conferenceseries.com 1271st Conference

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Scientific Tracks & Abstracts Day 1

Regenerative Medicine 2017

Page 27 Sessions: Day 1 October 02, 2017 Scaffolds in Regenerative Medicine|Stem Cells|Stem Treatments|Tissue Repair and |Tissue Engineering| Regenerative Medicine

Session Chair Session Co-Chair Pedro G Morouço Barbara Gawronska-Kozak Polytechnic Institute of Leiria, Portugal Institute of Animal Reproduction and Food Research, Poland

Session Introduction Session Introduction Title: Concerted interactions of and in promoting Ing-Ming Chiu, National Health Research Institutes, Taiwan Title: Peripheral regeneration using artificial nerve sheets composed of freeze-dried alginate gel Yoshihisa Suzuki, Shiga University of Medical Science, Japan Title: Response of post-ischemic heart to stem cells treatment: A case study Domingo Guerra, La Vega, Dominican Republic Title: From repair to regeneration – transcription factor FOXN1 directs the skin wound healing processes Barbara Gawronska-Kozak, Institute of Animal Reproduction and Food Research, Poland Title: Complete healing of a non-healing diabetic foot ulcer within one month and persisting for three years using autologous adipose tissue-derived Stromal Vascular Fraction and Platelet Rich Plasma Triantafyllos Didangelos, Diabetes Center, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Greece Title: Implant of autologous bone marrow mother stem cells in patients with congestive heart failure, functional class IV Fernandez Viña Matias, Clinica San Nicolás, Argentina Title: Behaviour of human mesenchymal-periosteal cells grown in contact with different scaffolds used in dentistry Gabriele Ceccarelli, University of Pavia, Italy Title: Translating repair into regeneration in wounds with PRP Sandeep Shrivastava, Datta Meghe Institute of Medical Sciences, India Title: The effect of polyglycerol sulfate-based hydrogels with tunable mechanical integrity on cartilage regeneration in osteoarthritis Shabnam Hemmati-Sadeghi, Free University of Berlin, Germany

Regenerative Medicine 2017

Page 28 Ing-Ming Chiu, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Concerted interactions of neurogenesis and myelinogenesis in promoting neuroregeneration Ing-Ming Chiu National Health Research Institutes, Taiwan

egeneration of injured peripheral and is a slow and complicated process that could be improved by Rimplantation of neural stem cells (NSCs) or nerve conduit. We previously showed that implantation of NSCs along with conduits promotes the regeneration of damaged nerve. The improvement is likely due to conduit supports and guides axonal growth from the proximal nerve stump to the distal one, while preventing fibrous tissue ingrowth and retaining ; and implanted NSCs differentiate into Schwann cells and maintain a -enriched microenvironment, which promotes nerve regeneration. We identified IL12p80 (homodimer of IL12p40) in the cell extracts of implanted nerve conduit combined with NSCs by using protein antibody array and western blotting analyses. Levels of IL12p80 in these conduits are 1.6-fold higher than those in conduits without NSCs. In the sciatic mouse model, implantation of NSCs combined with nerve conduit and IL12p80 improves motor recovery and increases the diameter up to 4.5-fold, at the medial site of the regenerated nerve. In vitro studies further revealed that IL12p80 stimulates the differentiation of mouse NSCs. Moreover, the cellular differentiation is enhanced through phosphorylation of signal transducer and activator of transcription 3 (Stat3). Our results suggest that IL12p80 could trigger Schwann cell differentiation of NSCs through Stat3 phosphorylation. Differentiation of myelinating Schwann cells increases the diameter of regenerated nerves and, in turn, enhances the functional recovery in a mouse sciatic nerve injury model.

Biography Ing-Ming Chiu is currently a Distinguished Investigator and Professor at the National Health Research Institutes in Taiwan. He is the Group Leader of a multidisciplinary research group in Stem Cell and Regenerative Medicine. He has earned his PhD in Biochemistry at Florida State University. He did Post-Doctoral training in National Cancer Institute in Bethesda, Maryland before joining as the Faculty in The Ohio State University in Columbus, Ohio in 1986. He served as the Director of the Brain Tumor Gene Therapy Program at The Ohio State University. In the National Health Research Institutes, his group developed a method to combine the use of nerve conduits, FGF1 and neural stem cells in the repair of sciatic nerve injury. He has published more than 140 papers and have received 14 patents.

[email protected]

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 29 Yoshihisa Suzuki, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Peripheral nerve regeneration using artificial nerve sheets composed of freeze-dried alginate gel Yoshihisa Suzuki Shiga University of Medical Science, Japan

ong injury gaps of the peripheral nerve are treated by autologous nerve transplantation, although sequelae, such as pain Land numbness, remain at the donor sites of nerves for transplantation. To solve this problem, tubular artificial nerves were developed. However, they have several drawbacks, such as the need for several types of materials with different diameters due to the tube structure, prolonged surgery time because of the need for suture, and inapplicability to the nerve branch and plexus because of the linear structure. Thus, we have initiated the development of products to overcome the above drawbacks, which can produce plant-derived alginate. Our new concept of nerve regeneration materials is to process a freeze-dried sponge into a sheet rather than a tube structure using sodium alginate as a covalently crosslinked gel. Its applicability to defects in the branched nerve was tested using rats. Nerve defect was made in the branched site from the sciatic nerve to the peroneal and the tibial nerves. Eight weeks after operation, regenerated were observed in both the peroneal and tibial nerves. The nerve axons were elongated and Schwann cells migrated in low-molecular-weight alginate after biodegradation. It can be considered that alginate gel is a potent material for promoting peripheral nerve regeneration at branched site, and that the non-tubular method is a promising approach for the repair of the peripheral nerve.

Biography Yoshihisa Suzuki obtained his MD and PhD degrees from Kyoto University, Faculty of Medicine, Kyoto, Japan during 1980-1986. Later, he joined Kyoto University, Faculty of Medicine, Plastic Surgery Department as a Staff Member in June, 1986. In May 1987, he joined Osaka Red Cross Hospital as a Staff Member. He later held various positions as Staff Member (1990.6-1998.1), Assistant Professor (1998.2-1999.12) and Associate Professor (2000.1-2006.6) at Kyoto University, Faculty of Medicine, Plastic Surgery Department. From July 2006 to present, he is the Director, Department of Plastic Surgery at Kitano Hospital, Osaka. He is also a Visiting Professor, Department of Stem Cell Biology and Regenerative Medicine and Specially Appointed Professor, Department of Plastic Surgery at Shiga University of Medical Science since 2015.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 30 Domingo Guerra et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-040

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Response of post-ischemic heart to stem cells treatment: a case study Domingo Guerra1 and Fiorella C Guerra2 1La Vega, Dominican Republic 2Policlinico La Vega, Dominican Republic

ase of a 68 years old male with past medical history of hypertension, ischemic cardiopathy, acute myocardial infarction C(AMI) 8 years ago, with stent placement; cardiac arrest (seven times after AMI) with pacemaker placement, and ischemic stroke (15 days after AMI) which resulted in complete left paralysis managed at the moment with fibrinolytic therapy, and later physical therapy. During this period of time, patient was under medication with Digoxin, Spironolactone, Ramipril and Potassium Supplement. Patient was received in our Regenerative medicine Unit on November 2015 presenting with fatigue, Ejection fraction (EF) of 30-35% (Figure 1.A), limitation of physical activity, erectile disfunction, left side motor disfunction, mild effort dyspnea, severe fatigue and not elective for stress test. At that moment patient received regenerative medicine therapy with bone marrow stem cells (activated), Platelet Rich Plasma, Cytokines, and Growth Hormone. A second session of treatment was performed two months after, same procedure and same products were given. Patient was now elective for a Stress Test with was performed on February 2016, test concluded negative for ischemic cardiopathy, with adequate cronotropic and pressure response, and exercise tolerance within regular range. Follow up of the patient demonstrated improve on fatigue, erectile disfunction, and a current ejection fraction of 42% (Figure 1.D), motor amelioration, patient now refers overall improvement. Digoxin and Spironolactone were removed by cardiologist from treatment, and is now been treated with Eiquinon and potassium supplement.

Biography Domingo Guerra is a Medical Doctor with a General Surgery specialty, and has Post-graduate fellowships of Advanced, Laser and Laparoscopic Surgery. In 1998, he became a Founder and Board Member (for 2 years) of the Policlinico La Vega SA in Dominican Republic. A few years later, in 2002, he started within this institution one of the first Regenerative Medicine units of the country. He has been practicing this field of medicine during the past 15 years, introducing to the country new therapeutic options. He has recently completed a Cell Culture (Stem Cell Oriented) workshop. By combining surgical knowledge with regenerative medicine knowledge, he has been able to apply this emerging field of the medicine to several types of diseases such as Diabetes, Hypertension, Renal Failure, Heart failure, Alzheimer’s, Parkinson’s, medullary problems (such as compression), joint problems (such as arthritis), inflammatory and autoimmune diseases, among others. He is now reorienting his work into research in order to document the work he has done. [email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 31 Barbara Gawronska-Kozak, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

From repair to regeneration – transcription factor FOXN1 directs the skin wound healing processes Barbara Gawronska-Kozak Institute of Animal Reproduction and Food Research, Poland

kin wound healing in mammals can be resolved through the common process of reparation (scar-forming) or sporadically Sobserved regeneration (scar-free). Regeneration is characterised by lack of scarring and reconstruction of normal tissues architecture and function. However, to date no mechanism has been proposed that allows the transformation of reparative healing into regeneration. We showed that the skin of nude mice, which is deficient in the transcriptional factor FOXN1, is capable of remarkable scar-free healing similar to mammalian fetuses. Interestingly, in both mammalian fetuses and nude mice lack in their skin FOXN1 activity coincide with regenerative wound healing. Next-generation high-throughout DNA sequencing data analysis comparing the uninjured skin of mouse fetuses during regenerative period (14th day of embryonic development; FOXN1 non-active) and adult nude mice (FOXN1-deficient) revealed the similarities in transcriptomic signature that predisposes them to regenerative skin healing. FOXN1 activity appears to be an essential condition to establish the adult skin phenotype and a key component of skin maturation. Analysis of post-injured skin from FOXN1::Egfp transgenic mice showed an intense FOXN1-eGFP signal at the wound margin and in the leading epithelial tongue, where it co-localized with keratin 16 and Mmp-9. Moreover, high levels of Snail1 and Mmp-9 expression, co-localization of vimentin/E-cadherin- positive cells and myofibroblast marker (αSMA) in dermis revealed the involvement of FOXN1-positive keratinocytes in an epithelial-mesenchymal transition (EMT). Together, our findings indicate that (i) FOXN1 inactivity in nude mice creates pro-regenerative conditions, (ii) FOXN1 is a potent factor in reparative (scar-forming) wound healing through engagement in re-epithelization and the EMT process.

Biography Barbara Gawronska-Kozak received her PhD from University of Warmia and Mazury in Olsztyn, Poland and her habilitation from Medical University in Bialystok, Poland. From 2005 to 2011, she worked as an Assistant Professor and Head of the Regenerative Biology Laboratory at Pennington Biomedical Research Center, Baton Rouge, USA. Currently she is Associate Professor at the Polish Academy of Sciences in Olsztyn, Poland and Leader of Regenerative Biology Team. She published more than 60 papers in reputed journals. Her study focuses on skin wound healing, adult stem cells and obesity.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 32 Triantafyllos Didangelos et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Complete healing of a non-healing diabetic foot ulcer within one month and persisting for three years using autologous adipose tissue-derived Stromal Vascular Fraction and Platelet Rich Plasma Triantafyllos Didangelos1, Georgios Koliakos2, Kokkona Kouzi-Koliakou3, Stamata eorga4, Katerina Kotzampassi5, Dimitrios Karamanos6, Konstantinos Tziomalos1, Georgios Arsos4 and Apostolos I. Hatzitolios1 1Diabetes Center, First Propedeutic Department of Internal Medicine, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Greece, 2Department of Biochemistry, Medical School, Aristotle University of Thessaloniki, Greece, 3Department of Histology and Embryology, Medical School, Aristotle University of Thessaloniki, Greece 4Laboratory of Nuclear Medicine, Papageorgiou Hospital, Medical School, Aristotle University of Thessaloniki, Greece 5First Propedeutic Department of Surgery, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Greece 6First Department of Surgery, Papageorgiou Hospital, Medical School, Aristotle University of Thessaloniki, Greece

he aim of the present study was to investigate the efficacy of the adipocyte tissue-derived Stromal Vascular Fraction (SVF) Tand Platelet Rich Plasma in a non-healing diabetic ulcer. A woman with Diabetes Mellitus (DM) type 1, aged 47 years old, with duration of DM 24 years, presented for first time two years ago, with a non-healing ulcer in the plantar surface of the right foot under the second metatarsal head. Osteomyelitis observed after the first visit of the patient in our outpatient clinic with Magnetic Resonance Imaging. The patient received treatment with antibiotics for six months and at the end of the treatment, the labelled white blood cells scan was normal. But, there was no evidence of closure of the ulcer for another six months, despite our efforts. So, we decided to apply autologus adipocyte tissue derived- SVFs to the site of the ulcer. A piece of about 60ml adipose tissue was collected by lipectomy performed by a surgeon from the abdominal subcutaneous area under local anesthesia. For the preparation of adipose tissue derived SVF and the method of collagenase digestion was applied. The cells were then rate controlled frozen and stored in liquid nitrogen until use. Before application the cells were rapidly defrozen at 40oC washed twice with PBS, and resuspended in 2mL of the patient’s serum. Injection of the SVFs in the ulcer site made in April 2013. After a month of the SVFs injection there was complete closure of the ulcer with normal dermal appearance. No side effects observed during the follow up period. After 3 years of follow up there is still complete healing of the ulcer. In conclusion, administration of autologus adipocyte tissue derived-Stromal Vascular Fraction and Platelet Rich Plasma achieved complete healing of the diabetic refractory to other treatment ulcer. This intervention may be helpful for other same cases in future.

Biography Prof. Triantafyllos Didangelos has completed his PhD at the age of 39 years from Aristotle University of Thessaloniki, Greece, and postdoctoral studies from Aristotle University of Thessaloniki, School of Medicine. He is the Head of Diabetes Center, First Propedeutic Department of Internal Medicine, AHEPA Hospital, Medical School, Aristotle University of Thessaloniki, Greece a premier outpatient clinic. He has published more than 50 papers in reputed journals and has been serving as an editorial board member of repute.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 33 Fernandez Viña Matias et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Implant of autologous bone marrow mother stem cells in patients with congestive heart failure, functional class IV Fernandez Viña Matias, Fernández Viña Roberto, Martínez San Juan Federico, Fernández Viña Federico, Camozzi Liliana and Saslavsky Jorge Clinica San Nicolás, Argentina

Objectives/Goals: The goals of the study are to: 1) Demonstrate that CD34+/CD38(-) adult stem cells in myocardial tissue generate a significant increase in the ejection fraction after implantation. 2) Observe the improvement in the quality of life of the implanted patients, evaluating the reduction of dyspnea and the number of hospitalizations. Material & Methods: A descriptive, observational study of 71 and 88-year-old patients with a history of CF IV heart failure with severe deteriorated FEY (<30%) who suffered hospitalizations every 15 days, due to biventricular dysfunction. They were submitted to implantation of bone marrow Stem Cells through retrograde venous technique in coronary sinus with balloon occlusion by femoral catheterization. A pre / post implant echocardiogram was indicated for comparisons (7 months) and a post-implant effort ergometry test was performed. Results: After a period of 210 days, a significant improvement in the ejection fraction of approximately 8% and 14% was observed with respect to the baseline (P:0.05), and a decrease in both hearts mass of 20% (grams). In addition, it was observed that the patients did not have recurrence in hospital admissions successively every 15-20 days, cause congestive heart failure and that there is a remarkable improvement in the quality of life rapidly after the implantation of stem cells. The post-implant ergometry test, at 210 days, turned out to be encouraging, since the patient tolerated the stress test greater than 6 minutes. Conclusion: The implantation of mother stem cells generated a favorable decrease in the number of hospital readmissions of the evaluated patients and showed improvement of their quality of life after no dyspnea CF IV. It was corroborated that there is a significant improvement of the ejection fraction, with decrease of the mass of implanted organ.

Biography Fernandez Viña Matias was educated at School of Medicine, Universidad Abierta Interamericana Rosario in 2014. He is a part of staff of Cardiology at Clinic San Nicolas and Professor at Universidad Abierta Interamericana.

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 34 Gabriele Ceccarelli et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Behaviour of human mesenchymal-periosteal cells grown in contact with different scaffolds used in dentistry Gabriele Ceccarelli, Rossella Presta, Laura Benedetti, Flavio Ronzoni, Saturnino Marco Lupi and Maria Gabriella Cusella De Angelis University of Pavia, Italy

one regeneration is currently one of the most important challenges for regenerative medicine and it is considered an ideal clinical Bstrategy in dentistry. Bone resorption of alveolar crest occurring after tooth extraction leads to several risks for future treatments, including dental implants. For this reason, alveolar ridge preservation (ARP) has become a key component of contemporary clinical dentistry. The main aim is to keep the shape and the size of the bone socket after tooth extraction allowing bone healing and installation of endosseous implants in a later time. For this reason, different types of organic (such as collagen) or inorganic biomaterials such as manufactured polymers (polyglycolic acid - PGA, polylactic acid - PLA, and polycaprolactone) are used in surgical techniques for dental defects. The goal of this study was to compare different scaffolds based on PLGA (Fisiograft®), PLGA + Hydroxyapatite (Alos®) and type-I Collagen (Sombrero®) in an in vitro model of Tissue Engineering. Human MSC cells derived from periosteum were cultured and characterized by FACS for the expression of mesenchymal antigens (CD90, CD105, CD29). Cells were successively seeded on scaffolds and, at different time points, adhesion and cell differentiation were analyzed by morphological and molecular biology analysis. Results showed the proliferation induction of Sombrero® material and the osteoconductive potential of PLGA+HA, as demonstrated by molecular biology analysis on osteogenic genes and proteins. Further studies will be focused on combined scaffolds, that matched the resorbable capability of PLGA-HA and the capacity of organic collagen to promote initial engraftment and cell proliferation.

Biography Gabriele Ceccarelli is a Post-doc Researcher at the University of Pavia, at the Department of Public Health, Experimental Medicine and Forensic, Human Anatomy section Unit. He has obtained his PhD in Bioengineering and Bioinformatics (2011) and he is involved in several Tissue Engineering and Regenerative Medicine research projects. His current research interests include stem cells for skin and bone repair. He has published more than 20 papers in reputed journals.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 35 Sandeep Shrivastava et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Translating repair into regeneration in wounds with PRP Sandeep Shrivastava, Deepti Shrivastava, C Mahakalkar and Anurag Gupta Datta Meghe Institute of Medical Sciences, India

t has never been possible to revive a dead/dying tissue in wounds including skin, muscles, tendons and bones. Rather they have Ialways been considered as threat towards conducive environment for wound healing specially towards infection. Hence during managements they are urgently excised, to enhance wound repairs. The recent advances in the regenerative medicine open a window of opportunity to translate the wound management. The author has developed a platelet rich plasma LED “STARS therapy” for management of wounds. With this management, it has been possible to revive these tissues from gross gangrenous/pre-gangrenous conditions, with minimum of further losses. A total of 162 wounds have been treated by STARS therapy. Out of these, 51 wounds had a dead/dying tissue including skin flaps, tendons, muscles, and bones. These were grossly identified by their clinical appearance such as sloughing, necrosis and blackening. These wounds are included in the study and results of such wounds towards tissue regeneration are studied. A few of them (randomly selected) also underwent histopathological examination at different stages of wound healing. The results are clinically evaluated in terms of four progressive stages: arrest of further necrosis and infections; appearance of regeneration (pink/red speckles in the tissue); absorption/spontaneous removal of blackened tissue and complete repair of wound. The neo-angiogenesis induced by PRP through its different growth factors have helped towards tissue regeneration and complete wound repairs. In this clinical study, we demonstrate the efficacy of such PRP LED treatment in wound care. The STARS therapy as developed and applied for wound healing has immense potentials to be a game changer for wound management and tissue regeneration. It is easily reproducible, safe and cost effective. This outcome could be the regenerative medicine’s finest application to a large clinical health care problem. This is perhaps the most awaited and turning point in history of wound healing, from era of local applications, drugs and devices to exciting era of regeneration.

Biography Sandeep Shrivastava is an Orthopaedic Surgeon. He has completed his MS, DNB and PhD from India. He is currently Dean of J N Medical College, Datta Meghe Institute of Medical Sciences. He is then Chairman to Limb Reconstruction and Deformity Clinic and Wound Care Clinic. He has 56 publications, gave 64 international lectures across the globe and 2 orations. He has 6 copyrights and is Developer of STARS therapy. He is in Editorial Board of Journal of DMIMSU, J Journal of Orthopaedics and Allied Sciences, IJ RJSMS and NJMR.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 36 Shabnam Hemmati-Sadeghi, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

The effect of polyglycerol sulfate-based hydrogels with tunable mechanical integrity on cartilage regeneration in osteoarthritis Shabnam Hemmati-Sadeghi Free University of Berlin, Germany

he current therapeutic approaches do not halt osteoarthritis (OA) progression or reverse joint damage caused by it. TFurthermore, hyaluronic acid (HA), a standard visco-supplement, injected for pain management in OA patients, has a rapid clearance. Our group has synthesized non-degradable hydrogels from a heparin-analogous polymer dendritic polyglycerol sulfate (dPGS), which can be tuned with respect to its rheological properties and has anti-inflammatory effects. In this study, we further characterized this hydrogel in our OA in vitro model to test its effect on OA regeneration. Importantly, several concentrations from 3.6 to 4.8 wt% of dPGS and, as standard visco-supplement, blends of commercially available HAs were investigated to find out a suitable concentration for intra-articular injections which mimic HA in terms of viscoelastic and mechanical properties. To test the dPGS potential for OA-treatment, recombinant porcine tumor necrosis factor alpha (TNF-α) was used to induce OA-like changes. To document ECM formation, cartilage-typical-sulfated glycosaminoglycans (GAG) were stained with Safranin O and cartilage-specific type II collagen was detected immunohistochemically. The rheological measurements were performed with a temperature-controlled Bohlin Gemini 200 HR nano rheometer. From oscillatory measurement data, storage (G') and loss (G'') modulus were deduced as function of the oscillating frequency ω. Our results show that dPGS prevents TNF-α induced GAG loss and therefore can play a role in cartilage regeneration. To further investigate the mechanism behind this protective effect of dPGS, these samples are currently under analysis by microarrays. Furthermore, in our rheological experiments, we found that the 4.0 wt% dPGS had comparable viscoelastic properties to HA. These findings suggest that dPGS hydrogels have similar mechanical properties, but might have an advantage in control of inflammation and of being much less easily disappear from its injection place.

Biography Shabnam Hemmati-Sadeghi has completed her Master’s degree from Ferdowsi University, Mashhad, Iran and she is currently a PhD student at Free University of Berlin, Germany. [email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 37 conferenceseries.com 1271st Conference

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Scientific Tracks & Abstracts Day 2

Regenerative Medicine 2017

Page 42 Sessions: Day 2 October 03, 2017 Regenerative Medicine|Regenerative Medicine | Models of Regeneration | Cell and Organ Regeneration | Regenerative Medicine Market

Session Chair Session Co-Chair Brian M Mehling Fumio Arai Blue Horizon International, USA Kyushu University, Japan

SessionSession Introduction Introduction Title: Notch-mediated conversion of activated T cells into stem cell -like T cells for adoptive immunotherapy Akihiko Yoshimura, Keio University School of Medicine, Japan Title: Identification of novel mesenchymal stromal cells that have the potential to support hematopoietic stem cell activity Fumio Arai, Kyushu University, Japan Title: Nanochannel-Based Electroporation Assisted Tissue Reprograming and Repair Durba Pal, Indian Institute of Technology Ropar, India Title: Comparative study between two ex vivo cell expansion models of CD34+ cells from cryopreserved umbilical cord blood co-cultured with encapsulated mesenchymal stem cells Avila-Portillo LuzM, CryoHoldco, Colombia

Regenerative Medicine 2017

Page 43 Akihiko Yoshimura, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Mesenchymal stem cells drive cell repopulation in an in vivo model of lung regeneration Akihiko Yoshimura Keio University School of Medicine, Japan

Rationale: The use of bone marrow mesenchymal stem cells (B-MSCs) to promote the recruitment of endogenous cells to a decellularized scaffold provides a novel approach for the generation of a functional organ for clinical use. Lung transplantation remains the only accepted treatment for end-stage lung, diseases however, long wait list times and scarcity of acceptable donor organs result in nearly 400,000 deaths per year for patients awaiting transplant. These reasons underscore the need for novel approaches to increase the number of organs suitable for transplant. Methods: Lungs from C57BL/6 wild type mice were decellularized in situ by perfusion of the pulmonary vasculature. In short, the pulmonary artery was cannulated through the right ventricle and the vasculature perfused with PBS, water and SDS. The matrix was then seeded with GFP B-MSCs and heterotopically transplanted into the dorsum of wild type mice for 1 month. Lungs containing DMEM were implanted on the opposite side of the dorsum of the same mouse to serve as an internal control. Alternatively, lungs were seeded with fibroblasts and placed in the dorsum of mice as a positive control. Revascularization of implanted lungs was imaged using two-photon microscopy prior to tissue retrieval. To determine the cellular makeup of the decellularized tissue, histological and immunofluorescent staining, qPCR and flow cytometry were used. Results: Lungs seeded with GFP B-MSCs and heterotopically placed in recipient mice exhibited macroscopic re-vascularization confirmed by two-photon microscopy compared to control lungs. Markers for CD45, CD4, CD8, CD19, GR1, CD11b, Cd73, CD44, CD106, Ter119, Cd31 and cytokeratin were used in both IF and flow cytometry to confirm the presence of endothelial, epithelial and smooth muscle as well as immune cells in lungs seeded with GFP B-MSCs compared to control lungs. A lack of co-localized GFP signal with cells indicates cells where recruited to the matrix from the recipient mouse, not differentiated GFP B-MSCs. Conclusions: These results indicate that decellularized lung matrix seeded with B-MSCs, serves as a viable scaffold for the recruitment of specific types of cells that will generate a functional and viable organ for transplant. Lack of co-localization of the GFP signal with cell markers and flow cytometry data indicate that repopulation of the decellularized matrix is by mesenchymal stem cell mediated recruitment of endogenous cells. Further studies are needed to interrogate the signaling pathways involved in this process.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 44 Fumio Arai, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Identification of novel mesenchymal stromal cells that have the potential to support hematopoietic stem cell activity Fumio Arai Kyushu University, Japan

nteraction of stem cells with their supportive microenvironment “niche” facilitate the signaling networks that control the Ibalance between self-renewal and differentiation. In the hematopoietic system, Nestin-GFP+ mesenchymal stromal cells (MSC), Leptin receptor (LepR)+ MSCs, NG2+ MSCs, CAR cells which located in the perivascular area have been implicated in the regulation of HSC maintenance. On the other hand, we identified that endosteal cells (bone lining cells) were composed of three populations: ALCAM+Sca-1– osteoblasts, ALCAM–Sca-1+ MSC, and ALCAM–Sca-1– cells osteoprogenitor/other stromal cells. All three fractions maintained long-term reconstitution (LTR) activity of hematopoietic stem cells (HSCs), and ALCAM+Sca-1– cells, in particular, showed robust supporting activity for HSCs. For characterization of the three endosteal cell populations, we performed the single cell gene expression analysis and identified the small subpopulation in ALCAM+Sca-1– cells that expressed pluripotent stem cell marker genes. These data indicate that ALCAM+Sca-1– cells are a heterogeneous population that contains immature cells. With the repeated single cell analysis, we found that the subpopulation of ALCAM+Sca-1– cells specifically expressed Cdh2. As expected, the gene expression pattern of ALCAM+Sca-1–Cdh2+ cells was similar to ES cell rather than bone marrow MSCs. Furthermore, ALCAM+Sca-1–Cdh2+ cells maintained LTR activity of HSCs after the coculture with HSCs. These data suggest that ALCAM+Sca-1–Cdh2+ cells are novel MSCs with niche cell activity for HSCs.

Biography Fumio Arai is a Professor of Department of stem cell biology and Medicine, Graduate School of Medical Sciences, Kyushu University. He has completed his Ph.D at the age of 28 years from Meikai University and postdoctoral studies from Keio University School of Medicine. His research interest is in studying the mechanisms of the cell fate regulation of HSCs at the single cell level for the establishment of the system that is able to expand HSCs.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 45 Durba Pal et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Nanochannel-Based Electroporation Assisted Tissue Reprograming and Repair Durba Pal, Daniel Gallego-Perez, Subhadip Ghatak, L. James Lee and Chandan K. Sen 1The Ohio State University, USA 2Indian Institute of Technology Ropar, India

ecent advances in nuclear cell reprogramming have opened up the possibility for the development of patient specific Rtherapies, and are thus a great leap forward in translational regenerative medicine. New evidence suggests that nuclear cell reprogramming can also be induced in vivo, which could potentially facilitate the transition from the lab bench to the clinic in some cases. However heavy reliance on viral methods is in conflict with clinical applications. Therefore, to realize the full potential of in vivo reprogramming, a safer, more efficient and better-controlled approach for the delivery of complex combinations of reprogramming factors is needed. Our newly developed nanochannel electroporation (NEP) patch technology allows for non-viral as well as in vivo gene delivery in a targeted, controlled and benign manner, which is not attainable by existing technologies. Plasmids encoding for specific reprogramming transcription factors were used as model cargo. Tissue transfection was characterized by in vivo imaging tool (Fig. 1). A specific cocktail of transcription factors Etv2, Fli1 and Foxc2 (EFF) was found to be enhanced by the transdifferentiation of adult fibroblast into functional reprogrammed endothelial cells bothin vitro (Fig. 2) as well as in vivo where ischemic tissues re- gained significant vascularization (Fig. 3) following NEP-based delivery of such reprogramming factors. Here we explored the tissue repair by reprogramming amenability of adult skin fibroblast to endothelial cells by the use of electrotransfection technology.

Figure 1: In vivo transfection of tagged DNA by non-viral nanoelectroporation approach. Electrotransfection of TFs was done on mouse skin with nanofabricated chip and validated with in vivo imaging system.

Figure 2: Nanoelectroporation of EFF cause dermal fibroblast to reprogram into endothelial cells by (A) by adapting cobblestone morphology of endothelial property. (B) functionally by showing capillary like tube formation on matrigel.

Figure 3: Improvement of ischemic hind limb (A) by pictorial representation of delivery of EFF or exosomes laden with EFF; (B) resulting revascularization of hindlimb within 14 days (Treated– EFF) or exosomes laden with EFF (Treated-EXO) as compared to control. Blood perfusion was evaluated by using laser Doppler blood flow meter of ischemic and non-ischemic limbs.

Biography Notes:Durba Pal recently joined as a faculty in the Center for Biomedical Engineering at Indian Institute of Technology Ropar, India. Her research area is on Tissue Engineering and Regenerative Medicine. After completion of her PhD from India in Molecular Biology, she joined The Ohio State University, Columbus OH, USA, as Post-doctoral Fellow where she began her journey in the field of cellular reprogramming and regenerative medicine. She has discovered the unique cocktail of transcription factors that can be able to convert dermal fibroblasts to endothelial cells when delivered in a simple-to-implement nano-electroporation approach. She had been successful in tissue repairing by in vivo reprogramming approach.

[email protected]

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 46 Avila-Portillo Luz M et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Comparative study between two ex vivo cell expansion models of CD34+ cells from cryopreserved umbilical cord blood co-cultured with encapsulated mesenchymal stem cells Avila-Portillo Luz M2,3, Riveros Angela2, Avila Jenniffer2, Franco Derly2, Ortiz Alejandra2, Godoy Marcela2 and Graciano Noiver1 1Universidad Militar Nueva Granada, Colombia 2CryoHoldco, Colombia 3Hospital Militar Central, Colombia

he expansion of primitive hematopoietic stem cells (HSCs) with long-term restocking capacity is a technological development Tnecessary to increase the use of umbilical cord blood units in HSC transplantation. At present, CD34+ and CD133+ HSC expansions are reported with adequate expansion factors, however a model is required in which maturation is not induced to compromise lineage progenitors. This work compares the expansion factor (EF) of HSC CD34+/CD90+/CD49f+ using two models of expansion (static and perfusion), the model incorporates mesenchymal cells of Wharton gelatin of CD146+ umbilical cord encapsulated in calcium alginate 2%, and recombinant human cytokines involved in self-renewal (SCF, TOP, FL, IL3, IL6), and the activation of the NOTCH pathway using the DLK-1 agonist in a 12-day culture. NOTCH uses the DLK-1 agonist in a 12-day culture. The perfusion culture vs the static yielded higher EFs for both CD34/45- (8.46 vs. 3.99) and CD49+/CXCR4+HSC (12.97 vs. 2.95) cells. When comparing the two models, it is evident that the perfusion model is statistically significant until day 8 with respect to the static one p<0.005. These results are superior to any study published to date. Comparing the expansion factor in the two culture models, this study suggests the importance of MSC CD146+ in co-cultures with HSC, by sustaining the selective expansion of cd34+/ CD90+CD49f+ without favoring the expansion of other subpopulations of Progenitors taken from unknown lineages. These findings need to be replicated in larger sample sizes in in vivo models and clinical trials of HSC transplantation in humans to demonstrate their importance in important clinical outcomes.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 47 conferenceseries.com 1271st Conference

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Scientific Tracks & Abstracts Day 3

Regenerative Medicine 2017

Page 48 Sessions: Day 2 October 04, 2017 Regenerative Medicine|Bone Tissue Engineering|Tissue Repair and Regeneration|Cellular Therapies|Scaffolds in Regenerative Medicine| Models of Regeneration

Session Chair Session Co-Chair Ing-Ming Chiu Alaa T alshareeda National Health Research Institutes, Taiwan King Abdullah International Medical Research Center, KSA

Session Introduction Title: Restoration of the corneal epithelium using a novel BioinkTM Hannah Frazer, University of Sydney, Australia Title: Regenerative implantable medical devices: An overview Hongman Wang, Peking University Health Science Center, China Title: Elabela, a novel hormone, involves in angiogenesis and cardiogenesis in vitro Zhi Wang, Nanjing Chest Hospital, China Title: Optimization of embryonic kidney organotypic renal organoid culture to study nepron development from individual cells to functional unit Ilya Skovorodkin, University of Oulu, Finland Title: Effect of pancreatic extract and signaling methods in direction of embryonic stem cells into β cells Elham Hoveizi, University of Ahvaz, Iran Title: Stem cell-based approach in nephropathy management: Pre-clinical study Hanaa H Ahmed, National Research Centre, Egypt Title: Lizards as model for organ regeneration in amniotes: Results and perspectives Lorenzo Alibardi, Comparative Histolab, Padova, Italy Title: Diabetes and regenerative medicine, a new strategy on rearrangement of amino acids consequences in insulin formation Emad Fawzy Eskander, National Research Center, Egypt Title: Limbo™ platform, an innovative methodological approach towards cryopreservation of cell therapy products Roberto Hernan, Cellulis Ltd., UK Title: In vivo repair of bone defect with PGLA/HA/CS scaffold and MSCs on rats Seyedeh Niloufar Rafiee Alavi ,Iran University of Medical Sciences, Iran

Regenerative Medicine 2017

Page 49 Hannah Frazer et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Restoration of the corneal epithelium using a novel BioinkTM Hannah Frazer1, Jingjing You1,2, Simon Cooper3, Chris Hodge4, Xiao Liu5, Zhi Chen5, Adam Taylor5, Erin McColl5, Gordon Wallace5 and Gerard Sutton1,3,4 1Sydney Medical School, Australia 2University of New South Wales, Australia 3Lions NSW Eye Bank, Australia 4Vision Eye Institute, New South Wales, Australia 5University of Wollongong, Australia

he cornea is the transparent outermost layer of the eye and plays both a tectonic and refractive role. Corneal trauma represents the Tmost common ophthalmic emergency presentation with approximately ¾ of all cases due to corneal foreign bodies or abrasions. These injuries are estimated to cost the Australian population more than $155 million per year and if not treated effectively, can lead to infection and scarring resulting in permanent, impaired vision. We have developed a novel, xenogeneic-free corneal BioinkTM using a human platelet lysate (hPL) base that promotes the proliferation and migration of corneal epithelial cells. hPL has previously been shown to promote mesenchymal stromal cell growth. Our BioinkTM sets in a matter of minutes as a transparent, gel-like substance. We performed cell proliferation and scratch wound-healing assays using human corneal epithelial cell lines (HCE-T), and rheology tests to examine its mechanical properties. Our preliminary results have shown the BioinkTM supports multidirectional growth and stratification of HEC-T and the cells completely biodegrade the BioinkTM once forming a complete monolayer. Performing flow cytometry using Ki67, we found that BioinkTM promotes proliferation in HCE-T at a rate comparable to foetal bovine serum (FBS), which is the current standard serum used to culture HCE-T. Scratch wound assays showed that the cells in the BioinkTM and FBS both promoted full-wound closure at a comparable rate. Rheology testing demonstrated the high gel-forming potential and shear-thinning property of our BioinkTM, which demonstrates its capability for extrusion bioprinting. Our BioinkTM represents a potential alternative to existing treatments.

Biography Hannah Frazer is currently completing her MPhil at Sydney Medical School, University of Sydney.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 50 Hongman Wang, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Regenerative implantable medical devices: an overview Hongman Wang Peking University Health Science Center, China

he objective of this study was to conduct a bibliometric evaluation and trend prediction of English literature on animal-derived Tregenerative implantable medical devices based on tissue engineering technology. Data identified by a search strategy with 11 combinations of keywords before 1 January, 2014 were downloaded from 8 databases on 25th November, 2014. The study analyzed publication year, journal preference, authors’ geographic location, and research topics. Research on animal-derived regenerative implantable medical devices is gradually increasing. The majority of the first authors are from colleges or universities. Approximate one third of the papers were the result of cooperation of different institutions. The top five productive countries are USA, China, UK, Germany and Italy. Biomaterials is the main literature source. Bradford's law analysis shows that a core journal area has formed. The active areas of research and future research directions are scaffold materials, biocompatibility, growth factors and extracellular matrix. Research of animal-derived regenerative implantable medical devices has attracted more and more attention from the academia. But most of the research achievements are generated by a few developed countries. Researchers around the world need to complement each other in knowledge and academic resources by communication and cooperation.

Biography Hongman Wang has received her PhD degree from Traditional Chinese Medicine of Peking University (China) in 1998 and served as a Post-doctoral Researcher at Peking University (China) in 1999-2000. Her research is focused on social decisions and social sciences, global health and social development. During the last 10 years, she has published over 100 papers, 4 monographs, and given about 100 invited lectures in various meetings and conferences. Currently, she is a Professor and the Head of the Center for Health and Social Development, Institute for the Medical Humanities of Peking University Health Science Center.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 51 Zhi Wang et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Elabela, a novel hormone, involves in angiogenesis and cardiogenesis in vitro Zhi Wang1, Huang Jin1, Daozhan Yu2 and Dawei Gong2 1Nanjing Chest Hospital, China 2University of Maryland School of Medicine, USA

Aim: Our previous study demonstrated that the Elabela (ELA), a recently discovered hormone, functionally activates apelin receptor (APJ) in mammalian system. However, the physiological effects of ELA in mammalian cardiovascular system remain unknown. The aim of the study was to investigate the role of ELA during human embryonic stem cells (hESCs) differentiation and proangiogenic effect in vitro. Methods: The process of cardiomyocytes differentiated from human embryonic stem cells (H9) with or without ELA treatment was observed continuously. Moreover, we investigated the physiological effect of ELA on angiogenesis of human umbilical vascular endothelial cells (HUVEC) in vitro. Results: ELA treatment at 1 μmol/L significantly increased the number of beating embryoid bodies (EBs) of differentiated hESCs. Furthermore, ELA up-regulated the expression of the cardiac-restricted transcription factors Nkx2.5and Tbx5 and factors involved in differentiated cardiac cells (αMHC, βMHC). Consistently, ELA significantly enhanced clear capillary-like tube formation of HUVEC. Conclusions: We delineate that ELA promotes cardiomyocyte differentiation of hESCs, and is a potent proangiogenic hormone that involves in angiogenesis in vitro. We speculate that ELA may be helpful in regenerative medicine, especially application in treatment of ischemia heart diseases.

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 52 Ilya Skovorodkin et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Optimization of embryonic kidney organotypic renal organoid culture to study nepron development from individual cells to functional unit Ilya Skovorodkin, Ulla Saarela, Aleksandra Rak-Raszewska, Yana Tarakanchikova, Zenglai Tan and Seppo J Vainio University of Oulu, Finland

ammalian embryonic kidneys and renal organoids can be cultured ex vivo to study different aspects of early nephrogenesis Mand applied as a model to analyse the origin of different congenital diseases (CACUT for example). In our laboratory we have earlier developed an experimental system where embryonic kidney cells can be dissociated, sorted (to exclude or add specific cell populations), genetically modified and reaggregated to form renal organoids. We will present our recent data concerning re- vascularisation of renal explants and organoids, optimization of imaging methods and comparison of several methods of genetical transformation of primary embryonic kidney cells to analyse the process of nephrogenesis and establishing of functional renal unit in vitro.

Biography Ilya Skovorodkin has completed his PhD from Institute of Cytology, Academy of Sciences of Russia in St.-Petresburg. After Post-doctoral studies from Tuebingen University (Germany) he is working as Senior Research Fellow in Laboratory of Developmental Biology, Oulu Center for Cell-Matrix Research, University of Oulu (Finland) under supervision of Professor Seppo Vainio. He has published 36 papers in reputed journals.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 53 Elham Hoveizi et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Effect of pancreatic extract and signaling methods in direction of embryonic stem cells into β cells Elham Hoveizi and Hashemitabar Mahmod University of Ahvaz, Iran

iabetes mellitus is caused by loss of insulin-secreting capacity due to an autoimmune destruction of the insulin-producing β cells. DInsufficient source of insulin-producing cells (IPCs) is the major limit in using transplantation for therapy diabetes. We present here a method for forming islet-like clusters of IPCs derived from mouse embryonic stem cells (mESCs). The protocol consisted of several steps. Embryoid bodies (EBs) were first cultured and plated in condition medium associated with activin and condition medium, followed by medium supplemented with basic fibroblast growth factor (bFGF). Next bFGF was withdrawn, and cyclopamine and noggin were added. Then the cells were treated with B27 and condition medium for maturation. Our results demonstrated that mESCs differentiated into IPCs. Immunofluorescence and qRT-PCR detected an enhanced expression of pancreatic genes in the differentiated cells and tests by ELISA showed an increased percentage of insulin-expressing cells in the differentiated cells. Moreover insulin, most cells also co expressed others markers of pancreatic cells. This method lead to induction of cells which exhibited higher insulin secretion and further improvement of this IPCs protocol may result in the formation of an unlimited source of cells suitable for transplantation. These evidences indicated that condition medium as critical components of the stem cell niche associated other factors had high potential to differentiate mESCs into IPCs.

Biography Elham Hoveizi has completed his PhD from Kharazmi University of Tehran. His major area of research is cell and developmental biology and currently he is working as an Assistant Professor in Shahid Chamran University of Ahvaz. He has published more than 24 papers in reputed journals. Currently his research is in the field of and beta cells differentiation.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 54 Hanaa H Ahmed et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Stem cell-based approach in nephropathy management: pre-clinical study Hanaa H Ahmed1, El-Sayed M E Mahdy2, Wafaa Gh Shousha3, Hatem A El-mezyn2 and Mustafa K EL-Sayed2 1National Research Centre, Egypt 2Helwan University, Egypt

his study aimed to assess the possible therapeutic role of mesenchymal stem cells (MSCs) isolated from bone marrow and adipose Ttissue in the management of nephropathy in male rats. Preparation and isolation of MSCs from bone marrow and adipose tissue were done. The isolated cells were identified via the morphological appearance, and the detection of some specific surface markers such as CD90, CD105 and CD45 using flow cytometry technology. The study included 100 adult male rats which were divided into 5 groups. Group (1) was control, group (2) was nephropathic rats, groups (3), (4) and (5) were the groups treated with a single dose of undifferentiated MSCs isolated from bone marrow, undifferentiated MSCs isolated from adipose tissue and lisinopril drug respectively. Our data indicated that the isolated cells were MSCs. This was evidenced from their spindle shape and CD markers. The efficacy of MSCs in kidney repair was evidenced from the significant decline in serum urea, creatinine MCP-1, NF-kB and ET-1 as well as the levels of NAG, in addition to the significant increase in serum nephrin level and GFR value compared with the untreated group. Interestingly, the histopathological investigation confirmed the biochemical data as stem cell therapy evoked great improvement in the structural organization of kidney tissue. This study clearly demonstrated the ability of MSCs, especially isolated from adipose tissue, in mitigating experimental nephropathy. The effectiveness of MSCs in this issue could be attributed to their proliferative capacity, anti-inflammatory activity and anti-apoptotic action.

Biography Hanaa H Ahmed has completed her PhD from Faculty of Science, Cairo University. She is the Head of Hormones Department, Medical Research Division, National Research Centre, one of the biggest research centers in Egypt. She has published more than 155 papers in reputed journals and 22 international books and serving as an Editorial Board Member of 31 international journals. She was awarded the Prize of Excellence at the National Research Centre in the Field of Science and Technology, the Advanced Medical Science (2013).

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 55 Lorenzo Alibardi, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Lizards as model for organ regeneration in amniotes: results and perspectives Lorenzo Alibardi Comparative Histolab, Padova, Italy

izards represent ectotherm amniotes with the broader tissues and organ regeneration, in the tail, and more limited recovery Lcapabilities present in vertebrae, knees, and spinal cord. After lumbar and initial paralysis, a limited and weak hindlimbs movement is often re-gained. While in the tail numerous tissues are reformed, the amputated limb incurs in a strong inflammation and eventually gives rise to a scarring outgrowth, like in mammals. The cellular processes of tail and limb regeneration are presented, indicating that FGFs and other growth factors sustain the process of tail regeneration in conjunction with the formation of an apical epidermal cap in the blastema, a leading microregion that is missing in the scarring limb. Administration of FGFs however improves limb regeneration in lizards, allowing tibia and fibula regeneration but no regeneration of the autopodium. Recent transcriptome data have indicated that the regenerating tail blastema is largely sustained by Wnt and snoRNAs that are expressed in an immuno-depressed environment while in the limb blastema an intense activation of inflammatory genes and no immuno- depression occurs, leading to scarring. The tail blastema appears as a temporary proliferating front with a strict control of cell division where no immune-reactivity and organ rejection occurs. The evolution of the mechanism of immuno-evasion in lizards remains to be analysed. In conclusion, the lizard model provides important clues to improve organ regeneration in amniotes, including mammals, providing that pharmacological therapies depressing immunity and controlling cell proliferation are utilized in concert.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 56 Emad Fawzy Eskander, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Diabetes and regenerative medicine, a new strategy on rearrangement of amino acids consequences in insulin formation Emad Fawzy Eskander National Research Center, Egypt

iabetes, also known as diabetes mellitus, is a group of metabolic diseases in which a person has high blood sugar either because Dthe body does not produce enough insulin, or because cells do not respond to the insulin that is produced. There are two primary types of diabetes. Type 1 diabetes, also known as insulin-dependent or juvenile diabetes, is an autoimmune disorder in which the immune system attacks and destroys the insulin-producing beta cells in the pancreas. Type 2 diabetes, known as adult onset or non- insulin dependent diabetes, is caused when either there is a deficiency in the insulin being produced, or when the cells of the body become resistant to the action of insulin. Diabetes is a chronic condition that requires constant monitoring and creates dangerous and debilitating secondary conditions. Long-term complications include increased risk of cardiovascular problems such as coronary artery disease, heart attack and stroke. Other complications include nerve damage in the limbs, kidney failure, blindness and nerve damage in the feet and legs that can cause diabetic foot ulcers which can lead to amputation, if not treated properly. There are a variety of regenerative medicine technologies in preclinical and clinical development that aim to reestablish insulin production and mediate the immune system’s attack on insulin producing beta cells. Presently, companies believe that multistem has the potential to regulate immune system function, and could thus work to protect the beta cells mesoblast and are using their patented human mesenchymal progenitor cells to target type 2 diabetes. In preclinical trials, the injection of a dose of MPCs into mice with diabetes resulted in a significant increase in blood insulin levels and sustained reduction in blood glucose levels during the follow-up period. Mesoblast is in the midst of a 60 patients’ Phase 2 clinical trial. Osiris Therapeutics has completed enrollment for a Phase 2 type 1 diabetes clinical trial evaluating the efficacy and safety of their product, Prochymal. Prochymal uses mesenchymal stem cells for their believed ability to delay the progression of type 1 diabetes by preserving beta cell function, and thus insulin production. The researchers are developing a stem cell based technology for the treatment of type 1 and type 2 diabetes derived from a human embryonic stem cell. In animal models, the cells differentiated into insulin producing and other endocrine cells that regulated blood glucose in a manner very similar to the normal pancreas when implanted under the skin. The study showed promising signs of long-term control of diabetes in large animals, current regenerative medicine therapies for diabetes.

Biography Emad Fawzy Eskander has completed his BSc in Biochemistry from Ain-Shams University, MSc in Organic Chemistry from the same university. He has obtained his PhD in Organic Chemistry from Cairo University. Then he has finished his Diploma of Medicinal Chemistry in the year of 1993 from Georgia (USA).

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 57 Roberto Hernan, J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

Limbo™ platform, an innovative methodological approach towards cryopreservation of cell therapy products Roberto Hernan Cellulis Ltd., UK

any cell therapy products need to be frozen to maintain product stability. Freezing and thawing cells correctly require several Mprocesses that need to be performed with care to avoid cell damage. The inadequate performance and lack of uniformity of these protocols may infer undesirable inconsistencies on the cellular products jeopardizing therapeutic efficacy. Despite current technological advances, cryopreservation protocols have been highly conserved during the last 65 years since the very first discovery of cryoprotectants. Hereby we propose a novel methodological approach to cryopreservation which introduces simple enclosed mechanisms that assure the correct standardization of freezing and thawing processes, as well as rendering a final product formulation for the cell therapy market. These mechanisms take place inside the Limbo™ vial, making the post-thawing cellular reconstitution process operator-independent. Limbo™ cryovials offer the opportunity to avoid sample washing to diminish the DMSO effect, reducing in turn expensive costs and resources at the point-of-care. Furthermore, this technology introduces a unique dry thawing system which not only enforces safe and correct thawing protocols but also eliminates contamination risks associated to water baths. In summary, this novel technology is a safeguard for most frozen cell therapies because it avoids sample handling at the point-of-care while addresses the need for appropriate cellular recovery standardization protocols in the clinic.

Biography Roberto Hernan has completed his Bachelor´s degree in Biology (1994). He worked as a Research Technician at the University of Newcastle Upon Tyne (UK), focused on his Cancer Research career at St. Jude´s Children Research Hospital, Memphis (USA) and in 2005 obtained his PhD in Pediatric Oncology at Newcastle University. He then led the Business Development of Pharmakine for 7 years. At present, he holds the position of Chief Scientific Officer at Cellulis, where he is also a partner. He has participated as a major contributor in ten scientific publications and has led the development of three different patents within the cryopreservation field.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 58 Seyedeh Niloufar Rafiee Alavi et al., J Tissue Sci Eng 2017, 8:4(Suppl) conferenceseries.com DOI: 10.4172/2157-7552-C1-039

7th International Conference on Tissue Engineering & Regenerative Medicine October 02-04, 2017 Barcelona, Spain

In vivo repair of bone defect with PGLA/HA/CS scaffold and MSCs on rats Seyedeh Niloufar Rafiee Alaviand Seyed Keihan Mostafavi Iran University of Medical Sciences, Iran

uman mesenchymal stem cells (hMSCs) are a very attractive option for cell engineering. In addition, as hMSCs does not cause Himmune response, its nonautologous application is also possible. This study discusses the application of hMSCs seeded in PGLA/ CS/HA scaffold in femur bone defect model in rats. After separating the cells from human bone marrow sample, they were cultured on the constructed scaffold. Then some tests were performed on the constructed tissue, which were: Electron microscopy studies, to determine cells adherence to scaffold and the tissue’s morphology, MTT test to determine the scaffold’s biocompatibility and measuring Alkaline-phosphatase amounts to check the quantity of differentiated hMSCs to osteoblasts. After forming the bone defect model on rats, the bone defect was filled with scaffold and MSCs in one group and with scaffold alone, in another. Also, there was a third, control group for comparison. After 12 weeks of implanting the tissue in rats, CT scan radiography revealed the new bone was only formed at the two ends of femur bone in the control group and filling was much more in the other two groups, one with scaffold alone and another with scaffold and cells. Although, the difference between the amount of newly formed bone in the group with both scaffolds and MSCs was statically significant from the two other groups. Electron microscopy revealed desirable adherence and polygonal morphology which indicates cell differentiation. MTT test’s results showed proper biocompatibility and increased cell proliferation. Alkaline phosphatase amounts indicated elevated osteoblast’s activity. Histological studies illustrated formation of lamellar bone in the two test groups and the thickness was more in the group with both scaffold and hMCSs. No immunological reaction was observed in the two groups. In conclusion, PLGA/HA/CS scaffold improves cell adherence, cell proliferation, biocompatibility, differentiation of MSCs to osteoblasts and osteogenesis in both in vitro and in vivo condition.

Biography Seyedeh Niloufar Rafiee Alavi studying medicine at Iran University Of Medical sciences.

[email protected]

Notes:

J Tissue Sci Eng, an open access journal Volume 8, Issue 4 (Suppl) ISSN: 2157-7552 Regenerative Medicine 2017 October 02-04, 2017

Page 59