Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 10th World Congress and Expo on Cell & Stem Cell Research New York, USA.

Day 3 :

Conference Series Stem Cell Research 2018 International Conference Keynote Speaker Sima T. Tarzami photo
Biography:

Sima T Tarzami has received her BSc and MSc degrees from Hofstra University, New York, and her PhD from Albert Einstein School of Medicine, New York in 2002. She was a Faculty in Mount Sinai School of Medicine from 2007 to 2015. She is currently an Associated Professor of Medicine at Howard University. Her laboratory studies the role of chemokines on cardiac myocyte biology. She focuses on cardiac physiology in both in vitro and in vivo models of heart failure. She is an author of 20 peer-reviewed papers and over 15 published abstracts.

Abstract:

An adult heart has an intrinsically limited capability to regenerate damaged myocardium, regardless of the underlying etiology. Embryonic and induced pluripotent stem cell (ESC/iPSC)- based therapies offer a unique strategy for developing cell replacement therapies for numerous, varied disorders including cardiac diseases. iPSCs hold great promise in the field of regenerative medicine because of their ability to grow indefinitely and give rise to all cells of the body. Recently, investigators shown that pluripotent stem cells produce tissue-specific lineages through the programmed acquisition of sequential gene expression patterns that function as a road map for organ formation, therefore, identifying a procardiogenic network that promotes iPSCs differentiation to favor a cardiac lineage is of great interest. Since adult human hearts have very little ability to regenerate postnatally, stem-cell-based cardiac regeneration has also been considered as a therapeutic approach to treat ischemic heart disease. Since these cells have been shown to migrate to sites of injury and inflammation in response to soluble mediators including the chemokine stromal cell derived factor-1 (SDF-1 also known as CXCL12). Here, we studied the role of SDF-1 and its receptors; CXCR4 and CXCR7 in transformation of pluripotent stem cells into IPSC-derived cardiomyocytes. This study demonstrates that CXCR4 and CXCR7 induce differential effects during cardiac lineage differentiation and β-adrenergic response in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). In engineered cardiac tissues, depletion of CXCR4 or CXCR7 had opposing effects on developed force and chronotropic response to β-agonists demonstrating distinct roles for the SDF-1/CXCR4 or CXCR7 network in hiPSC-derived ventricular cardiomyocyte specification, maturation and function.

  • Stem Cell Therapy | Tissue Engineering | Tumour Cell Science | Stem Cells | Regeneration and Therapeutics | Stem Cell Niches | Diseases And Stem Cell Treatment
Location: New York
Speaker
Biography:

Joel I Osorio is the CEO and Founder of Biotechnology and Regenerative Medicine at RegenerAge International ™ HYPERLINK "http://www.regenerage.clinic/"h(HYPERLINK"http://www.regenerage.clinic/" h www.regenerage.clinic). He is the Vice President of International Clinical Development for Bioquark, Inc. (HYPERLINK"http://www.bioquark.com/" h www.bioquark.comHYPERLINK "http://www.bioquark.com/" h ) and Chief Clinical Officer at ReAnima™ Advanced Biosciences(HYPERLINK "http://www.reanima.tech/" h www.reanima.tech) and Westhill University School of Medicine. Mexico. He pursued Advance Fellow by the American Board of Anti-Aging and Regenerative Medicine (A4M) and Visiting Scholar at University of North Carolina

Abstract:

As it has been previously demonstrated that coelectroporation of Xenopus laevis frog oocytes with normal cells and cancerous cell lines induces the expression of pluripotency markers and in experimental murine model studies that mRNA extract (Bioquantine® purified from intra and extra-oocyte liquid phases of electroporated oocytes) showed potential as a treatment for a wide range of conditions as Squint, Spinal Cord Injury (SCI) and Cerebral Palsy among others. The current study observed beneficial changes with Bioquantine® administration in a patient with a severe SCI. Pluripotent stem cells have therapeutic and regenerative potential in clinical situations CNS disorders even cancer. One method of reprogramming somatic cells into pluripotent stem cells is to expose them to extracts prepared from Xenopus laevis oocytes. We showed previously that coelectroporation of Xenopus laevis frog oocytes; with normal cells and cancerous cells lines, induces expression of markers of pluripotency. We also observed therapeutic effects of treatment with a purified extract (Bioquantine) of intra and extra-oocyte liquid phases derived from electroporated X. laevis oocytes, on experimentally induced pathologies including murine models of melanoma, traumatic brain injury and experimental skin wrinkling induced by squalene mono-hydroperoxide. The positive human findings for spinal cord injury and cerebral palsy with the results from previous animal studies with experimental models of traumatic brain injury, respectively. Because of ethical reasons, legal restrictions and a limited number of patients, we were able to treat only a very small number of patients. These results indicate that Bioquantine® may be safe and well tolerated for use in humans and deserves further study in a range of degenerative disorders. We propose that the mechanism of action of Bioquantine® in these various diseases derives from its unique pharmacology and combinatorial reprogramming properties.
In conclusion, these preliminary findings suggest that Bioquantine is safe and well tolerated on patients with cerebral palsy and spinal cord injury, among others. In addition to the regenerative therapy and due to the patient condition, we decided to include the RestoreSensor SureScan. Based on the electrical stimulation for rehabilitation and regeneration after spinal cord injury published by Hamid and MacEwan, we designed an improved delivery method for the in-situ application of MSCs and Bioquantine® in combination with the RestoreSensor® SureScan®. To the present day the patient who suffered a total section of spinal cord at T12-L1 shows an improvement in sensitivity, strength in striated muscle and smooth muscle connection, 11 months after the first therapy of cell regeneration and 3 months after the placement of RestoreSensor® at the level of the lesion, the patient with a complete medullary section shows an evident improvement on his therapy of physical rehabilitation on crawling from front to back by himself and standing on his feet for the first time and showing a progressively important functionality on the gluteal and legs sensitivity.

Biography:

Purwati Sumorejo has finished her General Practition from Airlangga University in 1997. She has completed her studies in Internal Medicine. She is a Specialist(2008) from Airlangga University and also pursued her Doctoral program in Airlangga University from 2010-2012. Her research interest is in stem cell field. Since 2008, she is a Secretary of Stem cell Laboratory of Airlangga University and also Secretary of Surabaya Regenerative Medicine Centre. From 2015, she holdsa position of Chairman of Stem Cell Research and Development Centre Airlangga University, Surabaya, Indonesia. She has almost 60 publications in journals, papers and seminars.

Abstract:

Autoimmune diseases (ADs) are the third most common disease in United States affecting 5 to 8% of population. The major treatment of ADs is immunosuppressive drugs but these are not effective and associated with substantial toxicities. Adipose tissue is one of the most potent and concentrated source of mesenchymal stem cells (MSCs) as an anti-inflammatory and tissue protecting agent which will promote healing and also minimal invasive. This study is conducted in 20 patients with autoimmune diseases in various ages between 22 to 70 years old. Patients were treated with autologous adipose-derived MSCs transplantation through catheterization. The laboratory analysis result of patients before and after MSCs application in 6 months were measured which include haemoglobin (Hb), white blood cells (WBC), erythrocyte sedimentation rate (ESR), protein and blood levels in urine, high sensitivity c-reactive protein (hsCRP), C3 and C4 complement, anti-nuclear antibodies (ANA) and anti-double stranded DNA (anti-dsDNA). MSCs are able to improve the performance of hemoglobin as shown in Hb which showed statistically significant increase (p=0.002). MSCs are able to reduce the inflammatory as shown in the number of leukocytes (p=0.015) and ESR (p=0.031) which showed statistically significant decrease. MSCs can repair the renal function with no presences of protein and blood in patient’s urine. MSCs are also able to augment the immune response as shown in hsCRP which statistically significant decrease (p≤0.001), while C3 and C4 complements statistically significant increase (p≤0.001). ANA and anti-dsDNA showed a negative result which means MSCs therapy may give a good response to heal the ADs.

Speaker
Biography:

Purwati Sumorejo has finished her General Practition from Airlangga University in 1997. She has completed her studies in Internal Medicine. She is a Specialist (2008) from Airlangga University and also pursued her Doctoral program in Airlangga University from 2010-2012. Her research interest is in stem cell field. Since 2008, she is a Secretary of Stem cell Laboratory of Airlangga University and also Secretary of Surabaya Regenerative Medicine Centre. From 2015, she holdsa position of Chairman of Stem Cell Research and Development Centre Airlangga University, Surabaya, Indonesia. She has almost 60 publications in journals,papers and seminars.

Abstract:

Autoimmune diseases (ADs) are the third most common disease in United States affecting 5 to 8% of population. The major treatment of ADs is immunosuppressive drugs but these are not effective and associated with substantial toxicities. Adipose tissue is one of the most potent and concentrated source of mesenchymal stem cells (MSCs) as an anti-inflammatory and tissue protecting agent which will promote healing and also minimal invasive. This study is conducted in 20 patients with autoimmune diseases in various ages between 22 to 70 years old. Patients were treated with autologous adipose-derived MSCs transplantation through catheterization. The laboratory analysis result of patients before and after MSCs application in 6 months were measured which include haemoglobin (Hb), white blood cells (WBC), erythrocyte sedimentation rate (ESR), protein and blood levels in urine, high sensitivity c-reactive protein (hsCRP), C3 and C4 complement, anti-nuclear antibodies (ANA) and anti-double stranded DNA (anti-dsDNA). MSCs are able to improve the performance of hemoglobin as shown in Hb which showed statistically significant increase (p=0.002). MSCs are able to reduce the inflammatory as shown in the number of leukocytes (p=0.015) and ESR (p=0.031) which showed statistically significant decrease. MSCs can repair the renal function with no presences of protein and blood in patient’s urine. MSCs are also able to augment the immune response as shown in hsCRP which statistically significant decrease (p≤0.001), while C3 and C4 complements statistically significant increase (p≤0.001). ANA and anti-dsDNA showed a negative result which means MSCs therapy may give a good response to heal the ADs.

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 Post-doctoral 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, widely published, organized and lectured at numerous national/international conferences, served on study sections for various federal and non-profit funding agencies, and is on the Editorial Board of a number of scientific journals. He is member of several national and international societies and recipient of various awards.

Abstract:

Allogenic islet transplantation is an important therapeutic approach for the treatment of type 1 diabetes (T1D). However, graft rejection initiated and perpetuated by pathogenic T effector (Teff) cells presents a major barrier. One approach that has proven successful for promoting graft tolerance is shifting the T cell balance away from the induction of pathogenic Teff cells and towards the generation of protective T regulatory (Treg) cells. PD-1/PD-L1 immune checkpoint pathway plays an important role in the Teff and Treg balance with demonstrated clinical efficacy in cancer immunotherapy. We generated a chimeric PD-L1 molecule that can be transiently displayed on the surface of pancreatic islets. In vitro, PD-L1 enhanced TGF-beta-induced conversion of Teff into Treg cells and effectively suppressed the proliferation of Teff cells in response to alloantigen stimulation. In vivo, PD-L1-engineered BALB/c islet grafts under a short window of rapamycin treatment achieved sustained long-term graft survival and function. These results provide strong proof-of-efficacy and feasibility for using PD-L1 protein as an immunomodulator to promote allogeneic islet graft survival in the absence of continued immunosuppression.

Biography:

Zita M Jessop is an Academic Clinical Lecturer in the Reconstructive Surgery and Regenerative Medicine Research Group at Swansea University and Registrar at the Welsh Centre for Burns and Plastic Surgery. After completion of Medicine at Cambridge University, she undertook surgical training in London Deanery before joining the Welsh Clinical Academic Training Pathway. Her Doctoral research is focusing on cartilage 3D bioprinting, has won significant awards from the Royal College of Surgeons of England, the British Association of Plastic, Reconstructive and Aesthetic Surgeons and the Medical Research Council. She is currently a Fulbrigth Scholar at the Wyss Institute, Harvard University.

Abstract:

Contemporary cartilage tissue engineered implants, many using unrelated adult stem cell sources do not produce stable, physiologically relevant cartilage. Although lacking intrinsic reparative ability, cartilage has been shown to contain a population of stem cells or progenitor cells, similar to other adult tissues, that are thought to be involved in maintenance of tissue homeostasis. Since they were first identified in articular cartilage, these so-called cartilage-derived stem/progenitor cells (CSPCs) have become the latest addition to the potential cell repertoire for cartilage tissue engineering. Unlike articular CSPCs, nasoseptal CSPCs have not been well studied, with definitive markers and molecular identity remaining obscure to date. We isolated human nasoseptal CSPCs using differential adhesion to fibronectin and undertook functional, genetic and immunophenotypic characterization using ac combination of RT-PCR, PCR array, Immunocytochemistry, Western blotting and Flow Cytometry. Fibronectin adhesive human nasoseptal CSPCs are a self-renewing subpopulation, positive for recognized stem cell markers CD24, CD44, CD73, CD90 and CD105 as well as integrins CD29 (beta1) and CD49e (alpha 5). Unlike previous reports for articular CSPCs, they are negative for STRO-1. Nasoseptal CSPCs are clonogenic with increased expression of neuroectodermal cell adhesion molecules and multilineage potential. Nasoseptal CSPCs are distinct from nasoseptal chondrocytes demonstrated by reduced expression of collagen type 1, collagen type 2 and aggrecan in monolayer culture. In addition, CSPCs can regain chondrogenic potential in 3D culture. This cell population may benefit future cell-based cartilage tissue engineering strategies due to its ability to maintain chondrogenicity and chromosomal stability following cell expansion.

Hina W Chaudhry

Icahn School of Medicine at Mount Sinai, USA

Title: Placental Cdx2 cells and cardiac regeneration
Biography:

Abstract:

Introduction: Placenta is an easily available and rich source of multipotent cells. We previously reported that fetal-placental cells “home” to injured maternal hearts with 40% of these cells expressing Cdx2.
Hypothesis: Placental Cdx2 cells could be a novel source for cardiac repair.
Methods: Fetal-derived Cdx2 cells were labeled with EGFP using a cre-lox strategy wherein female virgin B6; 129S6-gt (ROSA) 26Sor<tm1 (CAG-tdTomato*,-EGFP*) Ees>/J43 mice were crossed with male B6. Cg-Tg (Cdx2-cre) 101Erf/J mice. Myocardial infarction (MI) was induced in pregnant mice at mid-gestation. Maternal hearts were analyzed 5 weeks post-MI for Cdx2-derived cardiomyocytes. Cdx2 cells from end-gestation placenta were assayed for cardiac differentiation in vitro. Proteomic analysis, vascular differentiation and immune profiling were carried out and live cell imaging was done to capture spontaneous beating. Additionally, male WT mice was subjected to MI followed by MRI to confirm MI. 1 week post-MI, the test mice received 1 million Cdx2 cells and control mice received equal volume of PBS via tail vein. MRI was repeated 1 and 3 months later to assess the cardiac function.
Results: Cdx2 cells migrated to injured maternal hearts and differentiated into cardiomyocytes. Additionally, Cdx2 cells from the late placenta differentiated into spontaneously beating cardiomyocytes in vitro and expressed cTnt, α actinin and Cx43. The cells also differentiated into vascular cells indicative of multipotentiality. Proteomic analysis identified homing/survival signaling in Cdx2 cells compared to ES cells. Cdx2 cells further displayed a low expression of immune molecules, suggesting they can evade host immune surveillance. MRI analyses after 1 and 3 months showed a significant increase in ejection fraction (EF) in the cell-treated group [deltas: ctrl 0.355±3.421 vs test 17.18±1.05 (**p=.0093) in 1mo and ctrl 3.247±5.09 vs test 19.31±2.59 (*p=.045) in 3 mo]
Conclusion: We demonstrate for the first time that Cdx2 cells from placenta undergo trafficking upon cardiac injury to form cardiomyocytes. Our data imply that placental Cdx2 cells may represent a powerful new therapeutic strategy for cardiac repair.

Speaker
Biography:

Cevat Erisken has completed his PhD at Stevens Institute of Technology, Hoboken, NJ and served for Columbia University in the City of New York as a Research Assitant Professor. Currently, he is a faculty member in the Department of Chemical Engineering at Nazarbayev University, KZ, and doing research on Biomaterials and Regenerative Medicine. He has published more than 20 articles, lectured at universities and presented at international conferences.

Abstract:

Due to the limited capacity of current reconstruction procedures, there exists a need for new augmentation matrices to improve the biological fixation to obtain a scarless healing at the tendon-bone (TB) interface. The objectives of this study are to 1) fabricate electrospun polycaprolactone (PCL)-based scaffolds containing transforming growth factor (TGF)β3, connective tissue growth factor (CTGF) and nano-hydroxyapatite (nHA), where concentrations of CTGF and nHA change in opposite directions, while TGF-β3 is located in the middle portion of the nanofibrous composites [such organization is expected to contribute to generation of tendon (in CTGF rich zone), fibrocartilage (by TGFβ3) and bone (in nHA rich zone) upon positioning the scaffold in proper zones], 2) establish controlled release of TGFβ3 and CTGF from nanofiber scaffolds, and 3) to investigate human Bone Marrow Stem Cell (h-BMSC) behavior on these scaffolds. Such a design is proposed for the first time, and represents a significant departure from the conventional stratified approach, and is expected to contribute to scar-free TB interface regeneration. Findings show that linearly varying nHA distribution can be accomplished across the scaffold thickness that is also the case in native TB interface. Results also demonstrated that TGFβ3 and CTGF can be incorporated into nanofiber scaffolds with electrospinning, retain its bioactivity, and released in a sustained manner. The h-BMSCs proliferated similarly on graded and individual scaffolds at the end of 14 days. Collagen production by hBMSCs on different scaffold groups did not lead to any significant differences. This study not only reveals the importance of design and use of biomimetic scaffolds in tissue engineering but also yields new insights into the effect of bioactive molecules on interface regeneration by controlling their local availability.

Speaker
Biography:

Thazhumpal C Mathew has completed his PhD from the University of Alberta, Canada in 1992 and obtained FRCPath (UK) in 2003. In 1983, he had undergone aresearch training at the University of Lund, Sweden. After his Post-doctoral studies at the University of Alberta, he has worked as Assistant Scientist at NYU, USA. In 1993, he has joined the FAHS of Kuwait University. Currently, he is the Professor and Vice Dean for Research. He was also the Director of the Electron Microscope Unit. His research is in Molecular Neurobiology. He has received several awards and published more than 75 papers and attended over 100 conferences.

Abstract:

Neurogenesis from endogenous neural stem cells might contribute to functional recovery after brain injury. In this regard, the current study is focused on understanding the axonal injury induced neurogenesis at the supraependymal region of the third ventricle in rat using light and electron microscopy. The floor of the infundibulum is lined by a non-ciliated ependyma. Presence of supraependymal cells (SEC) and supraependymal nerve fibers (SEN) that are immersed in the cerebrospinal fluid is a prominent feature of the ventricular floor. In this study retrograde neuronal tracers, fast blue and fluorogold were used to understand the origin of SEN. For monitoring contralateral collateral sprouting and the changes in the SEC following unilateral cervical sympathectomy, sixteen Wistar rats weighing 150-200g were divided into four groups. Group I was used as normal control, group II and III for unilateral and bilateral cervical sympathectomy respectively and group IV as sham control. Fourteen days after the experiment, the ventricular floor was studied. For microscopic analysis, the animals were perfused under anesthesia transcardially with 3% glutaraldehyde and the brains were dissected out and processed. The SEN consist of a mixture of cholinergic, serotonergic and peptidergic nerve fibers. Retrograde tracer analysis showed that at least some of the SEN could originate from the superior cervical ganglia. Fourteen days after unilateral cervical sympathectomy, there was a profound increase in the number of SEN and supraependymal neurons of the infundibular floor. From these studies, it can be concluded that SEC of the third ventricular floor represent another neural stem cell niche that are induced to proliferate and differentiate following axotomy of the SEN.

Biography:

Abstract:

Background & Objectives: Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibition was proved in streptozotocin (STZ)-diabetic rats. The present study aimed at investigating and comparing the therapeutic effect of combined bone marrow mesenchymal stem cells (BMMSCs) with ascorbic acid (AA) and SERCA1a gene transfected MSCs in STZ induced skeletal myopathy of male albino rat.
Methods: Rats were divided into group I of 4 control rats, group II (Diabetic) of 7 rats given intraperitoneal (IP) injection of STZ 50 mg/kg, group III (BMMSCs) of 7 rats given STZ and BMMSCs intravenous (IV), group IV (BMMSCs and AA) of 7 rats given STZ, BMMSCs and AA 500 mg/Kg and group V (SERCA 1a transfected BMMSCs) of 7 rats given STZ andSERCA1a transfected BMMSCs. The rats were sacrificed after 8 weeks.Gastrocnemius specimens were subjected to histological,morphometric, biochemical and statistical studies.
Results: Diabetic rats revealed atypical widely separated and disorganized muscle fibers, loss of striations, some dark nuclei,markedly congested vessels and dense cellular infiltration. Disrupted atypical fibers with no obvious striations and multiple macrophages were evident. A significant increase in the area of atypical fibers, the mean blood glucose level and a significantdecrease in the mean area of pale nuclei, mean area % of CD105 and CD34 +ve cells and of mean PCR values were recorded.The morphological, morphometric and biochemical changes regressed by therapy.Conclusions: The therapeutic effect was found to be more remarkable by combined MSCs and AA administration and mostremarkable by SERCA1a modified MSCs injection.

Biography:

Abstract:

Background & Objectives: Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) inhibition was proved in streptozotocin
(STZ)-diabetic rats. The present study aimed at investigating and comparing the therapeutic effect of combined bone marrow
mesenchymal stem cells (BMMSCs) with ascorbic acid (AA) and SERCA1a gene transfected MSCs in STZ induced skeletal
myopathy of male albino rat.
Methods: Rats were divided into group I of 4 control rats, group II (Diabetic) of 7 rats given intraperitoneal (IP) injection
of STZ 50 mg/kg, group III (BMMSCs) of 7 rats given STZ and BMMSCs intravenous (IV), group IV (BMMSCs and AA)
of 7 rats given STZ, BMMSCs and AA 500 mg/Kg and group V (SERCA 1a transfected BMMSCs) of 7 rats given STZ and
SERCA1a transfected BMMSCs. The rats were sacrificed after 8 weeks. Gastrocnemius specimens were subjected to histological,
morphometric, biochemical and statistical studies.
Results: Diabetic rats revealed atypical widely separated and disorganized muscle fibers, loss of striations, some dark nuclei,
markedly congested vessels and dense cellular infiltration. Disrupted atypical fibers with no obvious striations and multiple
macrophages were evident. A significant increase in the area of atypical fibers, the mean blood glucose level and a significant
decrease in the mean area of pale nuclei, mean area % of CD105 and CD34 +ve cells and of mean PCR values were recorded.
The morphological, morphometric and biochemical changes regressed by therapy.
Conclusions: The therapeutic effect was found to be more remarkable by combined MSCs and AA administration and most
remarkable by SERCA1a modified MSCs injection.

Speaker
Biography:

Abstract:

In recent years, regenerative cell therapy has been widely explored for the treatment of myocardial infarction (MI). However, although transplantation of the stem cell population ADSC (adipose-derived stem cells) in chronic MI is associated with functional improvement, its therapeutic value is limited due to poor long-term cell engraftment and survival. Thus, the objective of our study was to examine whether transplantation of collagen patches seeded with ADSC could enhance cell engraftment and improve cardiac function in models of chronic MI. Chronically infarcted Sprague-Dawley rats (n=58) were divided into four groups and transplanted with media, collagen scaffold (CS), rat ADSC, or CS seeded with rat ADSC (CS-rADSC). Cell engraftment, histological changes, and cardiac function were assessed four months after transplantation. Functional and histological assessments were performed three months post-transplantation. Transplantation of CS-rADSC was associated with increased cell engraftment, significant improvement in cardiac function, myocardial remodeling, and revascularization. Next, their therapeutic potential was tested in a preclinical model of CMI. Gottingen minipigs (n=18) were subjected to MI and then transplanted two months later with CS or CS seeded with pig ADSC (CS-pADSC). transplantation of CS-pADSC in the preclinical swine model improved cardiac function and was associated with decreased fibrosis and increased vasculogenesis. In conclusion, transplantation of CS-ADSC resulted in enhanced cell engraftment and was associated with a significant improvement in cardiac function and myocardial remodeling.