Network


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

Hotspot


Dive into the research topics where Yizhou Huang is active.

Publication


Featured researches published by Yizhou Huang.


Cell | 2013

Orchestrated intron retention regulates normal granulocyte differentiation.

Justin Wong; William Ritchie; Olivia A. Ebner; Matthias Selbach; Jason Wong; Yizhou Huang; Dadi Gao; Natalia Pinello; Maria Gonzalez; Kinsha Baidya; Annora Thoeng; Teh-Liane Khoo; Charles G. Bailey; Jeff Holst; John E.J. Rasko

Intron retention (IR) is widely recognized as a consequence of mis-splicing that leads to failed excision of intronic sequences from pre-messenger RNAs. Our bioinformatic analyses of transcriptomic and proteomic data of normal white blood cell differentiation reveal IR as a physiological mechanism of gene expression control. IR regulates the expression of 86 functionally related genes, including those that determine the nuclear shape that is unique to granulocytes. Retention of introns in specific genes is associated with downregulation of splicing factors and higher GC content. IR, conserved between human and mouse, led to reduced mRNA and protein levels by triggering the nonsense-mediated decay (NMD) pathway. In contrast to the prevalent view that NMD is limited to mRNAs encoding aberrant proteins, our data establish that IR coupled with NMD is a conserved mechanism in normal granulopoiesis. Physiological IR may provide an energetically favorable level of dynamic gene expression control prior to sustained gene translation.


Leukemia | 2015

Overexpression of ERG in cord blood progenitors promotes expansion and recapitulates molecular signatures of high ERG leukemias

Melinda L. Tursky; Dominik Beck; Julie A.I. Thoms; Yizhou Huang; A. Kumari; Ashwin Unnikrishnan; Kathy Knezevic; Kathryn Evans; Laura A. Richards; Erwin M. Lee; Jonathan M. Morris; Liat Goldberg; Shai Izraeli; Jason Wong; Jake Olivier; Richard B. Lock; Karen L. MacKenzie; John E. Pimanda

High expression of the ETS family transcription factor ERG is associated with poor clinical outcome in acute myeloid leukemia (AML) and acute T-cell lymphoblastic leukemia (T-ALL). In murine models, high ERG expression induces both T-ALL and AML. However, no study to date has defined the effect of high ERG expression on primary human hematopoietic cells. In the present study, human CD34+ cells were transduced with retroviral vectors to elevate ERG gene expression to levels detected in high ERG AML. RNA sequencing was performed on purified populations of transduced cells to define the effects of high ERG on gene expression in human CD34+ cells. Integration of the genome-wide expression data with other data sets revealed that high ERG drives an expression signature that shares features of normal hematopoietic stem cells, high ERG AMLs, early T-cell precursor-ALLs and leukemic stem cell signatures associated with poor clinical outcome. Functional assays linked this gene expression profile to enhanced progenitor cell expansion. These results support a model whereby a stem cell gene expression network driven by high ERG in human cells enhances the expansion of the progenitor pool, providing opportunity for the acquisition and propagation of mutations and the development of leukemia.


Nature Communications | 2017

The non-coding RNA landscape of human hematopoiesis and leukemia

Adrian Schwarzer; Stephan Emmrich; Franziska Schmidt; Dominik Beck; Michelle Ng; Christina Reimer; Felix F. Adams; Sarah Grasedieck; Damian Witte; Sebastian Käbler; Jason Wong; Anushi Shah; Yizhou Huang; Razan Jammal; Aliaksandra Maroz; Mojca Jongen-Lavrencic; Axel Schambach; Florian Kuchenbauer; John E. Pimanda; Dirk Reinhardt; Dirk Heckl; Jan-Henning Klusmann

Non-coding RNAs have emerged as crucial regulators of gene expression and cell fate decisions. However, their expression patterns and regulatory functions during normal and malignant human hematopoiesis are incompletely understood. Here we present a comprehensive resource defining the non-coding RNA landscape of the human hematopoietic system. Based on highly specific non-coding RNA expression portraits per blood cell population, we identify unique fingerprint non-coding RNAs—such as LINC00173 in granulocytes—and assign these to critical regulatory circuits involved in blood homeostasis. Following the incorporation of acute myeloid leukemia samples into the landscape, we further uncover prognostically relevant non-coding RNA stem cell signatures shared between acute myeloid leukemia blasts and healthy hematopoietic stem cells. Our findings highlight the importance of the non-coding transcriptome in the formation and maintenance of the human blood hierarchy.While micro-RNAs are known regulators of haematopoiesis and leukemogenesis, the role of long non-coding RNAs is less clear. Here the authors provide a non-coding RNA expression landscape of the human hematopoietic system, highlighting their role in the formation and maintenance of the human blood hierarchy.


Leukemia | 2016

MAPK/ERK2 phosphorylates ERG at serine 283 in leukemic cells and promotes stem cell signatures and cell proliferation.

Yizhou Huang; Julie A.I. Thoms; Melinda L. Tursky; Kathy Knezevic; Dominik Beck; Chandrakanthan; Santi Suryani; Jake Olivier; Adam Boulton; Elias N. Glaros; Thomas; Richard B. Lock; Karen L. MacKenzie; John H. Bushweller; Jason Wong; John E. Pimanda

Aberrant ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) expression drives leukemic transformation in mice and high expression is associated with poor patient outcomes in acute myeloid leukemia (AML) and T-acute lymphoblastic leukemia (T-ALL). Protein phosphorylation regulates the activity of many ETS factors but little is known about ERG in leukemic cells. To characterize ERG phosphorylation in leukemic cells, we applied liquid chromatography coupled tandem mass spectrometry and identified five phosphorylated serines on endogenous ERG in T-ALL and AML cells. S283 was distinct as it was abundantly phosphorylated in leukemic cells but not in healthy hematopoietic stem and progenitor cells (HSPCs). Overexpression of a phosphoactive mutant (S283D) increased expansion and clonogenicity of primary HSPCs over and above wild-type ERG. Using a custom antibody, we screened a panel of primary leukemic xenografts and showed that ERG S283 phosphorylation was mediated by mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling and in turn regulated expression of components of this pathway. S283 phosphorylation facilitates ERG enrichment and transactivation at the ERG +85 HSPC enhancer that is active in AML and T-ALL with poor prognosis. Taken together, we have identified a specific post-translational modification in leukemic cells that promotes progenitor proliferation and is a potential target to modulate ERG-driven transcriptional programs in leukemia.


Nucleic Acids Research | 2016

A quantitative proteomics approach identifies ETV6 and IKZF1 as new regulators of an ERG-driven transcriptional network

Ashwin Unnikrishnan; Yi Fang Guan; Yizhou Huang; Dominik Beck; Julie A.I. Thoms; Sofie Peirs; Kathy Knezevic; Shiyong Ma; Inge Vande Walle; Ineke De Jong; Zara Ali; Ling Zhong; Mark J. Raftery; Tom Taghon; Jonas Larsson; Karen L. MacKenzie; Pieter Van Vlierberghe; Jason Wong; John E. Pimanda

Aberrant stem cell-like gene regulatory networks are a feature of leukaemogenesis. The ETS-related gene (ERG), an important regulator of normal haematopoiesis, is also highly expressed in T-ALL and acute myeloid leukaemia (AML). However, the transcriptional regulation of ERG in leukaemic cells remains poorly understood. In order to discover transcriptional regulators of ERG, we employed a quantitative mass spectrometry-based method to identify factors binding the 321 bp ERG +85 stem cell enhancer region in MOLT-4 T-ALL and KG-1 AML cells. Using this approach, we identified a number of known binders of the +85 enhancer in leukaemic cells along with previously unknown binders, including ETV6 and IKZF1. We confirmed that ETV6 and IKZF1 were also bound at the +85 enhancer in both leukaemic cells and in healthy human CD34+ haematopoietic stem and progenitor cells. Knockdown experiments confirmed that ETV6 and IKZF1 are transcriptional regulators not just of ERG, but also of a number of genes regulated by a densely interconnected network of seven transcription factors. At last, we show that ETV6 and IKZF1 expression levels are positively correlated with expression of a number of heptad genes in AML and high expression of all nine genes confers poorer overall prognosis.


Molecular and Cellular Biology | 2016

Arrested Hematopoiesis and Vascular Relaxation Defects in Mice with a Mutation in Dhfr

Julie A.I. Thoms; Kathy Knezevic; Jia Jenny Liu; Elias N. Glaros; Thuan Thai; Qiao Qiao; Heather Campbell; Deborah Packham; Yizhou Huang; Panagiotis Papathanasiou; Robert Tunningley; Belinda Whittle; Amanda W. S. Yeung; Vashe Chandrakanthan; Luke B. Hesson; Vivien M. Chen; Jason Wong; Louise E. Purton; Robyn L. Ward; Shane R. Thomas; John E. Pimanda

ABSTRACT Dihydrofolate reductase (DHFR) is a critical enzyme in the folate metabolism pathway and also plays a role in regulating nitric oxide (NO) signaling in endothelial cells. Although both coding and noncoding mutations with phenotypic effects have been identified in the human DHFR gene, no mouse model is currently available to study the consequences of perturbing DHFR in vivo. In order to identify genes involved in definitive hematopoiesis, we performed a forward genetic screen and produced a mouse line, here referred to as Orana, with a point mutation in the Dhfr locus leading to a Thr136Ala substitution in the DHFR protein. Homozygote Orana mice initiate definitive hematopoiesis, but expansion of progenitors in the fetal liver is compromised, and the animals die between embryonic day 13.5 (E13.5) and E14.5. Heterozygote Orana mice survive to adulthood but have tissue-specific alterations in folate abundance and distribution, perturbed stress erythropoiesis, and impaired endothelium-dependent relaxation of the aorta consistent with the role of DHFR in regulating NO signaling. Orana mice provide insight into the dual roles of DHFR and are a useful model for investigating the role of environmental and dietary factors in the context of vascular defects caused by altered NO signaling.


Leukemia | 2018

A four-gene LincRNA expression signature predicts risk in multiple cohorts of acute myeloid leukemia patients

Dominik Beck; Jai Thoms; Cintia Palu; Tobias Herold; Anushi Shah; Jake Olivier; Lies Boelen; Yizhou Huang; Diego Chacon; Alex Brown; Milena Babic; Chris Hahn; Michelle Perugini; Xiaobo Zhou; Brian J. P. Huntly; Adrian Schwarzer; J-H Klusmann; Wolfgang E. Berdel; Bernhard Wörmann; T. Büchner; Wolfgang Hiddemann; Stefan K. Bohlander; L. B. To; Hamish S. Scott; Ian D. Lewis; Richard J. D'Andrea; Jwh Wong; John E. Pimanda

Prognostic gene expression signatures have been proposed as clinical tools to clarify therapeutic options in acute myeloid leukemia (AML). However, these signatures rely on measuring large numbers of genes and often perform poorly when applied to independent cohorts or those with older patients. Long intergenic non-coding RNAs (lincRNAs) are emerging as important regulators of cell identity and oncogenesis, but knowledge of their utility as prognostic markers in AML is limited. Here we analyze transcriptomic data from multiple cohorts of clinically annotated AML patients and report that (i) microarrays designed for coding gene expression can be repurposed to yield robust lincRNA expression data, (ii) some lincRNA genes are located in close proximity to hematopoietic coding genes and show strong expression correlations in AML, (iii) lincRNA gene expression patterns distinguish cytogenetic and molecular subtypes of AML, (iv) lincRNA signatures composed of three or four genes are independent predictors of clinical outcome and further dichotomize survival in European Leukemia Net (ELN) risk groups and (v) an analytical tool based on logistic regression analysis of quantitative PCR measurement of four lincRNA genes (LINC4) can be used to determine risk in AML.


Development | 2018

A novel role for Lyl1 in primitive erythropoiesis

Sung K. Chiu; Jesslyn Saw; Yizhou Huang; Stefan E. Sonderegger; Nicholas C. Wong; David R. Powell; Dominic Beck; John E. Pimanda; Cedric Tremblay; David J. Curtis

ABSTRACT Stem cell leukemia (Scl or Tal1) and lymphoblastic leukemia 1 (Lyl1) encode highly related members of the basic helix-loop-helix family of transcription factors that are co-expressed in the erythroid lineage. Previous studies have suggested that Scl is essential for primitive erythropoiesis. However, analysis of single-cell RNA-seq data of early embryos showed that primitive erythroid cells express both Scl and Lyl1. Therefore, to determine whether Lyl1 can function in primitive erythropoiesis, we crossed conditional Scl knockout mice with mice expressing a Cre recombinase under the control of the Epo receptor, active in erythroid progenitors. Embryos with 20% expression of Scl from E9.5 survived to adulthood. However, mice with reduced expression of Scl and absence of Lyl1 (double knockout; DKO) died at E10.5 because of progressive loss of erythropoiesis. Gene expression profiling of DKO yolk sacs revealed loss of Gata1 and many of the known target genes of the SCL-GATA1 complex. ChIP-seq analyses in a human erythroleukemia cell line showed that LYL1 exclusively bound a small subset of SCL targets including GATA1. Together, these data show for the first time that Lyl1 can maintain primitive erythropoiesis. Summary: Using an erythroid-specific knockout of Scl has shown that Lyl1 can maintain yolk sac erythropoiesis by activation of common target genes including Gata1.


Clinical Cancer Research | 2018

Disruption of a −35 kb Enhancer Impairs CTCF Binding and MLH1 Expression in Colorectal Cells

Qing Liu; Julie A.I. Thoms; Andrea C. Nunez; Yizhou Huang; Kathy Knezevic; Deborah Packham; Rebecca C. Poulos; Rachel Williams; Dominik Beck; Nicholas J. Hawkins; Robyn L. Ward; Jason Wong; Luke B. Hesson; Mathew A. Sloane; John E. Pimanda

Purpose: MLH1 is a major tumor suppressor gene involved in the pathogenesis of Lynch syndrome and various sporadic cancers. Despite their potential pathogenic importance, genomic regions capable of regulating MLH1 expression over long distances have yet to be identified. Experimental Design: Here, we use chromosome conformation capture (3C) to screen a 650-kb region flanking the MLH1 locus to identify interactions between the MLH1 promoter and distal regions in MLH1-expressing and nonexpressing cells. Putative enhancers were functionally validated using luciferase reporter assays, chromatin immunoprecipitation, and CRISPR-Cas9–mediated deletion of endogenous regions. To evaluate whether germline variants in the enhancer might contribute to impaired MLH1 expression in patients with suspected Lynch syndrome, we also screened germline DNA from a cohort of 74 patients with no known coding mutations or epimutations at the MLH1 promoter. Results: A 1.8-kb DNA fragment, 35 kb upstream of the MLH1 transcription start site enhances MLH1 gene expression in colorectal cells. The enhancer was bound by CTCF and CRISPR-Cas9–mediated deletion of a core binding region impairs endogenous MLH1 expression. A total of 5.4% of suspected Lynch syndrome patients have a rare single-nucleotide variant (G > A; rs143969848; 2.5% in gnomAD European, non-Finnish) within a highly conserved CTCF-binding motif, which disrupts enhancer activity in SW620 colorectal carcinoma cells. Conclusions: A CTCF-bound region within the MLH1-35 enhancer regulates MLH1 expression in colorectal cells and is worthy of scrutiny in future genetic screening strategies for suspected Lynch syndrome associated with loss of MLH1 expression. Clin Cancer Res; 24(18); 4602–11. ©2018 AACR.


Cell Reports | 2017

Integrative Genomics Identifies the Molecular Basis of Resistance to Azacitidine Therapy in Myelodysplastic Syndromes

Ashwin Unnikrishnan; Elli Papaemmanuil; Dominik Beck; Nandan Deshpande; Arjun Verma; Ashu Kumari; Petter S. Woll; Laura A. Richards; Kathy Knezevic; Vashe Chandrakanthan; Julie A.I. Thoms; Melinda L. Tursky; Yizhou Huang; Zara Ali; Jake Olivier; Sally Galbraith; Austin Kulasekararaj; Magnus Tobiasson; Mohsen Karimi; Andrea Pellagatti; Susan R. Wilson; Robert Lindeman; Boris Young; Raj Ramakrishna; Christopher Arthur; Richard Stark; Philip Crispin; Jennifer Curnow; Pauline Warburton; Fernando Roncolato

Collaboration


Dive into the Yizhou Huang's collaboration.

Top Co-Authors

Avatar

John E. Pimanda

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Dominik Beck

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Jason Wong

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Julie A.I. Thoms

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Kathy Knezevic

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Ashwin Unnikrishnan

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Jake Olivier

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Melinda L. Tursky

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Vashe Chandrakanthan

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Karen L. MacKenzie

University of New South Wales

View shared research outputs
Researchain Logo
Decentralizing Knowledge