Network


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

Hotspot


Dive into the research topics where Cheul H. Cho is active.

Publication


Featured researches published by Cheul H. Cho.


Hepatology | 2008

Mesenchymal stem cell-derived molecules directly modulate hepatocellular death and regeneration in vitro and in vivo.

Daan van Poll; Biju Parekkadan; Cheul H. Cho; Francois Berthiaume; Yaakov Nahmias; Arno W. Tilles; Martin L. Yarmush

Orthotopic liver transplantation is the only proven effective treatment for fulminant hepatic failure (FHF), but its use is limited because of organ donor shortage, associated high costs, and the requirement for lifelong immunosuppression. FHF is usually accompanied by massive hepatocellular death with compensatory liver regeneration that fails to meet the cellular losses. Therefore, therapy aimed at inhibiting cell death and stimulating endogenous repair pathways could offer major benefits in the treatment of FHF. Recent studies have demonstrated that mesenchymal stem cell (MSC) therapy can prevent parenchymal cell loss and promote tissue repair in models of myocardial infarction, acute kidney failure, and stroke through the action of trophic secreted molecules. In this study, we investigated whether MSC therapy can protect the acutely injured liver and stimulate regeneration. In a D‐galactosamine–induced rat model of acute liver injury, we show that systemic infusion of MSC‐conditioned medium (MSC‐CM) provides a significant survival benefit and prevents the release of liver injury biomarkers. Furthermore, MSC‐CM therapy resulted in a 90% reduction of apoptotic hepatocellular death and a three‐fold increment in the number of proliferating hepatocytes. This was accompanied by a dramatic increase in the expression levels of 10 genes known to be up‐regulated during hepatocyte replication. Direct antiapoptotic and promitotic effects of MSC‐CM on hepatocytes were demonstrated using in vitro assays. Conclusion: These data provide the first clear evidence that MSC‐CM therapy provides trophic support to the injured liver by inhibiting hepatocellular death and stimulating regeneration, potentially creating new avenues for the treatment of FHF. (HEPATOLOGY 2008.)


Biochemical and Biophysical Research Communications | 2013

A multicellular 3D heterospheroid model of liver tumor and stromal cells in collagen gel for anti-cancer drug testing

Derek Yip; Cheul H. Cho

Two-dimensional (2D) monolayer cultures are the standard in vitro model for cancer research. However, they fail to recapitulate the three-dimensional (3D) environment and quickly lose their function. In this study, we developed a new 3D multicellular heterospheroid tumor model in a collagen hydrogel culture system that more closely mimics the in vivo tumor microenvironment for anti-cancer drug testing. Three aspects of cancer were chosen to be modeled based on their ability to resist anti-cancer drugs: 3D, multicellularity, and extracellular matrix (ECM) barrier. The hanging drop method and co-culture of liver carcinoma with stromal fibroblasts were used to form controlled and uniform heterospheroids. These heterospheroids were then encapsulated in collagen gel in order to create a 3D model of liver cancer that would act more similarly to in vivo ECM conditions. The 3D heterospheroid tumor model was tested with an anti-cancer drug to determine how each of the above aspects affects drug resistance. The results demonstrate that the 3D heterospheroid model is more resistant to drug over 2D monolayer and homospheroid cultures, indicating stromal fibroblasts and collagen hydrogel culture system provides more resistance to anti-cancer drug. This study will provide useful information toward the development of improved biomimetic tumor models in vitro for cancer research in pre-clinical drug development.


The FASEB Journal | 2008

Homogeneous differentiation of hepatocyte-like cells from embryonic stem cells: applications for the treatment of liver failure

Cheul H. Cho; Natesh Parashurama; Eric Y.H. Park; Kazuhiro Suganuma; Yaakov Nahmias; Jaesung Park; Arno W. Tilles; Francois Berthiaume; Martin L. Yarmush

One of the major hurdles of cellular therapies for the treatment of liver failure is the low availability of functional human hepatocytes. While embryonic stem (ES) cells represent a potential cell source for therapy, current methods for differentiation result in mixed cell populations or low yields of the cells of interest. Here we describe a rapid, direct differentiation method that yields a homogeneous population of endoderm‐like cells with 95% purity. Mouse ES cells cultured on top of collagen‐sandwiched hepa‐tocytes differentiated and proliferated into a uniform and homogeneous cell population of endoderm‐like cells. The endoderm‐like cell population was positive for Foxa2, Sox17, and AFP and could be further differentiated into hepatocyte‐like cells, demonstrating hepatic morphology, functionality, and gene and protein expression. Incorporating the hepatocyte‐like cells into a bioartificial liver device to treat fulminant hepatic failure improved animal survival, thereby underscoring the therapeutic potential of these cells.—Cho, C. H., Parashurama, N., Park, E. Y. H., Suganuma, K., Nahmias, Y., Park, J., Tilles, A. W., Berthiaume, F., Yarmush, M. L. Homogeneous differentiation of hep‐atocyte‐like cells from embryonic stem cells: applications for the treatment of liver failure. FASEB J. 22, 898–909 (2008)


BioTechniques | 2010

Layered patterning of hepatocytes in co-culture systems using microfabricated stencils

Cheul H. Cho; Jaesung Park; Arno W. Tilles; Francois Berthiaume; Mehmet Toner; Martin L. Yarmush

Microfabrication and micropatterning techniques in tissue engineering offer great potential for creating and controlling microenvironments in which cell behavior can be observed. Here we present a novel approach to generate layered patterning of hepatocytes on micropatterned fibroblast feeder layers using microfabricated polydimethylsiloxane (PDMS) stencils. We fabricated PDMS stencils to pattern circular holes with diameters of 500 microm. Hepatocytes were co-cultured with 3T3-J2 fibroblasts in two types of patterns to evaluate and characterize the cellular interactions in the co-culture systems. Results of this study demonstrated uniform intracellular albumin staining and E-cadherin expression, increased liver-specific functions, and active glycogen synthesis in the hepatocytes when the heterotypic interface between hepatocytes and fibroblasts was increased by the layered patterning technique. This patterning technique can be a useful experimental tool for applications in basic science, drug screening, and tissue engineering, as well as in the design of bioartificial liver devices.


Biotechnology and Bioengineering | 2013

Functional 3-D Cardiac Co-Culture Model Using Bioactive Chitosan Nanofiber Scaffolds

Ali Hussain; George L. Collins; Derek Yip; Cheul H. Cho

The in vitro generation of a three‐dimensional (3‐D) myocardial tissue‐like construct employing cells, biomaterials, and biomolecules is a promising strategy in cardiac tissue regeneration, drug testing, and tissue engineering applications. Despite significant progress in this field, current cardiac tissue models are not yet able to stably maintain functional characteristics of cardiomyocytes for long‐term culture and therapeutic purposes. The objective of this study was to fabricate bioactive 3‐D chitosan nanofiber scaffolds using an electrospinning technique and exploring its potential for long‐term cardiac function in the 3‐D co‐culture model. Chitosan is a natural polysaccharide biomaterial that is biocompatible, biodegradable, non‐toxic, and cost effective. Electrospun chitosan was utilized to provide structural scaffolding characterized by scale and architectural resemblance to the extracellular matrix (ECM) in vivo. The chitosan fibers were coated with fibronectin via adsorption in order to enhance cellular adhesion to the fibers and migration into the interfibrous milieu. Ventricular cardiomyocytes were harvested from neonatal rats and studied in various culture conditions (i.e., mono‐ and co‐cultures) for their viability and function. Cellular morphology and functionality were examined using immunofluorescent staining for alpha‐sarcomeric actin (SM‐actin) and gap junction protein, Connexin‐43 (Cx43). Scanning electron microscopy (SEM) and light microscopy were used to investigate cellular morphology, spatial organization, and contractions. Calcium indicator was used to monitor calcium ion flux of beating cardiomyocytes. The results demonstrate that the chitosan nanofibers retained their cylindrical morphology in long‐term cell cultures and exhibited good cellular attachment and spreading in the presence of adhesion molecule, fibronectin. Cardiomyocyte mono‐cultures resulted in loss of cardiomyocyte polarity and islands of non‐coherent contractions. However, the cardiomyocyte‐fibroblast co‐cultures resulted in polarized cardiomyocyte morphology and retained their morphology and function for long‐term culture. The Cx43 expression in the fibroblast co‐culture was higher than the cardiomyocytes mono‐culture and endothelial cells co‐culture. In addition, fibroblast co‐cultures demonstrated synchronized contractions involving large tissue‐like cellular networks. To our knowledge, this is the first attempt to test chitosan nanofiber scaffolds as a 3‐D cardiac co‐culture model. Our results demonstrate that chitosan nanofibers can serve as a potential scaffold that can retain cardiac structure and function. These studies will provide useful information to develop a strategy that allows us to generate engineered 3‐D cardiac tissue constructs using biocompatible and biodegradable chitosan nanofiber scaffolds for many tissue engineering applications. Biotechnol. Bioeng. 2013; 110: 637–647.


Acta Biomaterialia | 2013

Heparin crosslinked chitosan microspheres for the delivery of neural stem cells and growth factors for central nervous system repair.

Nolan B. Skop; Frances Calderon; Steven W. Levison; Chirag D. Gandhi; Cheul H. Cho

An effective paradigm for transplanting large numbers of neural stem cells after central nervous system (CNS) injury has yet to be established. Biomaterial scaffolds have shown promise in cell transplantation and in regenerative medicine, but improved scaffolds are needed. In this study we designed and optimized multifunctional and biocompatible chitosan-based films and microspheres for the delivery of neural stem cells and growth factors for CNS injuries. The chitosan microspheres were fabricated by coaxial airflow techniques, with the sphere size controlled by varying the syringe needle gauge and the airflow rate. When applying a coaxial airflow at 30 standard cubic feet per hour, ∼300μm diameter spheres were reproducibly generated that were physically stable yet susceptible to enzymatic degradation. Heparin was covalently crosslinked to the chitosan scaffolds using genipin, which bound fibroblast growth factor-2 (FGF-2) with high affinity while retaining its biological activity. At 1μgml(-1) approximately 80% of the FGF-2 bound to the scaffold. A neural stem cell line, GFP+RG3.6 derived from embryonic rat cortex, was used to evaluate cytocompatibility, attachment and survival on the crosslinked chitosan-heparin complex surfaces. The MTT assay and microscopic analysis revealed that the scaffold containing tethered FGF-2 was superior in sustaining survival and growth of neural stem cells compared to standard culture conditions. Altogether, our results demonstrate that this multifunctional scaffold possesses good cytocompatibility and can be used as a growth factor delivery vehicle while supporting neural stem cell attachment and survival.


Biotechnology and Bioengineering | 2008

A new technique for primary hepatocyte expansion in vitro

Cheul H. Cho; Francois Berthiaume; Arno W. Tilles; Martin L. Yarmush

The current application for many potential cell‐based treatments for liver failure is limited by the low availability of mature functional hepatocytes. Although adult hepatocytes have a remarkable ability to proliferate in vivo, attempts to proliferate adult hepatocytes in vitro have been less successful. In this study, we investigated the effect of coculture cell type on the proliferative response and the functional activities of hepatocytes. We show, for the first time, a robust proliferative response of primary adult rat hepatocytes when cocultured with mouse 3T3‐J2 fibroblasts. Hepatocytes cultured at low density on growth‐arrested 3T3‐J2 fibroblast feeder layers underwent significantly higher proliferation rates than when cultured on feeder layers made of four other cell types. Increasing colony size correlated with an increase in hepatocellular functions. The proliferating hepatocytes retained their morphologic, phenotypic, and functional characteristics. Using a cell patterning technique, we found that 3T3‐J2 fibroblasts stimulate DNA synthesis in hepatocytes by short‐range heterotypic cell–cell interactions. When hepatocytes that proliferated in cocultures were harvested and further subcultured either on 3T3‐J2 fibroblast feeders or in the collagen sandwich configuration, their behavior was similar to that of freshly isolated hepatocytes. We conclude that adult rat hepatocytes can proliferate in vitro in a coculture cell type‐dependent manner, and can be serially propagated by coculturing with 3T3‐J2 fibroblasts while maintaining their differentiated characteristics. Our results also suggest that one of the major reasons for the functional differences in hepatocyte cocultures may be due to the different proliferative responses of hepatocytes as a function of coculture cell type. This study provides new insights in the roles of coculture cell types and cell–cell interactions in the modulation of hepatic proliferation and function. Biotechnol. Bioeng. 2008;101: 345–356.


Molecular and Cellular Therapies | 2014

Improvements in biomaterial matrices for neural precursor cell transplantation

Nolan Skop; Frances Calderon; Cheul H. Cho; Chirag D. Gandhi; Steven W. Levison

Progress is being made in developing neuroprotective strategies for traumatic brain injuries; however, there will never be a therapy that will fully preserve neurons that are injured from moderate to severe head injuries. Therefore, to restore neurological function, regenerative strategies will be required. Given the limited regenerative capacity of the resident neural precursors of the CNS, many investigators have evaluated the regenerative potential of transplanted precursors. Unfortunately, these precursors do not thrive when engrafted without a biomaterial scaffold. In this article we review the types of natural and synthetic materials that are being used in brain tissue engineering applications for traumatic brain injury and stroke. We also analyze modifications of the scaffolds including immobilizing drugs, growth factors and extracellular matrix molecules to improve CNS regeneration and functional recovery. We conclude with a discussion of some of the challenges that remain to be solved towards repairing and regenerating the brain.


BioTechniques | 2012

Adhesive-tape soft lithography for patterning mammalian cells: application to wound-healing assays.

Anil Shrirao; Ali Hussain; Cheul H. Cho; Raquel Perez-Castillejos

This paper introduces a benchtop method for patterning mammalian cells-i.e., for culturing cells at specific locations-on planar substrates. Compared with standard cell culture techniques, which do not allow the control of what areas of a monolayer are populated by one type of cell or another, techniques of cell patterning open new routes to cell biology. Researchers interested in cell patterning, however, are often times hindered by limited access to photolithographic capabilities. This paper shows how cells can be patterned easily with sub-millimeter precision using a non-photolithographic technique that is based on the use of office adhesive tape and poly(dimethylsiloxane) (PDMS). This method is fast (~4 h to go from a layout to have the cells patterned in the shape of such layout) and only requires materials and tools readily available in a conventional biomedical laboratory. A wound-healing assay is presented here that illustrates the potential of the technique (which we call tape-based soft lithography) for patterning mammalian cells and studying biologically significant questions such as collective cellular migration.


Nature Communications | 2017

Long-term hepatitis B infection in a scalable hepatic co-culture system

Benjamin Y. Winer; Tiffany Huang; Eitan Pludwinski; Brigitte Heller; Felix Wojcik; Gabriel Lipkowitz; Amit Parekh; Cheul H. Cho; Anil Shrirao; Tom W. Muir; Eric Novik; Alexander Ploss

Hepatitis B virus causes chronic infections in 250 million people worldwide. Chronic hepatitis B virus carriers are at risk of developing fibrosis, cirrhosis, and hepatocellular carcinoma. A prophylactic vaccine exists and currently available antivirals can suppress but rarely cure chronic infections. The study of hepatitis B virus and development of curative antivirals are hampered by a scarcity of models that mimic infection in a physiologically relevant, cellular context. Here, we show that cell-culture and patient-derived hepatitis B virus can establish persistent infection for over 30 days in a self-assembling, primary hepatocyte co-culture system. Importantly, infection can be established without antiviral immune suppression, and susceptibility is not donor dependent. The platform is scalable to microwell formats, and we provide proof-of-concept for its use in testing entry inhibitors and antiviral compounds.The lack of models that mimic hepatitis B virus (HBV) infection in a physiologically relevant context has hampered drug development. Here, Winer et al. establish a self-assembling, primary hepatocyte co-culture system that can be infected with patient-derived HBV without further modifications.

Collaboration


Dive into the Cheul H. Cho's collaboration.

Top Co-Authors

Avatar

Arno W. Tilles

Shriners Hospitals for Children

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ali Hussain

New Jersey Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Amit Parekh

New Jersey Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Derek Yip

New Jersey Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge