Network


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

Hotspot


Dive into the research topics where Han Qin is active.

Publication


Featured researches published by Han Qin.


Hepatology | 2007

Directed differentiation of human embryonic stem cells into functional hepatic cells.

Jun Cai; Yang Zhao; Yanxia Liu; Fei Ye; Zhihua Song; Han Qin; Sha Meng; Yuezhou Chen; R.-L. Zhou; Xijun Song; Yushan Guo; Mingxiao Ding; Hongkui Deng

The differentiation capacity of human embryonic stem cells (hESCs) holds great promise for therapeutic applications. We report a novel three‐stage method to efficiently direct the differentiation of human embryonic stem cells into hepatic cells in serum‐free medium. Human ESCs were first differentiated into definitive endoderm cells by 3 days of Activin A treatment. Next, the presence of fibroblast growth factor‐4 and bone morphogenetic protein‐2 in the culture medium for 5 days induced efficient hepatic differentiation from definitive endoderm cells. Approximately 70% of the cells expressed the hepatic marker albumin. After 10 days of further in vitro maturation, these cells expressed the adult liver cell markers tyrosine aminotransferase, tryptophan oxygenase 2, phosphoenolpyruvate carboxykinase (PEPCK), Cyp7A1, Cyp3A4 and Cyp2B6. Furthermore, these cells exhibited functions associated with mature hepatocytes including albumin secretion, glycogen storage, indocyanine green, and low‐density lipoprotein uptake, and inducible cytochrome P450 activity. When transplanted into CCl4 injured severe combined immunodeficiency mice, these cells integrated into the mouse liver and expressed human alpha‐1 antitrypsin for at least 2 months. In addition, we found that the hESC‐derived hepatic cells were readily infected by human immunodeficiency virus‐hepatitis C virus pseudotype viruses. Conclusion: We have developed an efficient way to direct the differentiation of human embryonic stem cells into cells that exhibit characteristics of mature hepatocytes. Our studies should facilitate searching the molecular mechanisms underlying human liver development, and form the basis for hepatocyte transplantation and drug tests. (HEPATOLOGY 2007;45:1229–1239.)


Nature Cell Biology | 2011

Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells

Yuki Ohi; Han Qin; Chibo Hong; Laure Blouin; Jose M. Polo; Tingxia Guo; Zhongxia Qi; Sara L. Downey; Philip D. Manos; Derrick J. Rossi; Jingwei Yu; Matthias Hebrok; Joseph F. Costello; Jun S. Song; Miguel Ramalho-Santos

Human induced pluripotent stem (iPS) cells are remarkably similar to embryonic stem (ES) cells, but recent reports indicate that there may be important differences between them. We carried out a systematic comparison of human iPS cells generated from hepatocytes (representative of endoderm), skin fibroblasts (mesoderm) and melanocytes (ectoderm). All low-passage iPS cells analysed retain a transcriptional memory of the original cells. The persistent expression of somatic genes can be partially explained by incomplete promoter DNA methylation. This epigenetic mechanism underlies a robust form of memory that can be found in iPS cells generated by multiple laboratories using different methods, including RNA transfection. Incompletely silenced genes tend to be isolated from other genes that are repressed during reprogramming, indicating that recruitment of the silencing machinery may be inefficient at isolated genes. Knockdown of the incompletely reprogrammed gene C9orf64 (chromosome 9 open reading frame 64) reduces the efficiency of human iPS cell generation, indicating that somatic memory genes may be functionally relevant during reprogramming.


Cell Research | 2009

Efficient generation of hepatocyte-like cells from human induced pluripotent stem cells

Zhihua Song; Jun Cai; Yanxia Liu; Dongxin Zhao; Jun Yong; Shuguang Duo; Xijun Song; Yushan Guo; Yang Zhao; Han Qin; Xiaolei Yin; Chen Wu; Jie Che; Shichun Lu; Mingxiao Ding; Hongkui Deng

Human induced pluripotent stem (iPS) cells are similar to embryonic stem (ES) cells, and can proliferate intensively and differentiate into a variety of cell types. However, the hepatic differentiation of human iPS cells has not yet been reported. In this report, human iPS cells were induced to differentiate into hepatic cells by a stepwise protocol. The expression of liver cell markers and liver-related functions of the human iPS cell-derived cells were monitored and compared with that of differentiated human ES cells and primary human hepatocytes. Approximately 60% of the differentiated human iPS cells at day 7 expressed hepatic markers alpha fetoprotein and Alb. The differentiated cells at day 21 exhibited liver cell functions including albumin Asecretion, glycogen synthesis, urea production and inducible cytochrome P450 activity. The expression of hepatic markers and liver-related functions of the iPS cell-derived hepatic cells were comparable to that of the human ES cell-derived hepatic cells. These results show that human iPS cells, which are similar to human ES cells, can be efficiently induced to differentiate into hepatocyte-like cells.


Journal of Biological Chemistry | 2007

Regulation of Apoptosis and Differentiation by p53 in Human Embryonic Stem Cells

Han Qin; Tianxin Yu; Tingting Qing; Yanxia Liu; Yang Zhao; Jun Cai; Jian Li; Zhihua Song; Xiuxia Qu; Peng Zhou; Jiong Wu; Mingxiao Ding; Hongkui Deng

The essentially infinite expansion potential and pluripotency of human embryonic stem cells (hESCs) makes them attractive for cell-based therapeutics. In contrast to mouse embryonic stem cells (mESCs), hESCs normally undergo high rates of spontaneous apoptosis and differentiation, making them difficult to maintain in culture. Here we demonstrate that p53 protein accumulates in apoptotic hESCs induced by agents that damage DNA. However, despite the accumulation of p53, it nevertheless fails to activate the transcription of its target genes. This inability of p53 to activate its target genes has not been observed in other cell types, including mESCs. We further demonstrate that p53 induces apoptosis of hESCs through a mitochondrial pathway. Reducing p53 expression in hESCs in turn reduces both DNA damage-induced apoptosis as well as spontaneous apoptosis. Reducing p53 expression also reduces spontaneous differentiation and slows the differentiation rate of hESCs. Our studies reveal the important roles of p53 as a critical mediator of human embryonic stem cells survival and differentiation.


Human Molecular Genetics | 2012

Transcriptional Analysis of Pluripotency Reveals the Hippo Pathway as a Barrier to Reprogramming

Han Qin; Kathryn Blaschke; Grace Wei; Yuki Ohi; Laure Blouin; Zhongxia Qi; Jingwei Yu; Ru-Fang Yeh; Matthias Hebrok; Miguel Ramalho-Santos

Pluripotent stem cells are derived from culture of early embryos or the germline and can be induced by reprogramming of somatic cells. Barriers to reprogramming that stabilize the differentiated state and have tumor suppression functions are expected to exist. However, we have a limited understanding of what such barriers might be. To find novel barriers to reprogramming to pluripotency, we compared the transcriptional profiles of the mouse germline with pluripotent and somatic cells, in vivo and in vitro. There is a remarkable global expression of the transcriptional program for pluripotency in primordial germ cells (PGCs). We identify parallels between PGC reprogramming to pluripotency and human germ cell tumorigenesis, including the loss of LATS2, a tumor suppressor kinase of the Hippo pathway. We show that knockdown of LATS2 increases the efficiency of induction of pluripotency in human cells. LATS2 RNAi, unlike p53 RNAi, specifically enhances the generation of fully reprogrammed iPS cells without accelerating cell proliferation. We further show that LATS2 represses reprogramming in human cells by post-transcriptionally antagonizing TAZ but not YAP, two downstream effectors of the Hippo pathway. These results reveal transcriptional parallels between germ cell transformation and the generation of iPS cells and indicate that the Hippo pathway constitutes a barrier to cellular reprogramming.


Cell | 2014

Systematic Identification of Barriers to Human iPSC Generation

Han Qin; Aaron Diaz; Laure Blouin; Robert Jan Lebbink; Weronika Patena; Priscilia Tanbun; Emily LeProust; Michael T. McManus; Jun S. Song; Miguel Ramalho-Santos

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) holds enormous promise for regenerative medicine. To elucidate endogenous barriers limiting this process, we systematically dissected human cellular reprogramming by combining a genome-wide RNAi screen, innovative computational methods, extensive single-hit validation, and mechanistic investigation of relevant pathways and networks. We identify reprogramming barriers, including genes involved in transcription, chromatin regulation, ubiquitination, dephosphorylation, vesicular transport, and cell adhesion. Specific a disintegrin and metalloproteinase (ADAM) proteins inhibit reprogramming, and the disintegrin domain of ADAM29 is necessary and sufficient for this function. Clathrin-mediated endocytosis can be targeted with small molecules and opposes reprogramming by positively regulating TGF-β signaling. Genetic interaction studies of endocytosis or ubiquitination reveal that barrier pathways can act in linear, parallel, or feedforward loop architectures to antagonize reprogramming. These results provide a global view of barriers to human cellular reprogramming.


Cell Reports | 2016

YAP Induces Human Naive Pluripotency

Han Qin; Miroslav Hejna; Yanxia Liu; Michelle Percharde; Mark Wossidlo; Laure Blouin; Jens Durruthy-Durruthy; Priscilla Wong; Zhongxia Qi; Jingwei Yu; Lei S. Qi; Vittorio Sebastiano; Jun S. Song; Miguel Ramalho-Santos

SUMMARY The human naive pluripotent stem cell (PSC) state, corresponding to a pre-implantation stage of development, has been difficult to capture and sustain in vitro. We report that the Hippo pathway effector YAP is nuclearly localized in the inner cell mass of human blastocysts. Overexpression of YAP in human embryonic stem cells (ESCs) and induced PSCs (iPSCs) promotes the generation of naive PSCs. Lysophosphatidic acid (LPA) can partially substitute for YAP to generate transgene-free human naive PSCs. YAP- or LPA-induced naive PSCs have a rapid clonal growth rate, a normal karyotype, the ability to form teratomas, transcriptional similarities to human pre-implantation embryos, reduced heterochromatin levels, and other hallmarks of the naive state. YAP/LPA act in part by suppressing differentiation-inducing effects of GSK3 inhibition. CRISPR/Cas9-generated YAP−/− cells have an impaired ability to form colonies in naive but not primed conditions. These results uncover an unexpected role for YAP in the human naive state, with implications for early human embryology.


Nucleic Acids Research | 2015

HiTSelect: a comprehensive tool for high-complexity-pooled screen analysis

Aaron Diaz; Han Qin; Miguel Ramalho-Santos; Jun S. Song

Genetic screens of an unprecedented scale have recently been made possible by the availability of high-complexity libraries of synthetic oligonucleotides designed to mediate either gene knockdown or gene knockout, coupled with next-generation sequencing. However, several sources of random noise and statistical biases complicate the interpretation of the resulting high-throughput data. We developed HiTSelect, a comprehensive analysis pipeline for rigorously selecting screen hits and identifying functionally relevant genes and pathways by addressing off-target effects, controlling for variance in both gene silencing efficiency and sequencing depth of coverage and integrating relevant metadata. We document the superior performance of HiTSelect using data from both genome-wide RNAi and CRISPR/Cas9 screens. HiTSelect is implemented as an open-source package, with a user-friendly interface for data visualization and pathway exploration. Binary executables are available at http://sourceforge.net/projects/hitselect/, and the source code is available at https://github.com/diazlab/HiTSelect.


Cell Stem Cell | 2008

Two Supporting Factors Greatly Improve the Efficiency of Human iPSC Generation

Yang Zhao; Xiaolei Yin; Han Qin; Fangfang Zhu; Haisong Liu; Weifeng Yang; Qiang Zhang; Chengang Xiang; Pingping Hou; Zhihua Song; Yanxia Liu; Jun Yong; Pengbo Zhang; Jun Cai; Meng Liu; Honggang Li; Yanqin Li; Xiuxia Qu; Kai Cui; Weiqi Zhang; Tingting Xiang; Yetao Wu; Yiding Zhao; Chun Liu; Chen Yu; Kehu Yuan; Jinning Lou; Mingxiao Ding; Hongkui Deng


Biochemical and Biophysical Research Communications | 2006

A novel chemical-defined medium with bFGF and N2B27 supplements supports undifferentiated growth in human embryonic stem cells.

Yanxia Liu; Zhihua Song; Yang Zhao; Han Qin; Jun Cai; Hong Zhang; Tianxin Yu; Siming Jiang; Guangwen Wang; Mingxiao Ding; Hongkui Deng

Collaboration


Dive into the Han Qin'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
Top Co-Authors

Avatar

Laure Blouin

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge