Network


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

Hotspot


Dive into the research topics where Peili Gu is active.

Publication


Featured researches published by Peili Gu.


Nature Cell Biology | 2008

Nanog and Oct4 associate with unique transcriptional repression complexes in embryonic stem cells

Jiancong Liang; Ma Wan; Yi Zhang; Peili Gu; Huawei Xin; Sung Yun Jung; Jun Qin; Jiemin Wong; Austin J. Cooney; Dan Liu; Zhou Songyang

Nanog and Oct4 are essential transcription factors that regulate self-renewal and pluripotency of ES cells. However, the mechanisms by which Nanog and Oct4 modulate ES cell fate remain unknown. Through characterization of endogenous Nanog and Oct4 protein complexes in mouse ES cells, we found that these transcription factors interact with each other and associate with proteins from multiple repression complexes, including the NuRD, Sin3A and Pml complexes. In addition, Nanog, Oct4 and repressor proteins co-occupy Nanog-target genes in mouse ES cells, suggesting that Nanog and Oct4 together may communicate with distinct repression complexes to control gene transcription. To our surprise, of the various core components in the NuRD complex with which Nanog and Oct4 interact, Mta1 was preferred, whereas Mbd3 and Rbbp7 were either absent or present at sub-stoichiometric levels. We named this unique Hdac1/2- and Mta1/2-containing complex NODE (for Nanog and Oct4 associated deacetylase). Interestingly, NODE contained histone deacetylase (HDAC) activity that seemed to be comparable to NuRD, and retained its association with Nanog and Oct4 in Mbd3−/− ES cells. In contrast to Mbd3 loss-of-function, knockdown of NODE subunits led to increased expression of developmentally regulated genes and ES-cell differentiation. Our data collectively suggest that Nanog and Oct4 associate with unique repressor complexes on their target genes to control ES cell fate.


Molecular and Cellular Biology | 2005

Orphan Nuclear Receptor LRH-1 Is Required To Maintain Oct4 Expression at the Epiblast Stage of Embryonic Development

Peili Gu; Bryan Goodwin; Arthur C.-K. Chung; Xueping Xu; David A. Wheeler; Roger R. Price; Cristin M. Galardi; Li Peng; Anne M. Latour; Beverly H. Koller; Jan A. Gossen; Steven A. Kliewer; Austin J. Cooney

ABSTRACT Oct4 plays an essential role in maintaining the inner cell mass and pluripotence of embryonic stem (ES) cells. The expression of Oct4 is regulated by the proximal enhancer and promoter in the epiblast and by the distal enhancer and promoter at all other stages in the pluripotent cell lineage. Here we report that the orphan nuclear receptor LRH-1, which is expressed in undifferentiated ES cells, can bind to SF-1 response elements in the proximal promoter and proximal enhancer of the Oct4 gene and activate Oct4 reporter gene expression. LRH-1 is colocalized with Oct4 in the inner cell mass and the epiblast of embryos at early developmental stages. Disruption of the LRH-1 gene results in loss of Oct4 expression at the epiblast stage and early embryonic death. Using LRH-1 −/− ES cells, we also show that LRH-1 is required to maintain Oct4 expression at early differentiation time points. In vitro and in vivo results show that LRH-1 plays an essential role in the maintenance of Oct4 expression in ES cells at the epiblast stage of embryonic development, thereby maintaining pluripotence at this crucial developmental stage prior to segregation of the primordial germ cell lineage at gastrulation.


The EMBO Journal | 2003

GCNF‐dependent repression of BMP‐15 and GDF‐9 mediates gamete regulation of female fertility

Zi Jian Lan; Peili Gu; Xueping Xu; Kathy J. Jackson; Francesco J. DeMayo; Bert W. O'Malley; Austin J. Cooney

To determine the function of germ cell nuclear factor (GCNF) in female reproduction, we generated an oocyte‐specific GCNF knockout mouse model (GCNFfl/flZp3Cre+). These mice displayed hypofertility due to prolonged diestrus phase of the estrous cycle and aberrant steroidogenesis. These reproductive defects were secondary to a primary defect in the oocytes, in which expression of the paracrine transforming growth factor‐β signaling molecules, bone morphogenetic protein 15 (BMP‐15) and growth differentiation factor 9 (GDF‐9), were up‐regulated in GCNFfl/flZp3Cre+ females at diestrus. This was a direct effect of GCNF, as molecular studies showed that GCNF bound to DR0 elements within the BMP‐15 and GDF‐9 gene promoters and repressed their reporter activities. Consistent with these findings, abnormal double‐oocyte follicles, indicative of aberrant BMP‐15/GDF‐9 expression, were observed in GCNFfl/flZp3Cre+ females. The Cre/loxP knockout of GCNF in the oocyte has uncovered a new regulatory pathway in ovarian function. Our results show that GCNF directly regulates paracrine communication between the oocyte and somatic cells by regulating the expression of BMP‐15 and GDF‐9, to affect female fertility.


PLOS ONE | 2008

Novel MicroRNA Candidates and miRNA-mRNA Pairs in Embryonic Stem (ES) Cells

Peili Gu; Jeffrey G. Reid; Xiaolian Gao; Chad A. Shaw; Chad J. Creighton; Peter L. Tran; Xiaochuan Zhou; Rafal Drabek; David Steffen; David M. Hoang; Michelle K. Weiss; Arash O. Naghavi; Jad El-daye; Mahjabeen Khan; Glen B. Legge; David A. Wheeler; Richard A. Gibbs; Jonathan Miller; Austin J. Cooney; Preethi H. Gunaratne

Background MicroRNAs (miRNAs: a class of short non-coding RNAs) are emerging as important agents of post transcriptional gene regulation and integral components of gene networks. MiRNAs have been strongly linked to stem cells, which have a remarkable dual role in development. They can either continuously replenish themselves (self-renewal), or differentiate into cells that execute a limited number of specific actions (pluripotence). Methodology/Principal Findings In order to identify novel miRNAs from narrow windows of development we carried out an in silico search for micro-conserved elements (MCE) in adult tissue progenitor transcript sequences. A plethora of previously unknown miRNA candidates were revealed including 545 small RNAs that are enriched in embryonic stem (ES) cells over adult cells. Approximately 20% of these novel candidates are down-regulated in ES (Dicer −/−) ES cells that are impaired in miRNA maturation. The ES-enriched miRNA candidates exhibit distinct and opposite expression trends from mmu-mirs (an abundant class in adult tissues) during retinoic acid (RA)-induced ES cell differentiation. Significant perturbation of trends is found in both miRNAs and novel candidates in ES (GCNF −/−) cells, which display loss of repression of pluripotence genes upon differentiation. Conclusion/Significance Combining expression profile information with miRNA target prediction, we identified miRNA-mRNA pairs that correlate with ES cell pluripotence and differentiation. Perturbation of these pairs in the ES (GCNF −/−) mutant suggests a role for miRNAs in the core regulatory networks underlying ES cell self-renewal, pluripotence and differentiation.


Molecular and Cellular Biology | 2006

Differential Recruitment of Methylated CpG Binding Domains by the Orphan Receptor GCNF Initiates the Repression and Silencing of Oct4 Expression

Peili Gu; Damien Le Menuet; Arthur C.-K. Chung; Austin J. Cooney

ABSTRACT The pluripotent factor Oct4 is a key transcription factor that maintains embryonic stem (ES) cell self-renewal and is down-regulated upon the differentiation of ES cells and silenced in somatic cells. A combination of cis elements, transcription factors, and epigenetic modifications, such as DNA methylation, are involved in the regulation of Oct4 gene expression. Here we show that the orphan nuclear receptor GCNF initiates Oct4 repression and DNA methylation by the differential recruitment of MBD (methylated CpG binding domain) factors to the promoter. Compared with wild-type ES cells and gastrulating embryos, Oct4 repression is lost and its proximal promoter is significantly hypomethylated in RA-differentiated GCNF−/− ES cells. The Oct4 gene is reexpressed in some somatic cells of GCNF−/− embryos, showing that it has not been properly silenced coincident with reduced DNA methylation of its promoter. Efforts to characterize mediators of GCNFs repressive function and DNA methylation of the Oct4 promoter identified methyl-DNA binding proteins, MBD3 and MBD2, as GCNF-interacting factors. In P19 and ES cells, upon differentiation, endogenous GCNF binds to the Oct4 proximal promoter and differentially recruits MBD3 and MBD2. In differentiated GCNF−/− ES cells, recruitment of MBD3 and MBD2 to the Oct4 promoter is lost, and repression of Oct4 expression and DNA methylation fails to occur. RNA interference-mediated knockdown of MBD3 and/or MBD2 expression results in reduced Oct4 repression in differentiated P19 and ES cells. Repression of Oct4 expression and recruitment of MBD3 are maintained in de novo DNA methylation-deficient ES cells (Dnmt3A/3B-null cells), while MBD2 recruitment is lost. Thus, recruitment of MBD3 and MBD2 by GCNF links two events, gene-specific repression and DNA methylation, which occur differentially at the Oct4 promoter. GCNF initiates the repression and epigenetic modification of Oct4 gene during ES cell differentiation.


Ppar Research | 2007

Nuclear Receptors in Regulation of Mouse ES Cell Pluripotency and Differentiation

Eimear M. Mullen; Peili Gu; Austin J. Cooney

Embryonic stem (ES) cells have great therapeutic potential because they are capable of indefinite self-renewal and have the potential to differentiate into over 200 different cell types that compose the human body. The switch from the pluripotent phenotype to a differentiated cell involves many complex signaling pathways including those involving LIF/Stat3 and the transcription factors Sox2, Nanog and Oct-4. Many nuclear receptors play an important role in the maintenance of pluripotence (ERRβ, SF-1, LRH-1, DAX-1) repression of the ES cell phenotype (RAR, RXR, GCNF) and also the differentiation of ES cells (PPARγ). Here we review the roles of the nuclear receptors involved in regulating these important processes in ES cells.


Biology of Reproduction | 2003

Expression of the Orphan Nuclear Receptor, Germ Cell Nuclear Factor, in Mouse Gonads and Preimplantation Embryos

Zi Jian Lan; Peili Gu; Xueping Xu; Austin J. Cooney

Abstract Germ cell nuclear factor (GCNF, NR6A1) is an orphan member of the nuclear receptor superfamily and functions as a repressor of gene transcription. GCNF mRNA is expressed in postgastrulation mouse embryos and is required for normal mouse embryonic development. In adult mice, GCNF transcripts are predominantly expressed in spermatogenic cells and growing oocytes of the gonads. To extend this observation to the protein level, we generated and characterized a specific antibody against GCNF. Using this antibody we found that GCNF protein was exclusively present in postmeiotic spermatogenic cells of the testis in 21- and 56-day-old mice. In the ovary, GCNF protein was present in the cytoplasm of oocytes from primary to preovulatory follicles. GCNF protein was also present in unfertilized oocytes and preimplantation embryos. The presence of GCNF protein in adult mouse gonads indicates that GCNF may play a role during gametogenesis. Our results also show that GCNF in early embryos is a maternal protein and could be involved in the regulation of zygotic gene expression and preimplantation embryonic development.


Stem Cells | 2011

Differential Recruitment of Methyl CpG‐Binding Domain Factors and DNA Methyltransferases by the Orphan Receptor Germ Cell Nuclear Factor Initiates the Repression and Silencing of Oct4

Peili Gu; Xueping Xu; Damien Le Menuet; Arthur C.-K. Chung; Austin J. Cooney

The pluripotency gene Oct4 encodes a key transcription factor that maintains self‐renewal of embryonic stem cell (ESC) and is downregulated upon differentiation of ESCs and silenced in somatic cells. A combination of cis elements, transcription factors, and epigenetic modifications, such as DNA methylation, mediates Oct4 gene expression. Here, we show that the orphan nuclear receptor germ cell nuclear factor (GCNF) initiates Oct4 repression and DNA methylation by the differential recruitment of methyl‐CpG binding domain (MBD) and DNA methyltransferases (Dnmts) to the Oct4 promoter. When compared with wild‐type ESCs and gastrulating embryos, Oct4 repression is lost and its proximal promoter is significantly hypomethylated in retinoic acid (RA)‐differentiated GCNF−/− ESCs and GCNF−/− embryos. Efforts to characterize mediators of GCNFs repressive function and DNA methylation of the Oct4 promoter identified MBD3, MBD2, and de novo Dnmts as GCNF interacting factors. Upon differentiation, endogenous GCNF binds to the Oct4 proximal promoter and differentially recruits MBD3 and MBD2 as well as Dnmt3A. In differentiated GCNF−/− ESCs, recruitment of MBD3 and MBD2 as well as Dnmt3A to Oct4 promoter is lost and subsequently Oct4 repression and DNA methylation failed to occur. Hypomethylation of the Oct4 promoter is also observed in RA‐differentiated MBD3−/− and Dnmt3A−/− ESCs, but not in MBD2−/− and Dnmt3B−/− ESCs. Thus, recruitment of MBD3, MBD2, and Dnmt3A by GCNF links two events: gene‐specific repression and DNA methylation, which occur differentially at the Oct4 promoter. GCNF initiates the repression and epigenetic modification of Oct4 gene during ESC differentiation. STEM CELLS 2011;29:1041–1051


PLOS ONE | 2011

miRNA Regulatory Circuits in ES Cells Differentiation: A Chemical Kinetics Modeling Approach

Zijun Luo; Xuping Xu; Peili Gu; David M. Lonard; Preethi H. Gunaratne; Austin J. Cooney; Robert Azencott

MicroRNAs (miRNAs) play an important role in gene regulation for Embryonic Stem cells (ES cells), where they either down-regulate target mRNA genes by degradation or repress protein expression of these mRNA genes by inhibiting translation. Well known tables TargetScan and miRanda may predict quite long lists of potential miRNAs inhibitors for each mRNA gene, and one of our goals was to strongly narrow down the list of mRNA targets potentially repressed by a known large list of 400 miRNAs. Our paper focuses on algorithmic analysis of ES cells microarray data to reliably detect repressive interactions between miRNAs and mRNAs. We model, by chemical kinetics equations, the interaction architectures implementing the two basic silencing processes of miRNAs, namely “direct degradation” or “translation inhibition” of targeted mRNAs. For each pair (M,G) of potentially interacting miRMA gene M and mRNA gene G, we parameterize our associated kinetic equations by optimizing their fit with microarray data. When this fit is high enough, we validate the pair (M,G) as a highly probable repressive interaction. This approach leads to the computation of a highly selective and drastically reduced list of repressive pairs (M,G) involved in ES cells differentiation.


Journal of Molecular Biology | 2005

Correlated evolutionary pressure at interacting transcription factors and DNA response elements can guide the rational engineering of DNA binding specificity.

Michele Raviscioni; Peili Gu; Minawar Sattar; Austin J. Cooney; Olivier Lichtarge

Collaboration


Dive into the Peili Gu's collaboration.

Top Co-Authors

Avatar

Austin J. Cooney

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xueping Xu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Damien Le Menuet

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

David A. Wheeler

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Minawar Sattar

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Olivier Lichtarge

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Daniel H. Morgan

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Eimear M. Mullen

Northern Kentucky University

View shared research outputs
Researchain Logo
Decentralizing Knowledge