Network


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

Hotspot


Dive into the research topics where Junjiu Huang is active.

Publication


Featured researches published by Junjiu Huang.


Protein & Cell | 2015

CRISPR/Cas9-mediated gene editing in human tripronuclear zygotes

Puping Liang; Yanwen Xu; Xiya Zhang; Chenhui Ding; R. Huang; Zhen Zhang; Jie Lv; Xiaowei Xie; Yuxi Chen; Yujing Li; Ying Sun; Yaofu Bai; Zhou Songyang; Wenbin Ma; Canquan Zhou; Junjiu Huang

ABSTRACTGenome editing tools such as the clustered regularly interspaced short palindromic repeat (CRISPR)-associated system (Cas) have been widely used to modify genes in model systems including animal zygotes and human cells, and hold tremendous promise for both basic research and clinical applications. To date, a serious knowledge gap remains in our understanding of DNA repair mechanisms in human early embryos, and in the efficiency and potential off-target effects of using technologies such as CRISPR/Cas9 in human pre-implantation embryos. In this report, we used tripronuclear (3PN) zygotes to further investigate CRISPR/Cas9-mediated gene editing in human cells. We found that CRISPR/Cas9 could effectively cleave the endogenous β-globin gene (HBB). However, the efficiency of homologous recombination directed repair (HDR) of HBB was low and the edited embryos were mosaic. Off-target cleavage was also apparent in these 3PN zygotes as revealed by the T7E1 assay and whole-exome sequencing. Furthermore, the endogenous delta-globin gene (HBD), which is homologous to HBB, competed with exogenous donor oligos to act as the repair template, leading to untoward mutations. Our data also indicated that repair of the HBB locus in these embryos occurred preferentially through the non-crossover HDR pathway. Taken together, our work highlights the pressing need to further improve the fidelity and specificity of the CRISPR/Cas9 platform, a prerequisite for any clinical applications of CRSIPR/Cas9-mediated editing.


Cell Stem Cell | 2015

The Daxx/Atrx Complex Protects Tandem Repetitive Elements during DNA Hypomethylation by Promoting H3K9 Trimethylation.

Quanyuan He; Hyeung Kim; R. Huang; Weisi Lu; Mengfan Tang; Fengtao Shi; Dong Yang; Xiya Zhang; Junjiu Huang; Dan Liu; Zhou Songyang

In mammals, DNA methylation is essential for protecting repetitive sequences from aberrant transcription and recombination. In some developmental contexts (e.g., preimplantation embryos) DNA is hypomethylated but repetitive elements are not dysregulated, suggesting that alternative protection mechanisms exist. Here we explore the processes involved by investigating the role of the chromatin factors Daxx and Atrx. Using genome-wide binding and transcriptome analysis, we found that Daxx and Atrx have distinct chromatin-binding profiles and are co-enriched at tandem repetitive elements in wild-type mouse ESCs. Global DNA hypomethylation further promoted recruitment of the Daxx/Atrx complex to tandem repeat sequences, including retrotransposons and telomeres. Knockdown of Daxx/Atrx in cells with hypomethylated genomes exacerbated aberrant transcriptional de-repression of repeat elements and telomere dysfunction. Mechanistically, Daxx/Atrx-mediated repression seems to involve Suv39h recruitment and H3K9 trimethylation. Our data therefore suggest that Daxx and Atrx safeguard the genome by silencing repetitive elements when DNA methylation levels are low.


Protein & Cell | 2017

Correction of β-thalassemia mutant by base editor in human embryos

Puping Liang; Chenhui Ding; Hongwei Sun; Xiaowei Xie; Yanwen Xu; Xiya Zhang; Ying Sun; Yuanyan Xiong; Wenbin Ma; Yongxiang Liu; Yali Wang; Jianpei Fang; Dan Liu; Zhou Songyang; Canquan Zhou; Junjiu Huang

Abstractβ-Thalassemia is a global health issue, caused by mutations in the HBB gene. Among these mutations, HBB −28 (A>G) mutations is one of the three most common mutations in China and Southeast Asia patients with β-thalassemia. Correcting this mutation in human embryos may prevent the disease being passed onto future generations and cure anemia. Here we report the first study using base editor (BE) system to correct disease mutant in human embryos. Firstly, we produced a 293T cell line with an exogenous HBB −28 (A>G) mutant fragment for gRNAs and targeting efficiency evaluation. Then we collected primary skin fibroblast cells from a β-thalassemia patient with HBB −28 (A>G) homozygous mutation. Data showed that base editor could precisely correct HBB −28 (A>G) mutation in the patient’s primary cells. To model homozygous mutation disease embryos, we constructed nuclear transfer embryos by fusing the lymphocyte or skin fibroblast cells with enucleated in vitro matured (IVM) oocytes. Notably, the gene correction efficiency was over 23.0% in these embryos by base editor. Although these embryos were still mosaic, the percentage of repaired blastomeres was over 20.0%. In addition, we found that base editor variants, with narrowed deamination window, could promote G-to-A conversion at HBB −28 site precisely in human embryos. Collectively, this study demonstrated the feasibility of curing genetic disease in human somatic cells and embryos by base editor system.


Protein & Cell | 2014

Telomere regulation in pluripotent stem cells

Yan Huang; Puping Liang; Dan Liu; Junjiu Huang; Zhou Songyang

Pluripotent stem cells (PSCs) have the potential to produce any types of cells from all three basic germ layers and the capacity to self-renew and proliferate indefinitely in vitro. The two main types of PSCs, embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), share common features such as colony morphology, high expression of Oct4 and Nanog, and strong alkaline phosphatase activity. In recent years, increasing evidences suggest that telomere length represents another important internal factor in maintaining stem cell pluripotency. Telomere length homeostasis and its structural integrity help to protect chromosome ends from recombination, end fusion, and DNA damage responses, ensuring the divisional ability of mammalian cells. PSCs generally exhibit high telomerase activity to maintain their extremely long and stable telomeres, and emerging data indicate the alternative lengthening of telomeres (ALT) pathway may play an important role in telomere functions too. Such characteristics are likely key to their abilities to differentiate into diverse cell types in vivo. In this review, we will focus on the function and regulation of telomeres in ESCs and iPSCs, thereby shedding light on the importance of telomere length to pluripotency and the mechanisms that regulate telomeres in PSCs.


Protein & Cell | 2017

Effective gene editing by high-fidelity base editor 2 in mouse zygotes

Puping Liang; Hongwei Sun; Ying Sun; Xiya Zhang; Xiaowei Xie; Jinran Zhang; Zhen Zhang; Yuxi Chen; Chenhui Ding; Yuanyan Xiong; Wenbin Ma; Dan Liu; Junjiu Huang; Zhou Songyang

ABSTRACTTargeted point mutagenesis through homologous recombination has been widely used in genetic studies and holds considerable promise for repairing disease-causing mutations in patients. However, problems such as mosaicism and low mutagenesis efficiency continue to pose challenges to clinical application of such approaches. Recently, a base editor (BE) system built on cytidine (C) deaminase and CRISPR/Cas9 technology was developed as an alternative method for targeted point mutagenesis in plant, yeast, and human cells. Base editors convert C in the deamination window to thymidine (T) efficiently, however, it remains unclear whether targeted base editing in mouse embryos is feasible. In this report, we generated a modified high-fidelity version of base editor 2 (HF2-BE2), and investigated its base editing efficacy in mouse embryos. We found that HF2-BE2 could convert C to T efficiently, with up to 100% biallelic mutation efficiency in mouse embryos. Unlike BE3, HF2-BE2 could convert C to T on both the target and non-target strand, expanding the editing scope of base editors. Surprisingly, we found HF2-BE2 could also deaminate C that was proximal to the gRNA-binding region. Taken together, our work demonstrates the feasibility of generating point mutations in mouse by base editing, and underscores the need to carefully optimize base editing systems in order to eliminate proximal-site deamination.


Aging Cell | 2015

Mir-23a induces telomere dysfunction and cellular senescence by inhibiting TRF2 expression.

Zhenhua Luo; Xuyang Feng; Haoli Wang; Weiyi Xu; Yong Zhao; Wenbin Ma; Songshan Jiang; Dan Liu; Junjiu Huang; Zhou Songyang

Telomeric repeat binding factor 2 (TRF2) is essential for telomere maintenance and has been implicated in DNA damage response and aging. Telomere dysfunction induced by TRF2 inhibition can accelerate cellular senescence in human fibroblasts. While previous work has demonstrated that a variety of factors can regulate TRF2 expression transcriptionally and post‐translationally, whether microRNAs (miRNAs) also participate in post‐transcriptionally modulating TRF2 levels remains largely unknown. To better understand the regulatory pathways that control TRF2, we carried out a large‐scale luciferase reporter screen using a miRNA expression library and identified four miRNAs that could target human TRF2 and significantly reduce the level of endogenous TRF2 proteins. In particular, our data revealed that miR‐23a could directly target the 3′ untranslated region (3′UTR) of TRF2. Overexpression of miR‐23a not only reduced telomere‐bound TRF2 and increased telomere dysfunction‐induced foci (TIFs), but also accelerated senescence of human fibroblast cells, which could be rescued by ectopically expressed TRF2. Our findings demonstrate that TRF2 is a specific target of miR‐23a, and uncover a previously unknown role for miR‐23a in telomere regulation and cellular senescence.


Protein & Cell | 2016

Questions about NgAgo

Shawn M. Burgess; Linzhao Cheng; Feng Gu; Junjiu Huang; Zhiwei Huang; Shuo Lin; Jinsong Li; Wei Li; Wei Qin; Yujie Sun; Zhou Songyang; Wensheng Wei; Qiang Wu; Haoyi Wang; Xiaoqun Wang; Jing Wei Xiong; Jianzhong Xi; Hui Yang; Bin Zhou; Bo Zhang

Gao et al. published data in Nature Biotechnology (Nat Biotechnol. 2016 May 2) showing that DNA-guided genome editing using the Natronobacterium gregoryi Argonaute (NgAgo) protein targeted 47 mammalian genomic loci with a 100% success rate and an efficiency of 21.3%–41.3% at various targets. This report led us to test NgAgo’s utility in various cells and organisms such as mouse and zebrafish for gene editing. In most cases, a codon-optimized NgAgo for vertebrate animals was first synthesized and tested with appropriate guide oligos targeting specific genes using techniques similar to what has been utilized for the CRISPR/ Cas9 system. After failing to confirm any NgAgo induced genomic DNA editing in any experiments, some of us switched to use an NgAgo expression vector (CMV-NLS-NgAgoSK) used and provided by Han, the senior author of this paper, available from Addgene (#78253) since June or directly from his lab. Again, no success editing endogenous genomic DNA was achieved. As controls, the ability of this construct to induce indels was tested, targeting the same genes in cultured human 293T cells as those reported in Fig. 4 of Gao et al. Several researchers in different laboratories independently performed the experiments but no indels were observed at targeted loci, as assayed by T7E1 digestion, PAGE and/or sequencing. Representative data that directly repeat Fig. 4 of Gao et al from eight laboratories are shown in Fig. 1 and protocols used are detailed in supplementary information. We also include additional results from testing NgAgo in various systems by laboratories of signees of this letter in supplementary information. None of these studies proves that NgAgo has any genome editing activities. Han issued public statements suggesting that the reported findings require “superb experimental skills” and one needs to be able to repeat the result of Fig. 3C, which is the inhibition of GFP expression in plasmid DNA transfected cells. Indeed, plasmid GFP expression reduction by cotransfection of NgAgo and its targeting DNA oligo is reproducible in our hands. However, we cannot demonstrate by sequencing this reduction is a result of DNA mutation. Many factors can affect this type of GFP expression, including NgAgo’s ability to target RNA as well as non-specific stress induced by oligo and DNA transfection. More recently, Han added that the activity of NgAgo is very sensitive to mycoplasma or bacteria in the culture. However, it seems unlikely that independent laboratories would all have their cells contaminated, resulting in consistently negative results for DNA editing activity. In fact, several of the signees of this letter have made sure that our cells are free of mycoplasma by first testing them before performing replication experiments. The key point of paper by Gao et al is that DNA-guided NgAgo’s can efficiently target 47 genomic loci with a 100% success rate and a ≥20% efficiency. Neither the originally published protocol nor the newly released information on Addgene’s website involves any steps that seem to require “superb experimental skills”. To gain insights into NgAgo’s utility, some of us have even sent visiting researchers to Han’s laboratory but they were not allowed to perform genome editing experiments involving mammalian cells when they were there. Consequently, none of them returned with any information confirming Han’s data. Discussions on NgAgo have been frenzied in online forums, which cited some of the informal discussions in support of Han’s experimental data. Han also quoted David Cyranoski’s report (Nature, 2016 August 09) as evidence that NgAgo’s genome editing function had been confirmed. This further creates confusion because information in online forums is not accessible by the broader scientific community. We therefore urge the authors of the original paper to clarify the uncertainty surrounding NgAgo and provide all the necessary details for replicating the initial, very important results.


Nucleic Acids Research | 2016

Cold-inducible RNA-binding protein CIRP/hnRNP A18 regulates telomerase activity in a temperature-dependent manner

Youwei Zhang; Yangxiu Wu; Pingsu Mao; Feng Li; Xin Han; Yi Zhang; Shuai Jiang; Yuxi Chen; Junjiu Huang; Dan Liu; Yong Zhao; Wenbin Ma; Zhou Songyang

The telomerase is responsible for adding telomeric repeats to chromosomal ends and consists of the reverse transcriptase TERT and the RNA subunit TERC. The expression and activity of the telomerase are tightly regulated, and aberrant activation of the telomerase has been observed in >85% of human cancers. To better understand telomerase regulation, we performed immunoprecipitations coupled with mass spectrometry (IP-MS) and identified cold inducible RNA-binding protein (CIRP or hnRNP A18) as a telomerase-interacting factor. We have found that CIRP is necessary to maintain telomerase activities at both 32°C and 37°C. Furthermore, inhibition of CIRP by CRISPR-Cas9 or siRNA knockdown led to reduced telomerase activities and shortened telomere length, suggesting an important role of CIRP in telomere maintenance. We also provide evidence here that CIRP associates with the active telomerase complex through direct binding of TERC and regulates Cajal body localization of the telomerase. In addition, CIRP regulates the level of TERT mRNAs. At the lower temperature, TERT mRNA is upregulated in a CIRP-dependent manner to compensate for reduced telomerase activities. Taken together, these findings highlight the dual roles that CIRP plays in regulating TERT and TERC, and reveal a new class of telomerase modulators in response to hypothermia conditions.


Nucleic Acids Research | 2016

Preferential extension of short telomeres induced by low extracellular pH

Yuanlong Ge; Shu Wu; Yong Xue; Jun Tao; Feng Li; Yanlian Chen; Haiying Liu; Wenbin Ma; Junjiu Huang; Yong Zhao

The majority of tumor cells overcome proliferative limit by expressing telomerase. Whether or not telomerase preferentially extends the shortest telomeres is still under debate. When human cancer cells are cultured at neutral pH, telomerase extends telomeres in telomere length-independent manner. However, the microenvironment of tumor is slightly acidic, and it is not yet known how this influences telomerase action. Here, we examine telomere length homeostasis in tumor cells cultured at pHe 6.8. The results indicate that telomerase preferentially extends short telomeres, such that telomere length distribution narrows and telomeres become nearly uniform in size. After growth at pHe 6.8, the expression of telomerase, TRF1, TRF2 and TIN2 decreases, and the abundance of Cajal bodies decreases. Therefore, telomerase are insufficient for extending every telomere and shorter telomeres bearing less shelterin proteins are more accessible for telomerase recruitment. The findings support the ‘protein-counting mechanism’ in which extended and unextended state of telomere is determined by the number of associated shelterin proteins and the abundance of telomerase. Decreased expression of telomerase and preferential extension of short telomeres have important implications for tumor cell viability, and generate a strong rationale for research on telomerase-targeted anti-cancer therapeutics.


PLOS ONE | 2015

CRISPR/Cas9 Promotes Functional Study of Testis Specific X-Linked Gene In Vivo.

Minyan Li; R. Huang; Xue Jiang; Yuxi Chen; Zhen Zhang; Xiya Zhang; Puping Liang; Shaoquan Zhan; Shanbo Cao; Zhou Songyang; Junjiu Huang

Mammalian spermatogenesis is a highly regulated multistage process of sperm generation. It is hard to uncover the real function of a testis specific gene in vitro since the in vitro model is not yet mature. With the development of the CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated 9) system, we can now rapidly generate knockout mouse models of testis specific genes to study the process of spermatogenesis in vivo. SYCP3-like X-linked 2 (SLX2) is a germ cell specific component, which contains a Cor1 domain and belongs to the XLR (X-linked, lymphocyte regulated) family. Previous studies suggested that SLX2 might play an important role in mouse spermatogenesis based on its subcellular localization and interacting proteins. However, the function of SLX2 in vivo is still elusive. Here, to investigate the functions of SLX2 in spermatogenesis, we disrupted the Slx2 gene by using the CRISPR/Cas9 system. Since Slx2 is a testis specific X-linked gene, we obtained knockout male mice in the first generation and accelerated the study process. Compared with wild-type mice, Slx2 knockout mice have normal testis and epididymis. Histological observation of testes sections showed that Slx2 knockout affected none of the three main stages of spermatogenesis: mitosis, meiosis and spermiogenesis. In addition, we further confirmed that disruption of Slx2 did not affect the number of spermatogonial stem cells, meiosis progression or XY body formation by immunofluorescence analysis. As spermatogenesis was normal in Slx2 knockout mice, these mice were fertile. Taken together, we showed that Slx2 itself is not an essential gene for mouse spermatogenesis and CRISPR/Cas9 technique could speed up the functional study of testis specific X-linked gene in vivo.

Collaboration


Dive into the Junjiu Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiya Zhang

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Wenbin Ma

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Dan Liu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yong Zhao

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Yuxi Chen

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Hongwei Sun

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar

Zhen Zhang

Sun Yat-sen University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge