Network


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

Hotspot


Dive into the research topics where Duancheng Wen is active.

Publication


Featured researches published by Duancheng Wen.


Cell | 2010

Distinct Factors Control Histone Variant H3.3 Localization at Specific Genomic Regions

Aaron D. Goldberg; Laura A. Banaszynski; Kyung-Min Noh; Peter W. Lewis; Simon J. Elsaesser; Sonja C. Stadler; Scott Dewell; Martin Law; Xingyi Guo; Xuan Li; Duancheng Wen; Ariane Chapgier; Russell DeKelver; Jeffrey C. Miller; Ya Li Lee; Elizabeth A. Boydston; Michael C. Holmes; Philip D. Gregory; John M. Greally; Shahin Rafii; Chingwen Yang; Peter J. Scambler; David Garrick; Richard J. Gibbons; Douglas R. Higgs; Ileana M. Cristea; Fyodor D. Urnov; Deyou Zheng; C. David Allis

The incorporation of histone H3 variants has been implicated in the epigenetic memory of cellular state. Using genome editing with zinc-finger nucleases to tag endogenous H3.3, we report genome-wide profiles of H3 variants in mammalian embryonic stem cells and neuronal precursor cells. Genome-wide patterns of H3.3 are dependent on amino acid sequence and change with cellular differentiation at developmentally regulated loci. The H3.3 chaperone Hira is required for H3.3 enrichment at active and repressed genes. Strikingly, Hira is not essential for localization of H3.3 at telomeres and many transcription factor binding sites. Immunoaffinity purification and mass spectrometry reveal that the proteins Atrx and Daxx associate with H3.3 in a Hira-independent manner. Atrx is required for Hira-independent localization of H3.3 at telomeres and for the repression of telomeric RNA. Our data demonstrate that multiple and distinct factors are responsible for H3.3 localization at specific genomic locations in mammalian cells.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Histone variant H3.3 is an essential maternal factor for oocyte reprogramming

Duancheng Wen; Laura A. Banaszynski; Ying Liu; Fuqiang Geng; Kyung-Min Noh; Jenny Xiang; Olivier Elemento; Z. Rosenwaks; C. David Allis; Shahin Rafii

Significance A differentiated cell nucleus can be reprogrammed into the pluripotent state by maternal factors in ooplasm; the factors that are responsible for this reprogramming process have not yet been identified. In this paper, we show that histone variant H3.3 is one of the essential maternal factors involved in somatic nuclear reprogramming. Maternal H3.3, not H3.3 in the donor chromatin, is required for development and the reactivation of many key pluripotency genes in somatic cell nuclear transfer (SCNT) embryos. H3.3 facilitates reprogramming by remodeling the donor nuclear chromatin through replacement of donor H3 in chromatin with de novo synthesized maternal H3.3 at the beginning of reprogramming in SCNT embryos. Mature oocyte cytoplasm can reprogram somatic cell nuclei to the pluripotent state through a series of sequential events including protein exchange between the donor nucleus and ooplasm, chromatin remodeling, and pluripotency gene reactivation. Maternal factors that are responsible for this reprogramming process remain largely unidentified. Here, we demonstrate that knockdown of histone variant H3.3 in mouse oocytes results in compromised reprogramming and down-regulation of key pluripotency genes; and this compromised reprogramming for developmental potentials and transcription of pluripotency genes can be rescued by injecting exogenous H3.3 mRNA, but not H3.2 mRNA, into oocytes in somatic cell nuclear transfer embryos. We show that maternal H3.3, and not H3.3 in the donor nucleus, is essential for successful reprogramming of somatic cell nucleus into the pluripotent state. Furthermore, H3.3 is involved in this reprogramming process by remodeling the donor nuclear chromatin through replacement of donor nucleus-derived H3 with de novo synthesized maternal H3.3 protein. Our study shows that H3.3 is a crucial maternal factor for oocyte reprogramming and provides a practical model to directly dissect the oocyte for its reprogramming capacity.


Stem Cells | 2008

CD34+ Testicular Stromal Cells Support Long‐Term Expansion of Embryonic and Adult Stem and Progenitor Cells

Jiyeon Kim; Marco Seandel; Ilaria Falciatori; Duancheng Wen; Shahin Rafii

Stem cells reside in specialized microenvironments created by supporting stromal cells that orchestrate self‐renewal and lineage‐specific differentiation. However, the precise identity of the cellular and molecular pathways that support self‐renewal of stem cells is not known. For example, long‐term culture of prototypical stem cells, such as adult spermatogonial stem and progenitor cells (SPCs), in vitro has been impeded by the lack of an optimal stromal cell line that initiates and sustains proliferation of these cells. Indeed, current methods, including the use of mouse embryonic fibroblasts (MEFs), have not been efficient and have generally led to inconsistent results. Here, we report the establishment of a novel CD34‐positive cell line, referred to as JK1, derived from mouse testicular stromal cells that not only facilitated long‐term SPC culture but also allowed faithful generation of SPCs and multipotent stem cells. SPCs generated on JK1 maintained key features of germ line stem cells, including expression of PLZF, DAZL, and GCNA. Furthermore, these feeders also promoted the long‐term cultivation of other types of primitive cells including multipotent adult spermatogonial‐derived stem cells, pluripotent murine embryonic stem cells, and embryonic germ cells derived from primordial germ cells. Stem cells could be passaged serially and still maintained expression of characteristic markers such as OCT4 and NANOG in vitro, as well as the ability to generate all three germ layers in vivo. These results indicate that the JK1 cell line is capable of promoting long‐term culture of primitive cells. As such, this cell line allows for identification of stromal‐derived factors that support long‐term proliferation of various types of stem cells and constitutes a convenient alternative to other types of feeder layers.


Nucleus | 2014

H3.3 replacement facilitates epigenetic reprogramming of donor nuclei in somatic cell nuclear transfer embryos

Duancheng Wen; Laura A. Banaszynski; Z. Rosenwaks; C. David Allis; Shahin Rafii

Transfer of a somatic nucleus into an enucleated oocyte is the most efficient approach for somatic cell reprogramming. While this process is known to involve extensive chromatin remodeling of the donor nucleus, the maternal factors responsible and the underlying chromatin-based mechanisms remain largely unknown. Here we discuss our recent findings demonstrating that the histone variant H3.3 plays an essential role in reprogramming and is required for reactivation of key pluripotency genes in somatic cell nuclear transfer (SCNT) embryos. Maternal-derived H3.3 replaces H3 in the donor nucleus shortly after oocyte activation, with the amount of replacement directly related to the differentiation status of the donor nucleus in SCNT embryos. We provide additional evidence to suggest that de novo synthesized H3.3 replaces histone H3 carrying repressive modifications in the donor nuclei of SCNT embryos, and hypothesize that replacement may occur at specific loci that must be reprogrammed for gene reactivation.


PLOS ONE | 2014

Completely ES Cell-Derived Mice Produced by Tetraploid Complementation Using Inner Cell Mass (ICM) Deficient Blastocysts

Duancheng Wen; Néstor Saiz; Z. Rosenwaks; Anna-Katerina Hadjantonakis; Shahin Rafii

Tetraploid complementation is often used to produce mice from embryonic stem cells (ESCs) by injection of diploid (2n) ESCs into tetraploid (4n) blastocysts (ESC-derived mice). This method has also been adapted to mouse cloning and the derivation of mice from induced pluripotent stem (iPS) cells. However, the underlying mechanism(s) of the tetraploid complementation remains largely unclear. Whether this approach can give rise to completely ES cell-derived mice is an open question, and has not yet been unambiguously proven. Here, we show that mouse tetraploid blastocysts can be classified into two groups, according to the presence or absence of an inner cell mass (ICM). We designate these as type a (presence of ICM at blastocyst stage) or type b (absence of ICM). ESC lines were readily derived from type a blastocysts, suggesting that these embryos retain a pluripotent epiblast compartment; whereas the type b blastocysts possessed very low potential to give rise to ESC lines, suggesting that they had lost the pluripotent epiblast. When the type a blastocysts were used for tetraploid complementation, some of the resulting mice were found to be 2n/4n chimeric; whereas when type b blastocysts were used as hosts, the resulting mice are all completely ES cell-derived, with the newborn pups displaying a high frequency of abdominal hernias. Our results demonstrate that completely ES cell-derived mice can be produced using ICM-deficient 4n blastocysts, and provide evidence that the exclusion of tetraploid cells from the fetus in 2n/4n chimeras can largely be attributed to the formation of ICM-deficient blastocysts.


Nature Communications | 2016

Epigenetic profiles signify cell fate plasticity in unipotent spermatogonial stem and progenitor cells

Ying Liu; Eugenia G. Giannopoulou; Duancheng Wen; Ilaria Falciatori; Olivier Elemento; C. David Allis; Shahin Rafii; Marco Seandel

Spermatogonial stem and progenitor cells (SSCs) generate adult male gametes. During in vitro expansion, these unipotent murine cells spontaneously convert to multipotent adult spermatogonial-derived stem cells (MASCs). Here we investigate this conversion process through integrative transcriptomic and epigenomic analyses. We find in SSCs that promoters essential to maintenance and differentiation of embryonic stem cells (ESCs) are enriched with histone H3-lysine4 and -lysine 27 trimethylations. These bivalent modifications are maintained at most somatic promoters after conversion, bestowing MASCs an ESC-like promoter chromatin. At enhancers, the core pluripotency circuitry is activated partially in SSCs and completely in MASCs, concomitant with loss of germ cell-specific gene expression and initiation of embryonic-like programs. Furthermore, SSCs in vitro maintain the epigenomic characteristics of germ cells in vivo. Our observations suggest that SSCs encode innate plasticity through the epigenome and that both conversion of promoter chromatin states and activation of cell type-specific enhancers are prominent features of reprogramming.


Journal of Biological Chemistry | 2018

Histone variant H3.3-mediated chromatin remodeling is essential for paternal genome activation in mouse preimplantation embryos

Qingran Kong; Laura A. Banaszynski; Fuqiang Geng; Xiaolei Zhang; Jiaming Zhang; Heng Zhang; Claire L. O’Neill; Peidong Yan; Zhonghua Liu; Koji Shido; G.D. Palermo; C. David Allis; Shahin Rafii; Z. Rosenwaks; Duancheng Wen

Derepression of chromatin-mediated transcriptional repression of paternal and maternal genomes is considered the first major step that initiates zygotic gene expression after fertilization. The histone variant H3.3 is present in both male and female gametes and is thought to be important for remodeling the paternal and maternal genomes for activation during both fertilization and embryogenesis. However, the underlying mechanisms remain poorly understood. Using our H3.3B-HA–tagged mouse model, engineered to report H3.3 expression in live animals and to distinguish different sources of H3.3 protein in embryos, we show here that sperm-derived H3.3 (sH3.3) protein is removed from the sperm genome shortly after fertilization and extruded from the zygotes via the second polar bodies (PBII) during embryogenesis. We also found that the maternal H3.3 (mH3.3) protein is incorporated into the paternal genome as early as 2 h postfertilization and is detectable in the paternal genome until the morula stage. Knockdown of maternal H3.3 resulted in compromised embryonic development both of fertilized embryos and of androgenetic haploid embryos. Furthermore, we report that mH3.3 depletion in oocytes impairs both activation of the Oct4 pluripotency marker gene and global de novo transcription from the paternal genome important for early embryonic development. Our results suggest that H3.3-mediated paternal chromatin remodeling is essential for the development of preimplantation embryos and the activation of the paternal genome during embryogenesis.


Nature Cell Biology | 2018

Pluripotency transcription factors and Tet1/2 maintain Brd4-independent stem cell identity

Lydia W.S. Finley; Santosha Vardhana; Bryce W. Carey; Direna Alonso-Curbelo; Richard Koche; Yanyang Chen; Duancheng Wen; Bryan King; Megan R. Radler; Shahin Rafii; Scott W. Lowe; C. David Allis; Craig B. Thompson

A robust network of transcription factors and an open chromatin landscape are hallmarks of the naive pluripotent state. Recently, the acetyllysine reader Brd4 has been implicated in stem cell maintenance, but the relative contribution of Brd4 to pluripotency remains unclear. Here, we show that Brd4 is dispensable for self-renewal and pluripotency of embryonic stem cells (ESCs). When maintained in their ground state, ESCs retain transcription factor binding and chromatin accessibility independent of Brd4 function or expression. In metastable ESCs, Brd4 independence can be achieved by increased expression of pluripotency transcription factors, including STAT3, Nanog or Klf4, so long as the DNA methylcytosine oxidases Tet1 and Tet2 are present. These data reveal that Brd4 is not essential for ESC self-renewal. Rather, the levels of pluripotency transcription factor abundance and Tet1/2 function determine the extent to which bromodomain recognition of protein acetylation contributes to the maintenance of gene expression and cell identity.Finley et al. show that Brd4 is dispensable for self-renewal and pluripotency in murine embryonic stem cells (ESCs). In metastable ESCs, Brd4 independence can be achieved by increasing the expression of the pluripotency transcription factors Oct4, Sox2 and Nanog as long as Tet1/2 are present.


Cell | 2013

Hira-Dependent Histone H3.3 Deposition Facilitates PRC2 Recruitment at Developmental Loci in ES Cells

Laura A. Banaszynski; Duancheng Wen; Scott Dewell; Sarah J. Whitcomb; Mingyan Lin; Nichole Diaz; Simon J. Elsässer; Ariane Chapgier; Aaron D. Goldberg; Eli Canaani; Shahin Rafii; Deyou Zheng; C. David Allis


Neuron | 2015

Critical Role of Histone Turnover in Neuronal Transcription and Plasticity.

Ian Maze; Wendy Wenderski; Kyung-Min Noh; Rosemary C. Bagot; Nikos Tzavaras; Immanuel Purushothaman; Simon J. Elsässer; Yin Guo; Carolina Ionete; Yasmin L. Hurd; Carol A. Tamminga; Tobias Halene; Lorna Farrelly; Alexey A. Soshnev; Duancheng Wen; Shahin Rafii; Marc R. Birtwistle; Schahram Akbarian; Bruce A. Buchholz; Robert D. Blitzer; Eric J. Nestler; Zuo-Fei Yuan; Benjamin A. Garcia; Li Shen; Henrik Molina; C. David Allis

Collaboration


Dive into the Duancheng Wen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deyou Zheng

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Fuqiang Geng

Howard Hughes Medical Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge