91 results on '"Arefeh Rouhi"'
Search Results
2. The retinoic acid receptor co-factor NRIP1 is uniquely upregulated and represents a therapeutic target in acute myeloid leukemia with chromosome 3q rearrangements
- Author
-
Sarah Grasedieck, Ariene Cabantog, Liam MacPhee, Junbum Im, Christoph Ruess, Burcu Demir, Nadine Sperb, Frank G. Rücker, Konstanze Döhner, Tobias Herold, Jonathan R. Pollack, Lars Bullinger, Arefeh Rouhi, and Florian Kuchenbauer
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Aberrant expression of Ecotropic Viral Integration Site 1 (EVI1) is a hallmark of acute myeloid leukemia (AML) with inv(3) or t(3;3), which is a disease subtype with especially poor outcome. In studying transcriptomes from AML patients with chromosome 3q rearrangements, we identified a significant upregulation of the Nuclear Receptor Interacting Protein 1 (NRIP1) as well as its adjacent non-coding RNA LOC101927745. Utilizing transcriptomic and epigenomic data from over 900 primary samples from patients as well as genetic and transcriptional engineering approaches, we have identified several mechanisms that can lead to upregulation of NRIP1 in AML. We hypothesize that the LOC101927745 transcription start site harbors a context-dependent enhancer that is bound by EVI1, causing upregulation of NRIP1 in AML with chromosome 3 abnormalities. Furthermore, we showed that NRIP1 knockdown negatively affects the proliferation and survival of 3qrearranged AML cells and increases their sensitivity to all-trans retinoic acid, suggesting that NRIP1 is relevant for the pathogenesis of inv(3)/t(3;3) AML and could serve as a novel therapeutic target in myeloid malignancies with 3q abnormalities.
- Published
- 2021
- Full Text
- View/download PDF
3. The long non-coding RNA Cancer Susceptibility 15 (CASC15) is induced by isocitrate dehydrogenase (IDH) mutations and maintains an immature phenotype in adult acute myeloid leukemia
- Author
-
Sarah Grasedieck, Christoph Ruess, Kathrin Krowiorz, Susanne Lux, Nicole Pochert, Adrian Schwarzer, Jan-Henning Klusmann, Mojca Jongen-Lavrencic, Tobias Herold, Lars Bullinger, Jonathan R. Pollack, Arefeh Rouhi, and Florian Kuchenbauer
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
4. Micro-ribonucleic acid-155 is a direct target of Meis1, but not a driver in acute myeloid leukemia
- Author
-
Edith Schneider, Anna Staffas, Linda Röhner, Erik D. Malmberg, Arghavan Ashouri, Kathrin Krowiorz, Nicole Pochert, Christina Miller, Stella Yuan Wei, Laleh Arabanian, Christian Buske, Hartmut Döhner, Lars Bullinger, Linda Fogelstrand, Michael Heuser, Konstanze Döhner, Ping Xiang, Jens Ruschmann, Oleh I. Petriv, Alireza Heravi-Moussavi, Carl L. Hansen, Martin Hirst, R. Keith Humphries, Arefeh Rouhi, Lars Palmqvist, and Florian Kuchenbauer
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Micro-ribonucleic acid-155 (miR-155) is one of the first described oncogenic miRNAs. Although multiple direct targets of miR-155 have been identified, it is not clear how it contributes to the pathogenesis of acute myeloid leukemia. We found miR-155 to be a direct target of Meis1 in murine Hoxa9/Meis1 induced acute myeloid leukemia. The additional overexpression of miR-155 accelerated the formation of acute myeloid leukemia in Hoxa9 as well as in Hoxa9/Meis1 cells in vivo. However, in the absence or following the removal of miR-155, leukemia onset and progression were unaffected. Although miR-155 accelerated growth and homing in addition to impairing differentiation, our data underscore the pathophysiological relevance of miR-155 as an accelerator rather than a driver of leukemogenesis. This further highlights the complexity of the oncogenic program of Meis1 to compensate for the loss of a potent oncogene such as miR-155. These findings are highly relevant to current and developing approaches for targeting miR-155 in acute myeloid leukemia.
- Published
- 2018
- Full Text
- View/download PDF
5. Circular RNAs of the nucleophosmin (NPM1) gene in acute myeloid leukemia
- Author
-
Susanne Hirsch, Tamara J. Blätte, Sarah Grasedieck, Sibylle Cocciardi, Arefeh Rouhi, Mojca Jongen-Lavrencic, Peter Paschka, Jan Krönke, Verena I. Gaidzik, Hartmut Döhner, Richard F. Schlenk, Florian Kuchenbauer, Konstanze Döhner, Anna Dolnik, and Lars Bullinger
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
In acute myeloid leukemia, there is growing evidence for splicing pattern deregulation, including differential expression of linear splice isoforms of the commonly mutated gene nucleophosmin (NPM1). In this study, we detect circular RNAs of NPM1 and quantify circRNA hsa_circ_0075001 in a cohort of NPM1 wild-type and mutated acute myeloid leukemia (n=46). Hsa_circ_0075001 expression correlates positively with total NPM1 expression, but is independent of the NPM1 mutational status. High versus low hsa_circ_0075001 expression defines patient subgroups characterized by distinct gene expression patterns, such as lower expression of components of the Toll-like receptor signaling pathway in high hsa_circ_0075001 expression cases. Global evaluation of circRNA expression in sorted healthy hematopoietic controls (n=10) and acute myeloid leukemia (n=10) reveals circRNA transcripts for 47.9% of all highly expressed genes. While circRNA expression correlates globally with parental gene expression, we identify hematopoietic differentiation-associated as well as acute myeloid leukemia subgroup-specific circRNA signatures.
- Published
- 2017
- Full Text
- View/download PDF
6. Supplementary Figure S4 from HSP90 Supports Tumor Growth and Angiogenesis through PRKD2 Protein Stabilization
- Author
-
Thomas Seufferlein, Claudia Scholl, Stefan Fröhling, John Koren, Gabriela Chiosis, Johan van Lint, Hans Kestler, Alexander Becher, Felicitas Genze, Arefeh Rouhi, Cornelia Brunner, Kristina Diepold, and Ninel Azoitei
- Abstract
Supplementary Figure S4. HSP90 inhibition impairs tumor growth in vivo in a PRKD2-dependent manner.
- Published
- 2023
7. Supplementary Figure Legend from HSP90 Supports Tumor Growth and Angiogenesis through PRKD2 Protein Stabilization
- Author
-
Thomas Seufferlein, Claudia Scholl, Stefan Fröhling, John Koren, Gabriela Chiosis, Johan van Lint, Hans Kestler, Alexander Becher, Felicitas Genze, Arefeh Rouhi, Cornelia Brunner, Kristina Diepold, and Ninel Azoitei
- Abstract
Supplementary Figure Legend. Legend for Supplementary Figures S1-S7.
- Published
- 2023
8. Elucidating the importance and regulation of key enhancers for human MEIS1 expression
- Author
-
Ping Xiang, Xining Yang, Leo Escano, Ishpreet Dhillon, Edith Schneider, Jack Clemans-Gibbon, Wei Wei, Jasper Wong, Simon Xufeng Wang, Derek Tam, Yu Deng, Eric Yung, Gregg B. Morin, Pamela A. Hoodless, Martin Hirst, Aly Karsan, Florian Kuchenbauer, R. Keith Humphries, and Arefeh Rouhi
- Subjects
Homeodomain Proteins ,Leukemia, Myeloid, Acute ,Cancer Research ,Oncology ,Humans ,Hematology ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Neoplasm Proteins ,Transcription Factors - Abstract
Myeloid ecotropic virus insertion site 1 (MEIS1) is essential for normal hematopoiesis and is a critical factor in the pathogenesis of a large subset of acute myeloid leukemia (AML). Despite the clinical relevance of MEIS1, its regulation is largely unknown. To understand the transcriptional regulatory mechanisms contributing to human MEIS1 expression, we created a knock-in green florescent protein (GFP) reporter system at the endogenous MEIS1 locus in a human AML cell line. Using this model, we have delineated and dissected a critical enhancer region of the MEIS1 locus for transcription factor (TF) binding through in silico prediction in combination with oligo pull-down, mass-spectrometry and knockout analysis leading to the identification of FLI1, an E-twenty-six (ETS) transcription factor, as an important regulator of MEIS1 transcription. We further show direct binding of FLI1 to the MEIS1 locus in human AML cell lines as well as enrichment of histone acetylation in MEIS1-high healthy and leukemic cells. We also observe a positive correlation between high FLI1 transcript levels and worse overall survival in AML patients. Our study expands the role of ETS factors in AML and our model constitutes a feasible tool for a more detailed understanding of transcriptional regulatory elements and their interactome.
- Published
- 2022
9. The retinoic acid receptor co-factor NRIP1 is uniquely upregulated and represents a therapeutic target in acute myeloid leukemia with chromosome 3q rearrangements
- Author
-
Liam MacPhee, Florian Kuchenbauer, Sarah Grasedieck, Lars Bullinger, Ariene Cabantog, Junbum Im, Konstanze Döhner, Tobias Herold, Jonathan R. Pollack, Nadine Sperb, Christoph Ruess, Burcu Demir, Arefeh Rouhi, and Frank G. Rücker
- Subjects
Chromosome Aberrations ,Cancer Research ,Gene knockdown ,Myeloid ,Receptors, Retinoic Acid ,Myeloid leukemia ,Hematology ,Biology ,Chromosomes ,MDS1 and EVI1 Complex Locus Protein ,Nuclear Receptor Interacting Protein 1 ,Leukemia, Myeloid, Acute ,Retinoic acid receptor ,medicine.anatomical_structure ,Downregulation and upregulation ,Nuclear receptor ,hemic and lymphatic diseases ,Cancer research ,medicine ,Humans ,NRIP1 ,Enhancer - Abstract
Aberrant expression of Ecotropic Viral Integration Site 1 (EVI1) is a hallmark of acute myeloid leukemia (AML) with inv(3) or t(3;3), which is a disease subtype with especially poor outcome. In studying transcriptomes from AML patients with chromosome 3q rearrangements, we identified a significant upregulation of the Nuclear Receptor Interacting Protein 1 (NRIP1) as well as its adjacent non-coding RNA LOC101927745. Utilizing transcriptomic and epigenomic data from over 900 primary samples from patients as well as genetic and transcriptional engineering approaches, we have identified several mechanisms that can lead to upregulation of NRIP1 in AML. We hypothesize that the LOC101927745 transcription start site harbors a context-dependent enhancer that is bound by EVI1, causing upregulation of NRIP1 in AML with chromosome 3 abnormalities. Furthermore, we showed that NRIP1 knockdown negatively affects the proliferation and survival of 3qrearranged AML cells and increases their sensitivity to all-trans retinoic acid, suggesting that NRIP1 is relevant for the pathogenesis of inv(3)/t(3;3) AML and could serve as a novel therapeutic target in myeloid malignancies with 3q abnormalities.
- Published
- 2021
10. miR-148a-3p and DDX6 functional link promotes survival of myeloid leukemia cells
- Author
-
Maryam Ghashghaei, Cong Truc Le, Haya Shaalan, Leo Escano, Marty Yue, Aaremish Arsalan, Arefeh Rouhi, Tuan Anh Nguyen, and Ly Phuong Vu
- Subjects
Hematology - Abstract
Regulation of gene expression at the RNA level is an important regulatory mechanism in cancers. However, post-transcriptional molecular pathways underlying tumorigenesis remain largely unexplored. In this study, we uncovered a functional axis consisting of microRNA (miR)-148a-3p, RNA helicase DDX6, and its downstream target TXNIP in Acute Myeloid Leukemia (AML). Using a DROSHA-knockout cell system to evaluate miR-mediated gene expression control, we comprehensively profiled putative transcripts regulated by miR-148a-3p and identified DDX6 as a direct target of miR-148a-3p in AML cells. DDX6 depletion induced cell cycle arrest, apoptosis, and differentiation while delaying leukemia development in vivo. Genome-wide assessment of DDX6 binding transcripts and gene expression profiling of DDX6-depleted cells revealed TXNIP, a tumor suppressor, as the functional downstream target of DDX6. Overall, our study identified DDX6 as a post-transcriptional regulator that is required for AML survival. We proposed the regulatory link between miR-148a-3p and DDX6 as a potential therapeutic target in leukemia.
- Published
- 2022
11. An APOBEC/Inflammation-based classifier improves the stratification of multiple myeloma patients and identifies novel risk subgroups
- Author
-
Sarah Grasedieck, Afsaneh Panahi, Matthew C. Jarvis, Faezeh Borzooee, Reuben S. Harris, Mani Larijani, Hervé Avet-Loiseau, Mehmet Samur, Nikhil Munshi, Kevin Song, Arefeh Rouhi, and Florian Kuchenbauer
- Abstract
Background: Recent insights into the pathogenesis of multiple myeloma (MM) have highlighted inflammation and genome editing, e.g. by APOBEC enzymes, as major drivers of disease onset and progression. We hypothesized that inclusion of molecular features corresponding to these two mechanisms can be utilized to define novel MM risk groups at initial diagnosis. Methods: Using two independent patient cohorts (MMRF and IFM/DFCI 2009), we developed and validated an easy-to-calculate novel risk-score that is based on mRNA expression levels of APOBEC2 and APOBEC3B, as well as inflammatory cytokines (IL11, TGFB1 and TGFB3) and serum levels of ß2-microglobulin and LDH. Results: Performance of the Editor- and Inflammation-based score (EI-score) was superior to current cytogenetics-based risk classifiers. Moreover, the EI-score was able to identify previously unrecognized MM patients who experience favourable outcomes despite carrying adverse risk cytogenetics. Conclusions: Through accurate risk stratification we can identify patients who are currently over-or undertreated. The EI-score is a contemporary and superior prognostic score, calculated based on transcript levels at diagnosis, allowing the identification of unrecognized MM risk subgroups potentially leading to adjustment of clinical treatment and improvement of patient outcomes.
- Published
- 2022
12. Mitochondrial metabolism: powering new directions in acute myeloid leukemia
- Author
-
Florian Kuchenbauer, David Sanford, Arefeh Rouhi, Irina A. Maksakov, and Ryan J. Stubbins
- Subjects
Cancer Research ,Antineoplastic Agents ,Context (language use) ,Mitochondrion ,Oxidative Phosphorylation ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Humans ,neoplasms ,business.industry ,Venetoclax ,Myeloid leukemia ,Hematology ,medicine.disease ,Mitochondria ,3. Good health ,Lymphoma ,Leukemia, Myeloid, Acute ,Leukemia ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,business ,030215 immunology - Abstract
There has been an explosion of knowledge about the role of metabolism and the mitochondria in acute myeloid leukemia (AML). We have also recently seen several waves of novel therapies change the treatment landscape for AML, such as the selective B-cell lymphoma 2 (BCL-2) inhibitor venetoclax. In this new context, we review the rapidly advancing literature on the role of metabolism and the mitochondria in AML pathogenesis, and how these are interwoven with the mechanisms of action for novel therapeutics in AML. We also review the role of oxidative phosphorylation (OxPhos) in maintaining leukemia stem cells (LSCs), how recurrent genomic alterations in AML alter downstream metabolism, and focus on how the BCL-2 pathway and the mitochondria are inextricably linked in AML. Thus, we provide an overview of the mitochondria and metabolism in the context of our new therapeutic world for AML and outline how targeting these vulnerabilities may produce novel therapeutic strategies.
- Published
- 2021
13. Targeting AXL kinase sensitizes leukemic stem and progenitor cells to venetoclax treatment in acute myeloid leukemia
- Author
-
Yubin Ge, Hong Liu, Rick Li, German Novakovskiy, Jun Yan, Depei Wu, Florian Kuchenbauer, Min Chen, Katharina Rothe, Zaihui Zhang, Xiuyan Zhang, Sung-Eun Nam, Yun Zhao, Irina A Maksakova, Xiaojia Niu, Xiaoyan Jiang, Yehyeon Ahn, Arefeh Rouhi, Shenshen Lai, Wyeth W. Wasserman, Calvin K. Yip, Sarah Grasedieck, and Hong Zhang
- Subjects
Xenotransplantation ,medicine.medical_treatment ,Immunology ,Mice, SCID ,Biology ,Biochemistry ,Transcriptome ,Mice ,chemistry.chemical_compound ,Drug Delivery Systems ,Mice, Inbred NOD ,Cell Line, Tumor ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Progenitor cell ,Sulfonamides ,Kinase ,Venetoclax ,Receptor Protein-Tyrosine Kinases ,Myeloid leukemia ,Cell Biology ,Hematology ,Bridged Bicyclo Compounds, Heterocyclic ,Xenograft Model Antitumor Assays ,Axl Receptor Tyrosine Kinase ,Leukemia, Myeloid, Acute ,Cell killing ,chemistry ,Neoplastic Stem Cells ,Cancer research ,Stem cell - Abstract
The abundance of genetic abnormalities and phenotypic heterogeneities in acute myeloid leukemia (AML) poses significant challenges to the development of improved treatments. Here, we demonstrated that a key growth arrest-specific gene 6/AXL axis is highly activated in cells from patients with AML, particularly in stem/progenitor cells. We developed a potent selective AXL inhibitor that has favorable pharmaceutical properties and efficacy against preclinical patient-derived xenotransplantation (PDX) models of AML. Importantly, inhibition of AXL sensitized AML stem/progenitor cells to venetoclax treatment, with strong synergistic effects in vitro and in PDX models. Mechanistically, single-cell RNA-sequencing and functional validation studies uncovered that AXL inhibition, alone or in combination with venetoclax, potentially targets intrinsic metabolic vulnerabilities of AML stem/progenitor cells and shows a distinct transcriptomic profile and inhibits mitochondrial oxidative phosphorylation. Inhibition of AXL or BCL-2 also differentially targets key signaling proteins to synergize in leukemic cell killing. These findings have a direct translational impact on the treatment of AML and other cancers with high AXL activity.
- Published
- 2021
14. Prolonged Maintenance of Ex Vivo Functional Vk*MYC Multiple Myeloma Cells Allows for in Vitro Manipulation and Subsequent In Vivo Analysis
- Author
-
Frances E Lock, Afsaneh Panahi, Ariene Cabantog, Lorenzo Lindo, Daniel Waller, Kevin Hay, Marta Chesi, Arefeh Rouhi, and Florian Kuchenbauer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
15. Establishing an Easy-to-Use 3D BM Niche Model for the Co-Culture of Primary Bone Marrow Mesenchymal Stromal Cells (BM-MSC) with Hematopoietic Stem and Progenitor Cells (HSPC)
- Author
-
Frances E Lock, Alannah Wilson, Amanda Amoah, Sharon Gao, Carolina Tropini, Arefeh Rouhi, and Florian Kuchenbauer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
16. Targeting the WT1mut-Mir-193a Transcriptional Axis with INT-1B3, a Novel Lipid Nanoparticle-Formulated Mir-193a-3p Mimic in Acute Myeloid Leukemia
- Author
-
Xining Yang, Kathrin Krowiorz, Raj Bhayadia, Jessica Kohlschmidt, Deedra Nicolet, Tobias Maetzig, Caroline Pabst, Jeremy Hui, Arshia Shad, Sharon Gao, Maria Stephenson, Si Wei Wu, Marion van den Bosch, Daelynn Buelow, Sharyn Baker, Leo Escano, Frances E Lock, Afsaneh Panahi, Liam MacPhee, Irina Maksakova, Amanda Amoah, Konstanze Döhner, Linda Fogelstrand, Ann-Kathrin Eisfeld, Sanaz Yahyanejad, Michel Janicot, Christopher C. Oakes, Jan-Henning Klusmann, Arefeh Rouhi, and Florian Kuchenbauer
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
17. Sildenafil triggers tumor lethality through altered expression of HSP90 and degradation of PKD2
- Author
-
Evi Schmid, Kristina Diepold, A Fehn, Thomas Seufferlein, Yang Liu, Ninel Azoitei, Arefeh Rouhi, Lu Chen, Cornelia Brunner, Gabriela Chiosis, Alexander Becher, and Marcus V. Cronauer
- Subjects
0301 basic medicine ,Cancer Research ,TRPP Cation Channels ,Carcinogenesis ,Sildenafil ,Apoptosis ,Sildenafil Citrate ,Hsp90 inhibitor ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Cell Line, Tumor ,Neoplasms ,Humans ,Medicine ,HSP90 Heat-Shock Proteins ,Protein kinase A ,biology ,business.industry ,General Medicine ,Phosphodiesterase 5 Inhibitors ,Hsp90 ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,cGMP-specific phosphodiesterase type 5 ,Proteolysis ,Cancer cell ,biology.protein ,Cancer research ,business - Abstract
The repurposing of existing drugs has emerged as an attractive additional strategy to the development of novel compounds in the fight against cancerous diseases. Inhibition of phosphodiesterase 5 (PDE5) has been claimed as a potential approach to target various cancer subtypes in recent years. However, data on the treatment of tumors with PDE5 inhibitors as well as the underlying mechanisms are as yet very scarce. Here, we report that treatment of tumor cells with low concentrations of Sildenafil was associated with decreased cancer cell proliferation and augmented apoptosis in vitro and resulted in impaired tumor growth in vivo. Notably, incubation of cancer cells with Sildenafil was associated with altered expression of HSP90 chaperone followed by degradation of protein kinase D2, a client protein previously reported to be involved in tumor growth. Furthermore, the involvement of low doses of PU-H71, an HSP90 inhibitor currently under clinical evaluation, in combination with low concentrations of Sildenafil, synergistically and negatively impacted on the viability of cancer cells in vivo. Taken together, our study suggests that repurposing of already approved drugs, alone or in combination with oncology-dedicated compounds, may represent a novel cancer therapeutic strategy.
- Published
- 2020
18. P-150: Identification of misclassified multiple myeloma patient risk subgroups with a novel biological disease stratifier
- Author
-
Afsaneh Panahi, Sarah Grasedieck, Matthew Jarvis, Faezeh Borzooee, Reuben Harris, Mani Larijani, Kevin Song, Arefeh Rouhi, and Florian Kuchenbauer
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
19. MicroRNA-708 is a novel regulator of the Hoxa9 program in myeloid cells
- Author
-
Florian Kuchenbauer, Meena Kanduri, Lars Palmqvist, Reinhild Rösler, Christoph Ruess, Nadine Sperb, Christina Miller, Hartmut Döhner, Nicole Pochert, Erik Delsing Malmberg, Martin Hirst, Anna Staffas, Pradeep Kumar Kopparapu, Alireza Heravi-Moussavi, Alireza Lorzadeh, Ping Xiang, R. Keith Humphries, Sarah Grasedieck, Erik Larsson, Sebastian Wiese, Arghavan Ashouri, Liam MacPhee, Konstanze Döhner, Leo Escano, Linda Fogelstrand, Sebastian Iben, Edith Schneider, Kathrin Krowiorz, and Arefeh Rouhi
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Myeloid leukemia ,Hematology ,Biology ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,microRNA ,Transcriptional regulation ,medicine ,Homeobox ,Epigenetics ,Hox gene - Abstract
MicroRNAs (miRNAs) are commonly deregulated in acute myeloid leukemia (AML), affecting critical genes not only through direct targeting, but also through modulation of downstream effectors. Homeobox (Hox) genes balance self-renewal, proliferation, cell death, and differentiation in many tissues and aberrant Hox gene expression can create a predisposition to leukemogenesis in hematopoietic cells. However, possible linkages between the regulatory pathways of Hox genes and miRNAs are not yet fully resolved. We identified miR-708 to be upregulated in Hoxa9/Meis1 AML inducing cell lines as well as in AML patients. We further showed Meis1 directly targeting miR-708 and modulating its expression through epigenetic transcriptional regulation. CRISPR/Cas9 mediated knockout of miR-708 in Hoxa9/Meis1 cells delayed disease onset in vivo, demonstrating for the first time a pro-leukemic contribution of miR-708 in this context. Overexpression of miR-708 however strongly impeded Hoxa9 mediated transformation and homing capacity in vivo through modulation of adhesion factors and induction of myeloid differentiation. Taken together, we reveal miR-708, a putative tumor suppressor miRNA and direct target of Meis1, as a potent antagonist of the Hoxa9 phenotype but an effector of transformation in Hoxa9/Meis1. This unexpected finding highlights the yet unexplored role of miRNAs as indirect regulators of the Hox program during normal and aberrant hematopoiesis.
- Published
- 2019
20. The endothelin receptor type A is a downstream target of Hoxa9 and Meis1 in acute myeloid leukemia
- Author
-
Lars Palmqvist, Arefeh Rouhi, Laleh S. Arabanian, Tina Nilsson, Pegah Johansson, Anna Staffas, and Linda Fogelstrand
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Endothelin receptor type A ,Biology ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,Internal medicine ,medicine ,Animals ,Humans ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Homeodomain Proteins ,Hematology ,Gene Expression Regulation, Leukemic ,Cell growth ,Myeloid leukemia ,Receptor, Endothelin A ,medicine.disease ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,Cancer research ,Bone marrow - Abstract
Endothelin receptor type A (EDNRA) is known as a mediator of cell proliferation and survival. Aberrant regulation of EDNRA has been shown to play a role in tumor growth and metastasis. Using a global gene expression screen, we found that expression of Ednra was upregulated in murine leukemia inducing cells co-expressing Hoxa9 and Meis1 compared to cells only expressing Hoxa9. The aim of this study was to explore the role of Ednra in leukemogenesis further. In a murine bone marrow transplantation model, mice transplanted with cells overexpressing Ednra and Hoxa9 succumbed to leukemia significantly earlier than mice transplanted with cells overexpressing Hoxa9 only. Furthermore, overexpression of Ednra led to increased proliferation and resistance to apoptosis of bone marrow cells in vitro. We could also show that Meis1 binds to the Ednra promoter region, suggesting a regulatory role for Meis1 in Ednra expression. Taken together, our results suggest a role for Ednra in Hoxa9/Meis1-driven leukemogenesis.
- Published
- 2018
21. Micro-ribonucleic acid-155 is a direct target of Meis1, but not a driver in acute myeloid leukemia
- Author
-
Oleh Petriv, Jens Ruschmann, Edith Schneider, Lars Palmqvist, Kathrin Krowiorz, Anna Staffas, Erik Delsing Malmberg, Carl L. Hansen, Hartmut Döhner, Arefeh Rouhi, Florian Kuchenbauer, Martin Hirst, Arghavan Ashouri, Ping Xiang, Michael Heuser, Konstanze Döhner, Stella Yuan Wei, Lars Bullinger, Christina Miller, Linda Röhner, Nicole Pochert, R. Keith Humphries, Laleh S. Arabanian, Alireza Heravi-Moussavi, Christian Buske, and Linda Fogelstrand
- Subjects
0301 basic medicine ,Acute Myeloid Leukemia ,Carcinogenesis ,Article ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,hemic and lymphatic diseases ,microRNA ,Medicine ,Animals ,Humans ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Homeodomain Proteins ,Oncogene ,business.industry ,Gene Expression Regulation, Leukemic ,Myeloid leukemia ,Hematology ,medicine.disease ,Pathophysiology ,Leukemia ,Leukemia, Myeloid, Acute ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,business ,Homing (hematopoietic) - Abstract
Micro-ribonucleic acid-155 (miR-155) is one of the first described oncogenic miRNAs. Although multiple direct targets of miR-155 have been identified, it is not clear how it contributes to the pathogenesis of acute myeloid leukemia. We found miR-155 to be a direct target of Meis1 in murine Hoxa9/Meis1 induced acute myeloid leukemia. The additional overexpression of miR-155 accelerated the formation of acute myeloid leukemia in Hoxa9 as well as in Hoxa9/Meis1 cells in vivo. However, in the absence or following the removal of miR-155, leukemia onset and progression were unaffected. Although miR-155 accelerated growth and homing in addition to impairing differentiation, our data underscore the pathophysiological relevance of miR-155 as an accelerator rather than a driver of leukemogenesis. This further highlights the complexity of the oncogenic program of Meis1 to compensate for the loss of a potent oncogene such as miR-155. These findings are highly relevant to current and developing approaches for targeting miR-155 in acute myeloid leukemia.
- Published
- 2018
22. Thrombotic Microangiopathy after Allogeneic Stem Cell Transplantation: A Comparison of Eculizumab Therapy and Conventional Therapy
- Author
-
Florian Kuchenbauer, Donald Bunjes, Phyllis Schwarzwälder, Arefeh Rouhi, Hartmut Döhner, Stefanie von Harsdorf, Stefan S. Schönsteiner, Nadine Kloevekorn, Stephan R Bohl, and Martin Bommer
- Subjects
Adult ,medicine.medical_specialty ,Thrombotic microangiopathy ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,Defibrotide ,Antibodies, Monoclonal, Humanized ,Infections ,Gastroenterology ,Young Adult ,03 medical and health sciences ,Polydeoxyribonucleotides ,0302 clinical medicine ,Internal medicine ,Humans ,Transplantation, Homologous ,Medicine ,Aged ,Transplantation ,Thrombotic Microangiopathies ,business.industry ,Hematology ,Middle Aged ,Eculizumab ,medicine.disease ,Survival Analysis ,Thrombocytopenic purpura ,Surgery ,Treatment Outcome ,Cohort ,Plasmapheresis ,Rituximab ,business ,Stem Cell Transplantation ,030215 immunology ,medicine.drug - Abstract
We report the results of a single-center analysis of a cohort of 39 patients treated between 1997 and 2016 for transplantion-associated thrombotic microangiopathy. We evaluated 2 subgroups of patients: 24 patients treated between 1997 and 2014 who received conventional therapy and 15 patients treated with the complement-inhibiting monoclonal antibody eculizumab between 2014 and 2016. The conventional therapy group was treated predominantly with defibrotide alone or in combination with plasmapheresis or rituximab. Despite an initial response rate of 61%, only 4 patients (16%) were long-term survivors, 2 of whom had a low-risk thrombotic microangiopathy without multiorgan damage. Progression of thrombotic micorangiopathy and bacterial/fungal infections contributed equally to treatment failure. The overall response rate in the eculizumab group was significantly higher, at 93%. In addition, we were able to stop eculizumab treatment in 5 patients (33%), all of whom had high-risk thrombotic microangiopathy, due to sustained recovery. Despite the very good response in the eculizumab-treated group, we did not observe a significant improved overall survival, due primarily to a high rate of infection-related mortality (70%). Therefore, further studies are needed to identify the optimal therapeutic management approach for transplantation-associated thrombotic microangiopathy to improve its dismal outcome.
- Published
- 2017
23. Elucidating the Importance and Regulation of Key Enhancers for Human MEIS1 Expression
- Author
-
Edith Schneider, Gregg B. Morin, Xining Yang, Pamela A. Hoodless, R. Keith Humphries, Wei Wei, Leo Escano, Ishpreet Dhillon, Ping Xiang, Arefeh Rouhi, Florian Kuchenbauer, Jasper Wong, Martin Hirst, Aly Karsan, Simon Xufeng Wang, and Jack Clemans-Gibbon
- Subjects
Expression (architecture) ,Immunology ,Key (cryptography) ,Cell Biology ,Hematology ,Computational biology ,Biology ,Enhancer ,Biochemistry - Abstract
Myeloid ecotropic virus insertion site 1 (MEIS1) is essential for normal hematopoiesis and is deregulated in a large subset of acute myeloid leukemia (AML) by yet unknown mechanisms. We previously identified 3 candidate enhancer regions: enhancer region 1 (E1) at -2 kb upstream; enhancer region 2 (E2) at +10.6 kb downstream inside intron 6; and enhancer region 3 (E3) +140 kb downstream of the translation start site. In the current study, we utilized CRISPR-Cas9 genome editing to further characterize these enhancers in a human AML cell line and identify the key transcription factors (TFs) associated with their function. To efficiently track MEIS1 expression levels, a GFP reporter, a P2A self-cleaving peptide tag and a hemagglutinin tag at its translation start site was introduced in a MEIS1 high expressing human AML cell line, U937. Then we introduced random mutations (Indels) along the MEIS1 locus utilizing a CRISPR-Cas9 mediated genome editing vector system in mono-allelic MEIS1-GFP-tagged U937 cells with special focus on the previously identified enhancer regions to find the key sequences important to the function of the MEIS1 enhancer regions. Two targeted regions yielding the highest proportion of GFP - cells corresponded to the E2 enhancer region within intron 6 and were referred to as E2.1 and E2.2. Using chromosome conformation capture (3C) assay, we detected a significantly decreased interaction (p=0.0022) between the promoter and the intron 6 region surrounding the E2 region in E2.2 targeted cells compared to the parental cells. Moreover, our data indicated that the DNA sequence within E2.2 is highly critical to this region's enhancer function which is further influenced by the larger genomic region surrounding the E2.1 gRNA targeted site. To identify TFs binding to the E2 region, we further scrutinized the E2.2 indel region for loss of TF binding sites. We performed TF prediction analysis and performed a protein pull down-mass spectrometry experiment to identify TF candidates. The overlap yielded a list of 7 TFs, each of which we targeted via CRISPR/Cas9. Reduction in GFP levels was only observed for FLI1 locus targeting but not for the other 6 TFs. Concordant reduction in MEIS1 and FLI1 levels were confirmed by immunoblotting. Additionally, chromatin immunoprecipitation (ChIP) followed by quantitative PCR revealed significant FLI1 enrichment at the promoter and at 3 sites surrounding the E2.2 region (p=0.0004) compared to 4 control regions scattered along the MEIS1 locus. Given a previous study indicating MEIS1 upregulation of FLI1 in normal hematopoiesis, we hypothesised that a positive feedback loop may exist between FLI1 and MEIS1 in AML. Since MEIS1 levels are frequently elevated in normal karyotype AML (CN-AML), we used the murine Hoxa9/Meis1 AML model as a surrogate for CN-AML and performed Meis1 ChIP-seq analysis. We detected direct Meis1 binding to the intronic region of the mouse Fli1 gene as well as other ETS factor loci, in Hoxa9/Meis1 cells. To better understand the clinical relevance of FLI1 in AML, we analyzed the Beat AML dataset. High FLI1 transcript levels correlated with adverse overall survival in CN-AML (p=0.044). Additionally, we observed a trend towards worse outcome with high FLI1 in the NPM1-mutated CN-AML subtype (p=0.069). We also observed a similar correlation in CN-AML for another ETS factor, ELF1, which we had previously shown to bind and upregulate MEIS1 expression in AML, suggesting a broader unrecognized role for ETS factors in AML. In summary, we have developed a rapid flow cytometry-based readout for the fine dissection and characterization of the cis-regulatory elements and associated TFs critical for MEIS1 transcription via CRISPR-Cas9 genetic manipulation. Our study revealed FLI1 as the candidate key regulator of MEIS1 expression and a positive correlation between FLI1 mRNA levels and worse overall survival in MEIS1-high AML subgroups. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
24. Identification of Previously Unrecognized Multiple Myeloma Risk Subgroups with a Novel Biological Disease Stratifier
- Author
-
Matthew C. Jarvis, Reuben S. Harris, Sarah Grasedieck, Faezeh Borzooee, Arefeh Rouhi, Florian Kuchenbauer, Kevin W. Song, Mani Larijani, and Afsaneh M. Shariatpanahi
- Subjects
business.industry ,Immunology ,medicine ,Identification (biology) ,Cell Biology ,Hematology ,Disease ,medicine.disease ,business ,Bioinformatics ,Biochemistry ,Multiple myeloma - Abstract
Background: The prognosis of MM is determined by affected organs, tumor burden as measured by e.g., the international staging system (ISS), disease biology such as cytogenetic abnormalities, and response to therapy. The outcome of high-risk MM patients classified by ISS or adverse risk cytogenetics is not uniform and patients show heterogeneous survival. Recent insights into the pathogenesis of MM highlighted genome/transcriptome editing as well as inflammation as drivers for the onset and progression of MM. We hypothesized that inclusion of molecular features into risk stratification could potentially resolve the challenge of accurately distinguishing between high-risk and low-risk MM patients at initial diagnosis and improve outcome. Aim: We aimed to create a simple molecular risk score to identify unrecognized patient subgroups, who have been previously misclassified by current risk stratifiers. Method: The Multiple Myeloma Research Foundation CoMMpass study genomics dataset, combining mRNA Seq and clinical data from more than 700 MM patients, allowed us to evaluate the prognostic value of demographic and clinical parameters, cytogenetics, and gene expression levels of APOBEC genes as well as inflammation-modulating cytokines in MM patients. We calculated hazard ratios and Kaplan-Meier survival estimates for all extracted features. Combining clinical variables that were significantly associated with PFS and OS, we then applied machine learning approaches to identify the most accurate classification model to define a new risk score that is easy to compute and able to stratify NDMM patients more accurately than cytogenetics-based classifiers. Based on a Kaplan-Meier survival curve analysis, we then evaluated the performance of our newly built EI score in sub-classifying of current multiple myeloma risk stratifiers. Results: Based on machine learning models, we defined a weighted OS/PFS risk score (Editor-Inflammation (EI) score) based on mRNA expression of APOBEC2, APOBEC3B, IL11, TGFB1, TGFB3, as well as ß2-microglobulin and LDH serum levels. We showed that the EI score subclassified patients into high-risk, intermediate-risk, and low-risk prognostic groups and demonstrated superior performance (C-index: 0.76) compared to ISS (C-index: 0.66) and R-ISS (C-index: 0.64). We further showed that EI low-risk patients do not benefit from autograft and maintenance therapy. Re-classification of ISS (Figure 1a, b, c) and R-ISS risk groups further confirmed the superiority of the EI score. In addition, the EI score identified previously unrecognized distinct subgroups of MM patients with adverse risk cytogenetics but good prognosis (Figure 1d, e, f). For example, the EI score excellently subclassified del(17p) MM patients into three main risk subgroups including a super low-risk group (none of them has p53 mut) with 5-year OS of 100%, an intermediate-risk group (30% of these patients also have p53 mut) with 5-year OS rate of 75%, and a very poor prognosis group of patients (40% of these patients also have p53 mut) with 5-year OS rate of 0% (2y OS: 40%) (Figure 1f). In line, we could show that patients with del(17p) and high EI score exhibit an enrichment of APOBEC induced genomic mutations compared to intermediate-risk and low-risk patients supporting the hypothesis that del(17p) along with high APOBEC expression levels activate the APOBEC mutation program and thus create an optimal environment for tumor progression. These findings support the necessity of a prognostic score that more accurately reflects MM disease biology. Conclusion: Although MM is considered as an incurable disease, an improved risk stratification could help to identify previously unrecognized low- and high-risk patient subgroups that are over- or undertreated and lead to improved outcomes. Our EI score is a simple score that is based on recent insights into MM biology and accurately identifies high-risk and low-risk newly diagnosed MM patients as well as misclassified MM patients in different cytogenetic and ISS risk subgroups. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
25. P-120: Improving the risk stratification of multiple myeloma with a nucleotide editor/inflammation-based scoring system
- Author
-
Florian Kuchenbauer, Mani Larijani, Arefeh Rouhi, Matthew C. Jarvis, Kevin W. Song, Reuben S. Harris, Faezeh Borzooee, Afsaneh M. Shariatpanahi, and Sarah Grasedieck
- Subjects
APOBEC ,Oncology ,Cancer Research ,medicine.medical_specialty ,Framingham Risk Score ,business.industry ,Hazard ratio ,Cytogenetics ,Inflammation ,Genomics ,Hematology ,medicine.disease ,Maintenance therapy ,Internal medicine ,medicine ,medicine.symptom ,business ,Multiple myeloma - Abstract
Background Current clinical prognostication of newly diagnosed MM (NDMM) patients relies on clinical parameters and/or recurrent genetic changes, which mainly reflect early events in the development of MM. However, recent insights into the pathogenesis of MM highlighted genome/transcriptome editing through APOBEC genes as well as inflammation as drivers for the onset and progression of MM. Aims To build a superior nucleotide editor/inflammation-based risk scoring system reflecting biological processes that drive the progression of MM. Method We hypothesized that a prognostic score reflecting biological processes as well as clinical features is superior to the currently used classification systems for MM patients, such as ISS, R-ISS and the Mayo clinic classification. The Multiple Myeloma Research Foundation CoMMpass study genomics dataset, combining mRNA Seq and clinical data from more than 700 patients, allowed us to evaluate the prognostic value of demographic and clinical parameters, cytogenetics, and gene expression levels of APOBEC family as well as inflammation-modulating cytokines of MM patients. We calculated hazard ratios and Kaplan-Meier survival estimates for all extracted features. Combining clinical variables that were significantly associated with PFS and OS, we then applied machine learning approaches to identify the most accurate classification model to define a new risk score that is easy to compute and able to stratify NDMM patients more accurately than cytogenetics-based classifiers. Result Based on our machine learning models, we defined a weighted OS/PFS risk score (Editor-Inflammation score) based on expression of APOBEC2, APOBEC3B, IL11, TGFB1, TGFB3, as well as β-2-microglobulin and LDH serum levels, that achieved the best classification outcome and showed superior performance compared to ISS and R-ISS. Of note, cytogenetic abnormalities did not proof relevant have therefore not been included in the EI score. Besides superior overall risk stratification, the EI score further allowed to identify subgroups of MM patients with very good prognosis who do not significantly benefit from bone marrow transplantation maintenance therapy. Conclusion Our findings support the adoption of molecular biomarkers, reflecting dynamic biological processes rather than cytogenetics for a more accurate risk classification of MM. Considering that mRNA-seq is as cost effective as FISH and is being increasingly adopted by diagnostic centers, the EI score is a unique approach to shed light on underlying molecular mechanisms that drive disease progression and the development of true risk-adapted treatment strategies.
- Published
- 2021
26. NFATC1activation by DNA hypomethylation in chronic lymphocytic leukemia correlates with clinical staging and can be inhibited by ibrutinib
- Author
-
Sandra Robrecht, Rainer Claus, Barbara Eichhorst, Jasmin Bahlo, Hartmut Döhner, Dieter Weichenhan, Anja Weigel, Christine Wolf, Manuela Zucknick, Kirsten Fischer, Stephan Stilgenbauer, Natalia Becker, Katharina Filarsky, Arefeh Rouhi, Michael Hallek, Florian Kuchenbauer, Angela Garding, Christoph Plass, Daniel Mertens, and Peter Lichter
- Subjects
0301 basic medicine ,Cancer Research ,integumentary system ,Chronic lymphocytic leukemia ,B-cell receptor ,breakpoint cluster region ,NFAT ,Biology ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,chemistry ,hemic and lymphatic diseases ,Ibrutinib ,DNA methylation ,Cancer research ,medicine ,Transcription factor ,DNA hypomethylation - Abstract
B cell receptor (BCR) signaling is a key for survival of chronic lymphocytic leukemia (CLL) cells, and BCR signaling inhibitors are clinically active. However, relapse and resistance to treatment require novel treatment options. To detect novel candidate therapeutic targets, we performed a genome-wide DNA methylation screen with custom arrays and identified aberrant promoter DNA methylation in 2,192 genes. The transcription factor NFATC1 that is a downstream effector of BCR signaling was among the top hypomethylated genes and was concomitantly transcriptionally upregulated in CLL. Intriguingly, NFATC1 promoter DNA hypomethylation levels were significantly variant in clinical trial cohorts from different disease progression stages and furthermore correlated with Binet disease staging and thymidine kinase levels, strongly suggesting a central role of NFATC1 in CLL development. Functionally, DNA hypomethylation at NFATC1 promoter inversely correlated with RNA levels of NFATC1 and dysregulation correlated with expression of target genes BCL-2, CCND1 and CCR7. The inhibition of the NFAT regulator calcineurin with tacrolimus and cyclosporin A and the BCR signaling inhibitor ibrutinib significantly reduced NFAT activity in leukemic cell lines, and NFAT inhibition resulted in increased apoptosis of primary CLL cells. In summary, our results indicate that the aberrant activation of NFATC1 by DNA hypomethylation and BCR signaling plays a major role in the pathomechanism of CLL.
- Published
- 2017
27. STK33 participates to HSP90-supported angiogenic program in hypoxic tumors by regulating HIF-1α/VEGF signaling pathway
- Author
-
Thomas Simmet, Kristina Diepold, Ninel Azoitei, Arefeh Rouhi, Milena Armacki, Gabriela Chiosis, Yang Liu, Konrad Steinestel, and Thomas Seufferlein
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,education ,HIF-1α ,medicine.disease_cause ,VEGF-A ,03 medical and health sciences ,0302 clinical medicine ,VEGF Signaling Pathway ,medicine ,Serine/threonine-specific protein kinase ,hypoxia ,Kinase ,Molecular pathology ,business.industry ,STK33 ,tumor angiogenesis ,Molecular Pharmacology ,030104 developmental biology ,HIF1A ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Carcinogenesis ,business ,Research Paper - Abstract
// Yang Liu 1, 2 , Konrad Steinestel 3, 4 , Arefeh Rouhi 5 , Milena Armacki 1 , Kristina Diepold 1 , Gabriela Chiosis 6 , Thomas Simmet 7 , Thomas Seufferlein 1 and Ninel Azoitei 1 1 Center for Internal Medicine I, Ulm University, Ulm, Germany 2 Department of Gastroenterology and Hepatology, Zhongda Hospital, Southeast University, Nanjing, China 3 Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany 4 Gerhard-Domagk-Institute of Pathology, University of Munster, Munster, Germany 5 Center for Internal Medicine III, Ulm University, Ulm, Germany 6 Department of Molecular Pharmacology and Chemistry, Memorial Sloan-Kettering Institute, New York, NY, USA 7 Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany Correspondence to: Ninel Azoitei, email: ninel.azoitei@uni-ulm.de Keywords: tumor angiogenesis, hypoxia, VEGF-A, STK33, HIF-1α Received: May 13, 2017 Accepted: July 31, 2017 Published: August 24, 2017 ABSTRACT Lately, the HSP90 client serine/threonine kinase STK33 emerged to be required by cancer cells for their viability and proliferation. However, its mechanistic contribution to carcinogenesis is not clearly understood. Here we report that elevated STK33 expression correlates with advanced stages of human pancreatic and colorectal carcinomas. Impaired proliferation and augmented apoptosis associated with genetic abrogation of STK33 were paralleled by decreased vascularization in tumor xenografts. In line with this, ectopic STK33 not only promoted tumor growth after pharmacologic inhibition of HSP90 using structurally divergent small molecules currently in clinical development, but also restored blood vessel formation in vivo . Mechanistic studies demonstrated that HSP90-stabilized STK33 interacts with and regulates hypoxia-driven accumulation and activation of HIF-1α as well as secretion of VEGF-A in hypoxic cancer cells. In addition, our study reveals a putative cooperation between STK33 and other HSP90 client protein kinases involved in molecular and cellular events through which cancer cells ensure their survival by securing the oxygen and nutrient supply. Altogether, our findings indicate that STK33 interferes with signals from hypoxia and HSP90 to promote tumor angiogenesis and tumor growth.
- Published
- 2017
28. Circular RNAs of the nucleophosmin (NPM1) gene in acute myeloid leukemia
- Author
-
Florian Kuchenbauer, Susanne Hirsch, Tamara J. Blätte, Hartmut Döhner, Anna Dolnik, Jan Krönke, Richard F. Schlenk, Konstanze Döhner, Peter Paschka, Mojca Jongen-Lavrencic, Arefeh Rouhi, Sibylle Cocciardi, Verena I. Gaidzik, Sarah Grasedieck, Lars Bullinger, and Hematology
- Subjects
0301 basic medicine ,Acute Myeloid Leukemia ,NPM1 ,RNA Splicing ,Gene Expression ,Chromosomal translocation ,Biology ,Article ,03 medical and health sciences ,hemic and lymphatic diseases ,Gene expression ,Humans ,Gene ,Nucleophosmin ,Myeloid leukemia ,Nuclear Proteins ,Hematology ,RNA, Circular ,Molecular biology ,Haematopoiesis ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Case-Control Studies ,RNA splicing ,Cancer research ,RNA - Abstract
In acute myeloid leukemia, there is growing evidence for splicing pattern deregulation, including differential expression of linear splice isoforms of the commonly mutated gene nucleophosmin (NPM1). In this study, we detect circular RNAs of NPM1 and quantify circRNA hsa_circ_0075001 in a cohort of NPM1 wild-type and mutated acute myeloid leukemia (n=46). Hsa_circ_0075001 expression correlates positively with total NPM1 expression, but is independent of the NPM1 mutational status. High versus low hsa_circ_0075001 expression defines patient subgroups characterized by distinct gene expression patterns, such as lower expression of components of the Toll-like receptor signaling pathway in high hsa_circ_0075001 expression cases. Global evaluation of circRNA expression in sorted healthy hematopoietic controls (n=10) and acute myeloid leukemia (n=10) reveals circRNA transcripts for 47.9% of all highly expressed genes. While circRNA expression correlates globally with parental gene expression, we identify hematopoietic differentiation-associated as well as acute myeloid leukemia subgroup-specific circRNA signatures.
- Published
- 2017
29. Targeting miR-155 in FLT3-ITD mutated AML: ready for prime time?
- Author
-
Florian Kuchenbauer, Edith Schneider, and Arefeh Rouhi
- Subjects
Cancer Research ,Mutation ,Effector ,Cellular differentiation ,Biology ,medicine.disease_cause ,Bioinformatics ,Receptor tyrosine kinase ,miR-155 ,Transformation (genetics) ,Oncology ,In vivo ,hemic and lymphatic diseases ,medicine ,Cancer research ,biology.protein ,Radiology, Nuclear Medicine and imaging ,Receptor - Abstract
The most common genetic aberration in AML is a gain-of-function mutation in the FMS-like tyrosine kinase 3 (FLT3) receptor, which is present in about 30% of CN-AML and confers a poor prognosis (1). FLT3 encodes a receptor tyrosine kinase expressed on hematopoietic progenitor cells involved in stem cell differentiation and proliferation (2). FLT3 activating mutations such as internal tandem duplication (ITD) lead to constitutive, ligand-independent activation of this receptor, conferring a growth and survival advantage. The mutation itself has not been shown to independently drive leukemic transformation in vivo (1,3). Rather, FLT3-ITD must collaborate with additional oncogenic mutations to trigger hematopoietic malignancy (3). Despite extensive research throughout the last decades, only Midastaurin has been recently approved by the FDA as a first line treatment in combination with chemotherapy (4) highlighting the difficulties for establishing targeted therapies. Therefore, novel approaches such as targeting downstream effectors of FLT3-ITD signaling are relevant to explore new therapeutic targets.
- Published
- 2017
30. MicroRNA-708 is a novel regulator of the Hoxa9 program in myeloid cells
- Author
-
Edith, Schneider, Nicole, Pochert, Christoph, Ruess, Liam, MacPhee, Leo, Escano, Christina, Miller, Kathrin, Krowiorz, Erik, Delsing Malmberg, Alireza, Heravi-Moussavi, Alireza, Lorzadeh, Arghavan, Ashouri, Sarah, Grasedieck, Nadine, Sperb, Pradeep, Kumar Kopparapu, Sebastian, Iben, Anna, Staffas, Ping, Xiang, Reinhild, Rösler, Meena, Kanduri, Erik, Larsson, Linda, Fogelstrand, Hartmut, Döhner, Konstanze, Döhner, Sebastian, Wiese, Martin, Hirst, R, Keith Humphries, Lars, Palmqvist, Florian, Kuchenbauer, and Arefeh, Rouhi
- Subjects
Homeodomain Proteins ,Gene Expression Regulation, Leukemic ,Apoptosis ,Cell Differentiation ,Hematopoiesis ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,Mice ,MicroRNAs ,Tumor Cells, Cultured ,Animals ,Humans ,Female ,Myeloid Cells ,CRISPR-Cas Systems ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Cell Proliferation - Abstract
MicroRNAs (miRNAs) are commonly deregulated in acute myeloid leukemia (AML), affecting critical genes not only through direct targeting, but also through modulation of downstream effectors. Homeobox (Hox) genes balance self-renewal, proliferation, cell death, and differentiation in many tissues and aberrant Hox gene expression can create a predisposition to leukemogenesis in hematopoietic cells. However, possible linkages between the regulatory pathways of Hox genes and miRNAs are not yet fully resolved. We identified miR-708 to be upregulated in Hoxa9/Meis1 AML inducing cell lines as well as in AML patients. We further showed Meis1 directly targeting miR-708 and modulating its expression through epigenetic transcriptional regulation. CRISPR/Cas9 mediated knockout of miR-708 in Hoxa9/Meis1 cells delayed disease onset in vivo, demonstrating for the first time a pro-leukemic contribution of miR-708 in this context. Overexpression of miR-708 however strongly impeded Hoxa9 mediated transformation and homing capacity in vivo through modulation of adhesion factors and induction of myeloid differentiation. Taken together, we reveal miR-708, a putative tumor suppressor miRNA and direct target of Meis1, as a potent antagonist of the Hoxa9 phenotype but an effector of transformation in Hoxa9/Meis1. This unexpected finding highlights the yet unexplored role of miRNAs as indirect regulators of the Hox program during normal and aberrant hematopoiesis.
- Published
- 2018
31. Endogenous Tumor Suppressor microRNA-193b: Therapeutic and Prognostic Value in Acute Myeloid Leukemia
- Author
-
Hartmut Döhner, Jan-Henning Klusmann, Tobias Mätzig, C. Michel Zwaan, Stephan Emmrich, Kathrin Krowiorz, R. Keith Humphries, Thomas Thum, Michelle Ng, Dirk Reinhardt, Sabrina Bothur, Raj Bhayadia, Konstanze Döhner, Nadine Haetscher, Susanne Wingert, Marry M. Van den Heuvel Eibrink, Dirk Heckl, Michael Heuser, Razan Jammal, Arefeh Rouhi, Askar Obulkasim, Michael A. Rieger, Maarten Fornerod, Adrian Schwarzer, Florian Kuchenbauer, Jan Fiedler, and Pediatrics
- Subjects
0301 basic medicine ,Cancer Research ,MAP Kinase Signaling System ,Medizin ,Down-Regulation ,Endogeny ,Cell Growth Processes ,law.invention ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,law ,Mice, Inbred NOD ,microRNA ,Medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Polymerase chain reaction ,Homeodomain Proteins ,business.industry ,Myeloid leukemia ,Prognosis ,3. Good health ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia, Myeloid, Acute ,MicroRNAs ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Knockout mouse ,Cancer research ,Suppressor ,Heterografts ,business - Abstract
Purpose Dysregulated microRNAs are implicated in the pathogenesis and aggressiveness of acute myeloid leukemia (AML). We describe the effect of the hematopoietic stem-cell self-renewal regulating miR-193b on progression and prognosis of AML. Methods We profiled miR-193b-5p/3p expression in cytogenetically and clinically characterized de novo pediatric AML (n = 161) via quantitative real-time polymerase chain reaction and validated our findings in an independent cohort of 187 adult patients. We investigated the tumor suppressive function of miR-193b in human AML blasts, patient-derived xenografts, and miR-193b knockout mice in vitro and in vivo. Results miR-193b exerted important, endogenous, tumor-suppressive functions on the hematopoietic system. miR-193b-3p was downregulated in several cytogenetically defined subgroups of pediatric and adult AML, and low expression served as an independent indicator for poor prognosis in pediatric AML (risk ratio ± standard error, −0.56 ± 0.23; P = .016). miR-193b-3p expression improved the prognostic value of the European LeukemiaNet risk-group stratification or a 17-gene leukemic stemness score. In knockout mice, loss of miR-193b cooperated with Hoxa9/Meis1 during leukemogenesis, whereas restoring miR-193b expression impaired leukemic engraftment. Similarly, expression of miR-193b in AML blasts from patients diminished leukemic growth in vitro and in mouse xenografts. Mechanistically, miR-193b induced apoptosis and a G1/S-phase block in various human AML subgroups by targeting multiple factors of the KIT-RAS-RAF-MEK-ERK (MAPK) signaling cascade and the downstream cell cycle regulator CCND1. Conclusion The tumor-suppressive function is independent of patient age or genetics; therefore, restoring miR-193b would assure high antileukemic efficacy by blocking the entire MAPK signaling cascade while preventing the emergence of resistance mechanisms.
- Published
- 2018
32. MicroRNA-223 dose levels fine tune proliferation and differentiation in human cord blood progenitors and acute myeloid leukemia
- Author
-
Simon Leierseder, Arefeh Rouhi, Andrew L Muranyi, Stefan Engelhardt, Francesco Boccalatte, Jens Ruschmann, Daniel T. Starczynowski, Bob Argiropoulos, Aly Karsan, Christian Buske, Mojca Jongen-Lavrencic, Michael Heuser, Tiziana Plati, R. Keith Humphries, Hartmut Döhner, Bernhard Gentner, Nicole Pochert, Tobias Berg, Florian Kuchenbauer, Milijana mirkovic-Hosle, Su Ming Sun, Sarah M Mah, Donna E. Hogge, Luigi Naldini, Fernando D. Camargo, Gentner, B, Pochert, N, Rouhi, A, Boccalatte, F, Plati, T, Berg, T, Sun, Sm, Mah, Sm, Mirkovic Hösle, M, Ruschmann, J, Muranyi, A, Leierseder, S, Argiropoulos, B, Starczynowski, Dt, Karsan, A, Heuser, M, Hogge, D, Camargo, Fd, Engelhardt, S, Döhner, H, Buske, C, Jongen Lavrencic, M, Naldini, Luigi, Humphries, Rk, Kuchenbauer, F., and Hematology
- Subjects
Adult ,Male ,Cancer Research ,Myeloid ,CD34 ,Mice, SCID ,Biology ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,mir-223 ,Mice, Inbred NOD ,hemic and lymphatic diseases ,Genetics ,medicine ,Animals ,Humans ,Erythropoiesis ,Lymphopoiesis ,RNA, Neoplasm ,Progenitor cell ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,Mice, Knockout ,0303 health sciences ,Myeloid leukemia ,Cell Biology ,Hematology ,Neoplasms, Experimental ,Middle Aged ,medicine.disease ,Fetal Blood ,Hematopoietic Stem Cells ,3. Good health ,Leukemia ,Haematopoiesis ,Leukemia, Myeloid, Acute ,MicroRNAs ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Neoplastic Stem Cells ,Female - Abstract
A precise understanding of the role of miR-223 in human hematopoiesis and in the pathogenesis of acute myeloid leukemia (AML) is still lacking. By measuring miR-223 expression in blasts from 115 AML patients, we found significantly higher miR-223 levels in patients with favorable prognosis, whereas patients with low miR-223 expression levels were associated with worse outcome. Furthermore, miR-223 was hierarchically expressed in AML subpopulations, with lower expression in leukemic stem cell–containing fractions. Genetic depletion of miR-223 decreased the leukemia initiating cell (LIC) frequency in a myelomonocytic AML mouse model, but it was not mandatory for rapid-onset AML. To relate these observations to physiologic myeloid differentiation, we knocked down or ectopically expressed miR-223 in cord-blood CD34 + cells using lentiviral vectors. Although miR-223 knockdown delayed myeloerythroid precursor differentiation in vitro, it increased myeloid progenitors in vivo following serial xenotransplantation. Ectopic miR-223 expression increased erythropoiesis, T lymphopoiesis, and early B lymphopoiesis in vivo. These findings broaden the role of miR-223 as a regulator of the expansion/differentiation equilibrium in hematopoietic stem and progenitor cells where its impact is dose- and differentiation-stage-dependent. This also explains the complex yet minor role of miR-223 in AML, a heterogeneous disease with variable degree of myeloid differentiation.
- Published
- 2015
33. NFATC1 activation by DNA hypomethylation in chronic lymphocytic leukemia correlates with clinical staging and can be inhibited by ibrutinib
- Author
-
Christine, Wolf, Angela, Garding, Katharina, Filarsky, Jasmin, Bahlo, Sandra, Robrecht, Natalia, Becker, Manuela, Zucknick, Arefeh, Rouhi, Anja, Weigel, Rainer, Claus, Dieter, Weichenhan, Barbara, Eichhorst, Kirsten, Fischer, Michael, Hallek, Florian, Kuchenbauer, Christoph, Plass, Hartmut, Döhner, Stephan, Stilgenbauer, Peter, Lichter, and Daniel, Mertens
- Subjects
Male ,NFATC Transcription Factors ,Gene Expression Regulation, Leukemic ,Adenine ,DNA Methylation ,Leukemia, Lymphocytic, Chronic, B-Cell ,Pyrimidines ,Piperidines ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,Pyrazoles ,Female ,Neoplasm Recurrence, Local ,Promoter Regions, Genetic ,Protein Kinase Inhibitors ,Aged ,Neoplasm Staging ,Signal Transduction - Abstract
B cell receptor (BCR) signaling is a key for survival of chronic lymphocytic leukemia (CLL) cells, and BCR signaling inhibitors are clinically active. However, relapse and resistance to treatment require novel treatment options. To detect novel candidate therapeutic targets, we performed a genome-wide DNA methylation screen with custom arrays and identified aberrant promoter DNA methylation in 2,192 genes. The transcription factor NFATC1 that is a downstream effector of BCR signaling was among the top hypomethylated genes and was concomitantly transcriptionally upregulated in CLL. Intriguingly, NFATC1 promoter DNA hypomethylation levels were significantly variant in clinical trial cohorts from different disease progression stages and furthermore correlated with Binet disease staging and thymidine kinase levels, strongly suggesting a central role of NFATC1 in CLL development. Functionally, DNA hypomethylation at NFATC1 promoter inversely correlated with RNA levels of NFATC1 and dysregulation correlated with expression of target genes BCL-2, CCND1 and CCR7. The inhibition of the NFAT regulator calcineurin with tacrolimus and cyclosporin A and the BCR signaling inhibitor ibrutinib significantly reduced NFAT activity in leukemic cell lines, and NFAT inhibition resulted in increased apoptosis of primary CLL cells. In summary, our results indicate that the aberrant activation of NFATC1 by DNA hypomethylation and BCR signaling plays a major role in the pathomechanism of CLL.
- Published
- 2017
34. Stk33 is required for spermatid differentiation and male fertility in mice
- Author
-
Arefeh Rouhi, Ingrid Lohmann, Dorothee Terhardt, Bahtiyar Kurtulmus, Gislene Pereira, Stefan Fröhling, Laura Schwarzmüller, Stefan Koch, Hanno Glimm, Christof Niehrs, Claudia Scholl, Karsten Richter, Raffaela K. Bung, and Leila R. Martins
- Subjects
0301 basic medicine ,Male ,endocrine system ,Spermiogenesis ,Cellular differentiation ,Biology ,Protein Serine-Threonine Kinases ,Microtubules ,03 medical and health sciences ,Mice ,Microtubule ,Spermatocytes ,Testis ,Animals ,Spermatogenesis ,Molecular Biology ,reproductive and urinary physiology ,Genetics ,Mice, Knockout ,urogenital system ,Spermatid differentiation ,Cell Differentiation ,Cell Biology ,Sperm ,Spermatids ,Spermatozoa ,Cell biology ,030104 developmental biology ,Fertility ,Cytoplasm ,Developmental biology ,Developmental Biology - Abstract
Spermiogenesis is the final phase during sperm cell development in which round spermatids undergo dramatic morphological changes to generate spermatozoa. Here we report that the serine/threonine kinase Stk33 is essential for the differentiation of round spermatids into functional sperm cells and male fertility. Constitutive Stk33 deletion in mice results in severely malformed and immotile spermatozoa that are particularly characterized by disordered structural tail elements. Stk33 expression first appears in primary spermatocytes, and targeted deletion of Stk33 in these cells recapitulates the defects observed in constitutive knockout mice, confirming a germ cell-intrinsic function. Stk33 protein resides in the cytoplasm and partially co-localizes with the caudal end of the manchette, a transient structure that guides tail elongation, in elongating spermatids, and loss of Stk33 leads to the appearance of a tight, straight and elongated manchette. Together, these results identify Stk33 as an essential regulator of spermatid differentiation and male fertility.
- Published
- 2017
35. MicroRNA-155 promotes G-CSF-induced mobilization of murine hematopoietic stem and progenitor cells via propagation of CXCL12 signaling
- Author
-
Arefeh Rouhi, Florian Kuchenbauer, C Ludwig, Orit Kollet, Hartmut Geiger, Edith Schneider, Robert Brooks, Karin Golan, Tsvee Lapidot, William G. Kerr, Christopher M. Russo, Eran Hornstein, Eman Khatib-Massalha, Shiri Gur-Cohen, Tomer Itkin, Anju Kumari, and John D. Chisholm
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Biology ,Polymorphism, Single Nucleotide ,miR-155 ,03 medical and health sciences ,Mice ,microRNA ,Granulocyte Colony-Stimulating Factor ,Animals ,Progenitor cell ,Hematopoietic Stem Cell Mobilization ,Mice, Knockout ,Hematology ,Hematopoietic Stem Cells ,biological factors ,Chemokine CXCL12 ,Cell biology ,Haematopoiesis ,MicroRNAs ,030104 developmental biology ,Oncology ,embryonic structures ,Immunology ,biology.protein ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Stem cell ,Signal Transduction - Abstract
MicroRNA-155 promotes G-CSF-induced mobilization of murine hematopoietic stem and progenitor cells via propagation of CXCL12 signaling
- Published
- 2017
36. HSP90 Supports Tumor Growth and Angiogenesis through PRKD2 Protein Stabilization
- Author
-
Felicitas Genze, Kristina Diepold, Ninel Azoitei, Claudia Scholl, Stefan Fröhling, Alexander Becher, Arefeh Rouhi, Thomas Seufferlein, Johan Van Lint, Hans A. Kestler, Cornelia Brunner, John Koren, and Gabriela Chiosis
- Subjects
Vascular Endothelial Growth Factor A ,Proteasome Endopeptidase Complex ,Cancer Research ,Cell signaling ,Angiogenesis ,Mice, Nude ,Apoptosis ,Article ,Mice ,Cell Line, Tumor ,Animals ,Humans ,HSP90 Heat-Shock Proteins ,Protein Kinase C ,Neovascularization, Pathologic ,biology ,NF-kappa B ,HCT116 Cells ,Hypoxia-Inducible Factor 1, alpha Subunit ,Hsp90 ,Cell Hypoxia ,Vascular endothelial growth factor A ,Oncology ,Cancer cell ,Cancer research ,biology.protein ,Female ,Ectopic expression ,Signal transduction ,Protein stabilization ,Signal Transduction - Abstract
The kinase PRKD2 (protein kinase D) is a crucial regulator of tumor cell-endothelial cell communication in gastrointestinal tumors and glioblastomas, but its mechanistic contributions to malignant development are not understood. Here, we report that the oncogenic chaperone HSP90 binds to and stabilizes PRKD2 in human cancer cells. Pharmacologic inhibition of HSP90 with structurally divergent small molecules currently in clinical development triggered proteasome-dependent degradation of PRKD2, augmenting apoptosis in human cancer cells of various tissue origins. Conversely, ectopic expression of PRKD2 protected cancer cells from the apoptotic effects of HSP90 abrogation, restoring blood vessel formation in two preclinical models of solid tumors. Mechanistic studies revealed that PRKD2 is essential for hypoxia-induced accumulation of hypoxia-inducible factor-1α (HIF1α) and activation of NF-κB in tumor cells. Notably, ectopic expression of PRKD2 was able to partially restore HIF1α and secreted VEGF-A levels in hypoxic cancer cells treated with HSP90 inhibitors. Taken together, our findings indicate that signals from hypoxia and HSP90 pathways are interconnected and funneled by PRKD2 into the NF-κB/VEGF-A signaling axis to promote tumor angiogenesis and tumor growth. ispartof: Cancer Research vol:74 issue:23 pages:7125-36 ispartof: location:United States status: published
- Published
- 2014
37. Prospective identification of resistance mechanisms to HSP90 inhibition in KRAS mutant cancer cells
- Author
-
Hartmut Döhner, Christina Miller, Stefanie Reinhart, Lars Bullinger, Stefan Fröhling, Claudia Scholl, Florian Kuchenbauer, Britta Stolze, Sarah Grasedieck, and Arefeh Rouhi
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Ganetespib ,Apoptosis ,Drug resistance ,medicine.disease_cause ,Targeted therapy ,Hsp90 inhibitor ,0302 clinical medicine ,Neoplasms ,polycyclic compounds ,Benzoquinones ,Medicine ,Genetics ,HSP90 inhibition ,3. Good health ,Phenotype ,Oncology ,030220 oncology & carcinogenesis ,RNA Interference ,KRAS ,PU-H71 ,Research Paper ,ATP Binding Cassette Transporter, Subfamily B ,Cell Survival ,Lactams, Macrocyclic ,Mutation, Missense ,Antineoplastic Agents ,MDR1 ,Transfection ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,mutant KRAS ,Humans ,Genetic Predisposition to Disease ,Benzodioxoles ,HSP90 Heat-Shock Proteins ,Cell Proliferation ,drug resistance ,Oncogene ,Dose-Response Relationship, Drug ,business.industry ,Gene Amplification ,Triazoles ,030104 developmental biology ,A549 Cells ,Drug Resistance, Neoplasm ,Purines ,Cancer cell ,Mutation ,Cancer research ,business ,Carcinogenesis - Abstract
// Arefeh Rouhi 1 , Christina Miller 1 , Sarah Grasedieck 1 , Stefanie Reinhart 2 , Britta Stolze 2 , Hartmut Dohner 1 , Florian Kuchenbauer 1 , Lars Bullinger 1 , Stefan Frohling 2, 3, 4, * , Claudia Scholl 2, 4, * 1 Department of Internal Medicine III, Ulm University, Ulm, Germany 2 Department of Translational Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany 3 Section for Personalized Oncology, Heidelberg University Hospital, Heidelberg, Germany 4 German Cancer Consortium (DKTK), Heidelberg, Germany * shared senior authorship of this article Correspondence to: Claudia Scholl, email: claudia.scholl@nct-heidelberg.de Keywords: drug resistance, HSP90 inhibition, PU-H71, mutant KRAS, MDR1 Received: August 02, 2016 Accepted: November 30, 2016 Published: December 09, 2016 ABSTRACT Inhibition of the HSP90 chaperone results in depletion of many signaling proteins that drive tumorigenesis, such as downstream effectors of KRAS, the most commonly mutated human oncogene. As a consequence, several small-molecule HSP90 inhibitors are being evaluated in clinical trials as anticancer agents. To prospectively identify mechanisms through which HSP90-dependent cancer cells evade pharmacologic HSP90 blockade, we generated multiple mutant KRAS-driven cancer cell lines with acquired resistance to the purine-scaffold HSP90 inhibitor PU-H71. All cell lines retained dependence on HSP90 function, as evidenced by sensitivity to short hairpin RNA-mediated suppression of HSP90AA1 or HSP90AB1 (also called HSP90α and HSP90β, respectively), and exhibited two types of genomic alterations that interfere with the effects of PU-H71 on cell viability and proliferation: (i) a Y142N missense mutation in the ATP-binding domain of HSP90α that co-occurred with amplification of the HSP90AA1 locus, (ii) genomic amplification and overexpression of the ABCB1 gene encoding the MDR1 drug efflux pump. In support of a functional role for these alterations, exogenous expression of HSP90α Y142N conferred PU-H71 resistance to HSP90-dependent cells, and pharmacologic MDR1 inhibition with tariquidar or lowering ABCB1 expression restored sensitivity to PU-H71 in ABCB1-amplified cells. Finally, comparison with structurally distinct HSP90 inhibitors currently in clinical development revealed that PU-H71 resistance could be overcome, in part, by ganetespib (also known as STA9090) but not tanespimycin (also known as 17-AAG). Together, these data identify potential mechanisms of acquired resistance to small molecules targeting HSP90 that may warrant proactive screening for additional HSP90 inhibitors or rational combination therapies.
- Published
- 2016
38. Hoxa9 and Meis1 Cooperatively Induce Addiction to Syk Signaling by Suppressing miR-146a in Acute Myeloid Leukemia
- Author
-
Florian Kuchenbauer, Henning Urlaub, Hanibal Bohnenberger, R K Humphries, Carsten Müller-Tidow, Tobias Berg, Arefeh Rouhi, Federico Comoglio, Jasmin Corso, Lars Palmqvist, Thomas Oellerich, Anjali Cremer, Anthony R. Green, Kimberly Stegmaier, Philipp Ströbel, Halvard Bonig, Hubert Serve, Astrid Wachter, Frank Schnütgen, Joachim Beck, Ekkehard Schütz, Timm Schroeder, Sebastian Mohr, Tim Beissbarth, Stefanie Göllner, Michael A. Rieger, Nadine Haetscher, Carmen Doebele, Gabriela Alexe, Green, Tony [0000-0002-9795-0218], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,Hox genes ,Leukemia ,microRNA ,PU.1 ,Signal transduction ,Syk ,Context (language use) ,Kaplan-Meier Estimate ,Biology ,Article ,03 medical and health sciences ,Downregulation and upregulation ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Syk Kinase ,ddc:610 ,Myeloid Ecotropic Viral Integration Site 1 Protein ,Transcription factor ,Homeodomain Proteins ,Gene Expression Regulation, Leukemic ,Integrin beta3 ,leukemia ,Myeloid leukemia ,Hoxa9 ,Meis1 ,medicine.disease ,3. Good health ,Neoplasm Proteins ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cancer research ,signal transduction - Abstract
Summary The transcription factor Meis1 drives myeloid leukemogenesis in the context of Hox gene overexpression but is currently considered undruggable. We therefore investigated whether myeloid progenitor cells transformed by Hoxa9 and Meis1 become addicted to targetable signaling pathways. A comprehensive (phospho)proteomic analysis revealed that Meis1 increased Syk protein expression and activity. Syk upregulation occurs through a Meis1-dependent feedback loop. By dissecting this loop, we show that Syk is a direct target of miR-146a, whose expression is indirectly regulated by Meis1 through the transcription factor PU.1. In the context of Hoxa9 overexpression, Syk signaling induces Meis1, recapitulating several leukemogenic features of Hoxa9/Meis1-driven leukemia. Finally, Syk inhibition disrupts the identified regulatory loop, prolonging survival of mice with Hoxa9/Meis1-driven leukemia., Graphical Abstract, Highlights • Meis1 regulates expression and activation of Syk in Hoxa9-driven leukemia • Syk upregulation in Hoxa9/Meis1-driven leukemia is mediated by miR-146a • Increased Syk activity induces a Meis1 transcriptional program • In vivo, Meis1 sensitizes Hoxa9-driven leukemia to Syk inhibition, Mohr et al. identify a Meis1-dependent regulatory loop involving PU.1, miR-146a, and Syk, resulting in the addiction to Syk activity in Hoxa9/Meis1-transformed myeloid progenitor cells. Syk inhibition disrupts the regulatory loop and prolongs survival of mice with Hoxa9/Meis1-driven acute myeloid leukemia.
- Published
- 2016
39. Additional file 2: Figure S2. of PKM2 promotes tumor angiogenesis by regulating HIF-1Îą through NF-ÎşB activation
- Author
-
Azoitei, Ninel, Becher, Alexander, Steinestel, Konrad, Arefeh Rouhi, Diepold, Kristina, Genze, Felicitas, Simmet, Thomas, and Seufferlein, Thomas
- Subjects
hemic and lymphatic diseases ,circulatory and respiratory physiology - Abstract
IHC of pancreatic cancer cells growing on CAM using specific antibodies directed against desmin and von Willebrand factor (vWF) is presented. The images reveal both intratumoral and extratumoral immunoreactivity of the endothelial markers. (PPTX 4480 kb)
- Published
- 2016
- Full Text
- View/download PDF
40. Additional file 3: Figure S3. of PKM2 promotes tumor angiogenesis by regulating HIF-1Îą through NF-ÎşB activation
- Author
-
Azoitei, Ninel, Becher, Alexander, Steinestel, Konrad, Arefeh Rouhi, Diepold, Kristina, Genze, Felicitas, Simmet, Thomas, and Seufferlein, Thomas
- Subjects
hemic and lymphatic diseases ,cardiovascular system ,circulatory and respiratory physiology - Abstract
PKM2 abrogation results in decreased blood vessel formation in vivo. IHC of BxPC3 pancreatic cancer cells growing on CAM using specific antibodies for desmin and von Willebrand factor (vWF) is presented. (PPTX 3897 kb)
- Published
- 2016
- Full Text
- View/download PDF
41. Additional file 4: Figure S4. of PKM2 promotes tumor angiogenesis by regulating HIF-1α through NF-κB activation
- Author
-
Azoitei, Ninel, Becher, Alexander, Steinestel, Konrad, Arefeh Rouhi, Diepold, Kristina, Genze, Felicitas, Simmet, Thomas, and Seufferlein, Thomas
- Abstract
PKM2 regulates hypoxia-induced HIF-1α accumulation. A, B, pancreatic cancer cells transduced with a non-targeting control shRNA or PKM2-specific shRNAs were incubated under hypoxia or normoxia for 8 h. HIF-1α levels were determined using western blot analysis. Quantification of HIF-1α band intensity was conducted using ImageJ software (No – normoxia; Hy – hypoxia). (PPTX 72 kb)
- Published
- 2016
- Full Text
- View/download PDF
42. Additional file 5: Figure S5. of PKM2 promotes tumor angiogenesis by regulating HIF-1α through NF-κB activation
- Author
-
Azoitei, Ninel, Becher, Alexander, Steinestel, Konrad, Arefeh Rouhi, Diepold, Kristina, Genze, Felicitas, Simmet, Thomas, and Seufferlein, Thomas
- Subjects
animal structures - Abstract
PKM2 regulates hypoxia-induced VEGF promoter activity. A, PaTu2 cancer cells were transiently transfected with 3xHRE-luc and pTK-Renilla. Four hours after transfection cells were incubated under normoxic or hypoxic conditions in the absence or presence of 30 μM TEPP-46. Cell lysates were subjected to luciferase assay. Bars are the means +/- SEM of at least two independent experiments performed in duplicate. B, cancer cells were transiently transfected with VEGF-luc and pTK-Renilla. Four hours after transfection cells were incubated under normoxic or hypoxic conditions in the absence or presence of 30 μM TEPP-46. Cell lysates were subjected to luciferase assay. Bars are the means +/- SEM of at least two independent experiments performed in duplicate. C, D, PaTu2 and Capan1 cells were transiently transfected with VEGF-luc reporter. Four hours later cells were incubated under normoxic or hypoxic conditions in the absence or presence of 10 μM BAY 87-2243. Lysates were subjected to luciferase assay. Bars are the means +/- SEM of at least two independent experiments performed in duplicate (No – normoxia; Hy – hypoxia). (PPTX 138 kb)
- Published
- 2016
- Full Text
- View/download PDF
43. Additional file 1: Figure S1. of PKM2 promotes tumor angiogenesis by regulating HIF-1Îą through NF-ÎşB activation
- Author
-
Azoitei, Ninel, Becher, Alexander, Steinestel, Konrad, Arefeh Rouhi, Diepold, Kristina, Genze, Felicitas, Simmet, Thomas, and Seufferlein, Thomas
- Subjects
cardiovascular system - Abstract
34 human pancreatic specimens were stained with PKM2 and CD31 antibodies. Representative images are shown. (PPTX 760 kb)
- Published
- 2016
- Full Text
- View/download PDF
44. FOXtrotting with PUMILIOs
- Author
-
Florian Kuchenbauer and Arefeh Rouhi
- Subjects
0301 basic medicine ,Genetics ,Untranslated region ,Polyadenylation ,PUM1 ,Immunology ,Myeloid leukemia ,RNA-binding protein ,Cell Biology ,Hematology ,Cell cycle ,Biology ,medicine.disease ,Biochemistry ,03 medical and health sciences ,Haematopoiesis ,Leukemia ,030104 developmental biology ,hemic and lymphatic diseases ,medicine - Abstract
In this issue of Blood , [Naudin et al][1] investigated the functional relevance of PUMILIO 1 and 2 (PUM1 and PUM2), 2 highly conserved RNA-binding proteins (RBPs) and members of the PUM and FBF (PUF) family of posttranscriptional regulators, in early hematopoiesis and acute myeloid leukemia (AML).[
- Published
- 2017
45. Comprehensive analysis of mammalian miRNA* species and their role in myeloid cells
- Author
-
Yuzhuo Wang, Connie J. Eaves, Lars Bullinger, Michael Heuser, Akira Watahiki, Florian Kuchenbauer, R. Keith Humphries, Jürgen Krauter, Christian Buske, Samuel Aparicio, Bob Argiropoulos, Hartmut Döhner, Lars Palmqvist, Tobias Berg, Daniel T. Starczynowski, Aly Karsan, Arefeh Rouhi, Ryan D. Morin, Andrew McPherson, Marco A. Marra, Konstanze Döhner, Fernando D. Camargo, Jens Ruschmann, Sarah M Mah, Arnold Ganser, and David Lai
- Subjects
Adult ,Male ,Ribonuclease III ,DNA, Complementary ,Myeloid ,Adolescent ,Genetic Vectors ,Immunology ,Receptors, Cell Surface ,Biology ,Transfection ,Biochemistry ,DEAD-box RNA Helicases ,Small Molecule Libraries ,Mice ,Phosphatidylinositol 3-Kinases ,Genes, Reporter ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Gene silencing ,Insulin-Like Growth Factor I ,Luciferases ,Gene ,Myeloid Progenitor Cells ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Nucleic Acid Hybridization ,Myeloid leukemia ,Cell Differentiation ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Molecular biology ,Survival Rate ,Leukemia, Myeloid, Acute ,MicroRNAs ,Leukemia ,Retroviridae ,medicine.anatomical_structure ,Signal Transduction - Abstract
Processing of pre-miRNA through Dicer1 generates an miRNA duplex that consists of an miRNA and miRNA* strand. Despite the general view that miRNA*s have no functional role, we further investigated miRNA* species in 10 deep-sequencing libraries from mouse and human tissue. Comparisons of miRNA/miRNA* ratios across the miRNA sequence libraries revealed that 50% of the investigated miRNA duplexes exhibited a highly dominant strand. Conversely, 10% of miRNA duplexes showed a comparable expression of both strands, whereas the remaining 40% exhibited variable ratios across the examined libraries, as exemplified by miR-223/miR-223* in murine and human cell lines. Functional analyses revealed a regulatory role for miR-223* in myeloid progenitor cells, which implies an active role for both arms of the miR-223 duplex. This was further underscored by the demonstration that miR-223 and miR-223* targeted the insulin-like growth factor 1 receptor/phosphatidylinositol 3-kinase axis and that high miR-223* levels were associated with increased overall survival in patients with acute myeloid leukemia. Thus, we found a supporting role for miR-223* in differentiating myeloid cells in normal and leukemic cell states. The fact that the miR-223 duplex acts through both arms extends the complexity of miRNA-directed gene regulation of this myeloid key miRNA.
- Published
- 2011
46. Members of the microRNA-106a-363 Cluster Associate with Unfavorable Outcome in Adult Acute Myeloid Leukemia Patients and Promote Leukemogenesis invivo through Increased Metabolic Activity
- Author
-
Florian Kuchenbauer, Linda Fogelstrand, Sebastian Wiese, Tobias Maetzig, Hartmut Döhner, Nadine Sperb, Lars Palmqvist, Enrico Calzia, Nicole Pochert, Christoph Rueß, Sarah Grasedieck, Laleh Arabanian, Pegah Johansson, Edith Schneider, Kathrin Krowiorz, Erik Delsing Malmberg, Reinhild Rösler, and Arefeh Rouhi
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Immunology ,Complete remission ,Myeloid leukemia ,Adult Acute Myeloid Leukemia ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Transplantation ,03 medical and health sciences ,Leukemia ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,microRNA ,medicine ,Syngeneic transplantation ,Metabolic activity ,business ,030215 immunology - Abstract
The miR-106a-363 cluster, encoding six miRNAs (miR-106a, miR-18b, miR-20b, miR-19b, miR-92a and miR-363), is a paralogue of the oncogenic miR-17-92a polycistron and its role in leukemia is at present largely unknown. We aimed to investigate the putative oncogenic role of the miR-106a-363 cluster in adult acute myeloid leukemia (AML) and to dissect the contributions of its individual members to disease formation and progression. First, we analyzed the expression of each miRNA in AML patient samples as well as their clinical relevance. To determine the association of the miR-106a-363 cluster in AML with active disease, we quantified all six miRNAs individually in AML patient samples at initial diagnosis (n=33) and in AML patients in complete remission after chemotherapy (n=6). Hereof, miR-106a-5p, miR-19b-3p and miR-92a-3p levels were significantly lower in remission samples (p=0.0015, p=0.0013 and p=0.0004, respectively), confirming that these miRNAs are upregulated in AML. Stratifying AML patients within the LAML miRNA-Seq dataset of The Cancer Genome Atlas (TCGA) Research Network (n=187) (Ley et al., NEJM, 2013) according to their cytogenetic risk group demonstrated that all members of the cluster, except for miR-18b-5p, significantly associated with adverse cytogenetics. In addition, with the exception of miR-18b-5p, all members associated with an inferior overall survival (OS) in AML patients within the TCGA-LAML dataset, further supporting a pro-leukemogenic role for the cluster. Of note, miR-106a-5p was the most abundantly expressed unique miRNA of the polycistron, both in the TCGA patient cohort and in 11 myeloid leukemia cell lines quantified by quantitative real-time PCR (qRT-PCR). Since the miR-106a-363 cluster is associated with high risk AML, we hypothesized that increased levels of the entire cluster as well as individual members would significantly shorten the survival time in a murine transplantation model mimicking aggressive AML. Therefore, we engineered transplantable, primary murine AML cell lines based on retroviral overexpression of Hoxa9 and Meis1 exhibiting a median disease latency of 39 days (n=14) after syngeneic transplantation in mice. Enforced lentiviral expression of miR-106a-363 (n=13, p Based on these results, we focused on the mechanism by which miR-106a contributed to the pathogenesis of AML and performed a proteomics screen comparing Hoxa9/Meis1/miR-106a and Hoxa9/Meis1/control cells. In particular, mitochondrial respiration processes, such as oxidative phosphorylation and electron transport chain components were induced by miR-106a as shown by Gene Set Enrichment Analysis. Preliminary results using high-resolution respirometry further indicated an increased number of mitochondria in Hoxa9/Meis1/miR-106a cells, supporting these findings. In conclusion, we highlight the previously unrecognized oncogenic contribution of the miR-106a-363 polycistron in adult AML. Functional dissection of this cluster, in particular miR-106a, revealed a new therapeutic angle for high risk AML. Disclosures Döhner: Astellas: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria; Bristol Myers Squibb: Research Funding; Astex Pharmaceuticals: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Bristol Myers Squibb: Research Funding; Celator: Consultancy, Honoraria; Jazz: Consultancy, Honoraria; Astex Pharmaceuticals: Consultancy, Honoraria; Jazz: Consultancy, Honoraria; Celator: Consultancy, Honoraria; Pfizer: Research Funding; Sunesis: Consultancy, Honoraria, Research Funding; AROG Pharmaceuticals: Research Funding; Agios: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria; Astellas: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; AROG Pharmaceuticals: Research Funding; Seattle Genetics: Consultancy, Honoraria; Sunesis: Consultancy, Honoraria, Research Funding; Pfizer: Research Funding; Seattle Genetics: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding; Agios: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; Janssen: Consultancy, Honoraria.
- Published
- 2018
47. MiR-193a Is a Negative Regulator of Hematopoietic Stem Cells and Promotes Anti-Leukemic Effects in Acute Myeloid Leukemia
- Author
-
Florian Kuchenbauer, Kathrin Krowiorz, Hartmut Döhner, Lars Bullinger, Raj Bhayadia, R. Keith Humphries, Dirk Heckl, Arefeh Rouhi, Nicole Pochert, Vera C. Martins, Jan-Henning Klusmann, Christoph Rueß, Konstanze Döhner, Caroline Pabst, Sebastian Wiese, Stephan Emmrich, Christina Miller, Reinhild Rösler, Razan Jammal, and Tobias Maetzig
- Subjects
Myeloid ,business.industry ,Immunology ,CD34 ,Myeloid leukemia ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Biochemistry ,Granulocyte colony-stimulating factor ,Haematopoiesis ,medicine.anatomical_structure ,Cancer research ,Medicine ,Stem cell ,Progenitor cell ,business - Abstract
The microRNA (miRNA) family miR-193 consists of two members, miR-193a and miR-193b, which share identical seed regions and are therefore thought to be functionally redundant. However, the target spectrum of a miRNA is defined by the mRNA spectrum of the tissue, where it is expressed as well as dynamic strand preferences (miRNA-5p vs miRNA-3p) (Kuchenbauer et al., Blood, 2011). We and others previously showed that miR-193b is a tumor suppressor in acute myeloid leukemia (AML) (Bhayadia et al., JCO, 2018) and a potent regulator of hematopoietic stem cell expansion (Haetscher et al., Nat Com, 2015). However, the role of miR-193a in normal and malignant hematopoiesis is still unclear. First, we profiled the expression of miR-193a-5p and miR-193a-3p in human hematopoietic subpopulations from healthy donors via quantitative real-time PCR (qRT-PCR). Unlike miR-193b, which is enriched in hematopoietic stem cells (HSC) (Haetscher et al., Nat Com, 2015), miR-193a expression was restricted to differentiated cells of the myeloid lineage (granulocytes, monocytes, and erythrocytes). To investigate the function of miR-193a during normal hematopoiesis, we analyzed its effect on a murine myeloid progenitor cell line (32D cells) upon engineered overexpression of miR-193a in combination with granulocyte-stimulating growth factor (G-CSF) treatment (n=3-7). MiR-193a strongly promoted granulocytic differentiation of 32D cells already after two days compared to the control arm (p=0.006) as assessed by flow cytometry and morphological analysis. To analyze the effect of miR-193a on HSC function, we ectopically expressed miR-193a in highly purified E-SLAM (CD45+EPCR+CD48-CD150+) cells and transplanted them into lethally irradiated recipient mice (n=5 mice/arm). HSCs overexpressing miR-193a failed to reconstitute hematopoiesis (p=0.038). The negative effect on stem cell properties could be translated to human CD34+ cord blood (CB) cells, where miR-193a overexpression significantly reduced colony counts. Taken together, these results suggest, that miR-193a has pro-differentiation and anti-stemness functions. To assess the role of miR-193a in AML, we profiled miR-193a-5p and miR-193a-3p in two cohorts of de novo pediatric (n=187) and de novo adult AML patients (n=40) by qRT-PCR. We found that both miRNA strands (miR-193a-5p and miR-193a-3p) are present at similar levels, which is in contrast to the almost undetectable levels of miR-193b-5p strand, hinting at additional functional roles for miR-193a through its 5p arm. MiR-193a was significantly downregulated in adult and pediatric AML compared to healthy donor samples, suggesting a possible tumor suppressor function. To investigate the role of miR-193a in AML, we engineered transplantable, primary murine AML cell lines based on retroviral overexpression of Hoxa9/Meis1 (aggressive, short latency in vivo) with low endogenous miR-193a levels. Restoration of miR-193a by lentiviral overexpression delayed Hoxa9/Meis1 mediated leukemogenesis in vivo (p=0.01, n=8-10). Furthermore, miR-193a overexpression reduced leukemic growth of human AML cell lines (n=6) and decreased colony-forming capacity of primary AML patient samples (n=4; p=0.029) in vitro. To identify novel targets of miR-193a, we performed a proteomics screen in human NOMO1 AML cells overexpressing miR-193a compared to an empty control vector (n=5). We identified and verified stathmin (STMN1), a tubulin-associated, intracellular phosphoprotein previously linked to proliferation of AML cells, as a novel putative miR-193a target, further explaining its tumor suppressor effect. Taken together, we characterized miR-193a as a positive regulator of myeloid differentiation and negative modulator of HSCs. Based on the balanced presence of both miRNA arms, we hypothesize that each arm is functionally active and has different functions such as pro-differentiation and anti-stemness. Furthermore, this work is the first characterization of a miRNA family that exerts cooperative effects at both early and late hematopoietic differentiation stages, highlighting a novel mechanism of balancing anti-stemness and pro-differentiation. Disclosures Döhner: Bristol Myers Squibb: Research Funding; AROG Pharmaceuticals: Research Funding; Sunesis: Consultancy, Honoraria, Research Funding; Seattle Genetics: Consultancy, Honoraria; Agios: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Jazz: Consultancy, Honoraria; Agios: Consultancy, Honoraria; Pfizer: Research Funding; Janssen: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; Astellas: Consultancy, Honoraria; Astex Pharmaceuticals: Consultancy, Honoraria; AROG Pharmaceuticals: Research Funding; Astellas: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Celator: Consultancy, Honoraria; Astex Pharmaceuticals: Consultancy, Honoraria; Bristol Myers Squibb: Research Funding; Celator: Consultancy, Honoraria; Jazz: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Sunesis: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; Pfizer: Research Funding; Seattle Genetics: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Research Funding; Celgene: Consultancy, Honoraria, Research Funding. Bullinger:Bristol-Myers Squibb: Speakers Bureau; Pfizer: Speakers Bureau; Bayer Oncology: Research Funding; Sanofi: Research Funding, Speakers Bureau; Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen: Honoraria, Speakers Bureau; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Janssen: Speakers Bureau.
- Published
- 2018
48. Identification of Novel Lncrnas That Predict Survival in AML Patients and Modulate Leukemic Cells
- Author
-
Jonathan R. Pollack, Florian Kuchenbauer, Susanne Lux, Jan-Henning Klusmann, Arefeh Rouhi, Mojca Jongen-Lavrencic, Nicole Pochert, Christoph Rueß, Sarah Grasedieck, and Adrian Schwarzer
- Subjects
0301 basic medicine ,NPM1 ,Myeloid ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biology ,Core binding factor ,Biochemistry ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Tretinoin ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Bone marrow ,Myelopoiesis ,medicine.drug - Abstract
Long noncoding RNAs (lncRNAs) have complex, mainly chromatin-associated functions and their expression is highly coordinated and cell-type specific. Based on their tight regulation in normal differentiation, we set out to investigate whether lncRNAs are dysregulated in diseases where differentiation is impaired, such as in acute myeloid leukemia (AML). To identify lncRNAs that are essential for both normal hematopoiesis as well as AML maintenance, we sequenced the long polyA- and non-polyA-tagged transcriptome from successive stages of human myelopoiesis (myeloblasts, promyelocytes, metamyelocytes, and neutrophils) isolated from bone marrow of healthy donors (n=3). Applying a high-dimensional data portraying approach (OposSOM, Löffler-Wirth et al., BMC Bioinformatics, 2015), we identified functional expression modules of lncRNAs that are either positively or negatively associated with myeloid lineage commitment in our dataset. Seven out of the top15 differentiation-associated lncRNAs exhibit significant prognostic relevance in overall and event-free survival analyses of independent AML patient datasets and improve the predictive power of the current prognosis standards (cytogenetic risk/age/TP53-status). In particular, a combination of 3 transcripts, PROMYS (Promoter of Myelopoiesis, annotated as uncharacterized ncRNA LOC107985167), ANTAMY (Antagonist of Myelopoiesis, uncharacterized ncRNA LOC101927745) and LINC00677, outperformed the recently reported prognostic benefit of the LSC17high score (Ng et al, Nature, 2016) by a factor of Ø 22.7 based on concordance index score increase (Ø 4.8% vs. 0.21%). All three lncRNAs are highly conserved, expressed in 10 tested human AML cell lines as well as significantly differentially expressed in distinct cytogenetic patient subgroups of The Cancer Genome Atlas (TCGA) LAML cohort (n=171). PROMYS is downregulated in t(15;17) and t(8;21) cases, supporting its strong association with worse OS in the TCGA-LAML dataset (p=0.0001). In contrast, ANTAMY shows high expression in AML with t(8;21), and LINC00677 in NPM1+/FLT3- mutated AML patient samples with normal karyotype (CN-AML) and in core Binding factor (CBF) AMLs. Accordingly, high expression levels of both lncRNAs associate with a significantly better OS in the TCGA LAML dataset (p=0.01 and 0.02, respectively). To investigate their function in vitro, we knocked out each lncRNA individually in the human OCI/AML-5 AML cell line using CRISPR/Cas9. Loss of ANTAMY impaired proliferation (p=0.04) and increased both monocytic differentiation upon treatment with 2-0-tetradecanoylphorbol-13-acetate (TPA) (p=0.0001) and granulocytic differentiation with all-trans retinoic acid (ATRA) (p=0.0002) compared to the empty vector control. Loss of LINC00677 in OCI/AML-5 cells specifically increased granulocytic differentiation through ATRA (p=0.0002). In contrast, inactivation of PROMYS led to reduced differentiation induced by ATRA (p=0.00004) and TPA (p=0.002). Furthermore, we found that PROMYS is involved in the regulation of the Macrophage colony-stimulating factor 1 (CSF1), which is deregulated in ATRA- and TPA-induced differentiation in PROMYS knockout but not in control cells (p Disclosures No relevant conflicts of interest to declare.
- Published
- 2018
49. Variable DNA methylation of transposable elements: The case study of mouse Early Transposons
- Author
-
Max Reuter, Ying Zhang, Dixie L. Mager, Arefeh Rouhi, and Daphne Reiss
- Subjects
Male ,Cancer Research ,Endogenous retrovirus ,Locus (genetics) ,Retrotransposon ,Biology ,Epigenesis, Genetic ,Mice ,Genetic variation ,Animals ,Tissue Distribution ,Epigenetics ,Molecular Biology ,Genetics ,Stochastic Processes ,Genome ,Models, Genetic ,Genetic Variation ,Methylation ,DNA Methylation ,Mice, Inbred C57BL ,Phenotype ,CpG site ,DNA methylation ,DNA Transposable Elements ,RNA ,CpG Islands - Abstract
Phenotypic variation stems from both genetic and epigenetic differences between individuals. In order to elucidate how phenotypes are determined, it is necessary to understand the forces that generate variation in genome sequence as well as its epigenetic state. In both contexts, transposable elements (TEs) may play an important role. It is well established that TE activity is a major generator of genetic variation, but recent research also suggests that TEs contribute to epigenetic variation. Stochastic epigenetic silencing of some TE insertions in mice has been shown to cause phenotypic variability between individuals. However, the prevalence of this phenomenon has never been evaluated. Here, we use 18 insertions of a mouse Endogenous Retrovirus (ERV) family, the Early Transposons (ETns), to detect insertion-dependent determinants of DNA methylation levels and variability between both cells and individuals. We show that the structure and age of insertions influence methylation levels and variability, resulting in a subgroup of loci that displays unexpectedly high variability in methylation and suggesting stochastic events during methylation establishment. Despite variation in methylation according to the age and structure of each locus, homologous CpG sites show similar tendencies in methylation levels across loci, emphasizing the role of the insertion's sequence in methylation determination. Our results show that differences in methylation of ETns between individuals is not a sporadic phenomenon and support the hypothesis that ERVs contribute to phenotypic variability through their stochastic silencing.
- Published
- 2010
50. Evidence for high bi-allelic expression of activating Ly49 receptors
- Author
-
Wayne M. Yokoyama, Tammy P. Cheng, C. Benjamin Lai, Fumio Takei, Dixie L. Mager, and Arefeh Rouhi
- Subjects
Transcription, Genetic ,T-Lymphocytes ,Molecular Sequence Data ,Biology ,Gene Regulation, Chromatin and Epigenetics ,Major histocompatibility complex ,Genome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Sequence Homology, Nucleic Acid ,Genetics ,Transcriptional regulation ,Gene family ,Animals ,Allele ,Receptor ,Gene ,Alleles ,030304 developmental biology ,0303 health sciences ,Base Sequence ,DNA Methylation ,Mice, Inbred C57BL ,DNA methylation ,biology.protein ,5' Untranslated Regions ,NK Cell Lectin-Like Receptor Subfamily A ,030215 immunology - Abstract
Stochastic expression is a hallmark of the Ly49 family that encode the main MHC class-I-recognizing receptors of mouse natural killer (NK) cells. This highly polygenic and polymorphic family includes both activating and inhibitory receptor genes and is one of genome's fastest evolving loci. The inhibitory Ly49 genes are expressed in a stochastic mono-allelic manner, possibly under the control of an upstream bi-directional early promoter and show mono-allelic DNA methylation patterns. To date, no studies have directly addressed the transcriptional regulation of the activating Ly49 receptors. Our study shows differences in DNA methylation pattern between activating and inhibitory genes in C57BL/6 and F1 hybrid mouse strains. We also show a bias towards bi-allelic expression of the activating receptors based on allele-specific single-cell RT-PCR in F1 hybrid NK cells for Ly49d and Ly49H expression in Ly49h(+/-) mice. Furthermore, we have identified a region of high sequence identity with possible transcriptional regulatory capacity for the activating Ly49 genes. Our results also point to a likely difference between NK and T-cells in their ability to transcribe the activating Ly49 genes. These studies highlight the complex regulation of this rapidly evolving gene family of central importance in mouse NK cell function.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.