Network


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

Hotspot


Dive into the research topics where Guangjin Pan is active.

Publication


Featured researches published by Guangjin Pan.


Cell | 2009

MicroRNA-145 Regulates OCT4, SOX2, and KLF4 and Represses Pluripotency in Human Embryonic Stem Cells

Na Xu; Guangjin Pan; James A. Thomson; Kenneth S. Kosik

MicroRNAs (miRNAs) are posttranscriptional modulators of gene expression and play an important role in many developmental processes. We report here that expression of microRNA-145 (miR-145) is low in self-renewing human embryonic stem cells (hESCs) but highly upregulated during differentiation. We identify the pluripotency factors OCT4, SOX2, and KLF4 as direct targets of miR-145 and show that endogenous miR-145 represses the 3 untranslated regions of OCT4, SOX2, and KLF4. Increased miR-145 expression inhibits hESC self-renewal, represses expression of pluripotency genes, and induces lineage-restricted differentiation. Loss of miR-145 impairs differentiation and elevates OCT4, SOX2, and KLF4. Furthermore, we find that the miR-145 promoter is bound and repressed by OCT4 in hESCs. This work reveals a direct link between the core reprogramming factors and miR-145 and uncovers a double-negative feedback loop involving OCT4, SOX2, KLF4, and miR-145.


Cell Stem Cell | 2007

Whole-Genome Analysis of Histone H3 Lysine 4 and Lysine 27 Methylation in Human Embryonic Stem Cells

Guangjin Pan; Shulan Tian; Jeff Nie; Chuhu Yang; Victor Ruotti; Hairong Wei; Gudrun A. Jonsdottir; Ron Stewart; James A. Thomson

We mapped Polycomb-associated H3K27 trimethylation (H3K27me3) and Trithorax-associated H3K4 trimethylation (H3K4me3) across the whole genome in human embryonic stem (ES) cells. The vast majority of H3K27me3 colocalized on genes modified with H3K4me3. These commodified genes displayed low expression levels and were enriched in developmental function. Another significant set of genes lacked both modifications and was also expressed at low levels in ES cells but was enriched for gene function in physiological responses rather than development. Commodified genes could change expression levels rapidly during differentiation, but so could a substantial number of genes in other modification categories. SOX2, POU5F1, and NANOG, pluripotency-associated genes, shifted from modification by H3K4me3 alone to colocalization of both modifications as they were repressed during differentiation. Our results demonstrate that H3K27me3 modifications change during early differentiation, both relieving existing repressive domains and imparting new ones, and that colocalization with H3K4me3 is not restricted to pluripotent cells.


Cell Research | 2007

Nanog and transcriptional networks in embryonic stem cell pluripotency

Guangjin Pan; James A. Thomson

Several extrinsic signals such as LIF, BMP and Wnt can support the self-renewal and pluripotency of embryonic stem (ES) cells through regulating the “pluripotent genes.” A unique homeobox transcription factor, Nanog, is one of the key downstream effectors of these signals. Elevated level of Nanog can maintain the mouse ES cell self-renewal independent of LIF and enable human ES cell growth without feeder cells. In addition to the external signal pathways, intrinsic transcription factors such as FoxD3, P53 and Oct4 are also involved in regulating the expression of Nanog. Functionally, Nanog works together with other key pluripotent factors such as Oct4 and Sox2 to control a set of target genes that have important functions in ES cell pluripotency. These key factors form a regulatory network to support or limit each others expression level, which maintains the properties of ES cells.


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.


Cell Stem Cell | 2011

The Histone Demethylases Jhdm1a/1b Enhance Somatic Cell Reprogramming in a Vitamin-C-Dependent Manner

Tao Wang; Keshi Chen; Xiaoming Zeng; Jianguo Yang; Yun Wu; Xi Shi; Baoming Qin; Lingwen Zeng; Miguel A. Esteban; Guangjin Pan; Duanqing Pei

Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) resets the epigenome to an embryonic-like state. Vitamin C enhances the reprogramming process, but the underlying mechanisms are unclear. Here we show that the histone demethylases Jhdm1a/1b are key effectors of somatic cell reprogramming downstream of vitamin C. We first observed that vitamin C induces H3K36me2/3 demethylation in mouse embryonic fibroblasts in culture and during reprogramming. We then identified Jhdm1a/1b, two known vitamin-C-dependent H3K36 demethylases, as potent regulators of reprogramming through gain- and loss-of-function approaches. Furthermore, we found that Jhdm1b accelerates cell cycle progression and suppresses cell senescence during reprogramming by repressing the Ink4/Arf locus. Jhdm1b also cooperates with Oct4 to activate the microRNA cluster 302/367, an integral component of the pluripotency machinery. Our results therefore reveal a role for H3K36me2/3 in cell fate determination and establish a link between histone demethylases and vitamin-C-induced reprogramming.


Cell Research | 2002

Stem cell pluripotency and transcription factor Oct4

Guangjin Pan; Zengyi Chang; Hans R. Schöler; Duanqing Pei

ABSTRACTMammalian cell totipotency is a subject that has fascinated scientists for generations. A long lasting question whether some of the somatic cells retains totipotency was answered by the cloning of Dolly at the end of the 20th century. The dawn of the 21st has brought forward great expectations in harnessing the power of totipotentcy in medicine. Through stem cell biology, it is possible to generate any parts of the human body by stem cell engineering. Considerable resources will be devoted to harness the untapped potentials of stem cells in the foreseeable future which may transform medicine as we know today. At the molecular level, totipotency has been linked to a singular transcription factor and its expression appears to define whether a cell should be totipotent. Named Oct4, it can activate or repress the expression of various genes. Curiously, very little is known about Oct4 beyond its ability to regulate gene expression. The mechanism by which Oct4 specifies totipotency remains entirely unresolved. In this review, we summarize the structure and function of Oct4 and address issues related to Oct4 function in maintaining totipotency or pluripotency of embryonic stem cells.


The FASEB Journal | 2006

A negative feedback loop of transcription factors that controls stem cell pluripotency and self-renewal

Guangjin Pan; Jun Li; Yali Zhou; Hui Zheng; Duanqing Pei

Embryonic stem (ES) cells possess the ability to renew themselves while maintaining the capacity to differentiate into virtually all cell types of the body. Current evidence suggests that ES cells maintain their pluripotent state by expressing a battery of transcription factors including Oct4 and Nanog. However, little is known about how ES cells maintain the expression of these pluripotent factors in ES cells. Here we present evidence that Oct4, Nanog, and FoxD3 form a negative feedback loop to maintain their expression in pluripotent ES cells. First, Oct4 maintains Nanog activity by directly activating its promoter at sub‐steady‐state concentration but repressing it at or above steady‐state levels. On the other hand, FoxD3 behaves as a positive activator of Nanog to counter the repressive effect of Oct4. The expression of Oct4 is activated by FoxD3 and Nanog but repressed by Oct4 itself, thus, exerting an important negative feedback loop to limit its own activity. Indeed, overexpression of either FoxD3 or Nanog in ES cells failed to increase the concentration of Oct4 beyond the steady‐state concentration, whereas knocking down either FoxD3 or Nanog reduces the expression of Oct4 in ES cells. Finally, overexpression of Oct4 or Nanog failed to compensate the loss of Nanog or Oct4, respectively, suggesting that both are required for ES self‐renewal and pluripotency. Our results suggest the FoxD3‐Nanog‐Oct4 loop anchors an interdependent network of transcription factors that regulate stem cell pluripotency.—Pan, G., Li, J., Zhou, Y., Zheng, H., Pei, D. A negative feedback loop of transcription factors that controls stem cell pluripotency and self‐renewal. FASEB J. 20, E1094–E1102 (2006)


Cell Stem Cell | 2011

FGF2 sustains NANOG and switches the outcome of BMP4-induced human embryonic stem cell differentiation.

Pengzhi Yu; Guangjin Pan; Junying Yu; James A. Thomson

Here, we show that as human embryonic stem cells (ESCs) exit the pluripotent state, NANOG can play a key role in determining lineage outcome. It has previously been reported that BMPs induce differentiation of human ESCs into extraembryonic lineages. Here, we find that FGF2, acting through the MEK-ERK pathway, switches BMP4-induced human ESC differentiation outcome to mesendoderm, characterized by the uniform expression of T (brachyury) and other primitive streak markers. We also find that MEK-ERK signaling prolongs NANOG expression during BMP-induced differentiation, that forced NANOG expression results in FGF-independent BMP4 induction of mesendoderm, and that knockdown of NANOG greatly reduces T induction. Together, our results demonstrate that FGF2 signaling switches the outcome of BMP4-induced differentiation of human ESCs by maintaining NANOG levels through the MEK-ERK pathway.


Journal of Biological Chemistry | 2006

Regulation of the pluripotency marker Rex-1 by Nanog and Sox2.

Wenjing Shi; Hui Wang; Guangjin Pan; Yijie Geng; Yunqian Guo; Duanqing Pei

Rex-1 (Zfp-42) is a known marker for undifferentiated embryonic stem cells and teratocarcinoma cells. However, the mechanism by which Rex-1 is regulated in pluripotent cells remains unresolved. Here we report that Nanog, an Nk-2 homeodomain protein known for its role in maintaining stem cell pluripotency, is a transcription activator for the Rex-1 promoter. Knockdown of Nanog in embryonic stem cells resulted in a reduction of Rex-1 expression, whereas forced expression of Nanog in P19 stimulated Rex-1 expression. Employing a Rex-1 reporter, we demonstrate that Nanog transactivates Rex-1 directly. Serial deletion studies mapped the Nanog-responsive element between –187 and –286 of the Rex-1 promoter. Although Oct-3/4 and Sox2 can both transactivate Rex-1 promoter, only Sox2 cooperates with Nanog in up-regulating Rex-1. Furthermore, we demonstrate that the C terminus of Nanog is responsible for transactivating the Rex-1 promoter, a function that can be substituted for by a viral transactivator Vp16 efficiently in NIH3T3 cells but less so in P19 cells. Taking these findings together, we conclude that Rex-1 is a direct target of Nanog, which is augmented by Sox2 and Oct-3/4.


Nature Genetics | 2013

Vitamin C modulates TET1 function during somatic cell reprogramming

Jiekai Chen; Lin Guo; Lei Zhang; Haoyu Wu; Jiaqi Yang; He Liu; Xiaoshan Wang; Xiao Hu; Tian-Peng Gu; Zhiwei Zhou; Jing Liu; Jiadong Liu; Hongling Wu; Shi-Qing Mao; Kunlun Mo; Yingying Li; Keyu Lai; Jing Qi; Hongjie Yao; Guangjin Pan; Guoliang Xu; Duanqing Pei

Vitamin C, a micronutrient known for its anti-scurvy activity in humans, promotes the generation of induced pluripotent stem cells (iPSCs) through the activity of histone demethylating dioxygenases. TET hydroxylases are also dioxygenases implicated in active DNA demethylation. Here we report that TET1 either positively or negatively regulates somatic cell reprogramming depending on the absence or presence of vitamin C. TET1 deficiency enhances reprogramming, and its overexpression impairs reprogramming in the context of vitamin C by modulating the obligatory mesenchymal-to-epithelial transition (MET). In the absence of vitamin C, TET1 promotes somatic cell reprogramming independent of MET. Consistently, TET1 regulates 5-hydroxymethylcytosine (5hmC) formation at loci critical for MET in a vitamin C–dependent fashion. Our findings suggest that vitamin C has a vital role in determining the biological outcome of TET1 function at the cellular level. Given its benefit to human health, vitamin C should be investigated further for its role in epigenetic regulation.

Collaboration


Dive into the Guangjin Pan's collaboration.

Top Co-Authors

Avatar

Duanqing Pei

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Jiekai Chen

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Baojian Liao

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Ke Huang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Xiaoshan Wang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Jinyong Wang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Yongli Shan

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Lihui Wang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Wenhao Huang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Xiujuan Cai

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge