keyword
https://read.qxmd.com/read/33152396/runx1-eto-and-mutant-kit-both-contribute-to-programming-the-transcriptional-and-chromatin-landscape-in-t-8-21-acute-myeloid-leukemia
#21
JOURNAL ARTICLE
Paulynn Suyin Chin, Salam A Assi, Anetta Ptasinska, Maria Rosaria Imperato, Peter N Cockerill, Constanze Bonifer
Acute myeloid leukemia development occurs in a stepwise fashion whereby an original driver mutation is followed by additional mutations. The first type of mutations tends to be in genes encoding members of the epigenetic/transcription regulatory machinery (i.e., RUNX1, DNMT3A, TET2), while the secondary mutations often involve genes encoding members of signaling pathways that cause uncontrolled growth of such cells such as the growth factor receptors c-KIT of FLT3. Patients usually present with both types of mutations, but it is currently unclear how both mutational events shape the epigenome in developing AML cells...
December 2020: Experimental Hematology
https://read.qxmd.com/read/32460028/expression-of-runx1-eto-rapidly-alters-the-chromatin-landscape-and-growth-of-early-human-myeloid-precursor-cells
#22
JOURNAL ARTICLE
Monica Nafria, Peter Keane, Elizabeth S Ng, Edouard G Stanley, Andrew G Elefanty, Constanze Bonifer
Acute myeloid leukemia (AML) is a hematopoietic malignancy caused by recurrent mutations in genes encoding transcriptional, chromatin, and/or signaling regulators. The t(8;21) translocation generates the aberrant transcription factor RUNX1-ETO (RUNX1-RUNX1T1), which by itself is insufficient to cause disease. t(8;21) AML patients show extensive chromatin reprogramming and have acquired additional mutations. Therefore, the genomic and developmental effects directly and solely attributable to RUNX1-ETO expression are unclear...
May 26, 2020: Cell Reports
https://read.qxmd.com/read/32299907/runx1-evi1-disrupts-lineage-determination-and-the-cell-cycle-by-interfering-with-runx1-and-evi1-driven-gene-regulatory-networks
#23
JOURNAL ARTICLE
Sophie G Kellaway, Peter Keane, Ella Kennett, Constanze Bonifer
Hematological malignancies are characterised by a block in differentiation, which in many cases is caused by recurrent mutations affecting the activity of hematopoietic transcription factors. RUNX1-EVI1 is a fusion protein formed by the t(3;21) translocation linking two transcription factors required for normal hematopoiesis. RUNX1-EVI1 expression is found in myelodysplastic syndrome, secondary acute myeloid leukemia, and blast crisis of chronic myeloid leukemia; with clinical outcomes being worse than in patients with RUNX1-ETO, RUNX1 or EVI1 mutations alone...
April 16, 2020: Haematologica
https://read.qxmd.com/read/32029705/the-runx1-eto-target-gene-rassf2-suppresses-t-8-21-aml-development-and-regulates-rac-gtpase-signaling
#24
JOURNAL ARTICLE
Samuel A Stoner, Katherine Tin Heng Liu, Elizabeth T Andrews, Mengdan Liu, Kei-Ichiro Arimoto, Ming Yan, Amanda G Davis, Stephanie Weng, Michelle Dow, Su Xian, Russell C DeKelver, Hannah Carter, Dong-Er Zhang
Large-scale chromosomal translocations are frequent oncogenic drivers in acute myeloid leukemia (AML). These translocations often occur in critical transcriptional/epigenetic regulators and contribute to malignant cell growth through alteration of normal gene expression. Despite this knowledge, the specific gene expression alterations that contribute to the development of leukemia remain incompletely understood. Here, through characterization of transcriptional regulation by the RUNX1-ETO fusion protein, we have identified Ras-association domain family member 2 (RASSF2) as a critical gene that is aberrantly transcriptionally repressed in t(8;21)-associated AML...
February 6, 2020: Blood Cancer Journal
https://read.qxmd.com/read/31826021/stable-depletion-of-runx1-eto-in-kasumi-1-cells-induces-expression-and-enhanced-proteolytic-activity-of-cathepsin-g-and-neutrophil-elastase
#25
JOURNAL ARTICLE
Caroline Schoenherr, Katharina Wohlan, Iris Dallmann, Andreas Pich, Jan Hegermann, Arnold Ganser, Denise Hilfiker-Kleiner, Olaf Heidenreich, Michaela Scherr, Matthias Eder
The oncogenic fusion protein RUNX1-ETO is a product of the t(8;21) translocation and consists of the hematopoietic transcriptional master regulator RUNX1 and the repressor ETO. RUNX1-ETO is found in 10-15% of acute myeloid leukemia and interferes with the expression of genes that are essential for myeloid differentiation. The neutrophil serine protease Cathepsin G is one of the genes suppressed by RUNX1-ETO, but little is known about its impact on the regulation of other lysosomal proteases. By lentiviral transduction of the t(8;21) positive cell line Kasumi-1 with an RUNX1-ETO specific shRNA, we analyzed long-term effects of stable RUNX1-ETO silencing on cellular phenotypes and target gene expression...
2019: PloS One
https://read.qxmd.com/read/31817911/core-binding-factor-leukemia-chromatin-remodeling-moves-towards-oncogenic-transcription
#26
REVIEW
Alessandro Beghini
Acute myeloid leukemia (AML), the most common acute leukemia in adults, is a heterogeneous malignant clonal disorder arising from multipotent hematopoietic progenitor cells characterized by genetic and concerted epigenetic aberrations. Core binding factor-Leukemia (CBFL) is characterized by the recurrent reciprocal translocations t(8;21)(q22;q22) or inv(16)(p13;q22) that, expressing the distinctive RUNX1-RUNX1T1 (also known as Acute myeloid leukemia1-eight twenty-one, AML1-ETO or RUNX1/ETO) or CBFB-MYH11 (also known as CBFβ-ΣMMHX) translocation product respectively, disrupt the essential hematopoietic function of the CBF...
December 7, 2019: Cancers
https://read.qxmd.com/read/31722222/runx1-eto-depletion-in-t-8-21-aml-leads-to-c-ebp%C3%AE-and-ap-1-mediated-alterations-in-enhancer-promoter-interaction
#27
Anetta Ptasinska, Anna Pickin, Salam A Assi, Paulynn Suyin Chin, Luke Ames, Roberto Avellino, Stephan Gröschel, Ruud Delwel, Peter N Cockerill, Cameron S Osborne, Constanze Bonifer
No abstract text is available yet for this article.
November 12, 2019: Cell Reports
https://read.qxmd.com/read/31533028/runx1-eto-depletion-in-t-8-21-aml-leads-to-c-ebp%C3%AE-and-ap-1-mediated-alterations-in-enhancer-promoter-interaction
#28
JOURNAL ARTICLE
Anetta Ptasinska, Anna Pickin, Salam A Assi, Paulynn Suyin Chin, Luke Ames, Roberto Avellino, Stefan Gröschel, Ruud Delwel, Peter N Cockerill, Cameron S Osborne, Constanze Bonifer
Acute myeloid leukemia (AML) is associated with mutations in transcriptional and epigenetic regulator genes impairing myeloid differentiation. The t(8;21)(q22;q22) translocation generates the RUNX1-ETO fusion protein, which interferes with the hematopoietic master regulator RUNX1. We previously showed that the maintenance of t(8;21) AML is dependent on RUNX1-ETO expression. Its depletion causes extensive changes in transcription factor binding, as well as gene expression, and initiates myeloid differentiation...
September 17, 2019: Cell Reports
https://read.qxmd.com/read/30991028/the-oncogenic-transcription-factor-runx1-eto-corrupts-cell-cycle-regulation-to-drive-leukemic-transformation
#29
Natalia Martinez-Soria, Lynsey McKenzie, Julia Draper, Anetta Ptasinska, Hasan Issa, Sandeep Potluri, Helen J Blair, Anna Pickin, Asmida Isa, Paulynn Suyin Chin, Ricky Tirtakusuma, Daniel Coleman, Sirintra Nakjang, Salam Assi, Victoria Forster, Mojgan Reza, Ed Law, Philip Berry, Dorothee Mueller, Cameron Osborne, Alex Elder, Simon N Bomken, Deepali Pal, James M Allan, Gareth J Veal, Peter N Cockerill, Christian Wichmann, Josef Vormoor, Georges Lacaud, Constanze Bonifer, Olaf Heidenreich
No abstract text is available yet for this article.
April 15, 2019: Cancer Cell
https://read.qxmd.com/read/30792202/runx1-inhibits-proliferation-and-induces-apoptosis-of-t-8-21-leukemia-cells-via-klf4-mediated-transactivation-of-p57
#30
JOURNAL ARTICLE
Shuang Liu, Yanyan Xing, Wenting Lu, Shouyun Li, Zheng Tian, Haiyan Xing, Kejing Tang, Yingxi Xu, Qing Rao, Min Wang, Jianxiang Wang
RUNX1 is a key transcription factor in hematopoiesis and its disruption is one of the most common aberrations in acute myeloid leukemia. RUNX1 alterations affect its DNA binding capacity and transcriptional activities, leading to the deregulation of transcriptional targets and abnormality in proliferation and differentiation of myeloid cells. Identification of RUNX1 target genes and elucidation of their biological functions are of great importance to find new therapeutic strategies for RUNX1-altered leukemia...
February 21, 2019: Haematologica
https://read.qxmd.com/read/30654457/runx1-eto-attacking-the-epigenome-for-genomic-instable-leukemia
#31
REVIEW
Emiel van der Kouwe, Philipp Bernhard Staber
Oncogenic fusion protein RUNX1-ETO is the product of the t(8;21) translocation, responsible for the most common cytogenetic subtype of acute myeloid leukemia. RUNX1, a critical transcription factor in hematopoietic development, is fused with almost the entire ETO sequence with the ability to recruit a wide range of repressors. Past efforts in providing a comprehensive picture of the genome-wide localization and the target genes of RUNX1-ETO have been inconclusive in understanding the underlying mechanism by which it deregulates native RUNX1...
January 16, 2019: International Journal of Molecular Sciences
https://read.qxmd.com/read/30637949/the-runx1-eto-fusion-protein-trans-activates-c-kit-expression-by-recruiting-histone-acetyltransferase-p300-on-its-promoter
#32
JOURNAL ARTICLE
Guofeng Chen, Anqi Liu, Yihan Xu, Li Gao, Mengmeng Jiang, Yan Li, Na Lv, Lei Zhou, Lili Wang, Li Yu, Yonghui Li
The oncoprotein RUNX1-ETO is the fusion product of t(8;21)(q22;q22) and constitutes one of the most common genetic alterations in acute myeloid leukemia (AML). Abnormal c-KIT over-expression is considered an independent negative prognostic factor for relapse and survival in t(8;21) AML patients. However, the molecular mechanism of high c-KIT expression in t(8;21) AML remains unknown. In this study, we detected RUNX1-ETO and c-KIT gene expression in AML-M2 patients and verified the over-expression of c-KIT in t(8;21) AML patients...
January 13, 2019: FEBS Journal
https://read.qxmd.com/read/30341070/runx1-eto-upregulates-ccnd2-to-drive-leukemogenesis
#33
JOURNAL ARTICLE
(no author information available yet)
An RNAi screen identified CCND2 as a RUNX1-ETO target gene required for leukemia maintenance.
October 19, 2018: Cancer Discovery
https://read.qxmd.com/read/30300583/the-oncogenic-transcription-factor-runx1-eto-corrupts-cell-cycle-regulation-to-drive-leukemic-transformation
#34
JOURNAL ARTICLE
Natalia Martinez-Soria, Lynsey McKenzie, Julia Draper, Anetta Ptasinska, Hasan Issa, Sandeep Potluri, Helen J Blair, Anna Pickin, Asmida Isa, Paulynn Suyin Chin, Ricky Tirtakusuma, Daniel Coleman, Sirintra Nakjang, Salam Assi, Victoria Forster, Mojgan Reza, Ed Law, Philip Berry, Dorothee Mueller, Cameron Osborne, Alex Elder, Simon N Bomken, Deepali Pal, James M Allan, Gareth J Veal, Peter N Cockerill, Christian Wichmann, Josef Vormoor, Georges Lacaud, Constanze Bonifer, Olaf Heidenreich
Oncogenic transcription factors such as the leukemic fusion protein RUNX1/ETO, which drives t(8;21) acute myeloid leukemia (AML), constitute cancer-specific but highly challenging therapeutic targets. We used epigenomic profiling data for an RNAi screen to interrogate the transcriptional network maintaining t(8;21) AML. This strategy identified Cyclin D2 (CCND2) as a crucial transmitter of RUNX1/ETO-driven leukemic propagation. RUNX1/ETO cooperates with AP-1 to drive CCND2 expression. Knockdown or pharmacological inhibition of CCND2 by an approved drug significantly impairs leukemic expansion of patient-derived AML cells and engraftment in immunodeficient murine hosts...
October 8, 2018: Cancer Cell
https://read.qxmd.com/read/30093631/compatibility-of-runx1-eto-fusion-protein-modules-driving-cd34-human-progenitor-cell-expansion
#35
JOURNAL ARTICLE
Linping Chen-Wichmann, Marina Shvartsman, Caro Preiss, Colin Hockings, Roland Windisch, Enric Redondo Monte, Georg Leubolt, Karsten Spiekermann, Jörn Lausen, Christian Brendel, Manuel Grez, Philipp A Greif, Christian Wichmann
Chromosomal translocations represent frequent events in leukemia. In t(8;21)+ acute myeloid leukemia, RUNX1 is fused to nearly the entire ETO protein, which contains four conserved nervy homology regions, NHR1-4. Furthermore RUNX1/ETO interacts with ETO-homologous proteins via NHR2, thereby multiplying NHR domain contacts. As shown recently, RUNX1/ETO retains oncogenic activity upon either deletion of the NHR3 + 4 N-CoR/SMRT interaction domain or substitution of the NHR2 tetramer domain. Thus, we aimed to clarify the specificities of the NHR domains...
August 9, 2018: Oncogene
https://read.qxmd.com/read/30013160/mutant-asxl1-cooperates-with-bap1-to-promote-myeloid-leukaemogenesis
#36
JOURNAL ARTICLE
Shuhei Asada, Susumu Goyama, Daichi Inoue, Shiori Shikata, Reina Takeda, Tsuyoshi Fukushima, Taishi Yonezawa, Takeshi Fujino, Yasutaka Hayashi, Kimihito Cojin Kawabata, Tomofusa Fukuyama, Yosuke Tanaka, Akihiko Yokoyama, Satoshi Yamazaki, Hiroko Kozuka-Hata, Masaaki Oyama, Shinya Kojima, Masahito Kawazu, Hiroyuki Mano, Toshio Kitamura
ASXL1 mutations occur frequently in myeloid neoplasms and are associated with poor prognosis. However, the mechanisms by which mutant ASXL1 induces leukaemogenesis remain unclear. In this study, we report mutually reinforcing effects between a C-terminally truncated form of mutant ASXL1 (ASXL1-MT) and BAP1 in promoting myeloid leukaemogenesis. BAP1 expression results in increased monoubiquitination of ASXL1-MT, which in turn increases the catalytic function of BAP1. This hyperactive ASXL1-MT/BAP1 complex promotes aberrant myeloid differentiation of haematopoietic progenitor cells and accelerates RUNX1-ETO-driven leukaemogenesis...
July 16, 2018: Nature Communications
https://read.qxmd.com/read/29721072/glucocorticoids-inhibit-oncogenic-runx1-eto-in-acute-myeloid-leukemia-with-chromosome-translocation-t-8-21
#37
JOURNAL ARTICLE
Lianghao Lu, Yefei Wen, Yuan Yao, Fengju Chen, Guohui Wang, Fangrui Wu, Jingyu Wu, Padmini Narayanan, Michele Redell, Qianxing Mo, Yongcheng Song
Acute myeloid leukemia (AML) is a major blood cancer with poor prognosis. New therapies are needed to target oncogene-driven leukemia stem cells, which account for relapse and resistance. Chromosome translocation t(8;21), which produces RUNX1-ETO (R-E) fusion oncoprotein, is found in ~13% AML. R-E dominance negatively inhibits global gene expression regulated by RUNX1, a master transcription factor for hematopoiesis, causing increased self-renewal and blocked cell differentiation of hematopoietic progenitor cells, and eventually leukemia initiation...
2018: Theranostics
https://read.qxmd.com/read/29674496/gfi1-is-required-for-runx1-eto-positive-acute-myeloid-leukemia
#38
LETTER
Anna E Marneth, Lacramioara Botezatu, Judith M Hönes, Jimmy C L Israël, Judith Schütte, Lothar Vassen, Robert F Lams, Saskia M Bergevoet, Laura Groothuis, Amit Mandoli, Joost H A Martens, Gerwin Huls, Joop H Jansen, Ulrich Dührsen, Tobias Berg, Tarik Möröy, Christian Wichmann, Mia-Chia Lo, Dong-Er Zhang, Bert A van der Reijden, Cyrus Khandanpour
No abstract text is available yet for this article.
September 2018: Haematologica
https://read.qxmd.com/read/29431622/c-ebp%C3%AE-overrides-epigenetic-reprogramming-by-oncogenic-transcription-factors-in-acute-myeloid-leukemia
#39
JOURNAL ARTICLE
Justin Loke, Paulynn Suyin Chin, Peter Keane, Anna Pickin, Salam A Assi, Anetta Ptasinska, Maria Rosaria Imperato, Peter N Cockerill, Constanze Bonifer
Acute myeloid leukemia (AML) is a heterogeneous disease caused by recurrent mutations in the transcription regulatory machinery, resulting in abnormal growth and a block in differentiation. One type of recurrent mutations affects RUNX1 , which is subject to mutations and translocations, the latter giving rise to fusion proteins with aberrant transcriptional activities. We recently compared the mechanism by which the products of the t(8;21) and the t(3;21) translocation RUNX1-ETO and RUNX1-EVI1 reprogram the epigenome...
February 13, 2018: Blood Advances
https://read.qxmd.com/read/29105243/multiplex-fusion-gene-testing-in-pediatric-acute-myeloid-leukemia
#40
JOURNAL ARTICLE
Yuka Iijima-Yamashita, Hidemasa Matsuo, Miho Yamada, Takao Deguchi, Nobutaka Kiyokawa, Akira Shimada, Akio Tawa, Hiroyuki Takahashi, Daisuke Tomizawa, Takashi Taga, Akitoshi Kinoshita, Souichi Adachi, Keizo Horibe
BACKGROUND: Gene abnormalities, particularly chromosome rearrangements generating gene fusion, are associated with clinical characteristics and prognosis in pediatric acute myeloid leukemia (AML). Karyotyping is generally performed to enable risk stratification, but the results are not always consistent with those of reverse transcription-polymerase chain reaction (RT-PCR), and more accurate and rapid methods are required. METHODS: A total of 487 samples from de novo AML patients enrolled in the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) AML-05 study (n = 448), and from acute promyelocytic leukemia (APL) patients enrolled in the JPLSG AML-P05 study (n = 39) were available for this investigation...
January 2018: Pediatrics International: Official Journal of the Japan Pediatric Society
keyword
keyword
63142
2
3
Fetch more papers »
Fetching more papers... Fetching...
Remove bar
Read by QxMD icon Read
×

Save your favorite articles in one place with a free QxMD account.

×

Search Tips

Use Boolean operators: AND/OR

diabetic AND foot
diabetes OR diabetic

Exclude a word using the 'minus' sign

Virchow -triad

Use Parentheses

water AND (cup OR glass)

Add an asterisk (*) at end of a word to include word stems

Neuro* will search for Neurology, Neuroscientist, Neurological, and so on

Use quotes to search for an exact phrase

"primary prevention of cancer"
(heart or cardiac or cardio*) AND arrest -"American Heart Association"

We want to hear from doctors like you!

Take a second to answer a survey question.