Network


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

Hotspot


Dive into the research topics where Christina Benda is active.

Publication


Featured researches published by Christina Benda.


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 The American Society of Nephrology | 2011

Generation of induced pluripotent stem cells from urine

Ting Zhou; Christina Benda; Sarah Duzinger; Yinghua Huang; Xingyan Li; Yanhua Li; Xiangpeng Guo; Guokun Cao; Shen Chen; Lili Hao; Ys Chan; Kwong-Man Ng; Jenny Cy Ho; Matthias Wieser; Jiayan Wu; Heinz Redl; Hung-Fat Tse; Johannes Grillari; Regina Grillari-Voglauer; Duanqing Pei; Miguel A. Esteban

Forced expression of selected transcription factors can transform somatic cells into embryonic stem cell (ESC)-like cells, termed induced pluripotent stem cells (iPSCs). There is no consensus regarding the preferred tissue from which to harvest donor cells for reprogramming into iPSCs, and some donor cell types may be more prone than others to accumulation of epigenetic imprints and somatic cell mutations. Here, we present a simple, reproducible, noninvasive method for generating human iPSCs from renal tubular cells present in urine. This procedure eliminates many problems associated with other protocols, and the resulting iPSCs display an excellent ability to differentiate. These data suggest that urine may be a preferred source for generating iPSCs.


Current Opinion in Cell Biology | 2013

MicroRNAs in somatic cell reprogramming

Xichen Bao; Xihua Zhu; Baojian Liao; Christina Benda; Qiang Zhuang; Duanqing Pei; Baoming Qin; Miguel A. Esteban

The generation of induced pluripotent stem (iPS) cells by exogenous transcription factors involves a comprehensive rearrangement of cellular functions, including the microRNA profile. The resulting cell lines are similar to embryonic stem (ES) cells and have therefore raised much interest for in vitro studies and the perspective of clinical application. Yet, microRNAs are not mere listeners of the reprogramming orchestra but play an active role in the process. In consequence, overexpression or suppression of individual microRNAs has profound effects in colony formation efficiency, and in combination they can produce iPS cells without added transcription factors. Moreover, variations in microRNA expression of iPS/ES cells can predict their differentiation potential and may have consequences at other levels. Altogether, these findings highlight the relevance of pursuing further these studies.


Nature Cell Biology | 2015

Autophagy and mTORC1 regulate the stochastic phase of somatic cell reprogramming

Yasong Wu; Yuan Li; Hui Zhang; Yinghua Huang; Ping Zhao; Yujia Tang; Xiaohui Qiu; Yue Ying; Wen Li; Su Ni; Meng Zhang; Longqi Liu; Yan Xu; Qiang Zhuang; Zhiwei Luo; Christina Benda; Hong Song; Baohua Liu; Liangxue Lai; Xingguo Liu; Hung-Fat Tse; Xichen Bao; Wai-Yee Chan; Miguel A. Esteban; Baoming Qin; Duanqing Pei

We describe robust induction of autophagy during the reprogramming of mouse fibroblasts to induced pluripotent stem cells by four reprogramming factors (Sox2, Oct4, Klf4 and c-Myc), henceforth 4F. This process occurs independently of p53 activation, and is mediated by the synergistic downregulation of mechanistic target of rapamycin complex 1 (mTORC1) and the induction of autophagy-related genes. The 4F coordinately repress mTORC1, but bifurcate in their regulation of autophagy-related genes, with Klf4 and c-Myc inducing them but Sox2 and Oct4 inhibiting them. On one hand, inhibition of mTORC1 facilitates reprogramming by promoting cell reshaping (mitochondrial remodelling and cell size reduction). On the other hand, mTORC1 paradoxically impairs reprogramming by triggering autophagy. Autophagy does not participate in cell reshaping in reprogramming but instead degrades p62, whose accumulation in autophagy-deficient cells facilitates reprogramming. Our results thus reveal a complex signalling network involving mTORC1 inhibition and autophagy induction in the early phase of reprogramming, whose delicate balance ultimately determines reprogramming efficiency.


Cell Stem Cell | 2014

Transcriptional Pause Release Is a Rate-Limiting Step for Somatic Cell Reprogramming

Longqi Liu; Yan Xu; Minghui He; Meng Zhang; Fenggong Cui; Leina Lu; Mingze Yao; Weihua Tian; Christina Benda; Qiang Zhuang; Zhijian Huang; Wenjuan Li; Xiangchun Li; Ping Zhao; Wenxia Fan; Zhiwei Luo; Yuan Li; Yasong Wu; Andrew Paul Hutchins; Dongye Wang; Hung-Fat Tse; Axel Schambach; Jon Frampton; Baoming Qin; Xichen Bao; Hongjie Yao; Biliang Zhang; Hao Sun; Duanqing Pei; Huating Wang

Reactivation of the pluripotency network during somatic cell reprogramming by exogenous transcription factors involves chromatin remodeling and the recruitment of RNA polymerase II (Pol II) to target loci. Here, we report that Pol II is engaged at pluripotency promoters in reprogramming but remains paused and inefficiently released. We also show that bromodomain-containing protein 4 (BRD4) stimulates productive transcriptional elongation of pluripotency genes by dissociating the pause release factor P-TEFb from an inactive complex containing HEXIM1. Consequently, BRD4 overexpression enhances reprogramming efficiency and HEXIM1 suppresses it, whereas Brd4 and Hexim1 knockdown do the opposite. We further demonstrate that the reprogramming factor KLF4 helps recruit P-TEFb to pluripotency promoters. Our work thus provides a mechanism for explaining the reactivation of pluripotency genes in reprogramming and unveils an unanticipated role for KLF4 in transcriptional pause release.


Cell Regeneration | 2014

Characterization of a novel cell penetrating peptide derived from human Oct4

Eva Harreither; Hanna Rydberg; Helene L. Åmand; Vaibhav Jadhav; Lukas Fliedl; Christina Benda; Miguel A. Esteban; Duanqing Pei; Nicole Borth; Regina Grillari-Voglauer; Oliver Hommerding; Frank Edenhofer; Bengt Nordén; Johannes Grillari

BackgroundOct4 is a transcription factor that plays a major role for the preservation of the pluripotent state in embryonic stem cells as well as for efficient reprogramming of somatic cells to induced pluripotent stem cells (iPSC) or other progenitors. Protein-based reprogramming methods mainly rely on the addition of a fused cell penetrating peptide. This study describes that Oct4 inherently carries a protein transduction domain, which can translocate into human and mouse cells.ResultsA 16 amino acid peptide representing the third helix of the human Oct4 homeodomain, referred to as Oct4 protein transduction domain (Oct4-PTD), can internalize in mammalian cells upon conjugation to a fluorescence moiety thereby acting as a cell penetrating peptide (CPP). The cellular distribution of Oct4-PTD shows diffuse cytosolic and nuclear staining, whereas penetratin is strictly localized to a punctuate pattern in the cytoplasm. By using a Cre/loxP-based reporter system, we show that this peptide also drives translocation of a functionally active Oct4-PTD-Cre-fusion protein. We further provide evidence for translocation of full length Oct4 into human and mouse cell lines without the addition of any kind of cationic fusion tag. Finally, physico-chemical properties of the novel CPP are characterized, showing that in contrast to penetratin a helical structure of Oct4-PTD is only observed if the FITC label is present on the N-terminus of the peptide.ConclusionsOct4 is a key transcription factor in stem cell research and cellular reprogramming. Since it has been shown that recombinant Oct4 fused to a cationic fusion tag can drive generation of iPSCs, our finding might contribute to further development of protein-based methods to generate iPSCs.Moreover, our data support the idea that transcription factors might be part of an alternative paracrine signalling pathway, where the proteins are transferred to neighbouring cells thereby actively changing the behaviour of the recipient cell.


Journal of Biological Chemistry | 2013

Class IIa Histone Deacetylases and Myocyte Enhancer Factor 2 Proteins Regulate the Mesenchymal-to-Epithelial Transition of Somatic Cell Reprogramming

Qiang Zhuang; Xiaobing Qing; Yue Ying; Haitao Wu; Christina Benda; Jiao Lin; Zhijian Huang; Longqi Liu; Yan Xu; Xichen Bao; Baoming Qin; Duanqing Pei; Miguel A. Esteban

Background: Somatic cell reprogramming is an inefficient process because of the existence of roadblocks. Results: Class IIa histone deacetylases and MEF2 proteins increase during mouse fibroblast reprogramming and differentially regulate the expression of Tgfβ cytokines. Conclusion: This interplay regulates the mesenchymal-to-epithelial transition phase of reprogramming. Significance: Our findings help understand the mechanisms of reprogramming and may have implications in other contexts. Class IIa histone deacetylases (HDACs) and myocyte enhancer factor 2 (MEF2) proteins compose a signaling module that orchestrates lineage specification during embryogenesis. We show here that this module also regulates the generation of mouse induced pluripotent stem cells by defined transcription factors. Class IIa HDACs and MEF2 proteins rise steadily during fibroblast reprogramming to induced pluripotent stem cells. MEF2 proteins tend to block the process by inducing the expression of Tgfβ cytokines, which impairs the necessary phase of mesenchymal-to-epithelial transition (MET). Conversely, class IIa HDACs endeavor to suppress the activity of MEF2 proteins, thus enhancing the MET and colony formation efficiency. Our work highlights an unexpected role for a developmental axis in somatic cell reprogramming and provides new insight into how the MET is regulated in this context.


Advances in Biochemical Engineering \/ Biotechnology | 2012

Urine as a source of stem cells.

Christina Benda; Ting Zhou; Xianming Wang; Weihua Tian; Johannes Grillari; Hung-Fat Tse; Regina Grillari-Voglauer; Duanqing Pei; Miguel A. Esteban

Traditionally, clinicians and researchers have relied on a skin biopsy or blood extraction as relatively accessible supplies for in vitro cell expansion and biological studies. Perhaps surprisingly, limited attention has been given to a totally noninvasive source, urine, which eliminates the discomfort associated with other procedures. This may arise from the perception that urine is merely a body waste. Yet, the analysis of urine is a longstanding fundamental test for diagnostic purposes and nowadays there is growing interest in using urine for detecting biomarkers. In addition, recent work including ours reinforces the idea that urine contains a variety of viable cell types with relevant applications. In this review, we describe those cell types and their potential uses.


Nature Cell Biology | 2018

NCoR/SMRT co-repressors cooperate with c-MYC to create an epigenetic barrier to somatic cell reprogramming

Qiang Zhuang; Wenjuan Li; Christina Benda; Zhijian Huang; Tanveer Ahmed; Ping Liu; Xiangpeng Guo; David P. Ibañez; Zhiwei Luo; Meng Zhang; Mazid Md. Abdul; Zhongzhou Yang; Jiayin Yang; Yinghua Huang; Hui Zhang; Dehao Huang; Jianguo Zhou; Xiaofen Zhong; Xihua Zhu; Xiuling Fu; Wenxia Fan; Yulin Liu; Yan Xu; Carl Ward; Muhammad Jadoon Khan; Shahzina Kanwal; Bushra Mirza; Micky D. Tortorella; Hung-Fat Tse; Jiayu Chen

Somatic cell reprogramming by exogenous factors requires cooperation with transcriptional co-activators and co-repressors to effectively remodel the epigenetic environment. How this interplay is regulated remains poorly understood. Here, we demonstrate that NCoR/SMRT co-repressors bind to pluripotency loci to create a barrier to reprogramming with the four Yamanaka factors (OCT4, SOX2, KLF4 and c-MYC), and consequently, suppressing NCoR/SMRT significantly enhances reprogramming efficiency and kinetics. The core epigenetic subunit of the NCoR/SMRT complex, histone deacetylase 3 (HDAC3), contributes to the effects of NCoR/SMRT by inducing histone deacetylation at pluripotency loci. Among the Yamanaka factors, recruitment of NCoR/SMRT–HDAC3 to genomic loci is mostly facilitated by c-MYC. Hence, we describe how c-MYC is beneficial for the early phase of reprogramming but deleterious later. Overall, we uncover a role for NCoR/SMRT co-repressors in reprogramming and propose a dual function for c-MYC in this process.Zhuang et al. demonstrate that suppression of NCoR/SMRT enhances OSKM reprogramming efficiency, and that the barrier mechanism depends on the recruitment of HDAC3 to pluripotency loci by c-MYC.


Nature Cell Biology | 2018

Publisher Correction: NCoR/SMRT co-repressors cooperate with c-MYC to create an epigenetic barrier to somatic cell reprogramming

Qiang Zhuang; Wenjuan Li; Christina Benda; Zhijian Huang; Tanveer Ahmed; Ping Liu; Xiangpeng Guo; David P. Ibañez; Zhiwei Luo; Meng Zhang; Mazid Md. Abdul; Zhongzhou Yang; Jiayin Yang; Yinghua Huang; Hui Zhang; Dehao Huang; Jianguo Zhou; Xiaofen Zhong; Xihua Zhu; Xiuling Fu; Wenxia Fan; Yulin Liu; Yan Xu; Carl Ward; Muhammad Jadoon Khan; Shahzina Kanwal; Bushra Mirza; Micky D. Tortorella; Hung-Fat Tse; Jiayu Chen

In the version of this Article originally published, in Fig. 2c, the ‘+’ sign and ‘OSKM’ were superimposed in the label ‘+OSKM’. In Fig. 4e, in the labels, all instances of ‘Ant’ should have been ‘Anti-’. And, in Fig. 7a, the label ‘0.0’ was misplaced; it should have been on the colour scale bar. These figures have now been corrected in the online versions.

Collaboration


Dive into the Christina Benda's collaboration.

Top Co-Authors

Avatar

Duanqing Pei

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

Qiang Zhuang

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

Xichen Bao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yan Xu

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

Jiayin Yang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Meng Zhang

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Top Co-Authors

Avatar

Ting Zhou

Guangzhou Institutes of Biomedicine and Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge