Network


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

Hotspot


Dive into the research topics where Mei Zhong is active.

Publication


Featured researches published by Mei Zhong.


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.


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.


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.


PLOS ONE | 2013

Low Immunogenicity of Neural Progenitor Cells Differentiated from Induced Pluripotent Stem Cells Derived from Less Immunogenic Somatic Cells

Pengfei Liu; Shubin Chen; Xiang Li; Li Qin; Ke Huang; Lihui Wang; Wenhao Huang; Shengbiao Li; Bei Jia; Mei Zhong; Guangjin Pan; Jinglei Cai; Duanqing Pei

The groundbreaking discovery of induced pluripotent stem cells (iPS cells) provides a new source for cell therapy. However, whether the iPS derived functional lineages from different cell origins have different immunogenicity remains unknown. It had been known that the cells isolated from extra-embryonic tissues, such as umbilical cord mesenchymal cells (UMCs), are less immunogenic than other adult lineages such as skin fibroblasts (SFs). In this report, we differentiated iPS cells from human UMCs and SFs into neural progenitor cells (NPCs) and analyzed their immunogenicity. Through co-culture with allologous peripheral blood mononuclear cells (PBMCs), we showed that UMCs were indeed less immunogenic than skin cells to simulate proliferation of PBMCs. Surprisingly, we found that the NPCs differentiated from UMC-iPS cells retained low immunogenicity as the parental UMCs based on the PBMC proliferation assay. In cytotoxic expression assay, reactions in most kinds of immune effector cells showed more perforin and granzyme B expression with SF-NPCs stimulation than that with UMC-NPCs stimulation in PBMC co-culture system, in T cell co-culture system as well. Furthermore, through whole genome expression microarray analysis, we showed that over 70 immune genes, including all members of HLA-I, were expressed at lower levels in NPCs derived from UMC-iPS cells than that from SF-iPS cells. Our results demonstrated a phenomenon that the low immunogenicity of the less immunogenic cells could be retained after cell reprogramming and further differentiation, thus provide a new concept to generate functional lineages with lower immunogenicity for regenerative medicine.


Life Sciences | 2014

Modeling of hemophilia A using patient-specific induced pluripotent stem cells derived from urine cells

Bei Jia; Shen Chen; Zhiju Zhao; Pengfei Liu; Jinglei Cai; Dajiang Qin; Juan Du; Changwei Wu; Qianyu Chen; Xiujuan Cai; Hui Zhang; Yanhong Yu; Duanqing Pei; Mei Zhong; Guangjin Pan

AIMS Hemophilia A (HA) is a severe, congenital bleeding disorder caused by the deficiency of clotting factor VIII (FVIII). For years, traditional laboratory animals have been used to study HA and its therapies, although animal models may not entirely mirror the human pathophysiology. Human induced pluripotent stem cells (iPSCs) can undergo unlimited self-renewal and differentiate into all cell types. This study aims to generate hemophilia A (HA) patient-specific iPSCs that differentiate into disease-affected hepatocyte cells. These hepatocytes are potentially useful for in vitro disease modeling and provide an applicable cell source for autologous cell therapy after genetic correction. MAIN METHODS In this study, we mainly generated iPSCs from urine collected from HA patients with integration-free episomal vectors PEP4-EO2S-ET2K containing human genes OCT4, SOX2, SV40LT and KLF4, and differentiated these iPSCs into hepatocyte-like cells. We further identified the genetic phenotype of the FVIII genes and the FVIII activity in the patient-specific iPSC derived hepatic cells. KEY FINDINGS HA patient-specific iPSCs (HA-iPSCs) exhibited typical pluripotent properties evident by immunostaining, in vitro assays and in vivo assays. Importantly, we showed that HA-iPSCs could differentiate into functional hepatocyte-like cells and the HA-iPSC-derived hepatocytes failed to produce FVIII, but otherwise functioned normally, recapitulating the phenotype of HA disease in vitro. SIGNIFICANCE HA-iPSCs, particular those generated from the urine using a non-viral approach, provide an efficient way for modeling HA in vitro. Furthermore, HA-iPSCs and their derivatives serve as an invaluable cell source that can be used for gene and cell therapy in regenerative medicine.


Journal of Hematology & Oncology | 2015

Quantitative evaluation of the immunodeficiency of a mouse strain by tumor engraftments

Wei Ye; Zhiwu Jiang; Guan Xiong Li; Yiren Xiao; Simiao Lin; Yunxin Lai; Suna Wang; Baiheng Li; Bei Jia; Yin Li; Zhi Liang Huang; Jin Li; Fenglan Feng; Shuhua Li; Huihui Yao; Zixia Liu; Su Cao; Lin Xu; Yangqiu Li; Donghai Wu; Lingwen Zeng; Mei Zhong; Pentao Liu; Zhe Sheng Wen; Bing Xu; Yao Yao; Duanqing Pei; Peng Li

BackgroundThe mouse is an organism that is widely used as a mammalian model for studying human physiology or disease, and the development of immunodeficient mice has provided a valuable tool for basic and applied human disease research. Following the development of large-scale mouse knockout programs and genome-editing tools, it has become increasingly efficient to generate genetically modified mouse strains with immunodeficiency. However, due to the lack of a standardized system for evaluating the immuno-capacity that prevents tumor progression in mice, an objective choice of the appropriate immunodeficient mouse strains to be used for tumor engrafting experiments is difficult.MethodsIn this study, we developed a tumor engraftment index (TEI) to quantify the immunodeficiency response to hematologic malignant cells and solid tumor cells of six immunodeficient mouse strains and C57BL/6 wild-type mouse (WT).ResultsMice with a more severely impaired immune system attained a higher TEI score. We then validated that the NOD-scid-IL2Rg−/− (NSI) mice, which had the highest TEI score, were more suitable for xenograft and allograft experiments using multiple functional assays.ConclusionsThe TEI score was effectively able to reflect the immunodeficiency of a mouse strain.


Haematologica | 2015

ANGPTL7 regulates the expansion and repopulation of human hematopoietic stem and progenitor cells

Yiren Xiao; Zhiwu Jiang; Yin Li; Wei Ye; Bei Jia; Minjie Zhang; Yan Xu; Donghai Wu; Liangxue Lai; Yaoyu Chen; Yingjun Chang; Xiaojun Huang; Hudan Liu; Guoliang Qing; Pentao Liu; Yangqiu Li; Bing Xu; Mei Zhong; Yao Yao; Duanqing Pei; Peng Li

Successful expansion of hematopoietic stem cells would benefit the use of hematopoietic stem cell transplants in the clinic. Several angiopoietin-like proteins, including angiopoietin-like 7, can support the activity of hematopoietic stem cells. However, effects of ANGPTL7 on human hematopoietic stem cells and the downstream signaling cascade activated by ANGPTL7 are poorly understood. Here, we established a human hematopoietic stem and progenitor cell-supportive mouse fetal liver cell line that specifically expressed the Angptl7 protein. Furthermore, we found ANGPTL7 is capable of stimulating human hematopoietic stem and progenitor cell expansion and increasing the repopulation activities of human hematopoietic progenitors in xenografts. RNA-sequencing analysis showed that ANGPTL7 activated the expression of CXCR4, HOXB4 and Wnt downstream targets in human hematopoietic progenitors. In addition, chemical manipulation of Wnt signaling diminished the effects of ANGPTL7 on human hematopoietic stem and progenitor cells in culture. In summary, we identify the secreted growth factor ANGPTL7 as a regulator of both human hematopoietic stem and progenitor cell expansion and regeneration.


Ultrasound in Obstetrics & Gynecology | 2015

Traditional karyotyping vs copy number variation sequencing for detection of chromosomal abnormalities associated with spontaneous miscarriage

S. Liu; Song Ll; D. S. Cram; Xiong L; K. Wang; R. Wu; J. Liu; K. Deng; B. Jia; Mei Zhong; F. Yang

To compare the performance of traditional G‐banding karyotyping with that of copy number variation sequencing (CNV‐Seq) for detection of chromosomal abnormalities associated with miscarriage.


Molecular Cancer | 2015

Genome-wide analyses identify KLF4 as an important negative regulator in T-cell acute lymphoblastic leukemia through directly inhibiting T-cell associated genes

Wei Li; Zhiwu Jiang; Tianzhong Li; Xinru Wei; Yi Zheng; Donghai Wu; Lijian Yang; Shaohua Chen; Bing Xu; Mei Zhong; Jue Jiang; Yufeng Hu; Hexiu Su; Minjie Zhang; Xiaojun Huang; Suxia Geng; Jianyu Weng; Xin Du; Pentao Liu; Yangqiu Li; Hudan Liu; Yao Yao; Peng Li

BackgroundKruppel-like factor 4 (KLF4) induces tumorigenesis or suppresses tumor growth in a tissue-dependent manner. However, the roles of KLF4 in hematological malignancies and the mechanisms of action are not fully understood.MethodsInducible KLF4-overexpression Jurkat cell line combined with mouse models bearing cell-derived xenografts and primary T-cell acute lymphoblastic leukemia (T-ALL) cells from four patients were used to assess the functional role of KLF4 in T-ALL cells in vitro and in vivo. A genome-wide RNA-seq analysis was conducted to identify genes regulated by KLF4 in T-ALL cells. Chromatin immunoprecipitation (ChIP) PCR was used to determine direct binding sites of KLF4 in T-ALL cells.ResultsHere we reveal that KLF4 induced apoptosis through the BCL2/BCLXL pathway in human T-ALL cell lines and primary T-ALL specimens. In consistence, mice engrafted with KLF4-overexpressing T-ALL cells exhibited prolonged survival. Interestingly, the KLF4-induced apoptosis in T-ALL cells was compromised in xenografts but the invasion capacity of KLF4-expressing T-ALL cells to hosts was dramatically dampened. We found that KLF4 overexpression inhibited T cell-associated genes including NOTCH1, BCL11B, GATA3, and TCF7. Further mechanistic studies revealed that KLF4 directly bound to the promoters of NOTCH1, BCL2, and CXCR4 and suppressed their expression. Additionally, KLF4 induced SUMOylation and degradation of BCL11B.ConclusionsThese results suggest that KLF4 as a major transcription factor that suppresses the expression of T-cell associated genes, thus inhibiting T-ALL progression.

Collaboration


Dive into the Mei Zhong's collaboration.

Top Co-Authors

Avatar

Duanqing Pei

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Jinglei Cai

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Bei Jia

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Dajiang Qin

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Jiayin Yang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Miguel A. Esteban

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

Jianyong Xu

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Yanhong Yu

Southern Medical University

View shared research outputs
Top Co-Authors

Avatar

Bing Xu

Southern Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge