Network


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

Hotspot


Dive into the research topics where Beisi Xu is active.

Publication


Featured researches published by Beisi Xu.


Cell Stem Cell | 2015

Quantification of Retinogenesis in 3D Cultures Reveals Epigenetic Memory and Higher Efficiency in iPSCs Derived from Rod Photoreceptors

Daniel Hiler; Xiang Chen; Jennifer L. Hazen; Sergey Kupriyanov; Patrick A. Carroll; Chunxu Qu; Beisi Xu; Dianna A. Johnson; Lyra Griffiths; Sharon Frase; Alberto R. Rodriguez; Greg Martin; Jiakun Zhang; Jongrye Jeon; Yiping Fan; David Finkelstein; Robert N. Eisenman; Kristin K. Baldwin; Michael A. Dyer

Cell-based therapies to treat retinal degeneration are now being tested in clinical trials. However, it is not known whether the source of stem cells is important for the production of differentiated cells suitable for transplantation. To test this, we generated induced pluripotent stem cells (iPSCs) from murine rod photoreceptors (r-iPSCs) and scored their ability to make retinae by using a standardized quantitative protocol called STEM-RET. We discovered that r-iPSCs more efficiently produced differentiated retinae than did embryonic stem cells (ESCs) or fibroblast-derived iPSCs (f-iPSCs). Retinae derived from f-iPSCs had fewer amacrine cells and other inner nuclear layer cells. Integrated epigenetic analysis showed that DNA methylation contributes to the defects in f-iPSC retinogenesis and that rod-specific CTCF insulator protein-binding sites may promote r-iPSC retinogenesis. Together, our data suggest that the source of stem cells is important for producing retinal neurons in three-dimensional (3D) organ cultures.


Nature Genetics | 2016

Deregulation of DUX4 and ERG in acute lymphoblastic leukemia

Jinghui Zhang; Kelly McCastlain; Hiroki Yoshihara; Beisi Xu; Yunchao Chang; Michelle L. Churchman; Gang Wu; Yongjin Li; Lei Wei; Ilaria Iacobucci; Yu Liu; Chunxu Qu; Ji Wen; Michael Edmonson; Debbie Payne-Turner; Kerstin B Kaufmann; Shin-ichiro Takayanagi; Erno Wienholds; Esmé Waanders; Panagiotis Ntziachristos; Sofia Bakogianni; Jingjing Wang; Iannis Aifantis; Kathryn G. Roberts; Jing Ma; Guangchun Song; John Easton; Heather L. Mulder; Xiang Chen; Scott Newman

Chromosomal rearrangements deregulating hematopoietic transcription factors are common in acute lymphoblastic leukemia (ALL). Here we show that deregulation of the homeobox transcription factor gene DUX4 and the ETS transcription factor gene ERG is a hallmark of a subtype of B-progenitor ALL that comprises up to 7% of B-ALL. DUX4 rearrangement and overexpression was present in all cases and was accompanied by transcriptional deregulation of ERG, expression of a novel ERG isoform, ERGalt, and frequent ERG deletion. ERGalt uses a non-canonical first exon whose transcription was initiated by DUX4 binding. ERGalt retains the DNA-binding and transactivation domains of ERG, but it inhibits wild-type ERG transcriptional activity and is transforming. These results illustrate a unique paradigm of transcription factor deregulation in leukemia in which DUX4 deregulation results in loss of function of ERG, either by deletion or induced expression of an isoform that is a dominant-negative inhibitor of wild-type ERG function.


Neuron | 2017

The Dynamic Epigenetic Landscape of the Retina During Development, Reprogramming, and Tumorigenesis

Issam Aldiri; Beisi Xu; Lu Wang; Xiang Chen; Daniel Hiler; Lyra Griffiths; Marc Valentine; Abbas Shirinifard; Suresh Thiagarajan; András Sablauer; Marie-Elizabeth Barabas; Jiakun Zhang; Dianna A. Johnson; Sharon Frase; Xin Zhou; John Easton; Jinghui Zhang; Elaine R. Mardis; Richard Wilson; James R. Downing; Michael A. Dyer

SUMMARY In the developing retina, multipotent neural progenitors undergo unidirectional differentiation in a precise spatiotemporal order. Here we profile the epigenetic and transcriptional changes that occur during retinogenesis in mice and humans. Although some progenitor genes and cell cycle genes were epigenetically silenced during retinogenesis, the most dramatic change was derepression of cell type–specific differentiation programs. We identified developmental stage–specific super-enhancers and showed that most epigenetic changes are conserved in humans and mice. To determine how the epigenome changes during tumorigenesis and reprogramming, we performed integrated epigenetic analysis of murine and human retinoblastomas and induced pluripotent stem cells (iPSCs) derived from murine rod photoreceptors. The retinoblastoma epigenome mapped to the developmental stage when retinal progenitors switch from neurogenic to a terminal patterns of cell division. The epigenome of retinoblastomas was more similar to that of normal retina than was that of retina-derived iPSCs, and we identified retina-specific epigenetic memory.


Nature | 2017

Orthotopic patient-derived xenografts of paediatric solid tumours

Elizabeth Stewart; Sara M. Federico; Xiang Chen; Anang A. Shelat; Cori Bradley; Brittney Gordon; Åsa Karlström; Nathaniel R. Twarog; Michael R. Clay; Armita Bahrami; Burgess B. Freeman; Beisi Xu; Xin Zhou; Jianrong Wu; Victoria Honnell; Monica Ocarz; Kaley Blankenship; Jason Dapper; Elaine R. Mardis; Richard Wilson; James R. Downing; Jinghui Zhang; John Easton; Alberto S. Pappo; Michael A. Dyer

Paediatric solid tumours arise from endodermal, ectodermal, or mesodermal lineages. Although the overall survival of children with solid tumours is 75%, that of children with recurrent disease is below 30%. To capture the complexity and diversity of paediatric solid tumours and establish new models of recurrent disease, here we develop a protocol to produce orthotopic patient-derived xenografts at diagnosis, recurrence, and autopsy. Tumour specimens were received from 168 patients, and 67 orthotopic patient-derived xenografts were established for 12 types of cancer. The origins of the patient-derived xenograft tumours were reflected in their gene-expression profiles and epigenomes. Genomic profiling of the tumours, including detailed clonal analysis, was performed to determine whether the clonal population in the xenograft recapitulated the patient’s tumour. We identified several drug vulnerabilities and showed that the combination of a WEE1 inhibitor (AZD1775), irinotecan, and vincristine can lead to complete response in multiple rhabdomyosarcoma orthotopic patient-derived xenografts tumours in vivo.


Development | 2015

Brg1 coordinates multiple processes during retinogenesis and is a tumor suppressor in retinoblastoma.

Issam Aldiri; Itsuki Ajioka; Beisi Xu; Jiakun Zhang; Xiang Chen; Claudia A. Benavente; David Finkelstein; Dianna A. Johnson; Jennifer A. Akiyama; Len A. Pennacchio; Michael A. Dyer

Retinal development requires precise temporal and spatial coordination of cell cycle exit, cell fate specification, cell migration and differentiation. When this process is disrupted, retinoblastoma, a developmental tumor of the retina, can form. Epigenetic modulators are central to precisely coordinating developmental events, and many epigenetic processes have been implicated in cancer. Studying epigenetic mechanisms in development is challenging because they often regulate multiple cellular processes; therefore, elucidating the primary molecular mechanisms involved can be difficult. Here we explore the role of Brg1 (Smarca4) in retinal development and retinoblastoma in mice using molecular and cellular approaches. Brg1 was found to regulate retinal size by controlling cell cycle length, cell cycle exit and cell survival during development. Brg1 was not required for cell fate specification but was required for photoreceptor differentiation and cell adhesion/polarity programs that contribute to proper retinal lamination during development. The combination of defective cell differentiation and lamination led to retinal degeneration in Brg1-deficient retinae. Despite the hypocellularity, premature cell cycle exit, increased cell death and extended cell cycle length, retinal progenitor cells persisted in Brg1-deficient retinae, making them more susceptible to retinoblastoma. ChIP-Seq analysis suggests that Brg1 might regulate gene expression through multiple mechanisms. Summary: The SWI/SNF protein Brg1 controls cell cycle length, cell cycle exit and cell survival, and is required for cell differentiation and retinal lamination, in the developing mouse retina.


Cell Reports | 2018

Retinal Cell Type DNA Methylation and Histone Modifications Predict Reprogramming Efficiency and Retinogenesis in 3D Organoid Cultures

Lu Wang; Daniel Hiler; Beisi Xu; Issam Aldiri; Xiang Chen; Xin Zhou; Lyra Griffiths; Marc Valentine; Abbas Shirinifard; András Sablauer; Suresh Thiagarajan; Marie-Elizabeth Barabas; Jiakun Zhang; Dianna A. Johnson; Sharon Frase; Michael A. Dyer

SUMMARY Diverse cell types can be reprogrammed into pluripotent stem cells by ectopic expression of Oct4 (Pou5f1), Klf4, Sox3, and Myc. Many of these induced pluripotent stem cells (iPSCs) retain memory, in terms of DNA methylation and histone modifications (epigenetic memory), of their cellular origins, and this may bias subsequent differentiation. Neurons are difficult to reprogram, and there has not been a systematic side-by-side characterization of reprogramming efficiency or epigenetic memory across different neuronal subtypes. Here, we compare reprogramming efficiency of five different retinal cell types at two different stages of development. Retinal differentiation from each iPSC line was measured using a quantitative standardized scoring system called STEM-RET and compared to the epigenetic memory. Neurons with the lowest reprogramming efficiency produced iPSC lines with the best retinal differentiation and were more likely to retain epigenetic memory of their cellular origins. In addition, we identified biomarkers of iPSCs that are predictive of retinal differentiation.


Genes & Development | 2015

The glucose-sensing transcription factor MLX promotes myogenesis via myokine signaling

Liam Hunt; Beisi Xu; David Finkelstein; Yiping Fan; Patrick A. Carroll; Pei-Feng Cheng; Robert N. Eisenman; Fabio Demontis

Metabolic stress and changes in nutrient levels modulate many aspects of skeletal muscle function during aging and disease. Growth factors and cytokines secreted by skeletal muscle, known as myokines, are important signaling factors, but it is largely unknown whether they modulate muscle growth and differentiation in response to nutrients. Here, we found that changes in glucose levels increase the activity of the glucose-responsive transcription factor MLX (Max-like protein X), which promotes and is necessary for myoblast fusion. MLX promotes myogenesis not via an adjustment of glucose metabolism but rather by inducing the expression of several myokines, including insulin-like growth factor 2 (IGF2), whereas RNAi and dominant-negative MLX reduce IGF2 expression and block myogenesis. This phenotype is rescued by conditioned medium from control muscle cells and by recombinant IGF2, which activates the myogenic kinase Akt. Importantly, MLX-null mice display decreased IGF2 induction and diminished muscle regeneration in response to injury, indicating that the myogenic function of MLX is manifested in vivo. Thus, glucose is a signaling molecule that regulates myogenesis and muscle regeneration via MLX/IGF2/Akt signaling.


bioRxiv | 2018

The Nucleome of Developing Murine Rod Photoreceptors

Issam Al Diri; Marc Valentine; Beisi Xu; Daniel Putnam; Lyra Griffiths; Marybeth Lupo; Jackie Norrie; Jiakun Zhang; Dianna Johnson; John Easton; Abbas Shirinifard; Ying Shao; Victoria Honnell; Sharon Frase; Shondra Miller; Valerie Stewart; Xiang Chen; Michael A. Dyer

The nuclei of rod photoreceptors in mice and other nocturnal species have an unusual inverted chromatin structure: the heterochromatin is centrally located to help focus light and improve photosensitivity. To better understand this unique nuclear organization, we performed ultra-deep Hi-C analysis on murine retina at 3 stages of development and on purified rod photoreceptors. Predicted looping interactions from the Hi-C data were validated with fluorescence in situ hybridization (FISH). We discovered that a subset of retinal genes that are important for retinal development, cancer, and stress response are localized to the facultative heterochromatin domain. We also used machine learning to develop an algorithm based on our chromatin Hidden Markov Modeling (chromHMM) of retinal development to predict heterochromatin domains and study their dynamics during retinogenesis. FISH data for 264 genomic loci were used to train and validate the algorithm. The integrated data were then used to identify a developmental stage– and cell type-specific core regulatory circuit super-enhancer (CRC-SE) upstream of the Vsx2 gene, which is required for bipolar neuron expression. Deletion of the Vsx2 CRC-SE in mice led to the loss of bipolar neurons in the retina.


bioRxiv | 2018

MYCN Amplification and ATRX Mutations are Incompatible in Neuroblastoma

Maged Zeineldin; Sara M. Federico; Xiang Chen; Yiping Fan; Beisi Xu; Elizabeth Stewart; Xin Zhou; Jongrye Jeon; Lyra Griffiths; John Easton; Heather L. Mulder; Donald Yergeau; Yanling Liu; Jianrong Wu; Collin Van Ryn; Arlene Naranjo; Michael D. Hogarty; Marcin Kaminski; Marc Valentine; Shondra Miller; Alberto S. Pappo; Jinghui Zhang; Michael R. Clay; Armita Bahrami; Seungjae Lee; Anang A. Shelat; Rani George; Elaine R. Mardis; Richard K. Wilson; James R. Downing

Aggressive cancers often have activating mutations in growth-controlling oncogenes and inactivating mutations in tumor-suppressor genes. In neuroblastoma, amplification of the MYCN oncogene and inactivation of the ATRX tumor-suppressor gene correlate with high-risk disease and poor prognosis. Here we show that ATRX mutations and MYCN amplification are mutually exclusive across all ages and stages in neuroblastoma. Using human cell lines and mouse models, we found that elevated MYCN expression and ATRX mutations are incompatible. Elevated MYCN levels promote metabolic reprogramming, mitochondrial dysfunction, reactive-oxygen species generation, and DNA-replicative stress. The combination of replicative stress caused by defects in the ATRX–histone chaperone complex and that induced by MYCN-mediated metabolic reprogramming leads to synthetic lethality. Therefore, ATRX and MYCN represent an unusual example, where inactivation of a tumor-suppressor gene and activation of an oncogene are incompatible. This synthetic lethality may eventually be exploited to improve outcomes for patients with high-risk neuroblastoma.


Scientific Reports | 2018

Mouse medulloblastoma driven by CRISPR activation of cellular Myc

BaoHan T. Vo; Jin Ah Kwon; Chunliang Li; David Finkelstein; Beisi Xu; Brent A. Orr; Charles J. Sherr; Martine F. Roussel

MYC-driven Group 3 (G3) medulloblastoma (MB) is the most aggressive of four molecular subgroups classified by transcriptome, genomic landscape and clinical outcomes. Mouse models that recapitulate human G3 MB all rely on retroviral vector-induced Myc expression driven by viral regulatory elements (Retro-Myc tumors). We used nuclease-deficient CRISPR/dCas9-based gene activation with combinatorial single guide RNAs (sgRNAs) to enforce transcription of endogenous Myc in Trp53-null neurospheres that were orthotopically transplanted into the brains of naïve animals. Three combined sgRNAs linked to dCas9-VP160 induced cellular Myc expression and large cell anaplastic MBs (CRISPR-Myc tumors) which recapitulated the molecular characteristics of mouse and human G3 MBs. The BET inhibitor JQ1 suppressed MYC expression in a human G3 MB cell line (HD-MB03) and CRISPR-Myc, but not in Retro-Myc MBs. This G3 MB mouse model in which Myc expression is regulated by its own promoter will facilitate pre-clinical studies with drugs that regulate Myc transcription.

Collaboration


Dive into the Beisi Xu's collaboration.

Top Co-Authors

Avatar

Xiang Chen

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jinghui Zhang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

John Easton

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Yiping Fan

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

David Finkelstein

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Lyra Griffiths

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Michael A. Dyer

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Jiakun Zhang

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Xin Zhou

China Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Dianna A. Johnson

University of Tennessee Health Science Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge