Network


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

Hotspot


Dive into the research topics where Charles Y. Cho is active.

Publication


Featured researches published by Charles Y. Cho.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Reprogramming of murine fibroblasts to induced pluripotent stem cells with chemical complementation of Klf4.

Costas A. Lyssiotis; Ruth K. Foreman; Judith Staerk; Michael Garcia; Divya Mathur; Styliani Markoulaki; Jacob Hanna; Luke L. Lairson; Bradley D. Charette; Laure C. Bouchez; Michael Bollong; Conrad Kunick; Achim Brinker; Charles Y. Cho; Peter G. Schultz; Rudolf Jaenisch

Ectopic expression of defined transcription factors can reprogram somatic cells to induced pluripotent stem (iPS) cells, but the utility of iPS cells is hampered by the use of viral delivery systems. Small molecules offer an alternative to replace virally transduced transcription factors with chemical signaling cues responsible for reprogramming. In this report we describe a small-molecule screening platform applied to identify compounds that functionally replace the reprogramming factor Klf4. A series of small-molecule scaffolds were identified that activate Nanog expression in mouse fibroblasts transduced with a subset of reprogramming factors lacking Klf4. Application of one such molecule, kenpaullone, in lieu of Klf4 gave rise to iPS cells that are indistinguishable from murine embryonic stem cells. This experimental platform can be used to screen large chemical libraries in search of novel compounds to replace the reprogramming factors that induce pluripotency. Ultimately, such compounds may provide mechanistic insight into the reprogramming process.


Science | 2012

A Stem Cell–Based Approach to Cartilage Repair

Kristen Johnson; Shoutian Zhu; Matthew S. Tremblay; Joshua N. Payette; Jianing Wang; Laure C. Bouchez; Shelly Meeusen; Alana Althage; Charles Y. Cho; Xu Wu; Peter G. Schultz

Osteoarthritis and Kartogenin Osteoarthritis is characterized by progressive breakdown of articular cartilage and affects over 25 million people in the United States. Mesenchymal stem cells (MSCs), which reside in healthy and diseased joints, are multipotent adult stem cells that are able to differentiate into a variety of cell types. Johnson et al. (p. 717, published online 5 April) identified a small molecule, kartogenin, which was able to induce MSCs to differentiate into chondrocytes in vitro. When administered locally, kartogenin was efficacious in two animal models of osteoarthritis. A chemical screen using mesenchymal stem cells identifies a small molecule, kartogenin, that can promote chondrogenesis. Osteoarthritis (OA) is a degenerative joint disease that involves the destruction of articular cartilage and eventually leads to disability. Molecules that promote the selective differentiation of multipotent mesenchymal stem cells (MSCs) into chondrocytes may stimulate the repair of damaged cartilage. Using an image-based high-throughput screen, we identified the small molecule kartogenin, which promotes chondrocyte differentiation (median effective concentration = 100 nM), shows chondroprotective effects in vitro, and is efficacious in two OA animal models. Kartogenin binds filamin A, disrupts its interaction with the transcription factor core-binding factor β subunit (CBFβ), and induces chondrogenesis by regulating the CBFβ-RUNX1 transcriptional program. This work provides new insights into the control of chondrogenesis that may ultimately lead to a stem cell—based therapy for osteoarthritis.


Nature | 2011

Oxysterols direct immune cell migration via EBI2.

Sébastien Hannedouche; Juan Zhang; Tangsheng Yi; Weijun Shen; Deborah Nguyen; João P. Pereira; Danilo Guerini; Birgit Baumgarten; Silvio Roggo; Ben Wen; Richard Knochenmuss; Sophie Noël; François Gessier; Lisa M. Kelly; Mirka Vanek; Stephane Laurent; Inga Preuss; Charlotte Miault; Isabelle Christen; Ratna Karuna; Wei Li; Dong-In Koo; Thomas Suply; Christian Schmedt; Eric C. Peters; Rocco Falchetto; Andreas Katopodis; Carsten Spanka; Marie-Odile Roy; Michel Detheux

Epstein–Barr virus-induced gene 2 (EBI2, also known as GPR183) is a G-protein-coupled receptor that is required for humoral immune responses; polymorphisms in the receptor have been associated with inflammatory autoimmune diseases. The natural ligand for EBI2 has been unknown. Here we describe the identification of 7α,25-dihydroxycholesterol (also called 7α,25-OHC or 5-cholesten-3β,7α,25-triol) as a potent and selective agonist of EBI2. Functional activation of human EBI2 by 7α,25-OHC and closely related oxysterols was verified by monitoring second messenger readouts and saturable, high-affinity radioligand binding. Furthermore, we find that 7α,25-OHC and closely related oxysterols act as chemoattractants for immune cells expressing EBI2 by directing cell migration in vitro and in vivo. A critical enzyme required for the generation of 7α,25-OHC is cholesterol 25-hydroxylase (CH25H). Similar to EBI2 receptor knockout mice, mice deficient in CH25H fail to position activated B cells within the spleen to the outer follicle and mount a reduced plasma cell response after an immune challenge. This demonstrates that CH25H generates EBI2 biological activity in vivo and indicates that the EBI2–oxysterol signalling pathway has an important role in the adaptive immune response.


Cell Stem Cell | 2009

A Small Molecule Primes Embryonic Stem Cells for Differentiation

Shoutian Zhu; Heiko Wurdak; Jian Wang; Costas A. Lyssiotis; Eric C. Peters; Charles Y. Cho; Xu Wu; Peter G. Schultz

Embryonic stem cells (ESCs) are an attractive source of cells for disease modeling in vitro and may eventually provide access to cells/tissues for the treatment of many degenerative diseases. However, applications of ESC-derived cell types are largely hindered by the lack of highly efficient methods for lineage-specific differentiation. Using a high-content screen, we have identified a small molecule, named stauprimide, that increases the efficiency of the directed differentiation of mouse and human ESCs in synergy with defined extracellular signaling cues. Affinity-based methods revealed that stauprimide interacts with NME2 and inhibits its nuclear localization. This, in turn, leads to downregulation of c-Myc, a key regulator of the pluripotent state. Thus, our findings identify a chemical tool that primes ESCs for efficient differentiation through a mechanism that affects c-Myc expression, and this study points to an important role for NME2 in ESC self-renewal.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Genome-wide functional analysis of human cell-cycle regulators

Mridul Mukherji; Russell Bell; Lubica Supekova; Yan Wang; Anthony P. Orth; Serge Batalov; Loren Miraglia; Dieter Huesken; Joerg Lange; Chris Martin; Sudhir Sahasrabudhe; Mischa Reinhardt; Francois Natt; Jonathan Hall; Craig Mickanin; Mark Labow; Sumit K. Chanda; Charles Y. Cho; Peter G. Schultz

Human cells have evolved complex signaling networks to coordinate the cell cycle. A detailed understanding of the global regulation of this fundamental process requires comprehensive identification of the genes and pathways involved in the various stages of cell-cycle progression. To this end, we report a genome-wide analysis of the human cell cycle, cell size, and proliferation by targeting >95% of the protein-coding genes in the human genome using small interfering RNAs (siRNAs). Analysis of >2 million images, acquired by quantitative fluorescence microscopy, showed that depletion of 1,152 genes strongly affected cell-cycle progression. These genes clustered into eight distinct phenotypic categories based on phase of arrest, nuclear area, and nuclear morphology. Phase-specific networks were built by interrogating knowledge-based and physical interaction databases with identified genes. Genome-wide analysis of cell-cycle regulators revealed a number of kinase, phosphatase, and proteolytic proteins and also suggests that processes thought to regulate G1-S phase progression like receptor-mediated signaling, nutrient status, and translation also play important roles in the regulation of G2/M phase transition. Moreover, 15 genes that are integral to TNF/NF-κB signaling were found to regulate G2/M, a previously unanticipated role for this pathway. These analyses provide systems-level insight into both known and novel genes as well as pathways that regulate cell-cycle progression, a number of which may provide new therapeutic approaches for the treatment of cancer.


Clinical Cancer Research | 2010

Combination Therapy Targeting Both Tumor-Initiating and Differentiated Cell Populations in Prostate Carcinoma

Anna Dubrovska; Jimmy Elliott; Richard J. Salamone; Sungeun Kim; Lindsey Aimone; John R. Walker; James C. Watson; Maira Sauveur-Michel; Carlos Garcia-Echeverria; Charles Y. Cho; Venkateshwar A. Reddy; Peter G. Schultz

Purpose: The cancer stem cell hypothesis predicts that standard prostate cancer monotherapy eliminates bulk tumor cells but not a tumor-initiating cell population, eventually leading to relapse. Many studies have sought to determine the underlying differences between bulk tumor and cancer stem cells. Experimental Design: Our previous data suggest that the PTEN/PI3K/AKT pathway is critical for the in vitro maintenance of CD133+/CD44+ prostate cancer progenitors and, consequently, that targeting PI3K signaling may be beneficial in treatment of prostate cancer. Results: Here, we show that inhibition of PI3K activity by the dual PI3K/mTOR inhibitor NVP-BEZ235 leads to a decrease in the population of CD133+/CD44+ prostate cancer progenitor cells in vivo. Moreover, the combination of the PI3K/mTOR modulator NVP-BEZ235, which eliminates prostate cancer progenitor populations, and the chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors in a prostate cancer xenograft model than monotherapy. Conclusion: This combination treatment ultimately leads to the expansion of cancer progenitors with a PTEN E91D mutation, suggesting that the analysis of PTEN mutations could predict therapeutic response to the dual therapy. Clin Cancer Res; 16(23); 5692–702. ©2010 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2010

A small molecule accelerates neuronal differentiation in the adult rat.

Heiko Wurdak; Shoutian Zhu; Kyung-Hoon Min; Lindsey Aimone; Luke L. Lairson; James E. M. Watson; Gregory Chopiuk; James Demas; Bradley D. Charette; Rajkumar Halder; Eranthie Weerapana; Benjamin F. Cravatt; Hollis T. Cline; Eric C. Peters; Jay Zhang; John R. Walker; Chunlei Wu; Jonathan Chang; Tove Tuntland; Charles Y. Cho; Peter G. Schultz

Adult neurogenesis occurs in mammals and provides a mechanism for continuous neural plasticity in the brain. However, little is known about the molecular mechanisms regulating hippocampal neural progenitor cells (NPCs) and whether their fate can be pharmacologically modulated to improve neural plasticity and regeneration. Here, we report the characterization of a small molecule (KHS101) that selectively induces a neuronal differentiation phenotype. Mechanism of action studies revealed a link of KHS101 to cell cycle exit and specific binding to the TACC3 protein, whose knockdown in NPCs recapitulates the KHS101-induced phenotype. Upon systemic administration, KHS101 distributed to the brain and resulted in a significant increase in neuronal differentiation in vivo. Our findings indicate that KHS101 accelerates neuronal differentiation by interaction with TACC3 and may provide a basis for pharmacological intervention directed at endogenous NPCs.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Screening the mammalian extracellular proteome for regulators of embryonic human stem cell pluripotency

Rodolfo Gonzalez; Lori L. Jennings; Mark W. Knuth; Anthony P. Orth; Heath E. Klock; Weija Ou; Julie Feuerhelm; Mitchell V. Hull; Eric Koesema; Yuping Wang; Jia Zhang; Chunlei Wu; Charles Y. Cho; Andrew I. Su; Serge Batalov; Hong Chen; Kristen Johnson; Bryan A. Laffitte; Deborah G. Nguyen; Evan Y. Snyder; Peter G. Schultz; Jennifer L. Harris; Scott A. Lesley

Approximately 3,500 mammalian genes are predicted to be secreted or single-pass transmembrane proteins. The function of the majority of these genes is still unknown, and a number of the encoded proteins might find use as new therapeutic agents themselves or as targets for small molecule or antibody drug development. To analyze the physiological activities of the extracellular proteome, we developed a large-scale, high-throughput protein expression, purification, and screening platform. For this study, the complete human extracellular proteome was analyzed and prioritized based on genome-wide disease association studies to select 529 initial target genes. These genes were cloned into three expression vectors as native sequences and as N-terminal and C-terminal Fc fusions to create an initial collection of 806 purified secreted proteins. To determine its utility, this library was screened in an OCT4-based cellular assay to identify regulators of human embryonic stem-cell self-renewal. We found that the pigment epithelium-derived factor can promote long-term pluripotent growth of human embryonic stem cells without bFGF or TGFβ/Activin/Nodal ligand supplementation. Our results further indicate that activation of the pigment epithelium-derived factor receptor-Erk1/2 signaling pathway by the pigment epithelium-derived factor is sufficient to maintain the self-renewal of pluripotent human embryonic stem cells. These experiments illustrate the potential for discovering novel biological functions by directly screening protein diversity in cell-based phenotypic or reporter assays.


PLOS ONE | 2012

CXCR4 expression in prostate cancer progenitor cells.

Anna Dubrovska; Jimmy Elliott; Richard J. Salamone; Gennady D. Telegeev; Alexander E. Stakhovsky; Ihor B. Schepotin; Feng Yan; Yan Wang; Laure C. Bouchez; Sumith A. Kularatne; James Watson; Christopher Trussell; Venkateshwar A. Reddy; Charles Y. Cho; Peter G. Schultz

Tumor progenitor cells represent a population of drug-resistant cells that can survive conventional chemotherapy and lead to tumor relapse. However, little is known of the role of tumor progenitors in prostate cancer metastasis. The studies reported herein show that the CXCR4/CXCL12 axis, a key regulator of tumor dissemination, plays a role in the maintenance of prostate cancer stem-like cells. The CXCL4/CXCR12 pathway is activated in the CD44+/CD133+ prostate progenitor population and affects differentiation potential, cell adhesion, clonal growth and tumorigenicity. Furthermore, prostate tumor xenograft studies in mice showed that a combination of the CXCR4 receptor antagonist AMD3100, which targets prostate cancer stem-like cells, and the conventional chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors as compared to monotherapy.


ChemBioChem | 2007

A Small‐Molecule Antagonist of the Hedgehog Signaling Pathway

Jongkook Lee; Xu Wu; Marina Pasca di Magliano; Eric C. Peters; Yan Wang; Jiyong Hong; Metthias Hebrok; Sheng Ding; Charles Y. Cho; Peter G. Schultz

The Hedgehog (Hh) signaling pathway plays an important role in embryonic pattern formation and adult tissue maintenance by directing cell differentiation and proliferation. In mammals, three Hh genes, Sonic (Shh), Indian (Ihh), and Desert (Dhh), have been identified. Binding of Hh protein to the membrane receptor Patched (Ptc) releases its inhibitory effect on Smoothened (Smo). Activated Smo further transduces downstream signals to activate the Gli family of transcription factors, which promote the expression of Hh signaling target genes. Hh signaling has recently attracted considerable interest based on the discovery that aberrant activation of Shh signaling leads to the formation of various tumors, which include pancreatic cancer, medulloblastoma, basal cell carcinoma, small cell lung cancer, and prostate cancer. 5] Cell-based phenotypic assays and, more recently, pathway screens of natural products and synthetic small molecules have provided useful chemical tools for modulating and/or studying complex cellular processes, both in vitro and in vivo. Several Hh antagonists, including cyclopamine, CUR61414, and SANT1-4 have been reported. Some of these antagonists exert antiproliferative effects by binding directly to Smo. However, cancer cells with mutations downstream of Smo are resistant to these antagonists. Therefore, Hh antagonists that interrupt downstream Hh signaling could lead to antiproliferative agents with a broader spectrum of activity. Here, we report the identification and characterization of the Hh signaling antagonist, JK184, and initial studies to characterize its biological mechanism of action. To screen small molecule libraries for compounds that antagonize Hh signaling, we developed a protocol using mesenchymal progenitor (C3H10T1/2) cells derived from the mouse embryonic mesoderm. These cells were stably transfected with a reporter construct that encoded luciferase, which was driven by Gli responsive elements, along with a neomycin resistanceconferring gene. Stably transfected C3H10T1/2 cells were plated into 384-well plates, and treated with a library of approximately 20000 heterocycles (2 mm, final concentration). After treatment with compound for 36 h in the presence of recombinant peptide that corresponded to the N terminus of Shh (100 ngmL ), luciferase activity was assayed, and a number of active 2,4-disubstituted thiazole compounds were identified. One of these compounds, JK184 (Figure 1A), inhibited Gli-dependent transcriptional activity in a dose-dependent manner with an IC50 value of 30 nm. This effect was fur-

Collaboration


Dive into the Charles Y. Cho's collaboration.

Top Co-Authors

Avatar

Peter G. Schultz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Eric C. Peters

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Weijun Shen

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anthony P. Orth

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Xu Wu

Harvard University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John R. Walker

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Laure C. Bouchez

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Shoutian Zhu

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge