Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where So Gun Hong is active.

Publication


Featured researches published by So Gun Hong.


Cell Reports | 2014

Path to the Clinic: Assessment of iPSC-Based Cell Therapies In Vivo in a Nonhuman Primate Model

So Gun Hong; Thomas Winkler; Chuanfeng Wu; Vicky Guo; Stefania Pittaluga; Alina Nicolae; Robert E. Donahue; Mark E. Metzger; Sandra D. Price; Naoya Uchida; Sergei A. Kuznetsov; Tina M. Kilts; Li Li; Pamela Gehron Robey; Cynthia E. Dunbar

Induced pluripotent stem cell (iPSC)-based cell therapies have great potential for regenerative medicine but are also potentially associated with tumorigenic risks. Current rodent models are not optimal predictors of efficiency and safety for clinical application. Therefore, we developed a clinically relevant nonhuman primate model to assess the tumorigenic potential and in vivo efficacy of both undifferentiated and differentiated iPSCs in autologous settings without immunosuppression. Undifferentiated autologous iPSCs indeed form mature teratomas in a dose-dependent manner. However, tumor formation is accompanied by an inflammatory reaction. On the other hand, iPSC-derived mesodermal stromal-like cells form new bone in vivo without any evidence of teratoma formation. We therefore show in a large animal model that closely resembles human physiology that undifferentiated autologous iPSCs form teratomas, and that iPSC-derived progenitor cells can give rise to a functional tissue in vivo.


Molecular Therapy | 2013

Assessing the Risks of Genotoxicity in the Therapeutic Development of Induced Pluripotent Stem Cells

So Gun Hong; Cynthia E. Dunbar; Thomas Winkler

Induced pluripotent stem cells (iPSCs) have great potential for regenerative medicine as well as for basic and translational research. However, following the initial excitement over the enormous prospects of this technology, several reports uncovered serious concerns regarding its safety for clinical applications and reproducibility for laboratory applications such as disease modeling or drug screening. In particular, the genomic integrity of iPSCs is the focus of extensive research. Epigenetic remodeling, aberrant expression of reprogramming factors, clonal selection, and prolonged in vitro culture are potential pathways for acquiring genomic alterations. In this review, we will critically discuss current reprogramming technologies particularly in the context of genotoxicity, and the consequences of these alternations for the potential applications of reprogrammed cells. In addition, current strategies of genetic modification of iPSCs, as well as applicable suicide strategies to control the risk of iPSC-based therapies will be introduced.


Molecular therapy. Methods & clinical development | 2014

Development of an inducible caspase-9 safety switch for pluripotent stem cell–based therapies

Chuanfeng Wu; So Gun Hong; Thomas Winkler; David M Spencer; Alexander Jares; Brian Ichwan; Alina Nicolae; Vicky Guo; Andre Larochelle; Cynthia E. Dunbar

Induced pluripotent stem cell (iPSC) therapies offer a promising path for patient-specific regenerative medicine. However, tumor formation from residual undifferentiated iPSC or transformation of iPSC or their derivatives is a risk. Inclusion of a suicide gene is one approach to risk mitigation. We introduced a dimerizable-“inducible caspase-9” (iCasp9) suicide gene into mouse iPSC (miPSC) and rhesus iPSC (RhiPSC) via a lentivirus, driving expression from either a cytomegalovirus (CMV), elongation factor-1 α (EF1α) or pluripotency-specific EOS-C(3+) promoter. Exposure of the iPSC to the synthetic chemical dimerizer, AP1903, in vitro induced effective apoptosis in EF1α-iCasp9-expressing (EF1α)-iPSC, with less effective killing of EOS-C(3+)-iPSC and CMV-iPSC, proportional to transgene expression in these cells. AP1903 treatment of EF1α-iCasp9 miPSC in vitro delayed or prevented teratomas. AP1903 administration following subcutaneous or intravenous delivery of EF1α-iPSC resulted in delayed teratoma progression but did not ablate tumors. EF1α-iCasp9 expression was downregulated during in vitro and in vivo differentiation due to DNA methylation at CpG islands within the promoter, and methylation, and thus decreased expression, could be reversed by 5-azacytidine treatment. The level and stability of suicide gene expression will be important for the development of suicide gene strategies in iPSC regenerative medicine.


Molecular Therapy | 2016

The Role of Nonhuman Primate Animal Models in the Clinical Development of Pluripotent Stem Cell Therapies.

So Gun Hong; Yongshun Lin; Cynthia E. Dunbar; Jizhong Zou

On 28 September 2015, the Nonhuman Primate (NHP) Induced Pluripotent Stem Cells (iPSCs) Workshop was held at the National Institutes of Health (NIH) campus in Bethesda, Maryland. The workshop was sponsored by the Office of Scientific Director of the National Heart, Lung, and Blood Institute (NHLBI).


Blood | 2018

The impact of aging on primate hematopoiesis as interrogated by clonal tracking

Kyung-Rok Yu; Diego A. Espinoza; Chuanfeng Wu; Lauren Truitt; Tae-Hoon Shin; Shirley Chen; Xing Fan; Idalia Yabe; Sandhya R. Panch; So Gun Hong; Samson Koelle; Rong Lu; Aylin C. Bonifacino; Allen Krouse; Mark E. Metzger; Robert E. Donahue; Cynthia E. Dunbar

Age-associated changes in hematopoietic stem and progenitor cells (HSPCs) have been carefully documented in mouse models but poorly characterized in primates and humans. To investigate clinically relevant aspects of hematopoietic aging, we compared the clonal output of thousands of genetically barcoded HSPCs in aged vs young macaques after autologous transplantation. Aged macaques showed delayed emergence of output from multipotent (MP) clones, with persistence of lineage-biased clones for many months after engraftment. In contrast to murine aging models reporting persistence of myeloid-biased HSPCs, aged macaques demonstrated persistent output from both B-cell and myeloid-biased clones. Clonal expansions of MP, myeloid-biased, and B-biased clones occurred in aged macaques, providing a potential model for human clonal hematopoiesis of indeterminate prognosis. These results suggest that long-term MP HSPC output is impaired in aged macaques, resulting in differences in the kinetics and lineage reconstitution patterns between young and aged primates in an autologous transplantation setting.


Current protocols in stem cell biology | 2017

Rhesus Macaque iPSC Generation and Maintenance

Ravi Chandra Yada; So Gun Hong; Yongshun Lin; Thomas Winkler; Cynthia E. Dunbar

The rhesus macaque (Macaca mulatta) is physiologically and phylogenetically similar to humans, and therefore represents an invaluable model for the pre-clinical assessment of the safety and feasibility of iPSC-derived cell therapies. The use of an excisable polycistronic lentiviral STEMCCA vector to reprogram rhesus fibroblasts or bone marrow stromal cells (BMSCs) into RhiPSCs is described. After reprogramming, the pluripotency transgenes can be removed by transient expression of Cre, leaving a residual genetic tag that may be useful for identification of RhiPSC-derived tissues in vivo. Finally, the steps to maintain pluripotency during passaging of RhiPSCs, required for successful utilization of RhiPSCs, is described.


Scientific Reports | 2018

Efficient differentiation of cardiomyocytes and generation of calcium-sensor reporter lines from nonhuman primate iPSCs

Yongshun Lin; Huimin Liu; Michael G. Klein; John W. Ostrominski; So Gun Hong; Ravi Chandra Yada; Guibin Chen; Keron Navarengom; Robin Schwartzbeck; Hong San; Zu-Xi Yu; Chengyu Liu; Kaari Linask; Jeanette Beers; Lugui Qiu; Cynthia E. Dunbar; Manfred Boehm; Jizhong Zou

Nonhuman primate (NHP) models are more predictive than rodent models for developing induced pluripotent stem cell (iPSC)-based cell therapy, but robust and reproducible NHP iPSC-cardiomyocyte differentiation protocols are lacking for cardiomyopathies research. We developed a method to differentiate integration-free rhesus macaque iPSCs (RhiPSCs) into cardiomyocytes with >85% purity in 10 days, using fully chemically defined conditions. To enable visualization of intracellular calcium flux in beating cardiomyocytes, we used CRISPR/Cas9 to stably knock-in genetically encoded calcium indicators at the rhesus AAVS1 safe harbor locus. Rhesus cardiomyocytes derived by our stepwise differentiation method express signature cardiac markers and show normal electrochemical coupling. They are responsive to cardiorelevant drugs and can be successfully engrafted in a mouse myocardial infarction model. Our approach provides a powerful tool for generation of NHP iPSC-derived cardiomyocytes amenable to utilization in basic research and preclinical studies, including in vivo tissue regeneration models and drug screening.


Molecular therapy. Methods & clinical development | 2018

Barcoding of macaque hematopoietic stem and progenitor cells: a robust platform to assess vector genotoxicity

Idalia Yabe; Lauren Truitt; Diego A. Espinoza; Chuanfeng Wu; Samson Koelle; Sandhya R. Panch; Marcus A.F. Corat; Thomas Winkler; Kyung-Rok Yu; So Gun Hong; Aylin C. Bonifacino; Allen Krouse; Mark E. Metzger; Robert E. Donahue; Cynthia E. Dunbar

Gene therapies using integrating retrovirus vectors to modify hematopoietic stem and progenitor cells have shown great promise for the treatment of immune system and hematologic diseases. However, activation of proto-oncogenes via insertional mutagenesis has resulted in the development of leukemia. We have utilized cellular bar coding to investigate the impact of different vector designs on the clonal behavior of hematopoietic stem and progenitor cells (HSPCs) during in vivo expansion, as a quantitative surrogate assay for genotoxicity in a non-human primate model with high relevance for human biology. We transplanted two rhesus macaques with autologous CD34+ HSPCs transduced with three lentiviral vectors containing different promoters and/or enhancers of a predicted range of genotoxicities, each containing a high-diversity barcode library that uniquely tags each individual transduced HSPC. Analysis of clonal output from thousands of individual HSPCs transduced with these barcoded vectors revealed sustained clonal diversity, with no progressive dominance of clones containing any of the three vectors for up to almost 3 years post-transplantation. Our data support a low genotoxic risk for lentivirus vectors in HSPCs, even those containing strong promoters and/or enhancers. Additionally, this flexible system can be used for the testing of future vector designs.


Current protocols in stem cell biology | 2017

CRISPR/Cas9‐Based Safe‐Harbor Gene Editing in Rhesus iPSCs

Ravi Chandra Yada; John W. Ostrominski; I. Tunc; So Gun Hong; Jizhong Zou; Cynthia E. Dunbar

NHP iPSCs provide a unique opportunity to test safety and efficacy of iPSC-derived therapies in clinically relevant NHP models. To monitor these cells in vivo, there is a need for safe and efficient labeling methods. Gene insertion into genomic safe harbors (GSHs) supports reliable transgene expression while minimizing the risk the modification poses to the host genome or target cell. Specifically, this protocol demonstrates targeting of the adeno-associated virus site 1 (AAVS1), one of the most widely used GSH loci in the human genome, with CRISPR/Cas9, allowing targeted marker or therapeutic gene insertion in rhesus macaque induced pluripotent stem cells (RhiPSCs). Furthermore, detailed instructions for screening targeted clones and a tool for assessing potential off-target nuclease activity are provided.


Molecular Therapy | 2016

523. Human Umbilical Vein Endothelial Cell, HUVEC, Co-Culture Promotes Robust Expansion and Maintains Phenotypic Integrity of Rhesus Hematopoietic Stem and Progenitor Cells, HSPC, Prior to Autologous Tansplantation

Sandhya R. Panch; Idalia Yabe; Diego A. Espinoza; Chuanfeng Wu; So Gun Hong; Stephanie Sellers; Allen Krouse; Aylin C. Bonifacino; Robert E. Donahue; Cynthia E. Dunbar

The development of ex-vivo HSPC expansion techniques is particulary relevant for improving cord blood transplantation and gene therapy. Despite successful long-term, multilineage reconstitution of expanded human cord blood HSPC in immunodeficient mice, early phase clinical trials have failed to demonstrate improved outcomes. Thus, it is critical to develop a robust pre-clinical model to study ex-vivo expansion strategies, particularly function of long term HSPC and engrafment of all lineages, difficult in xenograft models. We have previously used retroviral insertion site retrieval and autologous competitive transplantation in rhesus macaques to track hematopoietic engraftment and ontogeny at a single HSPC level, in addition to comparing ex-vivo expanded and unexpanded HSPC (Gomes et al, Mol Ther). More recently we have developed retrieval of 31bp diverse barcodes as a more robust and quantitative in vivo HSPC tracking approach (Wu et al, Cell Stem Cell). Modified human endothelial cells (HUVEC) with the ability to be maintained in serum free media for prolonged periods have shown to support and expand human HSPC in murine Butler et al, Blood). We have developed a barcoded rhesus autologous transplant model to evaluate expansion of HPSC on HUVEC versus unexpanded or cytokine expanded rhesus HSPC. We first tested the feasibility of rhesus CD34+ cell expansion on HUVEC versus cytokines/fibronectin for 8 days. We also evaluated for the diversity of expanded HSPC by retrieving transduced viral barcode DNA tags from individual cells by low cycle PCR followed by Illumina sequencing. On average, rhesus CD34+ cells expanded 76 fold (± 53) on HUVEC versus 13 fold (±7) in cytokines (n=4; p=0.03). By morphologic assessment, the HUVEC expanded cell fraction contained higher numbers of progenitors and differentiating cells, with up to 97% CD34+CD45+ cells in the HUVEC expanded fraction and 65% in the cytokine expanded fraction. The HUVEC expanded rhesus CD34+ cells were capable of multi-lineage colony formation after 8 days of expansion. Within the HUVEC expanded cell fractions, fold expansion was similar whether CD34+ cells were frozen after (C1) or before (C2) transduction-expansion. However, viability and percentages of CD34+CD45+ cells were higher in C2 compared to C1 (55% versus 26%). GFP transduction efficiency was also higher in C2 compared to C1 (43% vs. 28%). We demonstrated a slightly lower percentage but higher absolute number of CD34+CD38-CD45RA-CD90+CD49f+Rho-low putative long-term HSC after expansion on HUVEC compared to in cytokines over 26 days in culture (0.018% vs. 0.016% pre and post expansion, respectively). Retrieved barcode analysis and quantitation revealed that in the HUVEC expanded fraction, the top 20 clones constituted up 15% of the total valid reads by day 8, in contrast to only 3.9% of total clones in the cytokine expanded fraction. Cells have been transplanted into autologous macaques, in a competitive model comparing non-expanded, cytokine-expanded and HUVEC-expanded conditions, and in vivo expansion and barcode analysis will be presented. Our data thus far show that peripheral blood CD34+ cells from rhesus macaques expand more robustly in the presence of HUVEC and cytokines as compared to expansion in cytokines alone, and barcode analysis suggests that the HUVEC fraction selectively expanded a subset of highly proliferative cells, which may represent true HSCs within the CD34+ cell fraction.

Collaboration


Dive into the So Gun Hong's collaboration.

Top Co-Authors

Avatar

Cynthia E. Dunbar

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Thomas Winkler

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Chuanfeng Wu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jizhong Zou

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ravi Chandra Yada

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert E. Donahue

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yongshun Lin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Allen Krouse

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Aylin C. Bonifacino

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Diego A. Espinoza

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge