Stanford Advisors


All Publications


  • GenomeMUSter mouse genetic variation service enables multitrait, multipopulation data integration and analysis. Genome research Ball, R. L., Bogue, M. A., Liang, H., Srivastava, A., Ashbrook, D. G., Lamoureux, A., Gerring, M. W., Hatoum, A. S., Kim, M. J., He, H., Emerson, J., Berger, A. K., Walton, D. O., Sheppard, K., El Kassaby, B., Castellanos, F., Kunde-Ramamoorthy, G., Lu, L., Bluis, J., Desai, S., Sundberg, B. A., Peltz, G., Fang, Z., Churchill, G. A., Williams, R. W., Agrawal, A., Bult, C. J., Philip, V. M., Chesler, E. J. 2024

    Abstract

    Hundreds of inbred mouse strains and intercross populations have been used to characterize the function of genetic variants that contribute to disease. Thousands of disease-relevant traits have been characterized in mice and made publicly available. New strains and populations including consomics, the collaborative cross, expanded BXD, and inbred wild-derived strains add to existing complex disease mouse models, mapping populations, and sensitized backgrounds for engineered mutations. The genome sequences of inbred strains, along with dense genotypes from others, enable integrated analysis of trait-variant associations across populations, but these analyses are hampered by the sparsity of genotypes available. Moreover, the data are not readily interoperable with other resources. To address these limitations, we created a uniformly dense variant resource by harmonizing multiple data sets. Missing genotypes were imputed using the Viterbi algorithm with a data-driven technique that incorporates local phylogenetic information, an approach that is extendable to other model organisms. The result is a web- and programmatically accessible data service called GenomeMUSter, comprising single-nucleotide variants covering 657 strains at 106.8 million segregating sites. Interoperation with phenotype databases, analytic tools, and other resources enable a wealth of applications, including multitrait, multipopulation meta-analysis. We show this in cross-species comparisons of type 2 diabetes and substance use disorder meta-analyses, leveraging mouse data to characterize the likely role of human variant effects in disease. Other applications include refinement of mapped loci and prioritization of strain backgrounds for disease modeling to further unlock extant mouse diversity for genetic and genomic studies in health and disease.

    View details for DOI 10.1101/gr.278157.123

    View details for PubMedID 38290977

  • BCG vaccination stimulates integrated organ immunity by feedback of the adaptive immune response to imprint prolonged innate antiviral resistance. Nature immunology Lee, A., Floyd, K., Wu, S., Fang, Z., Tan, T. K., Froggatt, H. M., Powers, J. M., Leist, S. R., Gully, K. L., Hubbard, M. L., Li, C., Hui, H., Scoville, D., Ruggiero, A. D., Liang, Y., Pavenko, A., Lujan, V., Baric, R. S., Nolan, G. P., Arunachalam, P. S., Suthar, M. S., Pulendran, B. 2023

    Abstract

    Bacille Calmette-Guérin (BCG) vaccination can confer nonspecific protection against heterologous pathogens. However, the underlying mechanisms remain mysterious. We show that mice vaccinated intravenously with BCG exhibited reduced weight loss and/or improved viral clearance when challenged with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 B.1.351) or PR8 influenza. Protection was first evident between 14 and 21 d post-vaccination and lasted ∼3 months. Notably, BCG induced a biphasic innate response and robust antigen-specific type 1 helper T cell (TH1 cell) responses in the lungs. MyD88 signaling was essential for innate and TH1 cell responses, and protection against SARS-CoV-2. Depletion of CD4+ T cells or interferon (IFN)-γ activity before infection obliterated innate activation and protection. Single-cell and spatial transcriptomics revealed CD4-dependent expression of IFN-stimulated genes in lung myeloid and epithelial cells. Notably, BCG also induced protection against weight loss after mouse-adapted SARS-CoV-2 BA.5, SARS-CoV and SHC014 coronavirus infections. Thus, BCG elicits integrated organ immunity, where CD4+ T cells feed back on tissue myeloid and epithelial cells to imprint prolonged and broad innate antiviral resistance.

    View details for DOI 10.1038/s41590-023-01700-0

    View details for PubMedID 38036767

    View details for PubMedCentralID 3253344

  • Neuron Navigator 1 (Nav1) regulates the response to cocaine in mice. Communications biology Bagley, J. R., Tan, Y., Zhu, W., Cheng, Z., Takeda, S., Fang, Z., Arslan, A., Wang, M., Guan, Y., Jiang, L., Jian, R., Gu, F., Parada, I., Prince, D., Jentsch, J. D., Peltz, G. 2023; 6 (1): 1053

    Abstract

    Genetic variation accounts for much of the risk for developing a substance use disorder, but the underlying genetic factors and their genetic effector mechanisms are mostly unknown. Inbred mouse strains exhibit substantial and heritable differences in the extent of voluntary cocaine self-administration. Computational genetic analysis of cocaine self-administration data obtained from twenty-one inbred strains identified Nav1, a member of the neuron navigator family that regulates dendrite formation and axonal guidance, as a candidate gene. To test this genetic hypothesis, we generated and characterized Nav1 knockout mice. Consistent with the genetic prediction, Nav1 knockout mice exhibited increased voluntary cocaine intake and had increased motivation for cocaine consumption. Immunohistochemistry, electrophysiology, and transcriptomic studies were performed as a starting point for investigating the mechanism for the Nav1 knockout effect. Nav1 knockout mice had a reduced inhibitory synapse density in their cortex, increased excitatory synaptic transmission in their cortex and hippocampus, and increased excitatory neurons in a deep cortical layer. Collectively, our results indicate that Nav1 regulates the response to cocaine, and we identified Nav1 knockout induced changes in the excitatory and inhibitory synaptic balance in the cortex and hippocampus that could contribute to this effect.

    View details for DOI 10.1038/s42003-023-05430-9

    View details for PubMedID 37853211

    View details for PubMedCentralID PMC10584906

  • PRPF8 controls alternative splicing of PIRH2 to modulate the p53 pathway and survival of human ESCs. Journal of cellular physiology Sun, Y., Zhang, L., Fang, Z., Liu, D., Shao, M., Liu, Y., Liao, B., Jin, Y. 2023

    Abstract

    Human embryonic stem cells (hESCs) have great potential for developmental biology and regenerative medicine. However, extensive apoptosis often occurs when hESCs respond to various stresses or injuries. Understanding the molecular control and identifying new factors associated with hESC survival are fundamental to ensure the high quality of hESCs. In this study, we report that PRPF8, an RNA spliceosome component, is essential for hESC survival. PRPF8 knockdown (KD) induces p53 protein accumulation and activates the p53 pathway, leading to apoptosis in hESCs. Strikingly, silencing of p53 rescues PRPF8 KD-induced apoptosis, indicating that PRPF8 KD triggers hESC apoptosis through activating the p53 pathway. In search for the mechanism by which p53 pathway is activated by PRPF8 KD, we find that PRPF8 KD alters alternative splicing of many genes, including PIRH2 which encodes an E3 ubiquitin ligase of p53. PIRH2 has several isoforms such as PIRH2A, PIRH2B, and PIRH2C. Intriguingly, PRPF8 KD specifically increases the transcript level of the PIRH2B isoform, which lacks a RING domain and E3 ligase activity. Functionally, PIRH2B KD partially rescues the reduction in cell numbers and upregulation of P21 caused by PRPF8 KD in hESCs. The finding suggests that PRPF8 controls alternative splicing of PIRH2 to maintain the balance of p53 pathway activity and survival of hESCs. The PRPF8/PIRH2/p53 axis identified here provides new insights into how p53 pathway and hESC survival are precisely regulated at multiple layers, highlighting an important role of posttranscriptional machinery in supporting hESC survival.

    View details for DOI 10.1002/jcp.31066

    View details for PubMedID 37357506

  • Targeting KDM2A Enhances T Cell Infiltration in NSD1-Deficient Head and Neck Squamous Cell Carcinoma. Cancer research Chen, C., Shin, J. H., Fang, Z., Brennan, K., Horowitz, N. B., Pfaff, K. L., Welsh, E. L., Rodig, S. J., Gevaert, O., Gozani, O., Uppaluri, R., Sunwoo, J. B. 2023

    Abstract

    In head and neck squamous cell carcinoma (HNSCC), a significant proportion of tumors have inactivating mutations in the histone methyltransferase NSD1. In these tumors, NSD1 inactivation is a driver of T cell exclusion from the tumor microenvironment (TME). A better understanding of the NSD1-mediated mechanism regulating infiltration of T cells into the TME could help identify approaches to overcome immununosuppression. Here, we demonstrated that NSD1 inactivation results in lower levels of H3K36 di-methylation and higher levels of H3K27 tri-methylation, the latter being a known repressive histone mark enriched on the promoters of key T cell chemokines CXCL9 and CXCL10. HNSCC with NSD1 mutations had lower levels of these chemokines and lacked responses to PD-1 immune checkpoint blockade. Inhibition of KDM2A, the primary lysine demethylase that is selective for H3K36, reversed the altered histone marks induced by NSD1 loss and restored T cell infiltration into the TME. Importantly, KDM2A suppression decreased growth of NSD1-deficient tumors in immunocompetent, but not in immunodeficient, mice. Together, these data indicate that KDM2A is an immunotherapeutic target for overcoming immune exclusion in HNSCC.

    View details for DOI 10.1158/0008-5472.CAN-22-3114

    View details for PubMedID 37311054

  • Innate immune cell activation causes lung fibrosis in a humanized model of long COVID. Proceedings of the National Academy of Sciences of the United States of America Cui, L., Fang, Z., De Souza, C. M., Lerbs, T., Guan, Y., Li, I., Charu, V., Chen, S. Y., Weissman, I., Wernig, G. 2023; 120 (10): e2217199120

    Abstract

    COVID-19 remains a global pandemic of an unprecedented magnitude with millions of people now developing "COVID lung fibrosis." Single-cell transcriptomics of lungs of patients with long COVID revealed a unique immune signature demonstrating the upregulation of key proinflammatory and innate immune effector genes CD47, IL-6, and JUN. We modeled the transition to lung fibrosis after COVID and profiled the immune response with single-cell mass cytometry in JUN mice. These studies revealed that COVID mediated chronic immune activation reminiscent to long COVID in humans. It was characterized by increased CD47, IL-6, and phospho-JUN (pJUN) expression which correlated with disease severity and pathogenic fibroblast populations. When we subsequently treated a humanized COVID lung fibrosis model by combined blockade of inflammation and fibrosis, we not only ameliorated fibrosis but also restored innate immune equilibrium indicating possible implications for clinical management of COVID lung fibrosis in patients.

    View details for DOI 10.1073/pnas.2217199120

    View details for PubMedID 36848564

  • Analysis of structural variation among inbred mouse strains. BMC genomics Arslan, A., Fang, Z., Wang, M., Tan, Y., Cheng, Z., Chen, X., Guan, Y., J Pisani, L., Yoo, B., Bejerano, G., Peltz, G. 2023; 24 (1): 97

    Abstract

    BACKGROUND: 'Long read' sequencing methods have been used to identify previously uncharacterized structural variants that cause human genetic diseases. Therefore, we investigated whether long read sequencing could facilitate genetic analysis of murine models for human diseases.RESULTS: The genomes of six inbred strains (BTBR T+Itpr3tf/J, 129Sv1/J, C57BL/6/J, Balb/c/J, A/J, SJL/J) were analyzed using long read sequencing. Our results revealed that (i) Structural variants are very abundant within the genome of inbred strains (4.8 per gene) and (ii) that we cannot accurately infer whether structural variants are present using conventional short read genomic sequence data, even when nearby SNP alleles are known. The advantage of having a more complete map was demonstrated by analyzing the genomic sequence of BTBR mice. Based upon this analysis, knockin mice were generated and used to characterize a BTBR-unique 8-bp deletion within Draxin that contributes to the BTBR neuroanatomic abnormalities, which resemble human autism spectrum disorder.CONCLUSION: A more complete map of the pattern of genetic variation among inbred strains, which is produced by long read genomic sequencing of the genomes of additional inbred strains, could facilitate genetic discovery when murine models of human diseases are analyzed.

    View details for DOI 10.1186/s12864-023-09197-5

    View details for PubMedID 36864393

  • GSEApy: a comprehensive package for performing gene set enrichment analysis in Python. Bioinformatics (Oxford, England) Fang, Z., Liu, X., Peltz, G. 2022

    Abstract

    Gene Set enrichment analysis (GSEA) is a commonly used algorithm for characterizing gene expression changes. However, the currently available tools used to perform GSEA have a limited ability to analyze large datasets, which is particularly problematic for the analysis of single-cell data. To overcome this limitation, we developed a GSEA package in Python (GSEApy), which could efficiently analyze large single-cell datasets.We present a package (GSEApy) that performs GSEA in either the command line or Python environment. GSEApy uses a Rust implementation to enable it to calculate the same enrichment statistic as GSEA for a collection of pathways. The Rust implementation of GSEApy is 3-fold faster than the Numpy version of GSEApy (v0.10.8) and uses >4-fold less memory. GSEApy also provides an interface between Python and Enrichr web services, as well as for BioMart. The Enrichr API enables GSEApy to perform over-representation analysis for an input gene list. Furthermore, GSEApy consists of several tools, each designed to facilitate a particular type of enrichment analysis.The new GSEApy with Rust extension is deposited in PyPI: https://pypi.org/project/gseapy/. The GSEApy source code is freely available at https://github.com/zqfang/GSEApy. Also, the documentation website is available at https://gseapy.rtfd.io/.is available online.

    View details for DOI 10.1093/bioinformatics/btac757

    View details for PubMedID 36426870

  • An Automated Multi-Modal Graph-Based Pipeline for Mouse Genetic Discovery. Bioinformatics (Oxford, England) Fang, Z., Peltz, G. 2022

    Abstract

    Our ability to identify causative genetic factors for mouse genetic models of human diseases and biomedical traits has been limited by the difficulties associated with identifying true causative factors, which are often obscured by the many false positive genetic associations produced by a GWAS.To accelerate the pace of genetic discovery, we developed a graph neural network (GNN)-based automated pipeline (GNNHap) that could rapidly analyze mouse genetic model data and identify high probability causal genetic factors for analyzed traits. After assessing the strength of allelic associations with the strain response pattern; this pipeline analyzes 29M published papers to assess candidate gene-phenotype relationships; and incorporates the information obtained from a protein-protein interaction network and protein sequence features into the analysis. The GNN model produces markedly improved results relative to that of a simple linear neural network. We demonstrate that GNNHap can identify novel causative genetic factors for murine models of diabetes/obesity and for cataract formation, which were validated by the phenotypes appearing in previously analyzed gene knockout mice. The diabetes/obesity results indicate how characterization of the underlying genetic architecture enables new therapies to be discovered and tested by applying 'precision medicine' principles to murine models.The GNNHap source code is freely available at https://github.com/zqfang/gnnhap, and the new version of the HBCGM program is available at https://github.com/zqfang/haplomap.Supplementary information is available online.

    View details for DOI 10.1093/bioinformatics/btac356

    View details for PubMedID 35608290

  • A human multi-lineage hepatic organoid model for liver fibrosis. Nature communications Guan, Y., Enejder, A., Wang, M., Fang, Z., Cui, L., Chen, S., Wang, J., Tan, Y., Wu, M., Chen, X., Johansson, P. K., Osman, I., Kunimoto, K., Russo, P., Heilshorn, S. C., Peltz, G. 2021; 12 (1): 6138

    Abstract

    To investigate the pathogenesis of a congenital form of hepatic fibrosis, human hepatic organoids were engineered to express the most common causative mutation for Autosomal Recessive Polycystic Kidney Disease (ARPKD). Here we show that these hepatic organoids develop the key features of ARPKD liver pathology (abnormal bile ducts and fibrosis) in only 21 days. The ARPKD mutation increases collagen abundance and thick collagen fiber production in hepatic organoids, which mirrors ARPKD liver tissue pathology. Transcriptomic and other analyses indicate that the ARPKD mutation generates cholangiocytes with increased TGFbeta pathway activation, which are actively involved stimulating myofibroblasts to form collagen fibers. There is also an expansion of collagen-producing myofibroblasts with markedly increased PDGFRB protein expression and an activated STAT3 signaling pathway. Moreover, the transcriptome of ARPKD organoid myofibroblasts resemble those present in commonly occurring forms of liver fibrosis. PDGFRB pathway involvement was confirmed by the anti-fibrotic effect observed when ARPKD organoids were treated with PDGFRB inhibitors. Besides providing insight into the pathogenesis of congenital (and possibly acquired) forms of liver fibrosis, ARPKD organoids could also be used to test the anti-fibrotic efficacy of potential anti-fibrotic therapies.

    View details for DOI 10.1038/s41467-021-26410-9

    View details for PubMedID 34686668

  • Calcineurin A gamma and NFATc3/SRPX2 axis contribute to human embryonic stem cell differentiation JOURNAL OF CELLULAR PHYSIOLOGY Chen, H., Zeng, Y., Shao, M., Zhao, H., Fang, Z., Gu, J., Liao, B., Jin, Y. 2021; 236 (8): 5698-5713

    Abstract

    Our understanding of signaling pathways regulating the cell fate of human embryonic stem cells (hESCs) is limited. Calcineurin-NFAT signaling is associated with a wide range of biological processes and diseases. However, its role in controlling hESC fate remains unclear. Here, we report that calcineurin A gamma and the NFATc3/SRPX2 axis control the expression of lineage and epithelial-mesenchymal transition (EMT) markers in hESCs. Knockdown of PPP3CC, the gene encoding calcineurin A gamma, or NFATC3, downregulates certain markers both at the self-renewal state and during differentiation of hESCs. Furthermore, NFATc3 interacts with c-JUN and regulates the expression of SRPX2, the gene encoding a secreted glycoprotein known as a ligand of uPAR. We show that SRPX2 is a downstream target of NFATc3. Both SRPX2 and uPAR participate in controlling expression of lineage and EMT markers. Importantly, SRPX2 knockdown diminishes the upregulation of multiple lineage and EMT markers induced by co-overexpression of NFATc3 and c-JUN in hESCs. Together, this study uncovers a previously unknown role of calcineurin A gamma and the NFATc3/SRPX2 axis in modulating the fate determination of hESCs.

    View details for DOI 10.1002/jcp.30255

    View details for Web of Science ID 000604253100001

    View details for PubMedID 33393109

  • The Effect of Population Structure on Murine Genome-Wide Association Studies. Frontiers in genetics Wang, M., Fang, Z., Yoo, B., Bejerano, G., Peltz, G. 2021; 12: 745361

    Abstract

    The ability to use genome-wide association studies (GWAS) for genetic discovery depends upon our ability to distinguish true causative from false positive association signals. Population structure (PS) has been shown to cause false positive signals in GWAS. PS correction is routinely used for analysis of human GWAS results, and it has been assumed that it also should be utilized for murine GWAS using inbred strains. Nevertheless, there are fundamental differences between murine and human GWAS, and the impact of PS on murine GWAS results has not been carefully investigated. To assess the impact of PS on murine GWAS, we examined 8223 datasets that characterized biomedical responses in panels of inbred mouse strains. Rather than treat PS as a confounding variable, we examined it as a response variable. Surprisingly, we found that PS had a minimal impact on datasets measuring responses in ≤20 strains; and had surprisingly little impact on most datasets characterizing 21 - 40 inbred strains. Moreover, we show that true positive association signals arising from haplotype blocks, SNPs or indels, which were experimentally demonstrated to be causative for trait differences, would be rejected if PS correction were applied to them. Our results indicate because of the special conditions created by GWAS (the use of inbred strains, small sample sizes) PS assessment results should be carefully evaluated in conjunction with other criteria, when murine GWAS results are evaluated.

    View details for DOI 10.3389/fgene.2021.745361

    View details for PubMedID 34589118

  • SOX1 Is Required for the Specification of Rostral Hindbrain Neural Progenitor Cells from Human Embryonic Stem Cells ISCIENCE Liu, X., Fang, Z., Wen, J., Tang, F., Liao, B., Jing, N., Lai, D., Jin, Y. 2020; 23 (9): 101475

    Abstract

    Region-specific neural progenitor cells (NPCs) can be generated from human embryonic stem cells (hESCs) by modulating signaling pathways. However, how intrinsic transcriptional factors contribute to the neural regionalization is not well characterized. Here, we generate region-specific NPCs from hESCs and find that SOX1 is highly expressed in NPCs with the rostral hindbrain identity. Moreover, we find that OTX2 inhibits SOX1 expression, displaying exclusive expression between the two factors. Furthermore, SOX1 knockout (KO) leads to the upregulation of midbrain genes and downregulation of rostral hindbrain genes, indicating that SOX1 is required for specification of rostral hindbrain NPCs. Our SOX1 chromatin immunoprecipitation sequencing analysis reveals that SOX1 binds to the distal region of GBX2 to activate its expression. Overexpression of GBX2 largely abrogates SOX1-KO-induced aberrant gene expression. Taken together, this study uncovers previously unappreciated role of SOX1 in early neural regionalization and provides new information for the precise control of the OTX2/GBX2 interface.

    View details for DOI 10.1016/j.isci.2020.101475

    View details for Web of Science ID 000577096400002

    View details for PubMedID 32905879

    View details for PubMedCentralID PMC7486433

  • SOX21 Ensures Rostral Forebrain Identity by Suppression of WNT8B during Neural Regionalization of Human Embryonic Stem Cells STEM CELL REPORTS Fang, Z., Liu, X., Wen, J., Tang, F., Zhou, Y., Jing, N., Jin, Y. 2019; 13 (6): 1038-1052

    Abstract

    The generation of brain region-specific progenitors from human embryonic stem cells (hESCs) is critical for their application. However, transcriptional regulation of neural regionalization in humans is poorly understood. Here, we applied a rostrocaudal patterning system from hESCs to dissect global transcriptional networks controlling early neural regionalization. We found that SOX21 is required for rostral forebrain fate specification. SOX21 knockout led to activation of Wnt signaling, resulting in caudalization of regional identity of rostral forebrain neural progenitor cells. Moreover, we identified WNT8B as a SOX21 direct target. Deletion of WNT8B or inhibition of Wnt signaling in SOX21 knockout neural progenitor cells restored rostral forebrain identity. Furthermore, SOX21 interacted with β-catenin, interfering with the binding of TCF4/β-catenin complex to the WNT8B enhancer. Collectively, these results unveil the unknown role of SOX21 and shed light on how a transcriptional factor modulates early neural regionalization through crosstalk with a key component of Wnt signaling.

    View details for DOI 10.1016/j.stemcr.2019.10.013

    View details for Web of Science ID 000502098700008

    View details for PubMedID 31761677

    View details for PubMedCentralID PMC6915843

  • CDK11 safeguards the identity of human embryonic stem cells via fine-tuning signaling pathways JOURNAL OF CELLULAR PHYSIOLOGY Ding, J., Fang, Z., Liu, X., Zhu, Z., Wen, C., Wang, H., Gu, J., Li, Q., Zeng, R., Li, H., Jin, Y. 2020; 235 (5): 4279-4290

    Abstract

    Signaling pathways transmit extracellular cues into cells and regulate transcriptome and epigenome to maintain or change the cell identity. Protein kinases and phosphatases are critical for signaling transduction and regulation. Here, we report that CDK11, a member of the CDK family, is required for the maintenance of human embryonic stem cell (hESC) self-renewal. Our results show that, among the three main isoforms of CDK11, CDK11p46 is the main isoform safeguarding the hESC identity. Mechanistically, CDK11 constrains two important mitogen-activated protein kinase (MAPK) signaling pathways (JNK and p38 signaling) through modulating the activity of protein phosphatase 1. Furthermore, CDK11 knockdown activates transforming growth factor β (TGF-β)/SMAD2/3 signaling and upregulates certain nonneural differentiation-associated genes. Taken together, this study uncovers a kinase required for hESC self-renewal through fine-tuning MAPK and TGF-β signaling at appropriate levels. The kinase-phosphatase axis reported here may shed new light on the molecular mechanism sustaining the identity of hESCs.

    View details for DOI 10.1002/jcp.29305

    View details for Web of Science ID 000489904800001

    View details for PubMedID 31612516

  • Stk40 deletion elevates c-JUN protein level and impairs mesoderm differentiation JOURNAL OF BIOLOGICAL CHEMISTRY Hu, J., Li, S., Sun, X., Fang, Z., Wang, L., Xiao, F., Shao, M., Ge, L., Tang, F., Gu, J., Yu, H., Guo, Y., Guo, X., Liao, B., Jin, Y. 2019; 294 (25): 9959-9972
  • Transcription coactivator Cited1 acts as an inducer of trophoblast-like state from mouse embryonic stem cells through the activation of BMP signaling CELL DEATH & DISEASE Xu, Y., Luo, X., Fang, Z., Zheng, X., Zeng, Y., Zhu, C., Gu, J., Tang, F., Hu, Y., Hu, G., Jin, Y., Li, H. 2018; 9
  • Single-cell analysis reveals lineage segregation in early post-implantation mouse embryos JOURNAL OF BIOLOGICAL CHEMISTRY Wen, J., Zeng, Y., Fang, Z., Gu, J., Ge, L., Tang, F., Qu, Z., Hu, J., Cui, Y., Zhang, K., Wang, J., Li, S., Sun, Y., Jin, Y. 2017; 292 (23): 9840-9854
  • Deletion of Stk40 impairs definitive erythropoiesis in the mouse fetal liver CELL DEATH & DISEASE Wang, L., Yu, H., Cheng, H., He, K., Fang, Z., Ge, L., Cheng, T., Jin, Y. 2017; 8
  • Itch, an E3 ligase of Oct4, is required for embryonic stem cell self-renewal and pluripotency induction JOURNAL OF CELLULAR PHYSIOLOGY Liao, B., Zhong, X., Xu, H., Xiao, F., Fang, Z., Gu, J., Chen, Y., Zhao, Y., Jin, Y. 2013; 228 (7): 1443-1451

    View details for DOI 10.1002/jcp.24297

    View details for Web of Science ID 000316681100010