Network


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

Hotspot


Dive into the research topics where Ren-He Xu is active.

Publication


Featured researches published by Ren-He Xu.


Nature Biotechnology | 2002

BMP4 initiates human embryonic stem cell differentiation to trophoblast.

Ren-He Xu; Xin Chen; Dong S. Li; Rui Li; Gregory C. Addicks; Clay Glennon; Thomas P. Zwaka; James A. Thomson

The excitement and controversy surrounding the potential role of human embryonic stem (ES) cells in transplantation therapy have often overshadowed their potentially more important use as a basic research tool for understanding the development and function of human tissues. Human ES cells can proliferate without a known limit and can form advanced derivatives of all three embryonic germ layers. What is less widely appreciated is that human ES cells can also form the extra-embryonic tissues that differentiate from the embryo before gastrulation. The use of human ES cells to derive early human trophoblast is particularly valuable, because it is difficult to obtain from other sources and is significantly different from mouse trophoblast. Here we show that bone morphogenetic protein 4 (BMP4), a member of the transforming growth factor-β (TGF-β) superfamily, induces the differentiation of human ES cells to trophoblast. DNA microarray, RT-PCR, and immunoassay analyses demonstrate that the differentiated cells express a range of trophoblast markers and secrete placental hormones. When plated at low density, the BMP4-treated cells form syncytia that express chorionic gonadotrophin (CG). These results underscore fundamental differences between human and mouse ES cells, which differentiate poorly, if at all, to trophoblast. Human ES cells thus provide a tool for studying the differentiation and function of early human trophoblast and could provide a new understanding of some of the earliest differentiation events of human postimplantation development.


Nature Methods | 2005

Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells.

Ren-He Xu; Ruthann M. Peck; Dong S. Li; Xuezhu Feng; Tenneille E. Ludwig; James A. Thomson

Human embryonic stem cells (hESCs) are routinely cultured on fibroblast feeder layers or in fibroblast-conditioned medium (CM). Bone morphogenetic proteins (BMPs) have previously been shown to induce hESC differentiation, in apparent contrast to mouse embryonic stem (ES) cells, in which BMP4 synergizes with leukemia inhibitory factor (LIF) to maintain self-renewal. Here we demonstrate that hESCs cultured in unconditioned medium (UM) are subjected to high levels of BMP signaling activity, which is reduced in CM. The BMP antagonist noggin synergizes with basic fibroblast growth factor (bFGF) to repress BMP signaling and sustain undifferentiated proliferation of hESCs in the absence of fibroblasts or CM. These findings suggest a basic difference in the self-renewal mechanism between mouse and human ES cells and simplify the culture of hESCs.


Stem Cells | 2006

Basic Fibroblast Growth Factor Support of Human Embryonic Stem Cell Self‐Renewal

Mark E. Levenstein; Tenneille E. Ludwig; Ren-He Xu; Rachel A. Llanas; Kaitlyn VanDenHeuvel‐Kramer; Daisy Manning; James A. Thomson

Human embryonic stem (ES) cells have most commonly been cultured in the presence of basic fibroblast growth factor (FGF2) either on fibroblast feeder layers or in fibroblast‐conditioned medium. It has recently been reported that elevated concentrations of FGF2 permit the culture of human ES cells in the absence of fibroblasts or fibroblast‐conditioned medium. Herein we compare the ability of unconditioned medium (UM) supplemented with 4, 24, 40, 80, 100, and 250 ng/ml FGF2 to sustain low‐density human ES cell cultures through multiple passages. In these stringent culture conditions, 4, 24, and 40 ng/ml FGF2 failed to sustain human ES cells through three passages, but 100 ng/ml sustained human ES cells with an effectiveness comparable to conditioned medium (CM). Two human ES cell lines (H1 and H9) were maintained for up to 164 population doublings (7 and 4 months) in UM supplemented with 100 ng/ml FGF2. After prolonged culture, the cells formed teratomas when injected into severe combined immunodeficient beige mice and expressed markers characteristic of undifferentiated human ES cells. We also demonstrate that FGF2 is degraded more rapidly in UM than in CM, partly explaining the need for higher concentrations of FGF2 in UM. These results further facilitate the large‐scale, routine culture of human ES cells and suggest that fibroblasts and fibro‐blast‐conditioned medium sustain human ES cells in part by stabilizing FGF signaling above a critical threshold.


Cell Stem Cell | 2008

NANOG Is a Direct Target of TGFβ/Activin-Mediated SMAD Signaling in Human ESCs

Ren-He Xu; Tori L. Sampsell-Barron; Feng Gu; Sierra H. Root; Ruthann M. Peck; Guangjin Pan; Junying Yu; Jessica Antosiewicz-Bourget; Shulan Tian; Ron Stewart; James A. Thomson

Self-renewal of human embryonic stem cells (ESCs) is promoted by FGF and TGFbeta/Activin signaling, and differentiation is promoted by BMP signaling, but how these signals regulate genes critical to the maintenance of pluripotency has been unclear. Using a defined medium, we show here that both TGFbeta and FGF signals synergize to inhibit BMP signaling; sustain expression of pluripotency-associated genes such as NANOG, OCT4, and SOX2; and promote long-term undifferentiated proliferation of human ESCs. We also show that both TGFbeta- and BMP-responsive SMADs can bind with the NANOG proximal promoter. NANOG promoter activity is enhanced by TGFbeta/Activin and FGF signaling and is decreased by BMP signaling. Mutation of putative SMAD binding elements reduces NANOG promoter activity to basal levels and makes NANOG unresponsive to BMP and TGFbeta signaling. These results suggest that direct binding of TGFbeta/Activin-responsive SMADs to the NANOG promoter plays an essential role in sustaining human ESC self-renewal.


Proceedings of the National Academy of Sciences of the United States of America | 2008

X-inactivation in female human embryonic stem cells is in a nonrandom pattern and prone to epigenetic alterations

Yin Shen; Youko Matsuno; Shaun D. Fouse; Nagesh Rao; Sierra Root; Ren-He Xu; Matteo Pellegrini; Arthur D. Riggs; Guoping Fan

X chromosome inactivation (XCI) is an essential mechanism for dosage compensation of X-linked genes in female cells. We report that subcultures from lines of female human embryonic stem cells (hESCs) exhibit variation (0–100%) for XCI markers, including XIST RNA expression and enrichment of histone H3 lysine 27 trimethylation (H3K27me3) on the inactive X chromosome (Xi). Surprisingly, regardless of the presence or absence of XCI markers in different cultures, all female hESCs we examined (H7, H9, and HSF6 cells) exhibit a monoallelic expression pattern for a majority of X-linked genes. Our results suggest that these established female hESCs have already completed XCI during the process of derivation and/or propagation, and the XCI pattern of lines we investigated is already not random. Moreover, XIST gene expression in subsets of cultured female hESCs is unstable and subject to stable epigenetic silencing by DNA methylation. In the absence of XIST expression, ≈12% of X-linked promoter CpG islands become hypomethylated and a portion of X-linked alleles on the Xi are reactivated. Because alterations in dosage compensation of X-linked genes could impair somatic cell function, we propose that XCI status should be routinely checked in subcultures of female hESCs, with cultures displaying XCI markers better suited for use in regenerative medicine.


PLOS ONE | 2010

Specification of region-specific neurons including forebrain glutamatergic neurons from human induced pluripotent stem cells.

Hui Zeng; Min Guo; Kristen Martins-Taylor; Xiaofang Wang; Zheng Zhang; Jung W. Park; Shuning Zhan; Mark S. Kronenberg; Alexander C. Lichtler; Hui Xia Liu; Fang Ping Chen; Lixia Yue; Xue Jun Li; Ren-He Xu

Background Directed differentiation of human induced pluripotent stem cells (hiPSC) into functional, region-specific neural cells is a key step to realizing their therapeutic promise to treat various neural disorders, which awaits detailed elucidation. Methodology/Principal Findings We analyzed neural differentiation from various hiPSC lines generated by others and ourselves. Although heterogeneity in efficiency of neuroepithelial (NE) cell differentiation was observed among different hiPSC lines, the NE differentiation process resembles that from human embryonic stem cells (hESC) in morphology, timing, transcriptional profile, and requirement for FGF signaling. NE cells differentiated from hiPSC, like those from hESC, can also form rostral phenotypes by default, and form the midbrain or spinal progenitors upon caudalization by morphogens. The rostrocaudal neural progenitors can further mature to develop forebrain glutamatergic projection neurons, midbrain dopaminergic neurons, and spinal motor neurons, respectively. Typical ion channels and action potentials were recorded in the hiPSC-derived neurons. Conclusions/Significance Our results demonstrate that hiPSC, regardless of how they were derived, can differentiate into a spectrum of rostrocaudal neurons with functionality, which supports the considerable value of hiPSC for study and treatment of patient-specific neural disorders.


Nature Biotechnology | 2011

Recurrent copy number variations in human induced pluripotent stem cells

Kristen Martins-Taylor; Benjamin S. Nisler; Seth M. Taapken; Tiwanna Compton; Leann Crandall; Karen Montgomery; Marc Lalande; Ren-He Xu

1. Wadman, M. Nature published online, doi:10.1038/473015a (3 May 2011). 2. Obama, B. Removing barriers to responsible scientific research involving human stem cells. Executive Order 13505 (9 March 2009). 3. Sudman, S. & Bradburn, n. Response Effects in Surveys (Aldine, Chicago; 1974). 4. Evans, M.D.R. & Kelley, J. Australian Economy and Society 2002: Religion, Morality, and Public Policy in International Perspective, 1984–2002. (Federation Press, Sydney; 2004). 5. Evans, M.D.R., Zanjani, E.D. & Kelley, J. J. Hematother. Stem Cell Res. 11, 711–717 (2002). 6. Evans, M.D.R., Kelley, J. & Zanjani, E.D. Fetal Diagn. Ther. 20, 223–234 (2005). 7. Bollen, K.A. Structural Equations with Latent Variables (Wiley, new York; 1989). 8. nisbet, M.C. Science 316, 56 (2007). 9. Ho, S., Brossard, D. & Scheufele, D. Int. J. Public Opin. Res. 20, 171–192 (2008). option because the general public is more familiar with stem cell procedures involving them. Comparison of levels of approval for iPSCs to those for fertilized eggs suggests that approval for therapeutic cloning involving unfertilized eggs is likely to be 1 to 2 points out of 100 higher than with fertilized eggs (Supplementary Methods, table S3). Thus, the American public has five distinct attitudes toward (i) cloning animals, (ii) cloning humans, (iii) curing serious diseases by therapeutic cloning or by using stem cells from an IVF embryo, (iv) curing serious diseases using adult stem cells and (v) using any of these for cosmetic purposes. They are strongly opposed to some, have mixed feelings about others and strongly support all that offer hope of curing serious diseases. US federal government policy largely conforms to public opinion, but with a striking exception. Funding for research on therapeutic cloning has long been banned. Nonetheless, public opinion strongly supports it. In a democratic society, deferring to objections from a small (mainly religious) minority and limiting research that has so much therapeutic promise may well be unethical.


Stem Cells | 2012

Concise Review: Genomic Stability of Human Induced Pluripotent Stem Cells

Kristen Martins-Taylor; Ren-He Xu

The usefulness of human induced pluripotent stem cells (hiPSCs) in research and therapeutic applications highly relies on their genomic integrity and stability. Many laboratories including ours have addressed this concern by comparing genomic (at both karyotypic and subkaryotypic levels) and epigenomic abnormalities of hiPSC lines (derived via either DNA‐ or non‐DNA‐based methods), as well as human embryonic stem cell lines during long‐term culture. A variety of methods have been used for this purpose, such as karyotyping and fluorescent in situ hybridization to detect karyotypic abnormalities, array‐based comparative genomic hybridization to detect copy number variations (CNVs), single‐nucleotide polymorphism‐based microarrays to detect both CNVs and loss of heterozygosity, analysis of integration sites in the genome, and whole genome sequencing for protein‐coding exome and DNA methylome profiling. Here, we summarize the progresses in this dynamically evolving field and also discuss how the findings apply to the study and application of hiPSCs. STEM CELLS 2012;30:22–27


Journal of Cellular Biochemistry | 2013

Efficient differentiation of human iPSC‐derived mesenchymal stem cells to chondroprogenitor cells

Rosa M. Guzzo; Jason D. Gibson; Ren-He Xu; Francis Y. Lee; Hicham Drissi

Induced pluripotent stem cells (iPSC) hold tremendous potential for personalized cell‐based repair strategies to treat musculoskeletal disorders. To establish human iPSCs as a potential source of viable chondroprogenitors for articular cartilage repair, we assessed the in vitro chondrogenic potential of the pluripotent population versus an iPSC‐derived mesenchymal‐like progenitor population. We found the direct plating of undifferentiated iPSCs into high‐density micromass cultures in the presence of BMP‐2 promoted chondrogenic differentiation, however these conditions resulted in a mixed population of cells resembling the phenotype of articular cartilage, transient cartilage, and fibrocartilage. The progenitor cells derived from human iPSCs exhibited immunophenotypic features of mesenchymal stem cells (MSCs) and developed along multiple mesenchymal lineages, including osteoblasts, adipocytes, and chondrocytes in vitro. The data indicate the derivation of a mesenchymal stem cell population from human iPSCs is necessary to limit culture heterogeneity as well as chondrocyte maturation in the differentiated progeny. Moreover, as compared to pellet culture differentiation, BMP‐2 treatment of iPSC‐derived MSC‐like (iPSC–MSC) micromass cultures resulted in a phenotype more typical of articular chondrocytes, characterized by the enrichment of cartilage‐specific type II collagen (Col2a1), decreased expression of type I collagen (Col1a1) as well as lack of chondrocyte hypertrophy. These studies represent a first step toward identifying the most suitable iPSC progeny for developing cell‐based approaches to repair joint cartilage damage. J. Cell. Biochem. 114: 480–490, 2013.


Journal of Biological Chemistry | 2009

Inhibition of Caspase-mediated Anoikis Is Critical for Basic Fibroblast Growth Factor-sustained Culture of Human Pluripotent Stem Cells

Xiaofang Wang; Ge Lin; Kristen Martins-Taylor; Hui Zeng; Ren-He Xu

Apoptosis and proliferation are two dynamically and tightly regulated processes that together maintain the homeostasis of renewable tissues. Anoikis is a subtype of apoptosis induced by detachment of adherent cells from the extracellular matrix. By using the defined mTeSR1 medium and collecting freshly detached cells, we found here that human pluripotent stem (PS) cells including embryonic stem (ES) cells and induced pluripotent stem cells are subject to constant anoikis in culture, which is escalated in the absence of basic fibroblast growth factor (bFGF). Withdrawal of bFGF also promotes apoptosis and differentiation of the remaining adherent cells without affecting their cell cycle progression. Insulin-like growth factor 2 (IGF2) has previously been reported to act downstream of FGF signaling to support self-renewal of human ES cells. However, we found that IGF2 cannot substitute bFGF in the TeSR1-supported culture, although endogenous IGF signaling is required to sustain self-renewal of human ES cells. On the other hand, all of the bFGF withdrawal effects observed here can be markedly prevented by the caspase inhibitor z-VAD-FMK. We further demonstrated that the bFGF-repressed anoikis is dependent on activation of ERK and AKT and associated with inhibition of Bcl-2-interacting mediator of cell death and the caspase-ROCK1-myosin signaling. Anoikis is independent of pre-detachment apoptosis and differentiation of the cells. Because previous studies of human PS cells have been focused on attached cells, our findings revealed a neglected role of bFGF in sustaining self-renewal of human PS cells: preventing them from anoikis via inhibition of caspase activation.

Collaboration


Dive into the Ren-He Xu's collaboration.

Top Co-Authors

Avatar

Ying Peng

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Kristen Martins-Taylor

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Hsiang-Fu Kung

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Jung W. Park

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Ge Lin

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Zeng

Central South University

View shared research outputs
Top Co-Authors

Avatar

Hicham Drissi

University of Connecticut Health Center

View shared research outputs
Top Co-Authors

Avatar

Wei Si

Kunming Institute of Zoology

View shared research outputs
Researchain Logo
Decentralizing Knowledge