesis. We report here that T4 stimulates PD-L1 gene expression by a mechanism that is initiated at the cell surface receptor for T4 on the extracellular domain of plasma membrane integrin αvβ3. T4 was studied in cultured human colon cancer HT-29 and HCT116 cells and triple-negative breast cancer MDA-MB-231 cells, at concentrations ranging from 10[-8] to 10[-6] M, where 10[-7] M yields physiological concentrations of free T4 in the culture system. T4 significantly increased PD-L1 mRNA abundance by 2-to-6-fold in HT-29 cells, by 2-fold in HCT116 cells and by 1.6-fold in breast cancer cells. An inhibitor of T4 actions at the integrin, tetraiodothyroacetic acid (tetrac), in a nanoparticulate formulation (Nano-diamino-tetrac, NDAT) intended to increase tetrac residence time at the integrin, blocked the actions of T4 on PD-L1 gene expression and significantly reduced basal PD-L1 expression. Basal expression is defined here as that which occurs in the absence of added T4. T4 also significantly increased PD-1 mRNA abundance in these tumor cell lines and NDAT blocked the T4 effect and significantly reduced basal levels of PD-1 mRNA. PD-1 is viewed primarily as T cell product, but has been reported to be expressed by a variety of solid tumor cells and we speculate in such cells, which may be anti-apoptotic. In summary, the components of the PD-1/PD-L1 check point are subject to non-immunologic modulation by T4 and by NDAT. The actions of NDAT in these in vitro studies have possible therapeutic implications

  • Stem Cells | Stem Cell Therapy | Cellular Therapies | Stem Cells and Cancer | Cell and Organ Regeneration
Location: Hall- A

Session Introduction

Haval Shirwan

University of Louisville, USA

Title: SA-4-1BBL as an adjuvant platform for the development of vaccines
Speaker
Biography:

Haval Shirwan is Dr. Michael and Joan Hamilton Endowed Chair in Autoimmune Disease, Professor of Microbiology and Immunology, Director of Molecular Immunomodulation Program at the Institute for Cellular Therapeutics. He conducted his Graduate studies at the University of California in Santa Barbara, CA, and Postdoctoral studies at California Institute of Technology in Pasadena, CA. He joined the University of Louisville in 1998, after holding academic appointments at various academic institutions in the United States. His research focuses on the modulation of immune system for the treatment of immune-based diseases with particular focus on type 1 diabetes, transplantation, and development of prophylactic and therapeutic vaccines against cancer and infectious diseases. He is an inventor of over a dozen of worldwide patents, Founder and CEO/CSO of FasCure Therapeutics, LLC. He widely published, organized and lectured at numerous national/international conferences, served on study sections for various federal and non-profit funding agencies, and is the Editorial Board Member of a number of scientific journals. He is the member of several national and international societies and recipient of various awards.

Abstract:

The costimulatory members of the TNFR family molecules are critical for the generation of cellular and humoral immune responses. In particular, the 4-1BB pathway regulates various cells of innate, adaptive, and regulatory immunity for a productive immune outcome. As such, the ligand, 4-1BBL, has a great potential as adjuvant component of prophylactic and therapeutic vaccines against cancer and infections. We generated a novel form of the ligand, SA-4-1BBL, and demonstrated its pleiotropic effects on various cells of the immune system. As adjuvant, SA-4- improved the protective efficacy of the lead rF1V subunit vaccine against Y. pestis in a mouse model by generating mixed Th1 cellular and humoral immune responses.

Biography:

Abstract:

Cancer-associated fibroblasts (CAFs) are one of the most abundant non-cancer cells in the tumor microenvironment that support melanoma to grow, migrate and develop drug resistance. β-catenin signaling is important for fibroblast activation and their biological functions. We discovered that CAFs that surround and infiltrate melanoma tumors express high levels of cytoplasmic and nuclear β-catenin. Ablation of β-catenin causes cell cycle arrest in stromal fibroblasts and reduces the production of autocrine and paracrine factors and extra cellular matrix (ECM) proteins. Thus, we designed a novel genetic approach to ablate β-catenin expression in melanoma-associated CAFs to evaluate their effects on the initiation and development of Braf-driven melanoma. To our surprise, stromal fibroblasts showed inhibitory effects on melanoma initiation in vivo. Conversely, Braf-activated Pten-deficient melanoma development was significantly suppressed after CAFs were deactivated by β-catenin ablation. Consistent with this observation, melanoma cell growth was significantly inhibited while cell death was increased. Meanwhile, decreased production of ECM protein collagen and fibronectin was found in Braf-driven melanoma tumors that contained β-catenin-deficient CAFs, suggesting the melanoma microenvironment was remodeled. Further analysis revealed that expression of phospho-Erk1/2 and phospho-Akt was greatly reduced, effectively abrogating the activating effects and abnormal cell cycle progression induced by Braf and Pten mutations. In addition, epithelial-mesenchymal transition (EMT) was also suppressed in melanoma cells. Taken together, our data highlight an important crosstalk between CAFs and the RAF-MEK-ERK signaling cascade in BRAF-activated melanoma and may offer a new approach to abrogate host-dependent drug resistance in targeted therapy.

Speaker
Biography:

Thomas Andl obtained his PhD from the University of Heidelberg, Germany and extended his training in squamous epithelia and hair follicle biology at the University of Pennsylvania and Vanderbilt University. He established mouse models to study BMP, Wnt and microRNA signaling and is interested in how squamous epithelia such as the epidermis or the oral mucosa are maintained under normal conditions and how these epithelia responds to stress, infection and carcinogenesis. His main focus is on the putative stem cells in these tissues and how they are organized, maintained, transformed into cancer and are responding to challenges.

Abstract:

Adult tissue stem cells maintain tissues throughout life. Therefore, there may be quantitative and qualitative differences in the stem cells of short-lived and long-lived mammals. We hypothesized that one adaptation to longevity could reside within the stem cell compartment, e.g. by enhancing the quality and integrity of the stem cell pool through increased stem cell quiescence and repair. Using squamous epithelia as model systems to explore our stem cell quality-longevity hypothesis, we show that the pattern of proliferation in human oral mucosa fundamentally differs from most other mammals. In contrast to mouse squamous epithelia, proliferation in humans is absent from the basal cell layer and over 90% of proliferating cells are found in the first suprabasal cell layer(s). Therefore, human squamous epithelia have a quiescent basal cell layer that traditionally is viewed as the stem cell layer. The relative proliferative activity of the basal cell layer is correlated with short-livedness in mammals and basal cell quiescence to our knowledge is the only other characteristic that correlates with longevity in addition to body size. Next, we defined the characteristics of this quiescent basal cell layer. Key features in addition to its quiescence are TGF beta expression signatures, polycomb repressor complex 1 (PRC1) expression, expression of a set of well know apoptosis protectors and Hippo signaling components. In a 3D squamous epithelial reconstruct TGF beta signaling was able to induce quiescent basal cell markers supporting a role of this signaling pathway in establishing or maintaining quiescent stem cells in human squamous epithelia.

George G Chen

The Chinese University of Hong Kong, Hong Kong

Title: Reduction of cytochrome P450 1A2 in hepatocellular carcinoma
Biography:

George G Chen is a Professor in the Department of Surgery, Director of Surgical Research Laboratories, The Chinese University of Hong Kong, China. He has extensive experience in cancer research, particularly in the area of hepato-carcinogenesis. He has authored or co-authored more than 190 papers and has written number of books or book chapters.

Abstract:

Clinically, hepatocellular carcinoma (HCC) occurs much more frequently to males than females, but, the underlying mechanisms remain largely unknown. Cytochrome P450 1A2 (CYP1A2) is involved in the metabolism of 17β-estradiol (E2) that is known to have a protective role in the development of hepatocellular carcinoma (HCC). In this study, experiments were carried out in cultured HCC cells and animal models to explore the significance of CYP1A2 in HCC. It was found that the level of CYP1A2 was significantly reduced in HCC. The overexpression of CYP1A2 could inhibit the growth of HCC in cultured HCC cells as well as the HCC tumor model in mice. Mechanically, the hypermethylation of the CYP1A2 promoter contributed to its reduction and the application of HDAC inhibitors recovered the loss of CYP1A2. In conclusion, the expression of CYP1A2 was significantly reduced in HCC and this reduction can be corrected by the inhibition of HDAC.

Biography:

Purwati 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. 2015 be a chairman of stem cell research and development Centre Universitas Airlangga Surabaya Indonesia. Have almost 50 publication in journals, papers, and seminar.

Abstract:

Infectious diseases still remains a health problem in both developing and developed countries. Based on epidemiological studies that have been conducted, sepsis is known as a major health problem, because it is still the leading cause of death in critical cases in the world. Dr. Soetomo Hospital reported that 32.18% new visitors are infected (Health Status Profile of East Java Province). In a research done at Dr. Moewardi Hospital in 1997, about 130 (97%) from 135 sepsis patients died, while at the Internal Medicine Dr. Moewardi Hospital in 2004, it was 74 (83.1%) patients. Armstrong and Pinner (1999) reported that recently in USA, there is an increase in the proportion of patients hospitalized for infection and mortality rate of infectious diseases. Three indicators that show an increase in the burden of infection are high mortality in infectious diseases, the proportion of infectious diseases hospitalization, and the number of ambulatory patients with infectious diseases. The most sepsis causes are derived from a toxin balk stimulation of gram-negative endotoxin or gram-positive exotoxin. Endotoxin can directly cause sepsis by the Lipo Poly Saccharide (LPS) and along with antibodies in the patient's blood serum form LPSab (Lipo Poly Saccharide antibody). LPSab in blood through the receptor CD14+ will react with macrophages. Makropfag will express Immunomodulatory IL-1, TNF-α, IL-6 and IL-8, which cause an inflammatory reaction. These reactions can only occur on gram-negative bacteria which have LPS on the walls. Stem cell therapy for sepsis includes Mesenchymal Stem Cells (MSCs) and Haematopoetic Stem Cells (HSCs), both types of stem cells as an immunomodulator in sepsis. MSCs inhibit maturation and decrease the expression of molecules presentation and co-stimulator from cells presenting antigen. MSCs also inhibit B-cell antibody production. In case of NK cells, MSCs can suppress proliferation, cytokine secretion and cytotoxicity. Furthermore, MSC does not only have the obstacles effect directly in T cells, but also affects the first critical stage of the immune response in which MSC can inhibit the differentiation and maturation of cells presenting antigen and causes dendritic cells to change the cytokine secretion profile to reduce secretion of proinflammatory cytokines such as TNF-α, IFN-γ and IL-12. The most important thing is to increase the production of IL-10, that is suppressive and tolerogenic and is a potent inducer of regulatory T cells (Treg). In addition, it has been reported that human MSC causes an increase in the proportion of Treg.

Kenneth K Wu

National Health Research Institutes and CMU, Taiwan

Title: Prevention of stress-induced MSC premature senescence by 5-methoxytryptophan
Speaker
Biography:

Kenneth K Wu, MD, PhD was the Professor and Director of Hematology and Vascular Biology Research Center at University of Texas Health Science Center at Houston, Texas, USA and served as the President of National Health Research Institutes of Taiwan. He is currently Chaired Professor at China Medical University (CMU) Taichung, Taiwan and National Tsing-Hua University Hsin-Chu, Taiwan. He has published more than 350 papers and served as Editorial Board Member or Associate Editor of several journals.

Abstract:

Premature senescence of Mesenchymal Stem Cells (MSCs) contributes to MSC growth arrest and secretory phenotype. It hampers MSC expansion in culture and MSC cell therapy. To develop strategies to prevent MSC senescence, we assessed 5-methoxytryptophan (5-MTP). We previously reported that human fibroblasts and endothelial cells produce and release 5-MTP into the extracellular milieu including circulating blood. Serum 5-MTP concentrations in healthy human subjects were 0.5-1.2 μM. 5-MTP inhibits stress-induced cyclooxygenase-2 (COX-2) and cytokine expressions in macrophages and protects endothelial barrier function and integrity. To determine whether 5-MTP controls MSC senescence, we pretreated bone marrow-derived MSC (BMMSC) with chemosynthetic pure L-5-MTP followed by high glucose (HG) or H2O2. HG metabolic stress or H2O2 oxidant stress induces typical premature senescence such as growth arrest, increased p16 and p21, SA-βgal and senescence-associated secretory phenotype (SASP). 5-MTP prevented all the senescence phenotypes induced by HG or H2O2. Our preliminary data suggest that 5-MTP exerted the anti-senescence effect by upregulating Foxo3a expression. We conclude that 5-MTP is a valuable lead compound to develop new agents to prevent MSC senescence and improve MSC cell therapy.

Speaker
Biography:

Yoshiaki Ito has obtained his MD and PhD from Tohoku University, Japan and studied in Duke University, USA, Imperial Cancer Research Fund Laboratories, UK and NIH, USA. He became Professor in 1984 in the Institute for Virus Research, Kyoto University and served as Director between 1995-2001. In 2002, he moved to the Institute of Molecular and Cell Biology in Singapore and also served as Director of Oncology Research Institute, National University of Singapore (NUS) between 2002 and 2008. He is currently a Senior Principal Investigator at the Cancer Science Institute of Singapore, NUS. He discovered the middle T of polyomavirus and RUNX gene.

Abstract:

The understanding of gastric carcinogenesis is still relatively poor, because of the lack of knowledge of stem cells in stomach. Stomach is roughly divided into the corpus (main body) and antrum (next to duodenum). Rapidly growing cells are located in the isthmus of both corpus and antrum. We reported earlier on the 270 bp RUNX1 enhancer element that drives the expression of RUNX1 in hematopoietic stem cells (HSC), termed eR1. We now show that eR1 also targets isthmus of both the corpus and antrum. A well-known stem cell marker, Lgr5 was reported to identify stem cells of stomach but never in the isthmus. In the antrum, Lgr5+ cells are located at the base of antrum below the location where eR1+ cells reside. Both Lgr5+ cells and eR1+ cells have the ability to form organoids which are considered to develop only from stem cells. I will discuss about the presence of more than one stem cell types in a given tissue. Small number of eR1+ cells is also detected in fully differentiated chief cells that express pepsinogen located at the base of corpus. There are several reports showing the ability of chief cells to proliferate. Currently, there are disagreements among the researchers as to whether chief cells have potential to have stem cell activity. The consequence of K-rasG12 expression in eR1+
cells will also be discussed.

Speaker
Biography:

Ganapathi Bhat M is a Senior Consultant Medical Oncologist & Stem Cell Transplant Physician at Jaslok Hospital & Research Centre, since 2006. He gained specialized training in stem cell transplantation as part of the ESH-EBMT (2007), 2011 (Labaule, France) and ICAS training program (2009) from ULM University, Germany. He is also a Member of academic organizations namely ESMO, IELSHG, EHA, Asia Pacific Bone Marrow Transplant and an Affiliate of American Association for Cancer Research and BITs Congress Tank. He served on the board of teaching faculty (Kuwait) for MRCP (Pathology & Hematology). He is also an Editorial Board Member of various international scientific journals.

Abstract:

Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) intents at abating malignant and benign diseases. However, Graft-versus-host disease (GVHD) still is the major cause of morbidity and mortality. GVHD demonstrates a heterogeneous clinical presentation and compromises the benefits associated with allo-HSCT. The complex pathophysiology of GVHD remains as a focus of unending research and it also outlines many existing and novel therapeutic targets. This has become a major objective of research in the transplant immunobiology. Standardized stratagem to understand the predisposing factors, pathophysiology and diagnosis for both acute and chronic forms of the disease, is still lacking. There is a need for better insight into the Pathobiology behind the GVHD process and widening more effective treatment strategies while maintaining the graft versus tumor effect to keep away a likely rise in relapse-related mortality. The studies should be directed towards assessment tools for disease severity, outcome, quality of life; variability factors influencing the outcome of the transplant and identification of potential biomarkers which predicts therapeutic responses as both acute and chronic GVHD are biologically and clinically heterogeneous. The increasing knowledge about host as well as the donor associated variables that aggravate GVHD and to develop drugs in relation to immune-modulation perhaps allows more effective treatment of GVHD in the future. Impending research strategies and the discovery of novel therapeutic targets may improve the outcome further. Well timed diagnosis, multidisciplinary working and good supportive care are undoubtedly important to optimize the response and survival.

Biography:

Mahsa Rashidi is MD, PhD at Children’s National Hospital, DC, affiliated to George Washington University of Medical Sciences, USA. She has finished her Medical degree at Shahid Beheshti University of Medical Sciences, Tehran, Iran. Her PhD is a joint program between George Washington University, USA and the University of Melbourne, Australia. This abstract is based on her PhD thesis, submitted to the University of Melbourne, Faculty of Medicine, Australia. Her work is focused on evaluating skin-stem-cells for cell-based therapies in muscular dystrophies. In her work, the effects of various dosages of several drugs/treatments on the stem cells differentiation is evaluated.

Abstract:

Duchenne-muscular-dystrophy (DMD) is the commonest muscular-dystrophy caused by the absence of dystrophin. Stem-celltherapy in DMD is one of the more promising-approaches for treatment. Multipotent-stem-cells residing in the hair-folliclepapilla are highly-plastic and are reprogrammable to bone, cartilage, haematopoietic and muscle. Dermal-papilla-cells (DPC) from the hair-follicles of mouse-whisker-pad were microdissected and cultured. We showed that DPC undergo myogenic-differentiation when co-cultured with different types of myoblasts including normal and dystrophic human-myoblasts. Lamin A/C staining was used to distinguish DPC and myoblast-derived myonuclei inside myotubes. DPC incorporated into myotubes and up-regulated the muscle-marker myogenin in co-culture with human-myoblasts, suggesting that DPC fully underwent myogenic-differentiation in these co-cultures. DPC incorporation-efficiency was low in all co-cultures and differed significantly between various types of myoblast; however, no significant difference was observed between normal and dystrophic human-myoblasts. These encouraging-findings suggested that the altered properties of dystrophic-myoblasts did not compromise the myogenic-differentiation of DPC in-vitro,supporting their in-vivo application and possible therapeutic-potential. The in-vitro effects of galectin-1, reversine and activation of the Shh signaling-pathway via. recombinant Shh and purmorphamine, on DPC myogenic-differentiation was also evaluated. None of the treatments increased myogenin-expression in DPC; but, triggering Shh-signaling produced a dose-dependent-pattern, whereby lower-levels of signaling promoted myogenic-differentiation while higher-levels inhibited it. Activating Shh-signaling upstream of Smo via. purmorphamine, induced a biphasic differentiative-response; however, the application of rShh hindered the differentiation of both cell types. Thus, murine-DPC are a readily-accessible-source of stem-cells that can undergo myogenic-differentiation in-vitro. We aim to improve their differentiation-efficacy to make them suitable-candidates for therapeutic-applications in muscle-wastingdisorders.

Biography:

Dr. Medet Jumabay has been serving as a doctor and researcher in the field of cardiovascular medicine for more than twenty years. Dr. Medet Jumabay has graduated from medical school and started her medical career at the Emergency Center of the First Affiliated Hospital of Xinjiang Medical University. Later she became an attendant physician and cardiologist in the department of medicine at the same university. Dr. Medet Jumabay received her Master’s degree from the Xinjiang Medical University and a PhD. degree from the Department of Medicine, Nihon University, Tokyo, Japan in medical science. Dr. Medet Jumabay first became fascinated by the power of cell plasticity when she saw beating cardiomyocytes, which are derived from adipose stem cells. Thus, she became interested in cell regeneration research immediately after completing her PhD. studies. In 2007, she was recruited to the Division of Cardiology, Department of Medicine, UCLA, as a researcher in the field of cardiovascular and stem cell biology.

Abstract:

The ability to identify an ideal cell source for tissue regeneration is still challenging researchers in the stem cell field. The possession of progeny cells to differentiate into other cell types is important for the processes of tissue reconstruction and tissue engineering and may have clinical, biochemical or molecular implications. Adaptation of stem cells from adipose tissue to use in regenerative medicine has created a new role for adipocytes. Mature adipocytes can easily be isolated from adipose cell suspensions and allowed to dedifferentiate into lipid-free multipotent cells, referred to as dedifferentiated fat (DFAT) cells. Compared with other adult stem cells, the DFAT cells have unique advantages in their abundance, ease of isolation and homogeneity. Under proper condition in vitro or in vivo, the DFAT cells have exhibited adipogenic, osteogenic, chondrogenic, cardiomyogenc, angiogenic, myogenic, and neurogenic potential. In addition, DFAT cells at early stage, spontaneously exhibit a high degree of pluripotency, which was responsive to changes in culture conditions and may benefit cell-based therapies. Thus, mature white adipocytes may be a new cell source for tissue regeneration. The unique features of DFAT cells are promising for clinical applications such as regenerative medicine.

Speaker
Biography:

Vincent S Gallicchio received his PhD in Hematology from New York University and his Diploma in Medicine from the University of Arad, Romania. He performed Postdoctoral studies at the University of Connecticut Medical Center and conducted his fellowship in Hematology from the Memorial Sloan Kettering Cancer Center. He has published more than 150 research articles in peer-reviewed journals on topics focused on Stem Cells and Hematology, seven books, many book chapters, and has been awarded eleven United States and one international patent for developmental therapeutics for AIDS and Cancer. He currently serves as a Director of Stem Cell Therapy for out-patient clinical use.

Abstract:

Lithium (Li) salts have been widely used in psychiatry as mood stabilizing agents for 60 years. Li found in variable amounts in foods, especially grains, vegetables, and in some areas, the drinking water provides a significant source of the element. Therefore, dietary intake in humans depends on location, type of foods consumed, and fluid intake. Traces of Li have been detected in humanorgans and tissues, leading to speculation that the element was responsible for specific functions in the human body. It was not until the 20th century that studies performed in the 1970’s and 1990’s, primarily in rats and goats, maintained on Li-deficient diets demonstrated higher mortality, altered reproductive and behavioural abnormalities. Such deficiencies have not been detected in humans; however, studies performed on populations living in areas with low Li levels in water supplies have been associated with higher rates of suicides, homicides, and the arrests rate for drug abuse and other crimes. Li appears to play a significant role in early fetal development as evidenced by high Li levels during the early gestational period. Biochemically, the mechanism of Li action involves multi-factor and interconnected pathways with enzymes, hormones, vitamins, and growth and transforming factors. Thisody of evidence now appears sufficient to label Li as an essential element with the recommended RDA for a 70 kg adult of 1000 mg/day. Of extreme importance for the future is the growing body of evidence indicating Li can be used effectively for the treatmentof acute brain injuries, e.g., ischemia and chronic neurodegenerative dis- eases such as Alzheimer’s disease, Parkinson’s disease, Tauopathies, and Huntington’s disease. This conclusion is based upon evidence showing Li as important in neurogenesis as well as protecting neurons from neurotoxicity. Li influences stem cells, both neuronal and marrow derived, thus, the additional therapeutic implications for this element in clinical medicine to treat disorders associated with the faulty production of blood and nerve cells or as a tool to enhance blood stem cell mobilization for transplantation are needed.

Biography:

Maliheh Nobakht is a full Professor of Medical Histology and Embryology in Iran University of Medical Sciences. She is a board certified Histologist and Embryologist and is engaged in education and is a basic Science Scientist known an Investigator at Iran University. She has expertise in wound healing and stem cell biology, scaffold and Neuroscience. She graduated from Tehran University in 1985, with a Bachelor of Science degree in Biology. Then, she received Master of Science degree on Histology and Embryology in 1987. She received her PhD degree from Tarbiat Modarres University on Histology in 1992. Subsequently, in 1994, she completed her Post-doctoral training in Molecular Biology and Electron Microscopy under supervision of Professor Leblond and Dr. Lee at McGill University and Shriner's Hospital in Montreal, Quebec, Canada. She became a full Professor in Histology Medicine in 2011 and at the same time, she was appointed to be the Co-director of the vice of education.

Abstract:

Introduction: The main aim of medicine is Wound healing in a shorter time and with fewer side effects. Cell therapy, especially treatment with stem cells has become as a possible solution for wound healing. In this study we evaluate the effect of hair follicle stem cells transplantation and SDF-1/CXCR4 axis in cutaneous wound healing in Rats. Materials and Methods: Animals (male Wistar rats) were divided into five groups: 1) Control (non-treated), 2) Vehicle (PBS), 3) AMD (treated with AMD), 4) HFSCs treated with AMD) , and 5) HFSCs (treated with HFSCs). (AMD is inhibitor of SDF-1/CXCR4 axis). The Bulge region of rat whiskers was isolated and cultured in DMEM/F12. Then transplanted to wound site. At the end of the treatment period, histological assessments (H&E, Masson's trichrome staining), and molecular assays (ELISA, q- PCR) were performed. Results: The diameter of epidermis, Amount of collagen, wound healing percent as well as expression of SDF-1, CXCR4 and VEGFR2 in HFSCs groups were significant compared with control group (P<0.05). In AMD group, these parameters were significantly low compared with control group (P<0.05). Conclusion: Transplantation of hair follicle stem cells had potential capability for cutaneous wound healing in Rats. However with increasing the SDF-1 expression level in in the injured area, HFSCs can recruit other stem cells and contribute to wound repair with
SDF-1/CXCR4 axis.

Biography:

Abstract:

Human bone morphogenetic protein 2 (hBMP2) through the other BMPs group family has been shown to have a significant role in regulating the odontogenic differentiation of dental tissue-derived stem cells. However, there are few studies on the effects of the BMP2 gene on the proliferation and odontogenic differentiation of human dental pulp stem cells (hDPSCs). DPSCs have many advantages in comparison to other types of dental stem cells including easy to access for isolation, high proliferation capacity, extensive capability to produce mineralized matrix and similarity to bone marrow MSCs phenotype. This study evaluated in vitro odontogenic differentiation potential of lentiviral-mediated BMP2 gene-transfected human DPSCs. DPSCs were isolated by enzymatic dissociation of primary and permanent teeth. The characteristics of DPSCs was evaluated by flow cytometry (FCM) then transduced with lentiviral vector to secretion of BMP2 along with expression level of green fluorescent protein (GFP). Transduced cells were analyzed for BMP2 secretion with RT PCR. Almost 70% of human dental pulp stem cells could express GFP marker following transduction. Graphical analysis and GFP assay confirmed BMP2 expression level in DPSCs transduced by recombinant lentiviral vectors was 6 times more than the native DPSCs. This result showed the induction odontogenic potential in DPSCs. In conclusion, DPSCs could be considered as one of the promising sources for tissue engineering including potential application for dental tissue, nerve and bone regeneration after tooth extraction or diseases and maxillofacial treatment failure. However, further studies should be performed to confirm this hypothesis. Also, lentiviral-mediated BMP2 gene transfection could enhance the in vitro odontogenic differentiation capacity of human DPSCs.

  • Stem Cells | Stem Cell Thearpy | Novel Stem Cell Technologies | Tissue Engineering | Gene Thearpy and Stem Cells
Biography:

Abstract:

The subgranular zone in the hippocampus and walls of the lateral ventricles are the established adult neurogenic niches in rat. Investigations on the occurrence of other adult neurogenic niches are being carried out in several laboratories. Recent studies from our laboratory showed that ependymal surface of the cerebral ventricles could be another neurogenic niche in the adult rat brain. Ependymal lining of cerebral ventricles lies at the interface between the ventricular cavities and the brain parenchyma. At several locations, at the ventricular surface, single and cluster of supraependymal structures consisting of neurons, axons and glial cells were observed. These supraependymal structures are immersed in the cerebrospinal fluid and enjoy a unique environment which is different from other regions of the mammalian brain. Transmission and scanning electron microscopic studies showed an extensive growth of the axons on the ventricular surface following axotomy of the supraependymal fibers. Interestigly, scanning electron microscopic studies showed a profound increase in the number of supraependymal neurons on the surface of the third ventricle following unilateral cervical sympathectomy. This increase was much larger at fifteen days following unilateral cervical sympathectomy as compared to that observed after seven days after unilateral cervical sympathectomy. From these studies, it can be concluded that supraependymal elements of the ventricular floor may represent a novel niche of neural stem cells in the adult mammalian brain that proliferate and differentiate in response to changes in sympathetic activity.

Biography:

Abstract:

Therapy resistance and inevitable post-treatment recurrence poses a persistant challenge for treating glioblastomas (GB). Recent advances in massively parallel sequencing have allowed to compare molecular landscapes between newly diagnosed and recurrent GBs. Previous studies have revealed that post-treatment recurrence in GBs is frequently accompanied by molecular drifts
manifest by considerable differences in the levels and spectrum of genomic aberrations, gene expression patterns and epigenetic profiles. The emergence of this fundamental trait necessitates in-depth investigations into the mechanisms that underlie changes in molecular patterns after (or during) therapy. The proposed project aims to elucidate the role of glioma stem cells implicated as a key driver of post-treatment recurrence in gliomas and the most clinically relevant type of glioma cells possessing both an inherent and acquired cytotoxic resistance. Up till now, genome-wide comparisons between untreated and recurrent GB have been exclusively performed on heterogeneous GB tissues with undefined proportions of glioma stem cells. In this study, a comparative analysis of gene expression landscapes was conducted in glioma stem cells isolated from patient-matched newly diagnosed and recurrent GB or
by using experimental models of radio/chemoresistant glioma stem cells. The main novelty of the proposed project lies in its focus on the specific contribution of glioma stem cells to GB recurrence under cytotoxic therapy. To our knowledge, this is the first study to address individual impacts of clinically relevant regimens of radiation and chemotherapy on gene expression patterns in glioma stem cells.

Biography:

Abstract:

Pancreatic β-cells are the predominant insulin producing cell types, within the Islets of Langerhans and insulin is the primary hormone which regulates carbohydrate and fat metabolism. Apoptosis of β-cells or insufficient insulin production leads to Diabetes Miletus (DM). Current therapy for diabetes includes either medical management or insulin replacement. Cell based therapies are being developed as a possible therapeutic option for Diabetes. Replacement of β- cells is an attractive treatment option for both Type-1 and Type-2 DM in view of the recent paper which indicates that β-cell apoptosis is the common underlying cause for both types of DM. In the present study, Human Umbilical Cord (HUC) samples were obtained after delivery with prior informed consent as approved by the Institutional Ethical Committee. Mesenchymal Stem Cells (MSCs) were successfully isolated from HUC (n=26)using a combination of mechanical and enzymatic, Collagenase-II treatment. Culturing in-vitro was done in L-DMEM with 10% FBS and 5% CO2 at 37oC. Cultured cells were characterized as MSC with flow-cytometry after reaching 80-90% confluency as they were CD90+, CD73+, CD105+, CD34-, CD45, HLA-DR-/Low and vimentin+ (Immunohistochemistry). These were differentiated to β-cells in 15 days using H-DMEM (Gibco), β-Mercaptoethanol (0.1mM, HiMedia), basic-fibroblast growth factor (10μg/L, Gibco) and Nicotinamide (10mmol/L HiMedia). Differentiated cells stained positive for Dithizone, a specific marker for pancreatic beta cells and their functionality was evaluated by insulin production with glucose stimulation (50mM/L). Amount of insulin released was four-fold higher when stimulated (8 IU/ml) compared with unstimulated (2 IU/ml) this is extremely promising for β-cell replacement therapy in diabetics.

Biography:

Maliheh Nobakht is a full Professor of Medical Histology and Embryology in Iran University of Medical Sciences. She is a board certified Histologist and Embryologist and is engaged in education and is a basic Science Scientist known an Investigator at Iran University. She has expertise in wound healing and stem cell biology, scaffold and Neuroscience. She graduated from Tehran University in 1985, with a Bachelor of Science degree in Biology. Then, she received Master of Science degree on Histology and Embryology in 1987. She received her PhD degree from Tarbiat Modarres University on Histology in 1992. Subsequently, in 1994, she completed her Post-doctoral training in Molecular Biology and Electron Microscopy under supervision of Professor Leblond and Dr. Lee at McGill University and Shriner's Hospital in Montreal, Quebec, Canada. She became a full Professor in Histology Medicine in 2011 and at the same time, she was appointed to be the Co-director of the vice of education.

Abstract:

Background: Tissue engineering is a new approach to reconstruction and/or regeneration of lost or damaged tissue. The purpose of this study was to fabricate the polycaprolactone (PCL) random nanofiber scaffold as well as evaluation of the cell viability, adhesion, and proliferation of rat nestin‑positive hair follicle stem cells (HFSCs) in the graft material using electrospun PCL nanofiber scaffold in regeneration medicine. Materials & Methods: The bulge HFSCs was isolated from rat whiskers and cultivated in Dulbecco’s modified Eagle’s medium/F12. To evaluate the biological nature of the bulge stem cells, flow cytometry using nestin, CD34 and K15 antibodies was performed.
Electrospinning was used for the production of PCL nanofiber scaffolds. Furthermore, scanning electron microscopy (SEM) for HFSCs attachment, infiltration, and morphology, 3‑(4, 5‑di‑methylthiazol‑2‑yl)‑2, 5‑diphenyltetrazolium bromide (MTT) assay for cell viability and cytotoxicity, tensile strength of the scaffolds mesh and histology analysis were used.
 
Results: Flow cytometry showed that HFSCs were nestin and CD34 positive but K15 negative. The results of the MTT assay showed cell viability and cell proliferation of the HFSCs on PCL nanofiber scaffolds. SEM microscopy photographs indicated that HFSCs are attached and spread on PCL nanofiber scaffolds. Furthermore, tensile strength of the scaffolds mesh was measured.
Conclusion: The results of this study revealed that modified PCL nanofiber scaffolds are suitable for HFSCs seeding attachment and proliferation. Furthermore, HFSCs are attached and proliferated on PCL nanofibers scaffolds.

Biography:

Mohamed Gamal El-Beltagy fifth year student at Faculty of Dentistry, Mansoura University, Egypt. I works as a Research Assistant in Mansoura Medical Research Center (MERC) in Faculty of Medicine, Mansoura University and I am a Director on a project entitled “Dental Pulp Stem Cell Banking” funded by the Ministry of Higher Education in Egypt under supervision of Professor Youssry elhawary I have an abstract conference publication entitled: Dental pulp stem cell isolation, tissue culture, characterization and cryopreservation from human tooth published Journal of Stem Cell Research & Therapy at 7th Annual Conference on Stem Cell and Regenerative Medicine August 04-05, 2016 Manchester, UK also I won the best poster award at the conference.

Abstract:

Human dental pulp represents a suitable alternative source of stem cells for the purpose of cell-based therapies in regenerative medicine. Spinal cord injuries at the cervical (neck) level result in full or partial tetraplegia ,low heart rate, low blood pressure, problems in regulating body temperature, and breathing dysfunction. We aim to recommend the best cell population and the best cell transplant criteria to treat cervical spinal cord injury. In this study we successfully isolate CD 24 positive cells from human dental apical papilla from impacted third molar of patient female 15 years using two different techniques enzymatic digestion using Collagenase 3 mg/ml / Dispase 4 mg/ml enzymes also with explant outgrowth method. Characterization done by using flowcytometry cell sorting and we confirm the viability of the sorted cells using trypan blue stain. Interestingly, despite the sorted cells was express
a limited present in the cell population only 5.9% but after culturing in Proliferation Media (PM): DMEM/F12 supplemented with 10% FBS, 100 μM L-ascorbic acid 2-phosphate, 2 mM L-glutamine, 100 units/ml penicillin, 100 mg/ml streptomycin, they give a high proliferation rate. The teeth of the study opened using Hand-held dental pulp isolator device Patent (WO 2016162041 A9). Based on the role of ENTPD1 gene in development of central nervous system during embryogenesis, hydrolyze ATP and other nucleotides to regulate purinergic neurotransmission, anti-inflammatory effect TNF-α-induced cytotoxicity and cell death also its high expression in dental derived stem cells so in the next steps we will compare between seven sub- populations CD24, CD39 (ENTPD1), CD90, CD73, CD105, Nestin and SSEA-3 positive cells from different dental sources as apical papilla, dental follicle and dental pulp of
human tooth. Moreover we will compare between whether cell transplant or whole tissue transplant of the dental pulp in spinal cord injury in sprague dawely rats is more effective. Regarding tissue transplant we will compare between graphene nano-scaffold with and without hydrogel, carbon nano-tubes and high or low porosity PCL Poly (ε-caprolactone) scaffolds and test the Adhesion, Cytotoxicity, Proliferation and neuron-differentiation capacities of the cells or the dental pulp tissue.

Biography:

Mohammadhadi Fartookzade has MSc degree in MBA and BSc degree in Microbiology. He is the Technician in Electron Microscope Unit. He is working in Iran University of Medical Sciences.

Abstract:

Neural stem cells (NSCs) as a heterogeneous multi-potent and self-renewal population find in the different areas in the developing mammalian nervous system, as well as the sub-ventricular zone (SVZ) and the hippocampus of the adult brain. NSCs can give rise to neurons, astrocytes and oligodendrocytes. Much evidence has accumulated for interactions between the noradrenergic-LC
(NA-LC) system and structures involved in sleep regulation. Locus Ceoruleus (LC) plays an important role in the sleep-wake cycle. Noradrenergic cells in LC participate in the process of cortical activation and behavioral arousal. 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 in the rat. Fourty-two adult male Wistar rats weighing between 250-275 g, obtained from Pasteur Institute of Tehran, were used as subjects. The 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. The cells were cultured in DMEM F12, B-27 supplemented with 20ng/ml (hFGF) and 20ng/ml (EGF) for 2 weeks. NSCs were differentiated in neurobasal medium, B-27 supplemented with BDNF (50 ng/ml) and GDNF (30 ng/ml) for 3 and 5 days. The animals received bilateral 6-hydroxydopamine (6-OHDA) [2μg/0.5μl in 0.1% ascorbic acid and 0.9% saline solution] lesion of the LC. For sleep-wake recording 3 EEG and 2 EMG electrodes were implanted respectively in the skull and dorsal neck muscle. After 7 weeks, following being anesthetized, brains were cut in 7μm serial sections and stained with cresyl violet. Cavity volume was evaluated through the stereological technique. In this study Nestin and Sox2 were expressed in
NSCs and neurospheres. NSCs were differentiated into noradrenergic-like cells (NACs) and Tyrosine hydroxylase was detected in these cells. Cavity volume caused by lesion was restricted to LC. A significant decrease (P≤0.05) was seen in NREM (Non Rapid Eye Movement) and PS (Paradoxical Sleep) stages and a significant increase (P≤0.05) were seen in wake and PS-A (Paradoxical Sleep without Atonia) in lesion group in comparison with control and sham groups. There was not significant differences in Wake, NREM, PS, PS-A between experimental groups. NSCs transplantation in experimental groups prevented of decrease in PS and increase in PS-A, So that a significant increase in PS and a significant decrease in PS-A in comparison with lesion group were seen in experimental groups (P≤0.05). The results of this study demonstrate NSCs have ability to differentiate into noradrenergic cells using of BDNF and
GDNF growth factors and NSCs transplantation improved disruption of the sleep-wake cycle after bilateral lesion of LC.