Network


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

Hotspot


Dive into the research topics where Jiayin Yang is active.

Publication


Featured researches published by Jiayin Yang.


Cell Stem Cell | 2010

A Mesenchymal-to-Epithelial Transition Initiates and Is Required for the Nuclear Reprogramming of Mouse Fibroblasts

Ronghui Li; Jialiang Liang; Su Ni; Ting Zhou; Xiaobing Qing; Huapeng Li; Wenzhi He; Jiekai Chen; Feng Li; Qiang Zhuang; Baoming Qin; Jianyong Xu; Wen Li; Jiayin Yang; Yi Gan; Dajiang Qin; Shipeng Feng; Hong Song; Dongshan Yang; Biliang Zhang; Lingwen Zeng; Liangxue Lai; Miguel A. Esteban; Duanqing Pei

Epithelial-to-mesenchymal transition (EMT) is a developmental process important for cell fate determination. Fibroblasts, a product of EMT, can be reset into induced pluripotent stem cells (iPSCs) via exogenous transcription factors but the underlying mechanism is unclear. Here we show that the generation of iPSCs from mouse fibroblasts requires a mesenchymal-to-epithelial transition (MET) orchestrated by suppressing pro-EMT signals from the culture medium and activating an epithelial program inside the cells. At the transcriptional level, Sox2/Oct4 suppress the EMT mediator Snail, c-Myc downregulates TGF-beta1 and TGF-beta receptor 2, and Klf4 induces epithelial genes including E-cadherin. Blocking MET impairs the reprogramming of fibroblasts whereas preventing EMT in epithelial cells cultured with serum can produce iPSCs without Klf4 and c-Myc. Our work not only establishes MET as a key cellular mechanism toward induced pluripotency, but also demonstrates iPSC generation as a cooperative process between the defined factors and the extracellular milieu. PAPERCLIP:


Cell Stem Cell | 2010

Vitamin C Enhances the Generation of Mouse and Human Induced Pluripotent Stem Cells

Miguel A. Esteban; Tao Wang; Baoming Qin; Jiayin Yang; Dajiang Qin; Jinglei Cai; Wen Li; Zhihui Weng; Jiekai Chen; Su Ni; Keshi Chen; Yuan Li; Xiaopeng Liu; Jianyong Xu; Shiqiang Zhang; Feng Li; Wenzhi He; Krystyna Labuda; Yancheng Song; Anja Peterbauer; Susanne Wolbank; Heinz Redl; Mei Zhong; Daozhang Cai; Lingwen Zeng; Duanqing Pei

Somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by defined factors. However, the low efficiency and slow kinetics of the reprogramming process have hampered progress with this technology. Here we report that a natural compound, vitamin C (Vc), enhances iPSC generation from both mouse and human somatic cells. Vc acts at least in part by alleviating cell senescence, a recently identified roadblock for reprogramming. In addition, Vc accelerates gene expression changes and promotes the transition of pre-iPSC colonies to a fully reprogrammed state. Our results therefore highlight a straightforward method for improving the speed and efficiency of iPSC generation and provide additional insights into the mechanistic basis of the reprogramming process.


Journal of Biological Chemistry | 2009

Generation of induced pluripotent stem cell lines from tibetan miniature pig

Miguel A. Esteban; Jianyong Xu; Jiayin Yang; Meixiu Peng; Dajiang Qin; Wen Li; Zhuoxin Jiang; Jiekai Chen; Kang Deng; Mei Zhong; Jinglei Cai; Liangxue Lai; Duanqing Pei

Induced pluripotent stem cell (iPS) technology appears to be a general strategy to generate pluripotent stem cells from any given mammalian species. So far, iPS cells have been reported for mouse, human, rat, and monkey. These four species have also established embryonic stem cell (ESC) lines that serve as the gold standard for pluripotency comparisons. Attempts have been made to generate porcine ESC by various means without success. Here we report the successful generation of pluripotent stem cells from fibroblasts isolated from the Tibetan miniature pig using a modified iPS protocol. The resulting iPS cell lines more closely resemble human ESC than cells from other species, have normal karyotype, stain positive for alkaline phosphatase, express high levels of ESC-like markers (Nanog, Rex1, Lin28, and SSEA4), and can differentiate into teratomas composed of the three germ layers. Because porcine physiology closely resembles human, the iPS cells reported here provide an attractive model to study certain human diseases or assess therapeutic applications of iPS in a large animal model.


Nature Protocols | 2012

Generation of human induced pluripotent stem cells from urine samples

Ting Zhou; Christina Benda; Sarah Dunzinger; Yinghua Huang; Jenny Cy Ho; Jiayin Yang; Yu Wang; Ya Zhang; Qiang Zhuang; Yanhua Li; Xichen Bao; Hung-Fat Tse; Johannes Grillari; Regina Grillari-Voglauer; Duanqing Pei; Miguel A. Esteban

Human induced pluripotent stem cells (iPSCs) have been generated with varied efficiencies from multiple tissues. Yet, acquiring donor cells is, in most instances, an invasive procedure that requires laborious isolation. Here we present a detailed protocol for generating human iPSCs from exfoliated renal epithelial cells present in urine. This method is advantageous in many circumstances, as the isolation of urinary cells is simple (30 ml of urine are sufficient), cost-effective and universal (can be applied to any age, gender and race). Moreover, the entire procedure is reasonably quick—around 2 weeks for the urinary cell culture and 3–4 weeks for the reprogramming—and the yield of iPSC colonies is generally high—up to 4% using retroviral delivery of exogenous factors. Urinary iPSCs (UiPSCs) also show excellent differentiation potential, and thus represent a good choice for producing pluripotent cells from normal individuals or patients with genetic diseases, including those affecting the kidney.


Journal of Biological Chemistry | 2010

Induced Pluripotent Stem Cells Can Be Used to Model the Genomic Imprinting Disorder Prader-Willi Syndrome

Jiayin Yang; Jie Cai; Ya Zhang; Xianming Wang; Wen Li; Jianyong Xu; Feng Li; Xiangpeng Guo; Kang Deng; Mei Zhong; Yonglong Chen; Liangxue Lai; Duanqing Pei; Miguel A. Esteban

The recent discovery of induced pluripotent stem cell (iPSC) technology provides an invaluable tool for creating in vitro representations of human genetic conditions. This is particularly relevant for those diseases that lack adequate animal models or where the species comparison is difficult, e.g. imprinting diseases such as the neurogenetic disorder Prader-Willi syndrome (PWS). However, recent reports have unveiled transcriptional and functional differences between iPSCs and embryonic stem cells that in cases are attributable to imprinting errors. This has suggested that human iPSCs may not be useful to model genetic imprinting diseases. Here, we describe the generation of iPSCs from a patient with PWS bearing a partial translocation of the paternally expressed chromosome 15q11-q13 region to chromosome 4. The resulting iPSCs match all standard criteria of bona fide reprogramming and could be readily differentiated into tissues derived from the three germ layers, including neurons. Moreover, these iPSCs retain a high level of DNA methylation in the imprinting center of the maternal allele and show concomitant reduced expression of the disease-associated small nucleolar RNA HBII-85/SNORD116. These results indicate that iPSCs may be a useful tool to study PWS and perhaps other genetic imprinting diseases as well.


Journal of The American Society of Nephrology | 2010

VHL inactivation induces HEF1 and Aurora kinase A.

Jianyong Xu; Huapeng Li; Bo Wang; Yan Xu; Jiayin Yang; Xiaofei Zhang; Sarah K. Harten; Deepa Shukla; Patrick H. Maxwell; Duanqing Pei; Miguel A. Esteban

The ciliary hypothesis for cystic renal diseases postulates that most of these conditions result from abnormalities in the primary cilium, a microtubule-based structure that acts as a sensor for extracellular cues. Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene predisposes to renal cysts and clear cell renal cell carcinoma. VHL plays a critical role in the formation of primary cilia in kidney epithelium, but the underlying mechanisms are poorly understood. Here, we demonstrate that VHL inactivation induces HEF1/Cas-L/NEDD9 and Aurora kinase A via the stabilization of hypoxia-inducible factors 1 and 2. Aurora kinase A is a mitotic kinase commonly upregulated in cancer that causes regression of the primary cilium by promoting histone deacetylase-dependent tubulin depolymerization of the ciliary axoneme. HEF1/Cas-L/NEDD9 is a component of focal adhesions that has a prominent role in inducing metastasis and that colocalizes with Aurora kinase A at the centrosome, thereby enhancing the harmful effect of Aurora kinase A on the cilium. Suppression of this pathway improved the formation of primary cilia and reduced cell motility in VHL-defective renal cancer cells. Our results highlight the gatekeeper role of VHL in the kidney epithelium.


Human Molecular Genetics | 2012

Modeling abnormal early development with induced pluripotent stem cells from aneuploid syndromes

Wen Li; Xianming Wang; Wenxia Fan; Ping Zhao; Ys Chan; Shen Chen; Shiqiang Zhang; Xiangpeng Guo; Ya Zhang; Yanhua Li; Jinglei Cai; Dajiang Qin; Xingyan Li; Jiayin Yang; Tianran Peng; Daniela Zychlinski; Dirk Hoffmann; Ruosi Zhang; Kang Deng; Kwong-Man Ng; Björn Menten; Mei Zhong; Jiayan Wu; Zhiyuan Li; Yonglong Chen; Axel Schambach; Hung-Fat Tse; Duanqing Pei; Miguel A. Esteban

Many human diseases share a developmental origin that manifests during childhood or maturity. Aneuploid syndromes are caused by supernumerary or reduced number of chromosomes and represent an extreme example of developmental disease, as they have devastating consequences before and after birth. Investigating how alterations in gene dosage drive these conditions is relevant because it might help treat some clinical aspects. It may also provide explanations as to how quantitative differences in gene expression determine phenotypic diversity and disease susceptibility among natural populations. Here, we aimed to produce induced pluripotent stem cell (iPSC) lines that can be used to improve our understanding of aneuploid syndromes. We have generated iPSCs from monosomy X [Turner syndrome (TS)], trisomy 8 (Warkany syndrome 2), trisomy 13 (Patau syndrome) and partial trisomy 11;22 (Emanuel syndrome), using either skin fibroblasts from affected individuals or amniocytes from antenatal diagnostic tests. These cell lines stably maintain the karyotype of the donors and behave like embryonic stem cells in all tested assays. TS iPSCs were used for further studies including global gene expression analysis and tissue-specific directed differentiation. Multiple clones displayed lower levels of the pseudoautosomal genes ASMTL and PPP2R3B than the controls. Moreover, they could be transformed into neural-like, hepatocyte-like and heart-like cells, but displayed insufficient up-regulation of the pseudoautosomal placental gene CSF2RA during embryoid body formation. These data support that abnormal organogenesis and early lethality in TS are not caused by a tissue-specific differentiation blockade, but rather involves other abnormalities including impaired placentation.


Iubmb Life | 2010

Porcine induced pluripotent stem cells may bridge the gap between mouse and human iPS

Miguel A. Esteban; Meixiu Peng; Zhang Deli; Jie Cai; Jiayin Yang; Jianyong Xu; Liangxue Lai; Duanqing Pei

Recently, three independent laboratories reported the generation of induced pluripotent stem cells (iPSCs) from pig (Sus scrofa). This finding sums to the growing list of species (mouse, human, monkey, and rat, in this order) for which successful reprogramming using exogenous factors has been achieved, and multiple others are possibly forthcoming. But apart from demonstrating the universality of the network identified by Shinya Yamanaka, what makes the porcine model so special? On one side, pigs are an agricultural commodity and have an easy and affordable maintenance compared with nonhuman primates that normally need to be imported. On the other side, resemblance (for example, size of organs) of porcine and human physiology is striking and because pigs are a regular source of food the ethical concerns that still remain in monkeys are not applicable. Besides, the prolonged lifespan of pigs compared with other domestic species can allow exhaustive follow up of side effects after transplantation. Porcine iPSCs may thus fill the gap between the mouse model, which due to its ease is preferred for mechanistic studies, and the first clinical trials using iPSCs in humans. However, although these studies are relevant and have created significant interest they face analogous problems that we discuss herein together with potential new directions.


Experimental Biology and Medicine | 2014

Solving the puzzle of Parkinson’s disease using induced pluripotent stem cells

Ping Zhao; Zhiwei Luo; Weihua Tian; Jiayin Yang; David P. Ibañez; Zhijian Huang; Micky D. Tortorella; Miguel A. Esteban; Wenxia Fan

The prevalence and incidence of Parkinson’s disease (PD) is increasing due to a prolonged life expectancy. This highlights the need for a better mechanistic understanding and new therapeutic approaches. However, traditional in vitro and in vivo experimental models to study PD are suboptimal, thus hampering the progress in the field. The epigenetic reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) offers a unique way to overcome this problem, as these cells share many properties of embryonic stem cells (ESCs) including the potential to be transformed into different lineages. PD modeling with iPSCs is nowadays facilitated by the growing availability of high-efficiency neural-specific differentiation protocols and the possibility to correct or induce mutations as well as creating marker cell lines using designer nucleases. These technologies, together with steady advances in human genetics, will likely introduce profound changes in the way we interpret PD and develop new treatments. Here, we summarize the different PD iPSCs reported so far and discuss the challenges for disease modeling using these cell lines.


Stem Cells | 2016

Generation of Induced Cardiospheres via Reprogramming of Skin Fibroblasts for Myocardial Regeneration

Jian Yong Xu; Yee Ki Lee; Xinru Ran; Song Yan Liao; Jiayin Yang; Ka Wing Au; Wing Hon Lai; Miguel A. Esteban; Hung-Fat Tse

Recent pre‐clinical and clinical studies have suggested that endogenous cardiospheres (eCS) are potentially safe and effective for cardiac regeneration following myocardial infarction (MI). Nevertheless the preparation of autologous eCS requires invasive myocardial biopsy with limited yield. We describe a novel approach to generate induced cardiospheres (iCS) from adult skin fibroblasts via somatic reprogramming. After infection with Sox2, Klf4, and Oct4, iCS were generated from mouse adult skin fibroblasts treated with Gsk3β inhibitor‐(2′Z,3′E)‐ 6‐Bromoindirubin‐3′‐oxime and Oncostatin M. They resembled eCS, but contained a higher percentage of cells expressing Mesp1, Isl1, and Nkx2.5. They were differentiated into functional cardiomyocytes in vitro with similar electrophysiological properties, calcium transient and contractile function to eCS and mouse embryonic stem cell‐derived cardiomyocytes. Transplantation of iCS (1 × 106 cells) into mouse myocardium following MI had similar effects to transplantation of eCS but significantly better than saline or fibroblast in improving left ventricular ejection fraction, increasing anterior/septal ventricular wall thickness and capillary density in the infarcted region 4 weeks after transplantation. No tumor formation was observed. iCS generated from adult skin fibroblasts by somatic reprogramming and a cocktail of Gsk3β inhibitor‐6‐Bromoindirubin‐3′‐oxime and Oncostatin M may represent a novel source for cell therapy in MI. Stem Cells 2016;34:2693–2706

Collaboration


Dive into the Jiayin Yang's collaboration.

Top Co-Authors

Avatar

Miguel A. Esteban

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Duanqing Pei

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Jianyong Xu

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Hung-Fat Tse

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Baoming Qin

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dajiang Qin

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Wen Li

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Zhiwei Luo

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

David P. Ibañez

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge