2015%Regenerative%Medicine%and%Cellular% Therapy

!
2015%Regenerative%Medicine%and%Cellular%
Therapy%Research%Symposium:%
%
“Clinical Translational Issues and Trials”
April%24th%2015%
%
Sponsored%by:%
The%GRU%Institute%for%Regenerative%and%
Reparative%Medicine%
Welcome to Members of the Regenerative Medicine, Tissue Engineering and Cellular Therapy
Research Community. The Georgia Regents University’s Institute for Regenerative and
Reparative Medicine is pleased to host this day long Symposium Friday April 24th 2015. We
appreciate your attendance and share your work with regional colleagues. Our focus is on moving
translational research into clinical trials. This will include talks from researchers and clinician
scientists who have ongoing regenerative medicine based clinical trials. We are targeting
researchers and clinicians who are involved in Regenerative Medicine, Tissue Engineering, and
Cellular Therapy in Georgia and regionally, including the Georgia Regenerative Engineering and
Medicine partnership. We would like to provide an opportunity to highlight the regional
translational and clinical trial research with the goal of facilitating collaborations between groups
at neighboring institutions and providing links to GRU clinical research programs.
We are pleased to also welcome our Keynote Speakers:
Meeting Keynote Speaker:
Arnold I. Caplan, Ph.D.
Case Western Reserve University
Arnold I. Caplan is professor of Biology and Director of the Skeletal
Research Center at Case Western Reserve University. He is widely
regarded as "The Father of the Mesenchymal Stem Cell". Dr. Caplan has
served as Chief Scientific Officer and founder of Osiris Therapeutics,Inc., co-founded Cell
Targeting Inc., and has served as Chief Scientific Officer of OrthoCyte Corporation, a subsidiary
of BioTime, Inc. He is active in developing clinical translational therapies and advocating
pathways
for
realizing
cell
therapeutic
regenerative
medicine.
Learn
more:
http://www.case.edu/artsci/biol/skeletal.
Symposium Keynote Speakers:
Eben Alsberg, Ph.D.
Case Western Reserve University
Dr. Alsberg took a faculty position in 2005 at Case Western Reserve
University, where he is currently an associate professor of Biomedical
Engineering and Orthopaedic Surgery and serves as Director of the Stem
Cell and Engineered Novel Therapeutics Laboratory. His lab focuses on
the engineering of new technologies to regenerate tissues and treat
diseases through the development of novel bio-materials and microenvironments. He’s co-authored over 75 peer-reviewed papers and book
chapters and 130 abstracts and conference proceedings. His work has
been recognized with the 2008 Ellison Medical Foundation New Scholar
in Aging Award and the Crain’s Cleveland Business 2009 Forty Under 40 Award. The NIH,
DOD, NSF, the Ellison Medical Foundation, the Coulter Foundation, the Musculoskeletal
Transplant Foundation, the State of Ohio and the AO Foundation have supported his lab’s
research. Learn more:http://bme.case.edu/alsberg.
Steve Stice, Ph.D.
University of Georgia
Dr. Steve Stice is a Georgia Research Alliance Eminent Scholar endowed
chair, Professor and Director of the Regenerative Bioscience Center at
the University of Georgia. He serves as the Director of the Regenerative
Engineering and Medicine partnership and also serves as Chief Scientific
Officer for ArunA Biomedical Inc. Dr. Stice co-founded five
biotechnology companies, including Advanced Cell Technology, and
CytoGenesis, Inc., which was later purchased by BresaGen. Throughout
his career, he has published and lectured on cloning and stem cell
technologies. His research focuses on developing innovative animal
cloning and stem cell technologies. He is also developing large animal models for use in
neurological and orthopedic transitioning from preclinical studies in rodents to human clinical
trials. Learn more:http://stice.uga.edu.
David C. Hess, M.D.
Georgia Regents University
David C. Hess holds the Presidential Distinguished Chair and is the Chair
of the Department of Neurology at MCG of GRU and Clinical Chair of
the Brain and Behavior Discovery Institute at GRU. He has been part of
the STEPS 1, 2, and 3 meetings to develop recommendations for clinical
trials of cell therapies in stroke. Dr. Hess has worked in both the pre-clinical arena and in clinical
trial design of cell therapies. He has worked closely with Athersys, Inc on the MASTERS clinical
trial in stroke. Dr Hess is a specialist in Cerebrovascular Disease and Stroke. He is actively
involved in the MCG Stroke Program Stroke Service, and sees outpatients with a wide variety of
neurological problems. Dr Hess's research interests include the use of bone marrow-derived stem
cells to regenerate the brain after stroke; and the planning and coordination of clinical trials to
prevent stroke. Learn more: http://www.gru.edu/mcg/neurology.
Johnna Temenoff, Ph.D.
Georgia Institute of Technology
Dr. Temenoff is currently an Associate Professor in the Coulter
Department of Biomedical Engineering at Georgia Tech/Emory
University in Atlanta, GA. Over the course of her career to date, Dr.
Temenoff has published over 40 peer-reviewed papers and 10 book
chapters, co-authored a biomaterials textbook published by PearsonPrentice Hall, and has produced ~90 scientific abstracts for national and
international conferences. Dr. Temenoff has received funding from a
wide range of sources, including federal agencies (NIH and NSF) and
groups such as the Aircast Foundation and National Football League
Charities, and currently serves at the Principle Investigator on a NIH T32 Predoctoral Training
grant in Biomaterials. She has been honored with several prestigious awards, such as AIMBE
Fellow, the NSF CAREER Award, and the Arthritis Foundation Investigator Award, and was
recently named the Co-Director for the Regenerative Engineering and Medicine Center, a
statewide initiative encompassing Georgia Tech, U. Georgia, and Emory University. Learn more:
http://temenoff.gatech.edu.
Liisa Kuhn, Ph.D.
University of Connecticut Health Center
Dr. Liisa Kuhn is a tenured Associate Professor in the Reconstructive
Sciences Department at the University of Connecticut Health Center
(UConn Health) and a faculty member of the UConn Biomedical
Engineering Department. Prior to joining the faculty of UConn in 2002,
Dr. Kuhn was Director of Orthopaedics Product Development for ETEX
Corporation in Boston, MA, and Director of Development for
NaturApatites Co., Inc., a bone graft substitute company which she cofounded. Her research in orthopedic biomaterials and drug delivery
systems has been supported by federal (NIH), state and private
foundations (e.g. Coulter Foundation, Komen Foundation). For her distinguished service and
exceptional contributions to medical product standards writing within the American Society of
Testing and Materials (ATSM), Dr. Kuhn been recognized with ASTM's top three awards,
including, in 2014, the Award of Merit with an accompanying honorary title of Fellow. Learn
more: http://kuhnbiomaterials.uconn.edu
April 24th 2015 Symposium Program:
8:00 Registration
•
Coffee & pastry
8:30 Opening remarks:
•
•
•
Greeting & Introduction: William D. Hill, Ph.D, GRU
Welcome IRRM: Carlos M. Isales, M.D. Director of IRRM
Welcome GRU: Michael Diamond, M.D. GRU VP for Research and Director of Clinical
& Translational Research
8:45 Meeting Keynote Lecture: Dr. Arnold I. Caplan, PhD - Case Western Reserve University:
"Adult MSCs: new logics for a new kind of Medicine"
9:45 - 10:40 - Engineered Stem Cell Microenvironments 1
•
9:45 Keynote Lecture: Eben Alsberg, Ph.D. - Case Western Reserve University:
"Inductive high-density stem cell systems for tissue regeneration"
•
10:15 Edward A. Botchwey, Ph.D. - Georgia Institute of Technology: "Sphingosine-1phosphate receptor-3 regulates hematopoietic stem cell retention in the bone marrow
niche"
10:35 Coffee break
10:55 - 11:50 - Engineered Stem Cell Microenvironments 2
•
10:55 Keynote Lecture: Johnna Temenoff, Ph.D. - Georgia Institute of Technology:
"Glycosaminoglycan-based Biomaterials for Protein Delivery in Orthopaedics"
•
11:25 Lohitash Karumbaiah, Ph.D. – University of Georgia: "Trophic Factor Enriching
Chondroitin Sulfate Glycosaminoglycan-Based Hydrogels for TBI"
11:45 - 12:15 - Tissue Engineered Structure Standards
•
11:45 Keynote Lecture: Lissa T. Kuhn, Ph.D. - University of Connecticut Health Center:
"Standardization of tissue engineered medical products from an ASTMperspective"
12:15 Lunch & Poster Session
1:30 - 3:00 - Pre-Clinical Trials in Large Animal Models
•
1:30 Keynote Lecture: Dr. Steve Stice, Ph.D. - Univ. of Georgia: "Dual purpose non
rodent models for human and animal medicine"
•
2:00 John F. Peroni, DVM, MS, Dip ACUS - Univ. of Georgia: "A practical review of
ovine fracture models for the purpose of bone repair."
•
2:20 Frank West, Ph.D. - Univ. of Georgia: "Regenerative Neural Stem Cell Therapy in a
Pig Stroke Model."
•
2:40 Samuel P. Franklin, DVM/Ph.D. - Univ. of Georgia: "Comparison of PRP and
Stromal Vascular Fraction Supplemented with a Novel Nanofiber Polymer for the
Treatment of Cartilage Pathology in Dogs"
3:00 Coffee break
3:20 – 5:00 Clinical Trials
•
3:20 Keynote Lecture: David C. Hess, M.D. - Georgia Regents University: "Cell
Therapy for Stroke: Results from early phase clinical trials"
•
3:50 Harold Solomon – Georgia Venture Lab: "Funding the Lean Approach to
Commercializing Health Technologies"
•
4:10 Changwon Park, Ph.D. – Emory University: "Function of endothelial ETV2 in
angiogenesis"
•
4:30 Adam Berman, M.D. - Georgia Regents University: "IxCell-DCM Clinical Trial and
Cardiac Regenerative Medicine Research"
•
4:50 Ayman Al-Hendy, M.D. - Georgia Regents University: "Gene and Stem cell
Therapy of Premature Ovarian Failure"
5:10 Closing Comments
5:15 – 6:00 Poster Session and hors d'oeuvres
Poster 1
Efficacy of Local Delivery of Mesenchymal Stem Cells for Large Animal Tendinopathy
Alexandra Scharf1, Shannon Holmes1,2, Merrilee Thoresen1, Jennifer Mumaw1, and John Peroni1
1
Department of Large Animal Medicine and Surgery, College of Veterinary Medicine, University
of Georgia 30605
2
Diagnostic Imaging, College of Veterinary Medicine, University of Georgia 30605
Tendinopathy is a career debilitating disease with a high rate of morbidity in both human
and equine athletes. The overall goal of our research is to enhance flexor tendon repair by the
local delivery of mesenchymal stem cell (MSC) therapy. Our hypothesis is that the therapeutic
benefits of MSCs correlate to the efficacy of cell delivery and cell survival. The goal of this study
was to a) evaluate the efficacy of MSC delivery following local delivery, with and without
ultrasound guidance and b) determine the degree of migration and persistence of cells over time.
Images of equine and ovine tendon lesions were acquired using a 1.5 Tesla Magnetic
Resonance Imaging (MRI) scanner and knee coil. These lesions were localized to the superficial
and deep digital flexor tendons in equine and ovine subjects, respectively. Lesions were treated
with 10*106 cells labeled with superparamagnetic iron oxide nanoparticles (SPIOs), which
produce a magnetic field perturbation detectable by MRI. Previous studies have been performed to
validate the biosafety and optimize imaging parameters for detection of labeled cells. Subjects
were imaged at the time of injection, 2 weeks, 6 weeks, and/or 4 months post-injection to assess
the location of cells and degree of healing.
The results of this study suggest that even under ultrasound guidance, localized cell
injections do not ensure that the entire payload is delivered to the site of injury. Of notable
concern is that the cells delivered to the paratenon and surrounding tissue demonstrate little
migratory capability. Any cells that do not make it into the lesion upon injection are unlikely
to migrate to the site of injury over time. Further studies need to be performed to determine if
the location of cells influences their survival and degree of healing. Future work on this project
will use histological and diagnostic imaging data to further evaluate the relationship between cell
delivery and tendon healing that can be obtained at 6 weeks and 4 months post-injection.
Poster 2
Chondroitin Sulfate Glycosaminoglycan Hydrogel-Based Neural Stem Cell Carriers for
Traumatic Brain Injury
Martha I. Betancur , Melissa Albarado , Meghan Logun , Ravi Bellamkonda , Lohitash
Karumbaiah
Regenerative Bioscience Center, Department of Animal and Dairy Science at The University of Georgia, Athens
GA 30602.
Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology, Atlanta, GA 30332
Chondroitin sulfate glycosaminoglycans (CS-GAGs) are integral components of the neural stem
cell (NSC) niche extracellular matrix (ECM), where they play key roles in the maintenance of
NSC self-renewal. We therefore hypothesized that the natural affinity of neurotrophins, stem cell
self- renewal, and differentiation inducing factors to CS-GAGs will help increase NSCs survival,
and help facilitate acute neuroprotection, and targeted cell differentiation when transplanted in a
rodent moderate traumatic brain injury (TBI) model. To achieve this, we designed CS-GAG
based hydrogels capable of carrying NSCs. We believe that CS-GAG based hydrogels have
significant advantages over synthetic polymer – based hydrogels owing to their ability to provide
biological recognition and function, and undergo natural degradation to facilitate tissue
remodeling. To test our hypothesis, we developed a moderate to severe TBI model and a
hydrogel intracortical injection model for histological and behavioral evaluation after treatment
with NSC laden CS-GAG hydrogels. Using a pneumatically actuated controlled cortical impactor
(CCI), we induced injuries of 3mm diameter by 2mm depth in a repeatable manner, using a
velocity of 2m/s, and a dwell time of 250 milliseconds. Two days after receiving a TBI, rats were
injected directly into the site of injury with CS-GAG hydrogels containing PKH26GL labeled
NSCs, and the trophic factors bFGF and BDNF. Histological analysis at 4 weeks post-injury
shows a significant difference in injury size between TBI control and TBI treated with CS-GAG
containing NSCs. In addition, quantification of cell survival shows a significant increase in cell
survival when NSCs were implanted in CS-GAG hydrogels when compared to cells implanted
with no hydrogel. The evaluation of cell survival at 12 weeks and the assessment of functional
recovery using standardized behavioral tests are currently ongoing.
Figure 1.
Figure 2.
Figure 1. CS-GAG hydrogel implants containing
NSCs and trophic factors significantly reduce
neural tissue loss. Cresyl violet stained coronal
sections taken at the epicenter of the injury were
imaged and quantified at the region of interest
(ROI) indicated by the white boxes. The percent
area occupied by neurons was quantified using
Image J. A pair wise comparison to the TBI group
using one-way ANOVA and a post hock Tukey
HSD Pairwise Comparison revealed a significantly
greater present of neuronal tissue area in the TBI
group treated with CS-GAG hydrogel encapsulated
NSCs+TF, as compare to the no-treatment TBI
group and the TBI-NSCs only group.
Figure 2. CS-GAG hydrogel implants containing
NSCs and trophic factors demonstrate a
significant increase in NSC presence within the
lesion 4 weeks post injury. Sections stained for
neurons (green) shows NeuN+ cells within injection
site. Zoomed in box of an area of the injury reveals
a large number of red PKH26GL+ NSCs.
Quantification shows that the percent PKH26GL+
area is greatest for group implanted with NSCs in
the CS-GAG-TF scaffold. A pairwise comparison to
the TBI-NSCs group using one-way ANOVA and a
posthock Tukey HSD Pairwise Comparison
revealed a significant difference between NSCsonly control and the two GAG-hydrogel-NSC
treatments.
Poster 3
GRU Regenerative Medicine and Cellular Therapy Research Symposium, 2015: Abstract
sHER3 Inhibits the Proliferation and Migration of Melanoma-derived Cells in a Tenascindependent Manner.
Chunlin Cai, Mojun Zhu*, Surendra Rajpurohit and Nita Maihle
Cancer Center, Georgia Regents University, Augusta, Georgia and *Yale School of Medicine, New
Haven, CT
The HER3 receptor tyrosine kinase is a member of the EGFR/HER family of cellular oncogenes.
While this receptor family has generally received wide attention and clinical success as a target in the
development of biologically-targeted cancer therapeutics, drugs targeting the HER receptor axis have to
date not been successful in the treatment of melanoma. Nonetheless, recent studies have shown that
HER3 receptor expression is common in both primary and metastatic melanoma, and that high levels of
HER3 expression are correlated with poor patient survival. However, no one has demonstrated the
mechanistic basis for HER3 as a poor prognostic determinant in melanoma patients, even though these
tumors poorly express other members of the EGFR/HER receptor family. In this study we present
evidence demonstrating that 13 of 14 cell lines tested express HER3, supporting previous reports
regarding the frequency of expression of this gene product in melanoma samples. We further report that
a soluble isoform of HER3 (sHER3) that we previously have shown to be a potent inhibitor of
neuregulin mediated, Akt-dependent breast cancer cell growth strongly inhibits the growth of HER3
expressing melanoma cells. Interestingly, however, the phosphorylation of Akt, a known mediator of
ligand-dependent HER3 signaling is not reduced in melanoma cells growth inhibited by sHER3. Since
the HER3 receptor lacks endogenous kinase activity by itself, it is typically dependent on co-expression
of other HER receptor family members for promotion of cancer cell growth. Together, these
observations suggested to us that the mechanism(s) by which HER3 promotes melanoma cell growth
and metastasis may be distinct. To explore alternative mechanisms of HER3 signaling in melanoma, we
identified sHER3 interacting partners using a mass spectrometry proteomic approach. Only one
prominent sHER3 binding partner was identified in melanoma cell conditioned media: tenascin. This
was an unexpected finding since ligand-dependent HER3 signaling has not previously been linked to
tenascin. Interestingly, tenascin expression has independently been identified as another negative
prognostic indicator in melanoma. Our initial studies to test the functional relationship between HER3
and tenascin suggest that tenascin expression is correlated with both HER3 expression and migration in
all of the melanoma lines tested. Moreover, sHER3 inhibition of melanoma migration was dependent on
the level of tenascin present. Treatment of melanoma cells with purified sHER3 inhibits the migration
only in the presence of high levels of tenascin. These results suggest the existence of an unanticipated
tenascin- and HER3-dependent signaling pathway in melanoma cells, and also suggest that this pathway
is functionally correlated with melanoma cell migratory potential and poor patient survival.
Since
tenascin expression has recently been shown to define a stem cell niche in other cancers, and also to
provide a protective signal in a therapy-resistant population of melanoma cells, together these results
identify a novel and targetable Achilles heel for prevention of melanoma cell growth and metastasis.
(These studies were supported by the Harry J. Lloyd Charitable Trust).
Poster 4
Role of the Hematopoietic Stem Cell During Osteogenesis and Fracture Repair
RR Kelly 1,2,3, MA McCrackin1,4, KR Wilson1,2, M Mehrotra1,2,3, AC LaRue1,2,3
1
Research Services, Ralph H. Johnson VAMC, 2Department of Pathology and Laboratory Medicine,
3
Hollings Cancer Center and Department of Comparative Medicine4, Medical University of South
Carolina, Charleston, South Carolina
This work is supported in part by the Biomedical Laboratory Research and Development Program* of
the Department of Veterans Affairs (ACL) and NIH/NCI RO1 CA148772 (ACL)
*The contents of this abstract do not represent the views of the Department of Veterans Affairs
or the United States Government.
Mesenchymal stem cells (MSC) have been the “gold standard” for cell-based fracture treatment.
However, multiple MSC-based trials have been hampered by low engraftment rates and an inability to
effectively and consistently isolate this population. Our studies and others have suggested
hematopoietic stem cells (HSCs) may also contribute to the osteogenic lineage. Using a single cellbased transplantation model, we showed that the HSC gives rise to osteoblasts, osteocytes, and
hypertrophic chondrocytes during non-stabilized fracture repair. We hypothesize that these HSCderived cells can be exploited to enhance fracture repair. In vitro assays were first conducted to identify
factors to promote osteogenesis from HSC-derived progenitors. Mineralized colonies formed when nonadherent BM, the fraction enriched for HSCs, or adherent BM, the fraction enriched for MSCs, was
cultured under osteogenic conditions. Exogenous BMP-2 and BMP-9 had a synergistic effect on this
mineralization. We then sought to determine if HSC-derived osteogenic precursors (CD34+OCN+)
could be mobilized from the bone marrow and found that delivery of AMD3100, a CXCR4 antagonist,
resulted in a 2-fold increase in CD34+OCN+ cells in circulation. Our ultimate goal is to test the
functional impact of HSC mobilization and/or delivery of pro-osteogenic factors on fracture healing in
both non-stabilized and atrophic non-union fracture. Towards this, we have generated an in vivo murine
model of surgical non-union. Preliminary X-ray and micro-CT analyses demonstrate delayed healing in
a subset of animals. Further longitudinal and histomorphometric analyses are underway to confirm nonunion. Animals with non-stabilized fracture or non-union will then be randomized to control (no
intervention) or experimental (+/-BMP, +/-AMD3100, +BMP/AMD3100) groups and temporal healing
assessed and correlated to HSC-derived osteogenesis. Given that the HSC is an earlier, more easily
mobilized, and better defined stem cell than the MSC, it may prove a more efficacious therapy for
treating fractures, particularly difficult to heal non-unions.
Poster 5
Hematopoietic Stem Cells Give Rise to Chondrocytes Through a Monocytic Precursor
Lineage
!
KR Wilson1,2,3, Y Xiong1,2,4, RR Kelly 1,2,3, AC LaRue1,2,4
1
Research Services, Ralph H. Johnson VAMC, 2Department of Pathology and Laboratory
Medicine, 3College of Graduate Studies, MUSC and 4Hollings Cancer Center, MUSC
This work is supported in part by the Biomedical Laboratory Research and Development
Program* of the Department of Veterans Affairs (ACL) and NIH/NCI RO1 CA148772 (ACL)
*The contents of this abstract do not represent the views of the Department of Veterans Affairs
or the United States Government.
Bone marrow consists of two types of stem cell populations, the mesenchymal stromal
cell (MSC) and the hematopoietic stem cell (HSC). While MSCs have been
demonstrated to have the capacity of differentiating into osteoblasts, chondrocytes, and
adipoctyes, recent studies are beginning to delve into the possibility of hematopoietic
stem cells also having this differential capability. Our laboratory has demonstrated an
HSC origin for cells such as osteoblasts, cancer-associated fibroblasts, and immature
adipocytes, revealing the ability of HSCs to give rise to cell types not typically
associated with these lineages. Of particular relevance to this study are our in vivo
studies which demonstrated that HSCs can give rise to hypertrophic chondrogenic cells
during non-stabilized fracture repair using a clonal cell transplantation model. This work
has led to the hypothesis that chondrocytes differentiate through an HSC lineage. To
initially address this hypothesis, in vitro studies of culture conditions for HSC-derived
chondroprogenitors was first elucidated. Past research has indicated an HSC and
myeloid lineage for other cells such as osteoblasts/cytes and adipocytes, respectively.
For this reason, the chondrogenic potential of monocytic precursor-derived cells was
examined in an effort to explore a new source of cells to be considered for cartilage
regeneration. Alcian Blue staining was used as an initial indicator of chondrogenic
potential in determining glycosaminoglycan production. In addition to this, nodule
formation was assessed in cultures in serum containing chondrogenic media. More
specific studies involved monocytic precursor cells cultured in serum-free conditions
and directed towards and chondrogenic lineage using TGF-β1. immunofluroescent
expression of cartilage specific markers Aggrecan and Collagen II after induction
indicated differentiation along with the negative expression of F4/80, a monocytic
marker. Determining the mechanisms behind HSC contribution to chondrogenic
lineages and the associated process of differentiation/maturation in future work has the
potential to enhance stem cell therapies for cartilage repair.
Poster 6
Development and Characterization of a Piglet Cortical Impact
Traumatic Brain Injury Model
Emily L. Wyatta,b, Holly A. Kindera,b, Jessica M. Hutchesona,b, Elizabeth W. Howerthc, W.
Matthew Hendersond, Kylee J. Dubersteina,b, and Franklin D. Westa,b
a
c
Regenerative Bioscience Center, bDepartment of Animal and Dairy Science,
Department of Veterinary Pathology, dU.S. Environmental Protection Agency
University of Georgia, Athens, GA 30602, USA
Traumatic brain injury (TBI) is a leading cause of death and long-term disability among
persons in the United States with toddler-aged children being one of the most affected age
groups. Despite the number of people who are affected by TBI, an FDA-approved treatment
remains elusive. Stem cell therapies offer new therapeutic potential as a treatment for brain
injury by producing regenerative and anti-inflammatory growth factors a well as functioning
as a cell replacement therapy. One of the difficulties in developing effective treatments for
TBI has been the poor translatability of therapies from the widely-used rodent model to
human patients. Therefore, the establishment of a more human-like and thus more predictive
animal model is crucial for the development of cell therapies. The piglet brain is more similar
to humans in anatomy and physiology with respect to neurodevelopmental sequence, size,
gyral pattern, and gray to white matter ratio making it an excellent candidate as a TBI model.
The objective of this study was to determine changes in lesion size, brain metabolism,
cellular composition, and motor function in piglets that experienced a controlled cortical
impact (CCI) at increasing impact velocities and penetration depths. In this study we
assessed 4 treatment groups: (1) 2 m/s and 6 mm, (2) 4 m/s and 6mm, (3) 4m/s and 12mm,
and (4) 4 m/s and 15mm impact velocity:penetration depth. Study results demonstrated a
direct correlation between increasing impact velocity and penetration depth with increased
brain lesion size, changes in brain metabolism, gliosis, and motor function deficits. At the
tissue level, gross histology showed a significant increase in brain lesion size as impact
velocity and penetration depth was increased. Animals receiving a CCI at 4 m/s impact
velocity with a 15mm penetration depth had the largest lesion volume of 1236 mm3 one week
post-TBI. At the cellular level, metabolomic analyses showed that there were significant
changes in brain metabolites after injury; and furthermore, gray and white matter in the piglet
brain exhibited differential responses to injury. Gliosis was quantified through
immunohistochemistical analysis of the GFAP astrocyte and Olig2 oligodendrocyte protein
markers. One week post-TBI, the amount of GFAP-positive astrocytes was significantly
increased on the ipsilateral hemisphere relative to the contralateral hemisphere in piglets
receiving a 4 m/s CCI with 6mm and 12mm penetration depths, while global increases of
Olig2-postive oligodendrocytes were noted in piglets receiving a 4 m/s CCI with 12mm and
15mm penetration depths. At the functional level, analysis of motor function kinematics in
piglets before and after injury showed that TBI resulted in motor instability characterized by
increased stance time and preference to remain in 3-limb support versus 2-limb support. This
study is the first to demonstrate that increasing velocity and penetration depth in a CCI piglet
TBI model results in progressive deficits at the cellular, tissue, and functional levels. These
quantifiable and reproducible changes in the piglet TBI model make it an attractive platform
for the testing and development of novel cell therapies.
Poster 7
!
FIRST! REPORT! OF! A! POTENTIALLY! NOVEL! ONCOGENIC! MECHANISM!
AND!THERAPEUTIC!TARGET!IN!ANAPLASTIC!THYROID!CANCER!
!
Leslie! Peard! BS1,! DeHuang! Guo! PhD1,2,! Kamran! Mohammed! BS2,!
Tiffany!Coleman!BS1,2,!and!Paul!Weinberger!MD1,2,3!
1
Department!of!Otolaryngology,!Medical!College!of!Georgia,!Georgia!Regents!
2
University;! Center! for! Biotechnology! and! Genomic! Medicine,! Georgia!
3
Regents! University,! and! GRU! Cancer! Center,! Georgia! Regents,! University,!
Augusta,!GA!
!
BACKGROUND:"Anaplastic"thyroid"cancer"(ATC)"is"a"rare"form"of"thyroid"
carcinoma"with"extremely"high"morbidity"and"fast"disease"progression."
There" are" no" effective" treatments," and" 5@year" overall" survival" is" 4.3%.""
We" and" others" have" noted" elevated" expression" of" keratin" 8" (CK8)" in"
several"ATC"cell"lines,"most"notably"the"highly"aggressive"lines"with"fast"
population" doubling" time" (Td)." Keratins" are" intermediate" filament"
proteins"with"known"role"as"a"structural"member"of"the"cytoskeleton"in"
epithelial"cells."METHODS:"Highly"characterized"thyroid"cancer"cell"lines"
were" obtained." Western" blot" and" immunohistochemistry" was" used" to"
determine" CK8" expression." Using" a" stable" lentiviral@CK8" shRNA"
construct," we" performed" a" knockdown" of" CK8" in" ATC1" cells." Following"
puromycin" selection," culture" wells" were" imaged" daily" to" determine"
effect." RESULTS:" Fast" growing" thyroid" cancer" cell" lines" ATC1," 29T," and"
11T" had" elevated" expression" of" CK8." Slow" growing" cell" lines" 16T,"
FTC133,"and"11T"had"undetectable"CK8"levels."In"ATC1"cells,"scrambled"
lentivirus" controls" demonstrated" expected" recovery" and" proliferation"
following" infection." Wells" with" no" virus" (negative" control)" underwent"
apoptosis." The" CK8" lentivirus" wells" showed" near" total" growth" arrest" of"
ATC" cells." CONCLUSION:" Rather" than" simply" being" a" biomarker" for"
epithelial"cancer,"CK8"may"play"a"direct"role"in"anaplastic"thyroid"cancer"
progression."If"true,"this"represents"a"novel"mechanism"of"action"and"a"
potentially"druggable"target"for"a"disease"that"currently"has"no"cure."
Poster 8
Assessment of Learning and Memory in a Piglet Model
Holly A. Kinder1,2, Emily L. Wyatt1,2, Franklin D. West1,2
1
Regenerative Bioscience Center, 2Department of Animal and Dairy Science,
University of Georgia, Athens GA, 30622
The use of the piglet in studying neurodevelopment has become of increased interest due to
similarities in brain structure and development to toddler-aged children. Several neural injury and
disease models have recently been developed creating a need for further validation of behavioral
tests to better assess learning and memory in piglet models. The present study evaluated the ability
of 5 week-old piglets to perform 4 behavioral tests, a spatial T-maze test, an object recognition test,
a social recognition test, and an open field test, to assess different aspects of cognition including
learning and spatial memory, spontaneous trial-unique memory, social memory, and
normal/abnormal behaviors, respectively. For spatial-T maze testing, piglets were trained to locate a
food reward at a constant place in space by using extra-visual cues, despite starting the task at
alternating locations. Piglets were significantly faster at making a reward arm choice by day 5 (p<
.05), indicating acquisition of the task. Additionally, piglets reached a performance criterion of at
least 80% correct reward arm choices by day 4 (p<0.05), indicating that piglets were able to create
and use spatial memories to find a food reward. Correct choices decreased after reversal of the
reward, but improved after several days of testing (p <0 .05). In object recognition testing, pigs were
exposed to two similar objects in a sample trial, followed by a 10 minute inter-phase interval, and
then exposed to one familiar and one novel object in a test trial. Piglets explored the novel object
significantly more than the familiar object (p<0.05) in the test trial. Additionally, piglets explored the
familiar object significantly less in the test trial than in the sample trial (p<0.05), indicating the piglet
formed a spontaneous trial-unique memory of the familiar object. In the social recognition test,
piglets were exposed to an unfamiliar pig and a novel object in a sociability test, followed by a 10
minute inter-phase interval, and then exposed to a familiar pig and a novel pig in a social recognition
test. Piglets spent significantly more time with the unfamiliar pig in the sociability trial than the novel
object (p<0.05) and tended to spend more time with the novel pig in the social recognition trial than
the familiar pig (p=0.01) indicating that the piglet had retained a social memory of the familiar piglet.
Finally, in the open field test, piglets were placed in an open arena for 10 minutes to monitor different
aspects of behavior such as ambulation, exploratory interest, and anxiety. Piglets generally explored
fewer zones and became less ambulatory over time, demonstrated significantly decreased
exploratory interest over time (p<0.05), and generally became less anxious over time. This test
overall suggests that piglets became habituated to the testing arena, a characteristic of normal piglet
behavior. Taken, together these tests provide an ideal platform with which to continue behavioral
testing in neural injury and disease models and to assess the effectiveness of different treatments on
improving learning, memory, and behavior.
Poster 9
A novel TNF-derived peptide activating alveolar liquid
clearance during acute lung injury
I. Czikora1, A. Alli2, D. Kaftan3, S. Sridhar1, Z. Bagi1, T. Chakraborty4,
D.C. Eaton2 and R. Lucas1.
1
Georgia Regents University - Augusta, GA/US, 2Emory University
School of Medicine - Atlanta, GA/US, 3University of South BohemiaCeske Budejovice/CR, 4Justus-Liebig University - Giessen/DE.
(Supported by an AHA post doc award to IC)
RATIONALE. Alveolar liquid clearance (ALC) is regulated by vectorial
Na+ transport through the apically expressed epithelial sodium channel
(ENaC) and basolateral Na+-K+-ATPase in type II alveolar epithelial
cells. ALC is critical to prevent life-threatening pulmonary edema
formation in the alveoli, for which no standard treatment exists to
date. Since the pneumococcal virulence factor pneumolysin impairs
ENaC activity1, it is of the utmost clinical importance to identify novel
mechanisms that increase the channel’s function, defined by both its
expression level and its open probability time, during pneumonia. The
lectin-like domain of the cytokine TNF, mimicked by the 17 residue TIP
peptide, activates ENaC-dependent Na+ uptake2-4. This study was
designed to determine the mechanism(s) by which the peptide
increases ENaC activity in the absence and presence of the
pneumococcal
pore-forming
toxin
pneumolysin.
RESULTS. The TIP peptide, upon caveolae-dependent internalization,
binds to the intracellular carboxy-terminal domain of the alpha subunit
of ENaC4, thus increasing the open probability of the channel. By
inhibiting pneumolysin-mediated activation of the enzymes protein
kinase C-alpha and ERK, which are known to blunt ENaC open
probability and expression, respectively, the lectin-like domain of TNF
restores the channel’s activity in H441 cells. As suggested by a
molecular docking study using ASIC-1 as a template4, mutation of
ENaC-alpha residues Val567, Glu568 and Glu571 to Ala significantly
blunted its interaction with the TIP peptide, indicating that these
amino acids are crucial for ENaC activation by TNF. The TIP peptide
significantly increased ALC in patients with acute lung injury in a
recent phase 2a clinical trial5.
References. 1Lucas et al., PNAS 2012; 2Lucas et al., Science 1994;
3
Elia et al., AJRCCM 2003; 4Czikora et al., AJRCCM 2014.5Sieck and
Wylann, editorial, AJRCCM 2014.
Poster 10
GILZ%Protects%TNF0α0induced%Bone%Loss%in%Mice%
Nianlan%Yang1,%Babak%Baban2,%William%D.%Hill3,%Mark%W.%Hamrick3,%Carlos%M.%Isales1,%4,%Xing0Ming%Shi1,%5%%
Departments%of%Neuroscience%&%Regenerative%Medicine1,%Pathology5,%Oral%Biology2,%Cellular%Biology%and%
Anatomy3,%Orthopaedic%Surgery4,%Georgia%Regents%University,%Augusta,%GA%USA%
%
Background:%Tumor%necrosis%factor0alpha%(TNF0α)%plays%a%key%role%in%the%pathogenesis%of%inflammatory%
diseases%such%as%rheumatoid%arthritis%(RA).%It%is%known%that%chronic%inflammation%causes%bone%loss.%We%
showed%previously%that%glucocorticoid%(GC)0induced%leucine%zipper%(GILZ),%a%GC%anti0inflammatory%effect%
mediator,%can%enhance%osteogenic%differentiation%of%bone%marrow%mesenchymal%stem%cells%(MSCs),%and%
antagonize%the%inhibitory%effect%of%TNF0a%on%MSC%differentiation.%However,%it%is%not%known%whether%GILZ%
will%have%the%same%effects%in%vivo%and%thus%serve%as%a%potential%new%anti0arthritis%drug%candidate.%%
Methods:%We%created%TNF0α:GILZ%double%transgenic%mice%by%crossing%a%human%TNF0α0expressing%
transgenic%mouse%with%a%GILZ%transgenic%mouse,%in%which%the%expression%of%GILZ%is%under%the%control%of%a%
3.6kb%type%I%collagen%promoter%fragment%(Col3.6).%The%expression%of%TNF0α%and%GILZ%in%double%transgenic%
mice%was%confirmed%by%RT0PCR,%Western%blot%and%ELISA,%and%the%bone%protective%effects%of%GILZ%were%
assessed%with%standard%bone%biology%and%histology%techniques.%%
Results:%%Overexpression%of%GILZ%blocked%TNF0α0mediated%bone%loss%in%double%transgenic%mice%as%
demonstrated%by%a%significantly%increased%bone%mineral%density%(BMD)%in%the%femur%(+13.5%,%p<0.01),%
tibia%(+12.9%,%p<0.05)%and%vertebra%(+14.5%,%p<0.05)%compared%with%that%in%the%TNF0α%transgenic%mice.%
GILZ%also%reduced%the%expression%levels%of%IL06%mRNA%and%serum%levels%of%RANKL,%as%well%as%the%levels%of%
overexpressed%human%TNF0α.%%Furthermore,%GILZ%antagonized%the%inhibitory%effect%of%TNF0α%on%
osteocalcin%expression%in%GILZ0TNF0α%double%transgenic%mice.%%As%predicted,%the%TNF0α%transgenic%mice%
developed%poly%arthritic%symptoms%between%week%809%along%with%significant%body%weight%loss%and%bone%
loss.%The%double%transgenic%mice,%somehow,%showed%little%effect%on%inflammatory%arthritic%symptoms%
compared%with%the%TNF0α%transgenic%mice.%This%could%be%due%to%the%restricted%GILZ%transgene%expression%
in%osteoblastic%lineage%cells%and%ubiquitous%and%high%level%of%TNF0α%transgene%expression%in%double%
transgenic%mice%because%significant%reductions%of%TNF0α%was%only%observed%in%bone%marrow%
mesenchymal%lineage%cells%(043.5%)%but%not%in%hematopoietic%(or%osteoclastic)%lineage%cells.%%
Conclusion:%Overexpression%of%GILZ%in%osteoprogenitor%cells%protects%bone%loss%in%mice%with%chronically%
elevated%levels%of%proinflammatory%cytokines%such%as%TNF0α.%%
Poster 11
Repurposing Dimethylfumarate for Parkinson's disease- Preclinical evidence
Manuj Ahuja1, Navneet Ammal Kaidery1, Lichuan Yang1,7, Noel Calingasan3, Natalia Smirnova4, Arsen
Gaysin5, Irina Gaysina6, Takao Iwawaki8, John Morgan2, Rajiv Ratan4, Irina Gazaryan4, Anatoly
Starkov3, Flint Beal3, Bobby Thomas1,2,*
1
Departments of Pharmacology & Toxicology and 2Neurology, Medical College of Georgia, Georgia
Regents University, Augusta, Georgia
3
Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, New York
4
Burke Medical Research Institute, White Plains, New York
5
Department of --- Northwestern University, Chicago
6
Department of Medicinal Chemistry, University of Illinois
7
Kunmig Biomed, China
8
Gunma University, Japan
Targeting oxidative stress either by providing exogenous antioxidants or by enhancing the endogenous
antioxidative capacity has been intensely investigated for PD therapies. The latter includes the activation
of nuclear-factor-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway which
regulates the expression of a battery of genes encoding anti-oxidative, anti-inflammatory, and cytoprotective genes. Tecfidera is an oral formulation of dimethylfumarate (DMF) approved for Multiple
sclerosis based on its promising beneficial effects. Fumaric acid esters such as dimethyl and mono-methyl
fumarate have been found to exert neuroprotective effects by activating the Nrf2/ARE signaling pathway.
We investigated in vivo pharmacokinetics, effects on Nrf2/ARE signaling both in vitro and in vivo and its
ability to block 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxicity and associated oxidative
damage, impaired mitochondrial function, and inflammation in mice. We found that dimethyl and monomethylfumarate selectively activate Nrf2 pathway using Neh2-luciferase reporter, and OK48-luc reporter
transgenic mice. Assessment of mRNA and protein levels showed upregulation of several cytoprotective
and antioxidative genes in discrete mouse brain regions commensurate with DMF pharmacologic levels in
vivo, and in wild type mouse embryonic fibroblasts but not in Nrf2 null fibroblasts in vitro. Oral administration of both dimethyl and mono-methylfumarate at (10, 25, 50, and 100mg/kg) dose dependently protected against acute MPTP neurotoxicity assessed by stereological cell counts of total and tyrosine
hydroxylase positive neurons of substantia nigra and striatal levels of catecholamines, employing HPLC
electrochemistry, in wild type but not Nrf2 null mice. Dimethyl and mono-methylfumarate blocked
against MPTP-induced oxidative damage assessed by 3-nitrotyrosine and inflammation determined CD68
immunoreactivity and expression of pro-inflammatory cytokines in the midbrains. Both dimethyl and
monomethylfumarate enhanced mitochondrial bioenergetics assessed by oxygen consumption rate in vitro
in mouse embryonic fibroblasts in an Nrf2 dependent fashion. Our results suggests that fumaric acid
esters protect against nigrostriatal dopaminergic neurotoxicity and associated oxidative damage, inflammation, and mitochondrial dysfunction by virtue of its ability to activate neuroprotective Nrf2/ARE genetic program. The current results clearly put forth pre-clinical evidence favoring DMF to be an ideal
therapeutic intervention that should be repurposed for PD.
Poster 12
Hdac3 regulates osteoblastic glucocorticoid and lipid metabolism during aging
Meghan E. McGee-Lawrence, Lomeli R. Carpio, Ryan J. Schulze, Mark A. McNiven, Sundeep
Khosla, Merry Jo Oursler, Jennifer J. Westendorf
Histone deacetylase 3 (Hdac3) removes acetyl groups from lysine residues in histones and other
proteins to epigenetically regulate gene expression. Hdac3 interacts with skeletal transcription
factors and is essential for bone development and maintenance. We previously reported that
conditional deletion of Hdac3 (Hdac3 CKO) in osteochondral progenitors, using Osx1-Cre,
caused osteopenia and increased marrow adiposity, both hallmarks of an aging skeleton. Here
we demonstrate that Hdac3 mRNA expression is reduced in bone cells from elderly women and
primary bone marrow stromal cells (BMSC) and osteocytes from aged mice. More significantly,
phosphorylation of S424-Hdac3, an event that stimulates deacetylase activity, is suppressed in
osseous cells from old (22-26 months) mice as compared to young (1-2 months) animals. Lipid
droplet formation was prevalent in the aged wildtype cultures as well as in osteogenic cultures of
young Hdac3-depleted BMSCs. Gene expression analyses revealed insignificant changes in
mRNA levels of Ppar gamma and fatty acid synthase, but Hdac3 CKO cultures demonstrated 10to 20-fold increases in expression of genes related to lipid storage (Cidec, Perilipin1) and 2 to 3fold changed in genes regulating glucocorticoid metabolism (11β-hydroxysteroid dehydrogenase
type 1 (Hsd11b1)), suggesting that Hdac3 regulates local osteoblastic activation of
glucocorticoids and lipid storage. In support of this hypothesis, lipid-containing cells in the
Hdac3 CKO cultures expressed Runx2. In vitro lipid droplet formation was dependent on
glucocorticoid signaling, as lipid storage and gene expression returned to near wildtype levels
when dexamethosone (Dex) was excluded from the culture medium. Hdac3 attenuated Dexinduced activation of the glucocorticoid-responsive MMTV-luciferase reporter. Taken together,
our data suggest that suppression of Hdac3 activity in bone cells contributes to reductions in
bone density and increases in marrow adiposity that are associated with aging and long-term
glucocorticoid treatment.
Poster 13
Early&Craniofacial&Defects&Occur&Following&Knockdown&of&the&Extracellular&Matrix&Protein,&
TINAGL1&
H.&Neiswender1,&S.&Navarre2,&D.J.&Kozlowski1,2,&and&E.K.&LeMosy1&
1Dept.'of'Cellular'Biology'and'Anatomy,'and'2Institute'of'Molecular'Medicine'and'Genetics,'Georgia'
Regents'University,'Augusta,'GA'30912'
&
Abstract&
Introduction:'The'TINAGL'family'of'secreted'basement'membrane'proteins'(TINAGL1'and'TINAG'in'
mammals;'TINAGL1'in'lower'species)'is'highly'conserved'but'functionally'opaque.'Limited'data'from'
humans'and'mice'has'suggested'functions'in'cell'adhesion,'renal'and'vascular'development,'cranial'
suture'closure,'and'suppression'of'metastasis,'while'the'fly'TINAGL1'appears'to'act'as'a'positive'Wnt'
(Wg)'cofactor.'''
Results/Methods:'We'are'using'morpholino'(MO)'knockdown'to'study'TINAGL1'function'in'zebrafish'
development.'A'consistent'pattern'of'pharyngeal'arch'cartilage'defects'is'observed'with'three'
independent'TINAGL1'MOs'but'not'their'misRmatch'controls,'and'is'observed'with'coRinjection'of'subR
threshold'doses'of'the'two'bestRcharacterized'MOs.'Substantial'rescue'(e.g.,'35%'vs'95%'larvae'
affected)'is'observed'by'coRinjection'of'TINAGL1'mRNA.'In'situ'hybridization'demonstrates'endogenous'
TINAGL1'expression'in'ventral'tissues'underlying'the'hindbrain'at'15R22'hpf.'In'24'hpf'morphant'
zebrafish,'the'neural'crest'cell'marker'dlx2a'is'reduced'or'absent'in'posterior'pharyngeal'arch'domains'
that'show'severe'defects.''
Conclusions:'These'results'suggest'that'TINAGL1'is'required'for'survival'or'migration'of'neural'crest'cells'
in'some'domains'of'the'pharyngeal'apparatus.''We'postulate'roles'involving'cell'adhesion'in'tissues'
through'which'the'neural'crest'cells'migrate,'or'involving'regulation'of'Wnt'localization'and'activity'
within'these'tissues.'Future'experiments'will'address'neural'crest'cell'behavior'in'these'morphants,'and'
whether'TINAGL1'genetically'and/or'physically'interacts'with'Wnts'important'during'early'craniofacial'
development.'
Translational&Impact:'Craniofacial'skeleton'development'is'a'model'for'birth'defects'and'
cartilage/bone/matrix'regulation.'In'future,'the'zebrafish'model'provides'a'platform'for'testing'drugs'
and'other'interventions'rationally'identified'via'mechanistic'studies.'
Poster 14
Dioleoylphosphatidylglycerol (DOPG) To Accelerate Corneal Wound Healing
Lawrence Olala, Xiaowen Lu, Mitchell Watsky, and Wendy B. Bollag
Charlie Norwood VA Medical Center, Augusta GA 30904 and Medical College of Georgia at
Georgia Regents University, Augusta, GA 30912
Primary roles of the cornea include maintenance of normal vision by refracting light onto the
lens and retina and provision of a barrier to the external environment. These functions are
sustained, in part, by the ability of the corneal epithelium, like the skin, to undergo continuous
renewal. Similarly to skin, corneal epithelial renewal is dependent on a highly integrated balance
between the processes of proliferation, differentiation, and cell death. Our laboratory has
previously described a novel cell signaling mechanism in skin epidermal keratinocytes composed
of the glycerol transporter, aquaporin-3 (AQP3) and the lipid-metabolizing enzyme
phospholipase D2 (PLD2), a member of the phospholipase D (PLD) family of enzymes. AQP3
and PLD2 function to produce phosphatidylglycerol (PG), which regulates keratinocyte
proliferation and differentiation. In particular, we have shown that PG species containing
monounsaturated fatty acids, e.g., dioleoylphosphatidylglycerol (DOPG), stimulate mouse
keratinocyte proliferation in slowly dividing cells, while PG species containing polyunsaturated
fatty acids, inhibit rapidly dividing keratinocytes. A mixture of PG species, such as PG derived
from egg, can normalize mouse keratinocyte proliferation in rapidly or slowly dividing
keratinocytes. We also have obtained in vivo data demonstrating that egg PG promotes healing of
a full-thickness skin wound. Interestingly, another group has shown that mice lacking the AQP3
gene (i.e., AQP3 knockout mice) show not only delayed skin wound healing but also impaired
corneal wound healing. Thus, we hypothesized that DOPG could regulate corneal epithelial
function in a similar manner to mouse skin and promote wound healing. Using AQP3 knockout
mice, we show here that 250µg/ml DOPG enhanced the rate at which corneal wounds healed. A
corneal epithelial scrape wound model was used to assess wound healing. By 12 hours there was
a significant reduction in wound size compared to vehicle (PBS)-treated controls, and the corneal
wounds were almost completely healed in the DOPG treatment group by 28 hours. We also
compared the rate of wound healing between genders, and observed no differences. Thus, our
results suggest that DOPG liposomes can be used to accelerate corneal wound healing,
particularly in individuals exhibiting wound healing defects.
Poster 15
SDF-1β / BMP-2 Co-Therapy Augments BMSC-Mediated Healing of Critical-Size
Mouse Calvarial Defects
Samuel Herberg (1), Alexandra Aguilar-Perez (2,3), R. Nicole Howie (3), Galina
Kondrikova (3), Sudharsan Periyasamy-Thandavan (3), Mohammed E. Elsalanty (3),
Xingming Shi (3), William D. Hill (3) and James J. Cray (4)
Case Western Reserve University, Cleveland, OH (1), Universidad Central del Caribe,
Bayamón, Puerto Rico (2),
Georgia Regents University, Augusta, GA (3), and Medical University of South Carolina,
Charleston, SC (4)
Bone is a dynamic and highly vascularized tissue that has the innate capacity for healing
upon damage. This regenerative process, however, often fails in patients with significant
co-morbidities, requiring surgical intervention [1]. Tissue engineering combining
biomaterial scaffolds, regenerative cells, and soluble growth factors may provide viable
alternatives to standard therapies (e.g., autografts, allografts, xenografts) [2-4]. We have
recently demonstrated that acellular DermaMatrix (ADM) scaffolds have native binding
affinities for relevant growth factors, facilitating bone healing [5]. Furthermore, we have
shown that stromal cell-derived factor-1β (SDF-1β) works in concert with bone
morphogenetic protein-2 (BMP-2) to potentiate osteogenic differentiation of bone
marrow-derived mesenchymal stem/stromal cells (BMSCs) in vitro [6], and to augment
bone formation in vivo using both mouse and rat models [5,7]. Here, we investigate the
regenerative capacity of BMSC/ SDF-1β/BMP-2 combination therapies delivered on
ADM relative to controls.
Poster 16
The Age-Associated Rise in miRNAs from Muscle Target SDF-1 and
Musculoskeletal Regulatory Genes is Reversed
with Caloric Restriction and Leptin
Sudharsan Periyasamy-Thandavan (1), Samuel Herberg (6), Phonepasong Arounleut
(1), Sunil Upadhyay (1), Galina Kondrikova (1), Amy Dukes (1), Colleen Davis (1),
Maribeth Johnson (5), Xing-Ming Shi (3,4), Carlos Isales (2,3,4), Mark W. Hamrick
(1,2,3,4), and William D. Hill (1,2,3,4).
1
Department of Cellular Biology & Anatomy (1), Department of Orthopaedic Surgery
(2), Institute of Molecular Medicine and Genetics (3), Institute for Regenerative and
Reparative Medicine (4), Department of Biostatistics (5), Georgia Regents University,
Augusta, GA, USA, Case Western Reserve University, Cleveland, OH, USA (6).
MicroRNAs (miRNAs) have the potential to regulate broad changes in connected
systems and pathways altering homeostatic gene expression. We have previously
identified age-related changes in the expression profiles of miRNAs isolated from human
bone marrow mesenchymal stem cells (BMSCs). These miRNAs targeted numerous
genes associated with musculoskeletal development, maintenance, aging and
osteoporosis. As well as, the chemokine SDF-1 (CXCL12), and its receptor CXCR4. The
SDF-1 axis is critical in the migration, survival, and engraftment of stem cells, including
BMSCs and muscle satellite cells (SCs). We have noted leptin can alter systemic and
muscle SDF-1 levels in an age-related manner suggesting nutrient signaling may effect
systemic, or tissue SDF-1 expression. We hypothesized that changes in nutrient signaling
pathways may modulate specific miRNA in an age specific manner. In turn these
miRNAs may alter SDF-1 and key osteogenic, or myogenic pathway genes. We tested
this hypothesis by examining changes in miRNAs, we had previously identified as linked
to aging in BMSCs, in muscles of mice aged 12 months and 20 months fed ad-libitum
(AL) and mice 20 months on caloric restriction (CR). We also treated other mice aged 20
months on caloric restriction with recombinant mouse leptin for 10 days at 10 mg/kg
body weight. Age-associated patterns of expression of miR-29b-1-3p, miR-29b-1-5p, &
miR-1244 in murine muscle (12 vs 20 months) mirrors that seen in human BMSCs
between young (under 45 years of age) and older (over 65 years of age) subjects. In both
cases the miRs 29b-1-5p, and 1244 increased, while miR-29b-1-p3 did not change. Of
interest the miR-29 family is well known to target muscluloskeletal and extracellular
matrix genes. CR reduced the miRs 29b-1-5p, 1244 and 141 in 20 month old mice to
levels equal to that of 12 month mice while miR-29b-1-3p was not altered. Of interest CR
with leptin treatment further reduced expression of miRs 29b-1-5p, 1244 & 141 well
below that seen in the 12 month mice and surprisingly the amount of miR-29b-1-3p rose
significantly. Therefore, it appears that food restriction is a potent regulator of miRNAs,
and suppresses miRNAs that target muscluloskeletal gene systems in aged muscle. Leptin
further drives this effect significantly below what is seen in younger muscle, which may
induce an inbalance in gene expression that might itself impair the potential for muscle
regeneration.
Poster 17
Alternative Splice Variants Of The Osteogenic Cytokine SDF-1 Differentially
Mediate CXCR4 and CXCR7 Expression in Bone Marrow MSCs
Aguilar-Pérez, A.1,3, Herberg, S.3, Periyasamy-Thandavan, S.3 Volkman B.F. 5,
Kondrikova, G. 3, Shi, X.6,7,8, Cubano, L. 1, Hamrick, M.W.3,6,7,8, Isales, C.M.3, 6,7,8, Hill,
W.D.2,3, 6,7,8
1Universidad Central del Caribe, Bayamón, Puerto Rico, USA, 2Charlie Norwood VA
Medical Center, Departments of 3Cellular Biology and Anatomy, 4Department of
Biochemistry, 5Medical College of Wisconsin
United States has one of the highest fracture rates associated with aging with up to 33%
of the over 50 population being affected. Aging-related osteopenic and osteoporosic
mechanisms remain poorly defined. It is a medical challenge to provide accurate and
early treatment in order to decrease morbidity, improve muscluloskeletal function and
increase independence among the elderly population. Our studies are focused on stromal
cell-derived factor-1 (SDF-1/CXCL12). SDF-1 is part of the CXC chemokine family and
can signal through CXCR4 and CXCR7. SDF-1 supports bone marrow-derived
stem/stromal cell (BMSC) survival, proliferation, and osteogenic differentiation (1,2).
There are six known alternative mRNA splice variants for SDF-1 (3). SDF-1α and SDF1β are the main isoforms in bone marrow. SDF-1α has been implicated in BMSC
mobilization and osteogenic differentiation. SDF-1β is more resistant to proteolytic
cleavage due to the four additional C-terminal amino acids compare with SDF-1α and has
also been shown to be important for BMSC survival and in regulating osteogenesis (1,2).
It is unclear whether there is a difference in the activation of gene expression and
signaling pathways between these two constitutively expressed isoforms in BMSCs.
CXCR4 second messenger signaling has recently been shown to be able to switch
between GPCR and β-Arrestin pathways in response to ligand concentration (4,5). We
assessed the effects of SDF-1α & β on the expression of SDF-1 ligands and receptors
using a dose range know to differentially affect SDF-1α mediated CXCR4 signaling
pathway choice due to ligand biased GPCR activation.
Poster 18
Caloric Restriction and the Adipokine Leptin alter the SDF-1 signaling axis and
autophagy in Bone and MSCs
Sudharsan Periyasamy-Thandavan (1), Samuel Herberg (6), Phonepasong Arounleut
(1), Sunil Upadhyay (1), Amy Dukes (1), Colleen Davis (1), Maribeth Johnson (5), Mark
W. Hamrick (1,2,3,4), Carlos Isales (2,3,4), and William D. Hill (1,2,3,4).
1
Department of Cellular Biology & Anatomy (1), Department of Orthopaedic Surgery
(2), Institute of Molecular Medicine and Genetics (3), Institute for Regenerative and
Reparative Medicine (4), Department of Biostatistics (5), Georgia Regents University,
Augusta, GA, USA, Case Western Reserve University, Cleveland, OH, USA (6).
Within the bone marrow (BM), both osteogenic and adipogenic lineage cells
originate from multipotent mesenchymal stromal/stem cells (MSCs). MSCs are a major
source of the secreted chemokine stromal cell-derived factor-1 (SDF-1), which is critical
in MSC differentiation and BM residence, largely via its receptor CXCR4. A major factor
in age-associated osteoporosis is the fate of MSCs. Growing evidence suggests that SDF1 is critical in regulating MSC differentiation resulting in either a pro-osteogenic fate, or
an adipogenic one that leads to reduced bone mass and mineral density, as well as
increased BM adipocytes. Leptin, a cytokine-like hormone is secreted in large part by
adipocytes, exhibits anti-osteogenic effects via hypothalamic receptors. However,
peripheral administration of leptin can demonstrate bone protective effects. Previous
studies in mice suggest that dietary restriction decreases circulating leptin while
increasing serum SDF-1 levels similar to the effect of aging. In contrast, the opposite
occurs with diet-induced obesity. This study investigates the effects of caloric restriction
(CR) and leptin on the SDF-1/CXCR4 axis in bone and BM tissues. For in vivo studies,
we collected bone, BM cells and BM interstitial fluid from 12 and 20 month-old C57Bl6
mice fed ad-libitum (AL), and 20 month-old mice on CR with, or without, leptin for 10
days (10mg/kg body weight). To mimic conditions of CR in vitro, murine BM derived
MSCs (BMSCs) were treated with 1) control (Ctrl): normal proliferation medium, 2) CR:
low glucose, low serum medium, or 3) CR+leptin: low glucose, low serum medium +
100 ng/ml rmLeptin for 6-72 h. Both SDF-1 and CXCR4 protein and mRNA expression
in MSCs were increased by CR and CR + leptin. In contrast, the alternate SDF-1
receptor CXCR7 was decreased, this supports a change in SDF-1 signaling due to a shift
in receptor availability. Additionally, autophagic flux was increased with CR and
attenuated with the addition of leptin. However, mRNA isolated from bone shows SDF-1
and CXCR4 & 7 increase with age and this is reversed with CR, but addition of leptin
returns this to the “aged” level. This suggests that in bone CR and leptin alter the nutrient
signaling pathways in different ways to affect autophagy and the osteogenic cytokine
SDF-1’s local action. Studies focusing on the molecular interaction between nutrient
signaling and autophagy mediated by CR, leptin and SDF-1 axis may help to address agerelated musculoskeletal changes.
Poster 19
Mediation of SDF-1/CXCR4 signaling in aged skeletal muscle by the adipokine
leptin.
* Samuel Herberg, Case Western Reserve University, USA, Sudharsan PeriyasamyThandavan, Georgia Regents University & Charlie Norwood VAMC, USA, Phonepasong
Arounleut, Georgia Regents University (formally Georgia Health Sciences University),
USA, Sunil Upadhyay, Georgia Regents University, USA, Amy Dukes, Georgia Regents
University, USA, Colleen Davis, Georgia Regents University, USA, Galina Kondrikova,
Georgia Regents University, USA, Maribeth Johnson, Georgia Regents University, USA,
Carlos Isales, Georgia Regents University, USA, William Hill, Georgia Regents
University & Charlie Norwood VAMC, USA, Mark Hamrick, Georgia Health Sciences
University, USA
Aging is associated with a decline in both muscle and bone mass, which is thought to
involve dysfunction in bone- and muscle-derived stem cells. The chemokine SDF-1
(CXCL12) and its receptor CXCR4 play important roles in the migration, survival, and
engraftment of stem cells, including bone marrow stromal cells (BMSCs) and muscle
satellite cells (SCs). Adult muscle constitutively expresses SDF-1, and CXCR4
expression in muscle is elevated during muscle regeneration. Previous work in mice has
shown that, with food restriction, leptin levels are decreased while SDF-1 levels are
increased, and the reverse is observed with diet-induced obesity. We therefore speculated
that leptin and the SDF-1/CXCR4 axis may interact throughout adulthood to mediate
musculoskeletal tissue repair and regeneration. We tested this hypothesis by examining
SDF-1 and CXCR4 expression in muscles of mice aged 12 months and 20 months fed adlibitum (AL) and mice 20 months on caloric restriction (CR). We also treated mice aged
20 months on caloric restriction with recombinant mouse leptin for 10 days at 10 mg/kg
body weight. Serum SDF-1 was increased in the aged mice, but leptin treatment reduced
serum SDF-1 to levels seen in the younger (12 month) animals. SDF-1alpha expression
in muscle decreased with age, but was increased with caloric restriction, and leptin
treatment reversed this increase. CXCR4 expression in muscle decreased slightly with
age, but was increased almost four-fold with caloric restriction, and this increase was
attenuated by leptin treatment. Together these data suggest that food restriction is a
potent stimulus for SDF-1 and CXCR4 activation in aged muscle, and that leptin can
antagonize this effect. Thus, while we have previously demonstrated that leptin can
increase muscle mass and IGF-1 in leptin-deficient rodents, leptin may also impair the
potential for muscle regeneration and satellite cell migration via its effects on SDF1/CXCR4 interactions.
Poster 20
TITLE: Therapeutic and Imaging Potential of Umbilical Cord Blood Derived Endothelial
Progenitor Cells in Stroke and Glioma
AUTHORS:
1,2
1,2
B.R Achyut, 2Branislava Janic, 2Nadimpalli Ravi S Varma,
1,2
ASM Iskander,
Adarsh Shankar 1,2Ali S Arbab
AFFILIATIONS: 1Biochemistry and Molecular Biology, Cancer Center, Georgia Regents
University, Augusta, GA. 2Cellular and Molecular Imaging Laboratory, Henry Ford Health
System, Detroit, MI
ABSTRACT
Nervous system has limited regenerative potential in disease conditions such as cancer,
neurodegeneration, stroke, and several neural injuries. Umbilical cord blood (UCB) derived
hematopoietic stem cells (HSCs) are capable of giving rise hematopoietic, epithelial, endothelial
and neural progenitor cells. Thus, suggested to significantly improve graft-versus-host disease
and represent the distinctive therapeutic option for brain diseases. Recent advances in strategies
to isolate, expand and shorten the timing of UCB stem cells engraftment have tremendously
improved the efficacy of transplantations. A subpopulation of CD34+ human HSCs identified by
the cell-surface molecule AC133+ (CD133+), have been shown to be more specific for
endothelial differentiation and vascular repair. In addition, emerging applications of UCB
derived AC133+ endothelial progenitor cells (EPCs) as imaging probe, regenerative agent, and
gene delivery vehicle have been studied by our lab and others. We have been exploited AC133+
EPCs for in vivo imaging modalities, importantly; magnetic resonance imaging (MRI) to monitor
the migration and engraftment efficacy of administered cells in stroke and glioma preclinical
animal models.
!
Poster 21
Establishment of Stro1/CD44 as new markers for human myometrial/ fibroid stem cell
Mas A1, Nair S1, Laknaur A1, Diamond MP1, Simon C2, 3, Al- Hendy A1.
1
Department of Obstetrics and Gynecology, Georgia Regents University (GRU), Augusta,
Georgia, USA.2Fundación Instituto Valenciano de Infertilidad, Instituto Universitario IVIUniversity of Valencia, INCLIVA, Valencia, Spain;3Department of Obstetrics and Gynecology,
School of Medicine, Stanford University, California, USA.
Keywords: Uterine fibroids, stem cells, Stro1/CD44.
Background: Uterine fibroids are benign monoclonal tumors, each arising from a single
dysregulated myometrial smooth muscle cell, likely a stem cell. To date, putative stem cells
have been isolated from several female reproductive organs, especially myometrium, using the
side population (SP) technique. However, given the limitations of this approach, further studies
are required to establish a more refined method of isolating stem cells based on the presence of
specific and unique surface markers.
Objective: To identify and characterize new specific myometrial/fibroid stem cell markers in
human myometrium to better understand their implication in the development of uterine
fibroids.
Methods:
Fibroids (F) and adjacent myometrium (MyoF) tissues were processed after signed informed
consent from patients undergoing hysterectomy. After specific tissue digestion with
collagenase, single cell suspensions were treated with CD44 and Stro1 antibodies-coated
biotinylated Dynabeads. CD44-/Stro1- and CD44+/Stro1+ cells were isolated and cultured under
hypoxic conditions (1-2% O2). Molecular analysis for stem cell markers, ABC transporters and
hormonal receptors were performed in isolated cells. Cell characterization was also achieved by
an immunophenotypic analysis using typical mesenchymal/ hematopoietic markers as well as in
vitro differentiation assays.Additionally, isolated cells were transfected with iron-RhoB
nanoparticles and injected under the renal capsule of female NOG mice with E2/P4 sex steroids
supplement for 8 weeks. During this period, MRI and fluorescent imaging were performed on
live animals and IHC assays were performed on the xenografts to assess the formation of
myometrial/fibroid-like tissues.
Results:
Using Stro1/CD44 markers, we were able to isolate stem cells from MyoF as well as from F
tissues. At mRNA level, these cells expressed ABCG2 transporter as well as other specific
stemness markers such as Oct4, Nanog and GDB3. However, they showed a low expression of
steroid receptors: ER-alpha and PR-A/PR-B, suggesting that they are not yet hormonally
committed.Presence of typical mesenchymal markers (CD90, CD105, CD73) and absence of
hematopoietic stem cell markers (CD34, CD45) supported their mesodermal origin. Moreover,
we demonstrated the ability of these cells to differentiate in vitro into adipocytes, osteocytes and
chondrocytes, further supporting mesodermal lineage derivation. Finally, their functional
capability to form fibroid-like lesions, was established in an animal model by MRI findings and
further confirmed by fluorescence imaging and histology staining, demonstrating the
regenerative potential of putative fibroid stem cells in vivo.
Conclusions:
We have identified and characterized new specific fibroid stem cell markers in human
myometrium, demonstrating the functional capacity of these Stro1+/CD44+ cells to induce
fibroid like tissue by in vitro and in vivo approaches. These findings offer a useful tool to better
understand the origin and initiation of uterine fibroids which can improve the development of
more effective therapeutic options.
Poster 22
Myometrial Tumor-Forming Stem Cells in a Murine Model of Uterine Fibroids Reside in Hypoxic
Niches
Mas A1, Laknaur A1, O’Connor P.M 2, Walker C.L3, Diamond MP1, Simon C4, 5, Al- Hendy A1.
1
Department of Obstetrics and Gynecology, Georgia Regents University (GRU), Augusta, Georgia, USA.
Department of Physiology, Georgia Regents University (GRU), Augusta, Georgia, USA. 3Center for
Translational Cancer Research, Houston, Texas, USA. 4Fundación Instituto Valenciano de Infertilidad,
Instituto Universitario IVI- University of Valencia, INCLIVA, Valencia, Spain; 5Department of
Obstetrics and Gynecology, School of Medicine, Stanford University, California, USA.
2
Keywords: Eker rat, stem cells, hypoxia.
Background: The Eker rat is a murine model for uterine fibroid formation. They carry a germ line
mutation in the Tsc2 tumor suppressor gene, producing uterine tumors with similar anatomic, histologic,
and biologic features to human uterine fibroids. To date, the potential role of stem cells in the formation
of uterine fibroids in Eker rats has not been investigated. Furthermore, prior studies revealed strong
regulatory links between O2 availability and function of stem cells.
Objective: In this study, we aim to identify and localize the myometrial stem cell population in the Eker
rat's uterus using specific surface markers, explore their role in fibroid tumor formation, as well as to
establish the role of hypoxia in the proliferation of myometrial stem cells.
Methods: Uterine horns, cervix and fibroid tissues from Eker rat uterus were firstly examined via double
immunohistochemistry (IHC) to co-localize the putative myometrial stem cell markers Stro1/CD44 with
the established markers of undifferentiation such as c-KIT, OCT-4 and NANOG. Subsequently, the Eker
rat uterus were digested at different points (cervix, lower, middle and upper horn) with collagenase to
obtain single cell suspensions in order to compute by flow cytometry the percentage of Stro1/CD44 stem
cells. To determine in vivo tissue oxygen tension (pO2) in different anatomical locations of Eker rat
uterus, Clark type oxygen micronsensors (Unisense) were used and changes in hypoxia were also
examined by injection of hypoxia-detecting dye pimonidazole followed 24 hours later by IHC as well as
specific staining for the well-known hypoxic markers: CAIX and HIF-α.
Results:
In the Eker rat model, uterine fibroid lesions occur predominantly in the cervix in 85% of cases versus
15% in the uterine horns. We demonstrate the co-localization of Stro-1/ CD44 with other specific stem
cell markers (Oct-4, c-kit, nanog) validating their undifferentiated status and their distribution in Eker
fibroids, uterus horns and cervix. Interestingly, IHC quantification demonstrated that significantly higher
number of stem cells are present in the cervical region compared to uterine horns. Flow cytometric
analysis corroborated that the percentage of stem cells in cervix is significantly higher than in uterine
horns (P<0.05). The quantification of in vivo oxygenation demonstrates that the pO2 levels in cervix
(13.24 mmHg) were significantly lower than in uterine horns (29.47mmHg; pANOVA=0.0014).
Moreover, pimonidazole, CAIX and HIF-α staining demonstrating stronger expression in cervix than in
uterine horns confirm the hypoxic status of cervix region when compared to other regions of the uterus.
Conclusions:
Eker rats serve as a preclinical model to screen for and evaluate the origin of uterine fibroids. For the first
time, we have identified the myometrial stem cell niche by stem cell markers and we have establish an
association of hypoxic niches in uterine cervix which is the primary location of fibroid lesions in this
model. These observations could provide a putative mechanism by which myometrial stem cells regulate
transformations into tumor initiating cells.
Contact information for Symposium Participants
City
Augusta
Augusta
Augusta
State
GA
GA
GA
Phone Number
Email
706-721-4375 [email protected]
706-721-4797 [email protected]
706-721-0905 [email protected]
Augusta
GA
706-721-3833
[email protected]
Cleveland
Augusta
Athens
Augusta
Augusta
Augusta
OH
GA
GA
GA
GA
GA
216-368-6425
706-721-8909
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
Atlanta
Augusta
Augusta
Martinez
GA
GA
Ga
GA
228-343-3914
706-721-3391
706-284-6941
[email protected]
[email protected]
[email protected]
[email protected]
Augusta
GA
706-721-7815
[email protected]
Athens
GA
678-896-0248
[email protected]
Augusta
GA
706-721-0698
[email protected]
Atlanta
Augusta
Augusta
GA
GA
GA
434-249-6040
504-578-8804
706-721-3383
[email protected]
[email protected]
[email protected]
Cleveland
Augusta
Augusta
OH
GA
GA
216-368-3562
706-721-4055
706-721-6759
[email protected]
[email protected]
[email protected]
Augusta
Augusta
Augusta
GA
GA
GA
706-721-3591
706-721-9680
706-589-4276
[email protected]
[email protected]
[email protected]
University of Georgia & GRU
Institution Department/Program
Biochemistry and Molecular Biology, Cancer Center
Cellular Biology and Anatomy
Pharmacology & Toxicology
Dean for Global Translational Research, Professor and
Director Division of Translational Research, Obstetrics
and Gynecology
Director of the Stem Cell and Engineered Novel
Therapeutics Laboratory, Dept. Biomedical Engineering
and Dept. Orthopaedic Surgery
Cancer Research Center
Bioengineering
Biochemistry and Molecular Biology
Neuroscience & Regenerative Medicine
Cancer Research Center
George W. Woodruff School of Mechanical
Engineering/IBB
OB/GYN
Medical College of Georgia
Biological Sciences
Director, Cardiac Arrhythmia Ablation Services, Dept.
Medicine, Institute for Regenerative and Reparative
Medicine
Animal and Dairy Science, Regenerative Bioscience
Center
Physiology, Institute for Regenerative and Reparative
Medicine
Parker H. Petit Institute for Bioengineering and
Biosciences, Biomedical Engineering
OB/GYN
Pharmacology and Toxicology
Professor of Biology and Director of the Skeletal
Research Center
Office of Innovation Commercialization
Vascular Biology Center
Sr. VP for Research, Assoc. Dean, the Brooks Chair, and
Chair of OB/GYN, Director - Clinical & Translational
Research
OB/GYN
OB/GYN
Associate Professor, Director, Clinical and Experimental
Therapeutics Graduate Program
706-721-6760
[email protected]
BA/BS
Georgia Regents University
Department of Neuroscience & Regenerative Medicine
Augusta
GA
[email protected]
PhD
Georgia Regents University
Department of Neuroscience & Regenerative Medicine,
Institute for Regenerative and Reparative Medicine
Augusta
GA
[email protected]
Last Name
Achyut
Aguilar-Perez
Ahuja
First Name
Degree
Institution Name
Bhagelu
PhD
Georgia Regents University
Alexandra
BA/BS
Georgia Regents University
Manuj
PhD
Georgia Regents University
Al-Hendy
Ayman
MD/Ph.D Georgia Regents University
Alsberg
Alsulami
Andrews
Angara
Aoued
Arbab
Eben
Meshal
Seth
Kartik
Hadj
Ali
PhD
MD
Averett
Badr
Balotin
Benson
Rodney
Marwa
Jeanette
Krista
PhD
MD
MPA MA
BA/BS
Georgia Institute of Technology
Georgia Regents University
Georgia Regents University
Georgia Regents University
Berman
Adam
MD
Georgia Regents University
Betancur
Martha
BA/BS
University of Georgia
Bollag
Wendy
PhD
Georgia Regents University
Botchwey
Brakta
Caldwell
Edward
Soumia
R. William
PhD
MD
PhD
Georgia Institute of Technology
Georgia Regents University
Georgia Regents University
Caplin
Clark
Czikora
Arnold I.
Carl
Istvan
PhD
PhD
Case Western Reserve University
Georgia Regents University
Georgia Regents University
Diamond
Edwards
Elhusseini
Michael P.
Connie
Heba
MD
RN
MD
Georgia Regents University
Georgia Regents University
Georgia Regents University
El-Remessy
Azza
PhD
Erion
Joanna
Eroglu
Ali
Case Western Reserve University
Georgia Regents University
University of Georgia
BA/BS
Georgia Regents University
M.S.
Georgia Regents University
MD/Ph.D Georgia Regents University
706-721-4375
Contact information for Symposium Participants
Last Name
Franklin
First Name
Sam
Degree
Institution Name
Institution Department/Program
Department of Small Animal Med & Surg., College of
Veterinary Medicine, Regen Biosci Ctr
DVM/PhD University of Georgia
Fulzele
Galipeau
Gavalas
Ghoshal-Gupta
Gomillion
Guo
Halder
Han
Handa
Hardigan
Hayman
Helwa
Sadanand
Jacques
Charlotte
Sampa
Cheryl
De-Huang
Sunil
Qimei
Hitesh
Trevor
Joy
Inas
PhD
MD
BA/BS
PhD
PhD
PhD
PhD
MD
PhD
BA/BS
M.S.
PhD
Georgia Regents University
Emory University
Georgia Regents University
Georgia Regents University
University of Georgia
Georgia Regents University
Georgia Regents University
Georgia Regents University
University of Georgia
Georgia Regents University
Georgia Regents University
Georgia Regents University
Hess
David C.
MD
Georgia Regents University
Orthopaedic Surgery, Institute for Regenerative and
Reparative Medicine
EPIC
Clinical Trials Office/Orthopaedic Surgery
Pathology
College of Engineering/Biological Engineering
Department of Obstetrics and Gynecology
College of Graduate Studies
RDS Research support
Cellular Biology and Anatomy
Presidential Distinguished Chair, Chair Dept of
Neurology
Kuhn
LaRue
Laserna
LeMosy
Professor Depts. Cellular Biology &
Anatomy, Orthopaedic Surgery, Neurology and Institute
for Regenerative and Reparative Medicine
William D.
PhD
Georgia Regents University
Clinical and Translational Sciences
Joan
BA/BS
Georgia Regents University
Chair, Dept Orthopaedic Surgery
Monte
MD
Georgia Regents University
Regents' Professor, Vice Chair for Clinical Affairs, DNRM,
Vice Chair of Clinical and Translational Research
Orthopaedic Surgery, Director of Institute for
Regenerative and Reparative Medicine
Carlos
MD
Georgia Regents University
Cancer Research Center
Meenu
PhD
Georgia Regents University
Assistant Professor, Regenerative Bioscience Center,
ADS
Lohitash
PhD
The University of Georgia
VAMC Research Services/Department of Pathology and
Ryan
BA/BS
Medical University of South Carolina Laboratory Medicine
Neurology
Mohammad B PhD
Georgia Regents University
Vascular Biology Center
Ha Won
PhD
Georgia Regents University
Regenerative Bioscience Center
Holly
BA/BS
University of Georgia
Cell Biology and Anatomy
Galina
BA/BS
Georgia Regents University
Reconstructive Sciences Department, Biomedical
Liisa T.
PhD
University of Connecticut Health Ctr Engineering Department
Amanda
PhD
Medical University of South CarolinaPathology and Laboratory Medicine
Clinical Trials Office
Candelario
BA/BS
Georgia Regents University
Cellular Biology and Anatomy
Ellen
MD/Ph.D Georgia Regents University
Lin
Liu
Locklin
Lucas
Sen
Yutao
Jason
Rudolf
Hill
Holloway
Hunter
Isales
Jain
Karumbaiah
Kelly
Khan
Kim
Kinder
Kondrikova
PhD
PhD
PhD
PhD
Georgia Regents University
Georgia Regents University
University of Georgia
Georgia Regents University
Department of Neuroscience & Regenerative Medicine
Cellular Biology and Anatomy
Chemistry/Engineering
Vascular Biology Center
City
State Phone Number
Email
Athens
GA
706-206-7752
[email protected]
CB2509
Atlanta
Augusta
Augusta
Athens
Augusta
Augusta
Augusta
Athens
Augusta
Augusta
Augusta
GA
GA
GA
GA
GA
GA
GA
GA
GA
GA
GA
GA
706-721-4850
404-831-1209
706-721-2199
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
Augusta
GA
(706) 721-1691 [email protected]
Augusta
Augusta
Augusta
GA
GA
GA
706-721-2019
706-721-9942
706-721-6172
Augusta
Martinez
GA
GA
(706) 721-0692 [email protected]
301-204-7490 [email protected]
Athens
GA
706-542-2017
[email protected]
Charleston
Augusta
Augusta
Athens
Augusta
SC
GA
GA
GA
GA
410-409-2558
706-721-1671
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
Farmington
Charleston
Augusta
Augusta
CT
SC
GA
GA
860-679-3922
843-789-6713
706-721-0876
[email protected]
[email protected]
[email protected]
[email protected]
Augusta
Augusta
Athens
Augusta
GA
GA
GA
GA
706-721-5780
706-721-2015
706-542-2359
706-721-9470
[email protected]
[email protected]
[email protected]
[email protected]
706-542-0918
706-721-1470
706-721-8907
205-240-9206
706-542-8109
706-721-0925
706-721-7436
706-721-4843
7702861810
[email protected]
[email protected]
[email protected]
Contact information for Symposium Participants
Last Name
First Name
Degree
Institution Name
Institution Department/Program
City
State Phone Number
Email
Augusta
Augusta
GA
GA
706-721-8910
[email protected]
[email protected]
PhD
M.S.
PhD
PhD
BA/BS
PhD
PhD
Department of Neuroscience & Regenerative Medicine
OB/GYN
Cellular Biology and Anatomy, Institute for Regenerative
and Reparative Medicine
Georgia Regents University
Environmental Health Science
University of Georgia
Medical University of South CarolinaRegenerative Medicine
Regenerative Bioscience Center
University of Georgia
Cell Biology and Anatomy
Georgia Regents University
Pediatrics
Emory University
Physiology
Georgia Regents University
Augusta
Athens
Charleston
Athens
Augusta
Atlanta
Augusta
GA
GA
SC
GA
GA
GA
GA
706-446-0128
678-642-8547
843-647-0996
706-721-0893
404-727-9961
706-721-0704
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
Jinxiu
MD
Georgia Regents University
Augusta
GA
706-721-5780
[email protected]
Park
Changwon
PhD
Emory University
Atlanta
GA
404-727-7143
[email protected]
Park
Peard
Mary Anne
Leslie
M.S.
BA/BS
Georgia Regents University
Georgia Regents University
Augusta
Augusta
GA
GA
706-721-0193
404-403-0505
[email protected]
[email protected]
Periyasamy-Thandavan
Sudharsan
PhD
Georgia Regents University
Augusta
GA
706-721-4797
[email protected]
Peroni
Pierce
John
Jessica
Department of Neuroscience & Regenerative Medicine
Assistant Professor, Department of Pediatrics, Center
for Cardiovascular Biology
Manager Clinical Trials Office, Clinical & Translational
Sciences/CTO
Medical College of GA
Cellular Biology and Anatomy, Institute for Regenerative
and Reparative Medicine
Dept. Large Animal Medicine, College of Veterinary
Medicine, Reg. Biosci. Ctr.
Cellular Biology & Anatomy
Athens
Augusta
GA
GA
678-467-8244
[email protected]
[email protected]
Platt
Rajpurohit
Rosson
Scharf
Sen
Seremwe
Shaaban
Shalaby
Shankar
Shin
Solomon
Simon
Surendra
Brenda
Alex
Nilkantha
Mutsa
Sherif
Shahinaz
Adarsh
Eric
Harold
Athens
Augusta
Augusta
Athens
Augusta
Augusta
Augusta
Augusta
Augusta
Atlanta
Atlanta
GA
GA
GA
GA
GA
GA
GA
GA
GA
GA
GA
706-206-6692
706-267-4590
706-721-9680
216-632-2986
615-419-5718
706-721-4375
765-532-6707
404-939-6271
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
Stevens
Mark
Augsta
GA
706-721-2411
[email protected]
Stice
Su
Tang
Steve
Yun
Yaoliang
DVM, MS, Dip
University
ACUS of Georgia
BA/BS
Georgia Regents University
BVM&S
MRCVS
Dipl.
ACVIM,
Dipl.
Neurology & Neurosurgery Svc, Dept. of Small Animal
ECVN
Med. & Surg./ College of Vet Med
University of Georgia
Cancer Center
PhD
Georgia Regents University
Clinical Trials Office
BSN
Georgia Regents University
College of Veterinary Medicine
University of Georgia
PhD
Georgia Regents University
Neuroscience & Regnerative Medicine
Joint Physiology & Cell Biology and Anatomy
PhD
Georgia Regents University
Biochemistry and Molecular Biology
MD
Georgia Regents University Cancer Center
OB/GYN
MD
Georgia Regents University
Biochemistry and Molecular Biology
BA/BS
Georgia Regents University
Cardiology
MD
Emory University
VentureLab
BA/BS
Georgia Institute of Technology
College of Dental Medicine/Oral and Maxillofacial
Surgery
DDS
Georgia Regents University
Georgia Research Alliance Eminent Scholar Chair,
Professor and Director of the Regenerative Bioscience
Center, UGA Director of the Regenerative Engineering
and Medicine partnership
University of Georgia
Institute for Regenerative and Reparative Medicine
PhD
Georgia Regents University
Vascular Biology Center
MD/Ph.D Georgia Regents University
Athens
Augusta
Augusta
GA
GA
GA
706-583-0071
706-721-7920
909-643-7388
[email protected]
[email protected]
[email protected]
Luo
Mas
Tong
Aymara
PhD
PhD
McGee-Lawrence
McKenzie
Moreno
Mortensen
Neiswender
Oh
Olala
Meghan
Marie
Ricardo
Luke
Hannah
Se-Yeong
Lawrence
Pan
Georgia Regents University
Georgia Regents University
706-721-8185
Contact information for Symposium Participants
Last Name
First Name
Temenoff
Thomas
West
Johnna
Bobby
Franklin
Wikesjö
Wilson
Wilson
Wu
Wyatt
Yang
Yoon
Zhang
Zhang
Zhao
Zimmerman
Ulf
Katie
Kyle
Dona
Emily
Nianlan
Young-Sup
Maoxiang
Mingzhen
Qun
Arthur
Degree
Institution Name
Institution Department/Program
Associate Professor, Coulter Department of Biomedical
Engineering at Georgia Tech/Emory, GIT Director of the
Regenerative Engineering and Medicine partnership
Georgia Institute of Technology
Pharmacology, Toxicology and Neurology
PhD
Georgia Regents University
Assistant Professor, Regenerative Bioscience Center
PhD
University of Georgia
Oral Biology, Institute for Regenerative and Reparative
Medicine
DDS
Georgia Regents University
M.S.
Medical University of South CarolinaPathology and Laboratory Medicine
Department of Medicine, Hematology/Oncology
BA/BS
Georgia Regents University
Microbiology & Immunology / Vaccine Center
MD/Ph.D Emory University
Regenerative Bioscience Center
BA/BS
University of Georgia
DNRM
MD/Ph.D Georgia Regents University
Stem Cell Research Group Cardiology
Emory University
PHAMARCOLOGY
PhD
Georgia Regents University
PhD
University of Georgia
Regenerative Bioscience Center
PhD
University of Georgia
Department of Cellular Biology and Anatomy
M.S.
Georgia Regents University
City
State Phone Number
Email
Atlanta
Augusta
Athens
GA
GA
GA
404-385-5026 [email protected]
706-721-6356 [email protected]
(706) 542-0988 [email protected]
Augusta
Charleston
Augusta
Atlanta
Athens
Augusta
Atlanta
Augusta
Athens
Athens
Augusta
GA
SC
GA
GA
GA
GA
GA
GA
GA
GA
GA
706288-8649
706-871-6591
404-712-0563
706-721-0519
404-712-1733
706-306-6423
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]
[email protected]