Network


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

Hotspot


Dive into the research topics where John E. Pimanda is active.

Publication


Featured researches published by John E. Pimanda.


Cell Stem Cell | 2010

Combinatorial Transcriptional Control In Blood Stem/Progenitor Cells: Genome-wide Analysis of Ten Major Transcriptional Regulators

Nicola K. Wilson; Samuel D. Foster; Xiaonan Wang; Kathy Knezevic; Judith Schütte; Polynikis Kaimakis; Paulina M. Chilarska; Sarah Kinston; Willem H. Ouwehand; Elaine Dzierzak; John E. Pimanda; Marella de Bruijn; Berthold Göttgens

Combinatorial transcription factor (TF) interactions control cellular phenotypes and, therefore, underpin stem cell formation, maintenance, and differentiation. Here, we report the genome-wide binding patterns and combinatorial interactions for ten key regulators of blood stem/progenitor cells (SCL/TAL1, LYL1, LMO2, GATA2, RUNX1, MEIS1, PU.1, ERG, FLI-1, and GFI1B), thus providing the most comprehensive TF data set for any adult stem/progenitor cell type to date. Genome-wide computational analysis of complex binding patterns, followed by functional validation, revealed the following: first, a previously unrecognized combinatorial interaction between a heptad of TFs (SCL, LYL1, LMO2, GATA2, RUNX1, ERG, and FLI-1). Second, we implicate direct protein-protein interactions between four key regulators (RUNX1, GATA2, SCL, and ERG) in stabilizing complex binding to DNA. Third, Runx1(+/-)::Gata2(+/-) compound heterozygous mice are not viable with severe hematopoietic defects at midgestation. Taken together, this study demonstrates the power of genome-wide analysis in generating novel functional insights into the transcriptional control of stem and progenitor cells.


Cell Stem Cell | 2011

Adult Cardiac-Resident MSC-like Stem Cells with a Proepicardial Origin

James J.H. Chong; Vashe Chandrakanthan; Munira Xaymardan; Naisana S. Asli; Joan Li; Ishtiaq Ahmed; Corey Heffernan; Mary K. Menon; Christopher J. Scarlett; Amirsalar Rashidianfar; Christine Biben; Hans Zoellner; Emily K. Colvin; John E. Pimanda; Andrew V. Biankin; Bin Zhou; William T. Pu; Owen W.J. Prall; Richard P. Harvey

Colony-forming units - fibroblast (CFU-Fs), analogous to those giving rise to bone marrow (BM) mesenchymal stem cells (MSCs), are present in many organs, although the relationship between BM and organ-specific CFU-Fs in homeostasis and tissue repair is unknown. Here we describe a population of adult cardiac-resident CFU-Fs (cCFU-Fs) that occupy a perivascular, adventitial niche and show broad trans-germ layer potency in vitro and in vivo. CRE lineage tracing and embryo analysis demonstrated a proepicardial origin for cCFU-Fs. Furthermore, in BM transplantation chimeras, we found no interchange between BM and cCFU-Fs after aging, myocardial infarction, or BM stem cell mobilization. BM and cardiac and aortic CFU-Fs had distinct CRE lineage signatures, indicating that they arise from different progenitor beds during development. These diverse origins for CFU-Fs suggest an underlying basis for differentiation biases seen in different CFU-F populations, and could also influence their capacity for participating in tissue repair.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Gata2, Fli1, and Scl form a recursively wired gene-regulatory circuit during early hematopoietic development

John E. Pimanda; Katrin Ottersbach; Kathy Knezevic; Sarah Kinston; Wan Y I Chan; Nicola K. Wilson; Josette Renée Landry; Andrew Wood; Anja Kolb-Kokocinski; Anthony R. Green; David Tannahill; Georges Lacaud; Valerie Kouskoff; Berthold Göttgens

Conservation of the vertebrate body plan has been attributed to the evolutionary stability of gene-regulatory networks (GRNs). We describe a regulatory circuit made up of Gata2, Fli1, and Scl/Tal1 and their enhancers, Gata2-3, Fli1+12, and Scl+19, that operates during specification of hematopoiesis in the mouse embryo. We show that the Fli1+12 enhancer, like the Gata2-3 and Scl+19 enhancers, targets hematopoietic stem cells (HSCs) and relies on a combination of Ets, Gata, and E-Box motifs. We show that the Gata2-3 enhancer also uses a similar cluster of motifs and that Gata2, Fli1, and Scl are expressed in embryonic day-11.5 dorsal aorta where HSCs originate and in fetal liver where they multiply. The three HSC enhancers in these tissues and in ES cell-derived hemangioblast equivalents are bound by each of these transcription factors (TFs) and form a fully connected triad that constitutes a previously undescribed example of both this network motif in mammalian development and a GRN kernel operating during the specification of a mammalian stem cell.


eLife | 2014

Origins and functional consequences of somatic mitochondrial DNA mutations in human cancer.

Young Seok Ju; Ludmil B. Alexandrov; Moritz Gerstung; Inigo Martincorena; Serena Nik-Zainal; Manasa Ramakrishna; Helen Davies; Elli Papaemmanuil; Gunes Gundem; Adam Shlien; Niccolo Bolli; Sam Behjati; Patrick Tarpey; Jyoti Nangalia; C E Massie; Adam Butler; J Teague; George S. Vassiliou; Anthony R. Green; M Q Du; Ashwin Unnikrishnan; John E. Pimanda; Bin Tean Teh; Nikhil C. Munshi; Mel Greaves; Paresh Vyas; Adel K. El-Naggar; Thomas Santarius; V P Collins; Richard Grundy

Recent sequencing studies have extensively explored the somatic alterations present in the nuclear genomes of cancers. Although mitochondria control energy metabolism and apoptosis, the origins and impact of cancer-associated mutations in mtDNA are unclear. In this study, we analyzed somatic alterations in mtDNA from 1675 tumors. We identified 1907 somatic substitutions, which exhibited dramatic replicative strand bias, predominantly C > T and A > G on the mitochondrial heavy strand. This strand-asymmetric signature differs from those found in nuclear cancer genomes but matches the inferred germline process shaping primate mtDNA sequence content. A number of mtDNA mutations showed considerable heterogeneity across tumor types. Missense mutations were selectively neutral and often gradually drifted towards homoplasmy over time. In contrast, mutations resulting in protein truncation undergo negative selection and were almost exclusively heteroplasmic. Our findings indicate that the endogenous mutational mechanism has far greater impact than any other external mutagens in mitochondria and is fundamentally linked to mtDNA replication. DOI: http://dx.doi.org/10.7554/eLife.02935.001


Cancer Cell | 2014

Myelodysplastic Syndromes Are Propagated by Rare and Distinct Human Cancer Stem Cells In Vivo.

Petter S. Woll; Una Kjällquist; Onima Chowdhury; Helen Doolittle; David C. Wedge; Supat Thongjuea; Mtakai Ngara; Kristina Anderson; Qiaolin Deng; Adam Mead; L Stenson; Alice Giustacchini; Eleni Giannoulatou; Stephen Taylor; Mohsen Karimi; Christian Scharenberg; Teresa Mortera-Blanco; Iain C Macaulay; Sally Ann Clark; Ingunn Dybedal; Dag Josefsen; Pierre Fenaux; Peter Hokland; Mette Holm; Mario Cazzola; Luca Malcovati; Sudhir Tauro; David G. Bowen; Jacqueline Boultwood; Andrea Pellagatti

Evidence for distinct human cancer stem cells (CSCs) remains contentious and the degree to which different cancer cells contribute to propagating malignancies in patients remains unexplored. In low- to intermediate-risk myelodysplastic syndromes (MDS), we establish the existence of rare multipotent MDS stem cells (MDS-SCs), and their hierarchical relationship to lineage-restricted MDS progenitors. All identified somatically acquired genetic lesions were backtracked to distinct MDS-SCs, establishing their distinct MDS-propagating function in vivo. In isolated del(5q)-MDS, acquisition of del(5q) preceded diverse recurrent driver mutations. Sequential analysis in del(5q)-MDS revealed genetic evolution in MDS-SCs and MDS-progenitors prior to leukemic transformation. These findings provide definitive evidence for rare human MDS-SCs in vivo, with extensive implications for the targeting of the cells required and sufficient for MDS-propagation.


Blood | 2010

Epigenetic silencing of BIM in glucocorticoid poor-responsive pediatric acute lymphoblastic leukemia, and its reversal by histone deacetylase inhibition.

Petra S. Bachmann; Rocco Piazza; Mary E. Janes; Nicholas C. Wong; Carwyn Davies; A Mogavero; Vivek A. Bhadri; Barbara Szymanska; Greta Geninson; Vera Magistroni; Giovanni Cazzaniga; Andrea Biondi; Diego Miranda-Saavedra; Berthold Göttgens; Richard Saffery; Jeffrey M. Craig; Glenn M. Marshall; Carlo Gambacorti-Passerini; John E. Pimanda; Richard B. Lock

Glucocorticoids play a critical role in the therapy of lymphoid malignancies, including pediatric acute lymphoblastic leukemia (ALL), although the mechanisms underlying cellular resistance remain unclear. We report glucocorticoid resistance attributable to epigenetic silencing of the BIM gene in pediatric ALL biopsies and xenografts established in immune-deficient mice from direct patient explants as well as a therapeutic approach to reverse resistance in vivo. Glucocorticoid resistance in ALL xenografts was consistently associated with failure to up-regulate BIM expression after dexamethasone exposure despite confirmation of a functional glucocorticoid receptor. Although a comprehensive assessment of BIM CpG island methylation revealed no consistent changes, glucocorticoid resistance in xenografts and patient biopsies significantly correlated with decreased histone H3 acetylation. Moreover, the histone deacetylase inhibitor vorinostat relieved BIM repression and exerted synergistic antileukemic efficacy with dexamethasone in vitro and in vivo. These findings provide a novel therapeutic strategy to reverse glucocorticoid resistance and improve outcome for high-risk pediatric ALL.


Proceedings of the National Academy of Sciences of the United States of America | 2007

The SCL transcriptional network and BMP signaling pathway interact to regulate RUNX1 activity

John E. Pimanda; Ian J. Donaldson; Marella de Bruijn; Sarah Kinston; Kathy Knezevic; Liz Huckle; Sandie Piltz; Josette Renée Landry; Anthony R. Green; David Tannahill; Berthold Göttgens

Hematopoietic stem cell (HSC) development is regulated by several signaling pathways and a number of key transcription factors, which include Scl/Tal1, Runx1, and members of the Smad family. However, it remains unclear how these various determinants interact. Using a genome-wide computational screen based on the well characterized Scl +19 HSC enhancer, we have identified a related Smad6 enhancer that also targets expression to blood and endothelial cells in transgenic mice. Smad6, Bmp4, and Runx1 transcripts are concentrated along the ventral aspect of the E10.5 dorsal aorta in the aorta–gonad–mesonephros region from which HSCs originate. Moreover, Smad6, an inhibitor of Bmp4 signaling, binds and inhibits Runx1 activity, whereas Smad1, a positive mediator of Bmp4 signaling, transactivates the Runx1 promoter. Taken together, our results integrate three key determinants of HSC development; the Scl transcriptional network, Runx1 activity, and the Bmp4/Smad signaling pathway.


Cancer Research | 2005

The proto-oncogene ERG in megakaryoblastic leukemias.

Liat Rainis; Tsutomu Toki; John E. Pimanda; Ester Rosenthal; Keren Machol; Sabine Strehl; Berthold Göttgens; Etsuro Ito; Shai Izraeli

Aneuploidy is one of the hallmarks of cancer. Acquired additions of chromosome 21 are a common finding in leukemias, suggesting a contributory role to leukemogenesis. About 10% of patients with a germ line trisomy 21 (Down syndrome) are born with transient megakaryoblastic leukemia. We and others have shown acquired mutations in the X chromosome gene GATA1 in all these cases. The gene or genes on chromosome 21 whose overexpression promote the megakaryoblastic phenotype are presently unknown. We propose that ERG, an Ets transcription factor situated on chromosome 21, is one such candidate. We show that ERG is expressed in hematopoietic stem cells, megakaryoblastic cell lines, and in primary leukemic cells from Down syndrome patients. ERG expression is induced upon megakaryocytic differentiation of the erythroleukemia cell lines K562 and UT-7, and forced expression of ERG in K562 cells induces erythroid to megakaryoblastic phenotypic switch. We also show that ERG activates the gpIb megakaryocytic promoter and binds the gpIIb promoter in vivo. Furthermore, both ERG and ETS2 bind in vivo the hematopoietic enhancer of SCL/TAL1, a key regulator of hematopoietic stem cell and megakaryocytic development. We propose that trisomy 21 facilitates the occurrence of megakaryoblastic leukemias through a shift toward the megakaryoblastic lineage caused by the excess expression of ERG, and possibly by other chromosome 21 genes, such as RUNX1 and ETS2, in hematopoietic progenitor cells, coupled with a differentiation arrest caused by the acquisition of mutations in GATA1.


Blood | 2008

Runx genes are direct targets of Scl/Tal1 in the yolk sac and fetal liver

Josette-Renee Landry; Sarah Kinston; Kathy Knezevic; Marella de Bruijn; Nicola K. Wilson; Wade T Nottingham; Michael Peitz; Frank Edenhofer; John E. Pimanda; Katrin Ottersbach; Berthold Göttgens

Transcription factors such as Scl/Tal1, Lmo2, and Runx1 are essential for the development of hematopoietic stem cells (HSCs). However, the precise mechanisms by which these factors interact to form transcriptional networks, as well as the identity of the genes downstream of these regulatory cascades, remain largely unknown. To this end, we generated an Scl(-/-) yolk sac cell line to identify candidate Scl target genes by global expression profiling after reintroduction of a TAT-Scl fusion protein. Bioinformatics analysis resulted in the identification of 9 candidate Scl target transcription factor genes, including Runx1 and Runx3. Chromatin immunoprecipitation confirmed that both Runx genes are direct targets of Scl in the fetal liver and that Runx1 is also occupied by Scl in the yolk sac. Furthermore, binding of an Scl-Lmo2-Gata2 complex was demonstrated to occur on the regions flanking the conserved E-boxes of the Runx1 loci and was shown to transactivate the Runx1 element. Together, our data provide a key component of the transcriptional network of early hematopoiesis by identifying downstream targets of Scl that can explain key aspects of the early Scl(-/-) phenotype.


Cancer Research | 2013

Cancerous Inhibitor of Protein Phosphatase 2A, an Emerging Human Oncoprotein and a Potential Cancer Therapy Target

Anchit Khanna; John E. Pimanda; Jukka Westermarck

Protein phosphatase 2A (PP2A) complexes function as tumor suppressors by inhibiting the activity of several critical oncogenic signaling pathways. Consequently, inhibition of the PP2A phosphatase activity is one of many prerequisites for the transformation of normal human cells into cancerous cells. However, mechanisms for PP2A inactivation in human cancers are poorly understood. The aberrant expression of cancerous inhibitor of protein phosphatase 2A (CIP2A), a recently identified endogenous PP2A inhibitor in malignant cells, is one such mechanism. Various independent studies have validated CIP2As role in promoting tumor growth and resistance to apoptosis and senescence-inducing therapies. Notably, high CIP2A expression predicts poor patient prognosis in several human cancer types. Among the oncogenic proteins dephosphorylated by PP2A, the MYC oncoprotein, which is phosphorylated at serine 62, has surfaced as a marker for the oncogenic activity of CIP2A. The positive-feedback loop between CIP2A and MYC augments the activity of MYC in cancer cells. In addition, CIP2A promotes the phosphorylation and activity of additional oncoproteins, including E2F1 and AKT. However, CIP2A is not essential for normal mouse growth and development. These findings indicate that CIP2A is a novel anticancer target based on PP2A reactivation and inhibition of the oncogenic activity of its downstream effectors. The potential approaches and feasibility of targeting CIP2A are discussed here.

Collaboration


Dive into the John E. Pimanda's collaboration.

Top Co-Authors

Avatar

Kathy Knezevic

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
Top Co-Authors

Avatar

Julie A.I. Thoms

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

Vashe Chandrakanthan

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar

Luke B. Hesson

University of New South Wales

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yizhou Huang

University of New South Wales

View shared research outputs
Researchain Logo
Decentralizing Knowledge