29 results on '"R. Hoogenboezem"'
Search Results
2. S887 DISSECTING THE ROLE OF CXCL4 IN PRIMARY MYELOFIBROSIS
- Author
-
J. Saez-Rodriguez, H. Gleitz, S. Fuchs, A. Dugourd, J.E. Pritchard, Eric M.J. Bindels, Rebekka K. Schneider, R. Hoogenboezem, I. Snoeren, R. Kramann, B. Banjanin, and N. Leimkühler
- Subjects
Oncology ,medicine.medical_specialty ,Primary (chemistry) ,business.industry ,Internal medicine ,Medicine ,Hematology ,business ,Myelofibrosis ,medicine.disease - Published
- 2019
- Full Text
- View/download PDF
3. Introduction of an embolus detection system based on analysis of the transcranial Doppler audio-signal
- Author
-
R. W. M. Keunen, R. G. A. Ackerstaff, R. Wijnands, R. Hoogenboezem, and A. C. M. Van den Hengel
- Subjects
Sound Spectrography ,Audio signal ,Artificial neural network ,Ultrasonography, Doppler, Transcranial ,business.industry ,Biomedical Engineering ,Process (computing) ,Reproducibility of Results ,General Medicine ,Transient ischaemic attacks ,Sensitivity and Specificity ,Signal ,Transcranial Doppler ,Software ,Intracranial Embolism ,Image Interpretation, Computer-Assisted ,Data file ,Humans ,Medicine ,Computer vision ,Artificial intelligence ,business ,Algorithms ,Biomedical engineering - Abstract
A new embolus detection system (EDS) is presented, built with the intention of detecting ongoing cerebral embolization in patients at risk of transient ischaemic attacks or stroke. It is based on the analysis of the audio-Doppler signal of a transcranial Doppler machine. The algorithm of the EDS estimates the intensity, duration and zero-crossing dynamics of the audio signal. The EDS has a multi-layer neural network which classifies events into micro-emboli signals (MES) or artefacts. The decision-making component of the software has been validated against human experts. Data from patients in the post-operative phase of carotid surgery were used for the validation process. The results showed agreement in MES and artefact classification of93%. Apart from a monitoring display, the monitoring system includes a verification unit that allows the user to listen and to look at all data of individual MES and artefacts. Moreover, the system allows the user to record, store and re-calculate all data files. Data are stored using European Data Format, which allows data transportation over the Internet. The EDS may have a potential in stroke risk stratification, evaluating the effect of novel anti-thrombotic therapies, and in peri-operative and remote monitoring of carotid endarterectomy.
- Published
- 2008
- Full Text
- View/download PDF
4. M3P sequencing panel identifies TP53 mutational status as a prognostic factor in chemotherapy-naive multiple myeloma
- Author
-
D.H., Op Bruinink, K.M., Kortum, M., Van Duin, M.A., Sanders, R., Hoogenboezem, M., Vermeulen, L., Bruins, J., Ahmann, A., Broijl, S., Zweegman, E., Braggio, B., Van Der Holt, A.K., Stewart, CCA - Treatment and quality of life, CCA - Imaging and biomarkers, CCA - Cancer biology and immunology, and Hematology
- Abstract
Embedded Image Introduction Multiple myeloma (MM) is characterized by a highly variable disease course, which can be traced to initiating and acquired genomic events. Whole exome analysis of matched tumor and germline DNA from 287 MM patients identified recurrently somatically mutated genes (RSMGs) (Lohr et al. - Cancer Cell 2014, Bolli et al. - Nat Commun 2014). Despite the fact that these RSMGs affect pathways that are biologically important in MM, the clinical relevance of many of these genes in the context of conventional prognostic markers remains to be elucidated. Aims The aims of this pilot study were: (1) To validate the prevalence of RSMGs in our newly diagnosed MM patient cohort; (2) To assess the correlation between RSMGs, clinical parameters and outcome; (3) To thereby identify the potential clinical usefulness of introducing RSMG mutational profiling in larger MM trial cohorts. Material and Methods CD138+ enriched MM cells and peripheral blood were obtained with informed consent from chemotherapy-naive patients, participating in 3 clinical trials: HOVON-65/GMMG-HD4, HOVON-87/NMSG-18 and Carthadex (EudraCT number 2004-000944-26, 2007-004007-34 and 2009-014922-40, respectively). Matched tumor and germline DNA were sequenced on an Ion Torrent sequencing platform (PGM, Life Technologies), using the M3 P Mutational Panel v3.0, comprising 1327 customized oligos (Life Technologies), targeted at the coding sequences of 88 MM-relevant genes, including the RSMGs. Somatic mutations were considered positive when present in >=10% of tumor reads and 1 year (n=23 versus n=79), only showed a significant correlation with TP53 mutational status (adj. p-value=0,012). TP53 mutational status remained the only significant prognostic factor when comparing patients with an OS 1 year (adj. p-value=0,003; n=13 versus n=89). When comparing the number of mutated genes, del17p and t(4;14) status, EMC92 score, transplant versus non-transplant protocol and ISS stage between TP53 mutated and wildtype MM, TP53 mutated patients had a significantly higher number of mutated genes in the M3 P panel (adj. p-value=0,001). Conclusions (1) With the M3 P Mutational Panel, we confirm the published prevalence of RSMGs in MM in our cohort of chemotherapy-naive patients. NRAS, KRAS, DIS3, FAM46C, TP53 and BRAF are the most frequently mutated genes. (2) TP53 mutational status is the strongest unfavorable prognostic factor in our cohort and it seems to be associated with greater mutational burden. Validation in a more extensive population is planned. (3) This warrants further investigation of the mutational status of these genes in larger clinical trial cohorts, enabling a more robust comparison with conventional prognostic markers in a multivariate analysis.
- Published
- 2015
5. The Association of Mutations in RUNX1 and CSF3R with the Development of Leukemia in Severe Congenital Neutropenia: A unique pathway in leukemogenesis
- Author
-
M Ünalan, Siarhei Kandabarau, D Steinemann, Martin Stanulla, Gudrun Göhring, R Hoogenboezem, Susanne Schnittger, Brigitte Schlegelberger, Karl Welte, A Kohlmann, C. Zeidler, P Vandenberghe, R Beekman, David C. Dale, Julia Skokowa, Jean Donadieu, Maksim Klimiankou, MG Valkhof, V. Makaryan, and IP Touw
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.disease ,chemistry.chemical_compound ,Leukemia ,RUNX1 ,chemistry ,Internal medicine ,Pediatrics, Perinatology and Child Health ,medicine ,Cancer research ,business ,Congenital Neutropenia - Published
- 2013
- Full Text
- View/download PDF
6. Cooperativity Of RUNX1 and CSF3R Mutations In The Development Of Leukemia In Severe Congenital Neutropenia: A Unique Pathway In Myeloid Leukemogenesis
- Author
-
Julia Skokowa, Doris Steinemann, Jenny Katsman-Kuipers, Cornelia Zeidler, Olga Klimenkova, Maksim Klimiankou, Murat Uenalan, Siarhei Kandabarau, Vahagn Makaryan, Renee Beekman, Carol Stocking, J Obenauer, Susanne Schnittger, Alexander Kohlmann, M Valkhof, R Hoogenboezem, Gudrun Göhring, Dirk Reinhardt, Brigitte Schlegelberger, Martin Stanulla, Peter Vandenberghe, Jean Donadieu, Michel Zwaan, Ivo P. Touw, Marry M. van den Heuvel-Eibrink, David C. Dale, and Karl Welte
- Subjects
hemic and lymphatic diseases ,embryonic structures ,Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Congenital neutropenia (CN) is a rare inherited disorder of hematopoiesis with a 20% risk of evolving into acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Using next-generation sequencing in 31 CN patients who developed leukemia we found that 20 of the 31 patients (64.5%) had mutations in RUNX1 (runt-related transcription factor 1). Of these 20 patients, 19 had inherited mutations associated with CN. Intriguingly, the majority of patients with RUNX1 mutations (80.5%) also had acquired CSF3R (colony stimulating factor 3 receptor) mutations. Other leukemia-associated mutations (EP300, FLT3-ITD, CBL, and SUZ12) were less frequent. In eight patients, we detected two distinct heterozygous RUNX1 mutations. These mutations were localized to the splice-acceptor site of intron 4, affecting splicing of exons 3 and 4, which encode the Runt homology/DNA binding domain (RHD) of RUNX1, or solely in the RHD or were present in both RHD and trans-activation domain (TAD). In two patients, we were able to perform allele-specific analysis of RUNX1 mutations. Patient #10 had an Phe13TrpfsX14 deletion on one allele of RUNX1 and an Arg139ProfsX47 deletion on the other allele. In Patient #14, two RUNX1 mutations were on the same allele; one of the mutations (Met240Ile) was inherited from the mother and was localized two amino acids before the TAD, and the second acquired mutation (Arg139Gly) was in the RHD of RUNX1. Ten patients with RUNX1 mutations developed monosomy 7 and six patients developed trisomy 21 at diagnosis of leukemia. In contrast to their high frequency in CN evolving into AML, RUNX1 mutations were found in only 9 of 307 (2.9%) patients with de novo pediatric AML. RUNX1 mutations were mainly found in pediatric AML patients with an adverse prognosis. A sequential analysis at stages prior to overt leukemia in ten CN/AML patients showed that RUNX1 mutation is a late event in leukemogenic transformation. In 6 of 10 patients, a CSF3R mutation occurred prior to RUNX1 mutations (24-192 months prior to CN/AML for CSF3R mutations vs. 1-36 months prior to CN/AML for RUNX1 mutations). Interestingly, monosomy 7 or trisomy 21 appeared after acquisition of RUNX1 mutations and no additional chromosomal aberrations were detected by array-CGH. Single-cell analyses in two patients revealed that RUNX1 and CSF3R mutations were segregated in the same malignant clone. Moreover, functional studies demonstrated elevated G-CSF-induced proliferation with diminished myeloid differentiation of hematopoietic CD34+ cells after co-transduction with mutated RUNX1 and CSF3R, in comparison to cells transduced with mutated RUNX1 or mutated CSF3R only. The importance of RUNX1 mutations in leukemogenic transformation was substantially strengthened by the analysis of a unique family with two siblings suffering from CN that subsequently transformed to AML. In both children, cooperating RUNX1 and CSF3Rmutations were detected that were not present in healthy family members. Taken together, the high frequency and the time course of cooperating RUNX1 and CSF3R mutations in CN patients who developed leukemia suggests a unique molecular pathway of leukemogenesis similar as has been reported in the Gilliland-Griffin two-hit hypothesis for AML development. The concomitant detection of RUNX1 and CSF3Rmutations represents a useful biomarker for identifying CN patients with a high risk of progressing to leukemia or MDS. Disclosures: Schnittger: MLL Munich Leukemia Laboratory: Employment, Equity Ownership. Kohlmann:MLL Munich Leukemia Laboratory: Employment.
- Published
- 2013
- Full Text
- View/download PDF
7. Interferon gamma-mediated prevention of tumor progression in a mouse model of multiple myeloma.
- Author
-
Kellermayer Z, Tahri S, de Jong MME, Papazian N, Fokkema C, Stoetman ECG, Hoogenboezem R, van Beek G, Sanders MA, Boon L, Den Hollander C, Broijl A, Sonneveld P, and Cupedo T
- Abstract
Malignant plasma cells in multiple myeloma patients reside in the bone marrow and continuously interact with local immune cells. Progression and therapy response are influenced by this immune environment, highlighting the need for a detailed understanding of endogenous immune responses to malignant plasma cells. Here we used the 5TGM1 murine transfer model of multiple myeloma to dissect early immune responses to myeloma cells. We modeled stable and progressive disease by transferring 5TGM1 murine myeloma cells into C57Bl/6 mice and KaLwRij mice, respectively. We used flow cytometry and single-cell and bulk transcriptomic analyses to characterize differential immune responses in stable and progressive disease. Transfer of 5TGM1 cells in C57Bl/6 mice led to stable disease with low tumor burden in a subset of animals. Stable disease was associated with sustained activation and expansion of NK cells, ILC1, and CD8
+ T cells, a response that was lost upon disease progression. Single-cell RNA-sequencing of immune cells and bulk RNA sequencing of immune and mesenchymal stromal cells implicated the activation of interferon responses as a central immune pathway during stable disease. Experimentally, neutralization of IFNγ significantly increased myeloma development and progression in C57Bl/6 mice, testifying to the importance of this pathway in early disease control. In conclusion, we provide a framework for studying immune responses to multiple myeloma progression in immunocompetent and genetically modifiable mice and highlight the importance of bone marrow immunity in tumor control., Competing Interests: Annemiek Broijl consults for BMS/Celgene, Janssen, Amgen, and Sanofi. Pieter Sonneveld is on the advisory board for Amgen, BMS/Celgene, Janssen, Seagen, and Pfizer and receives research support from Janssen, BMS/Celgene, and Karyopharm., (© 2024 The Author(s). HemaSphere published by John Wiley & Sons Ltd on behalf of European Hematology Association.)- Published
- 2024
- Full Text
- View/download PDF
8. GATA2 heterozygosity causes an epigenetic feedback mechanism resulting in myeloid and erythroid dysplasia.
- Author
-
Gioacchino E, Zhang W, Koyunlar C, Zink J, de Looper H, Gussinklo KJ, Hoogenboezem R, Bosch D, Bindels E, Touw IP, and de Pater E
- Subjects
- Animals, Zebrafish Proteins genetics, Erythroid Cells metabolism, Erythroid Cells pathology, Myeloid Cells metabolism, Myeloid Cells pathology, Erythropoiesis genetics, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology, Myelodysplastic Syndromes metabolism, Zebrafish, GATA2 Transcription Factor genetics, Epigenesis, Genetic, Heterozygote
- Abstract
The transcription factor GATA2 has a pivotal role in haematopoiesis. Heterozygous germline GATA2 mutations result in a syndrome characterized by immunodeficiency, bone marrow failure and predispositions to myelodysplastic syndrome (MDS) and acute myeloid leukaemia. Clinical symptoms in these patients are diverse and mechanisms driving GATA2-related phenotypes are largely unknown. To explore the impact of GATA2 haploinsufficiency on haematopoiesis, we generated a zebrafish model carrying a heterozygous mutation of gata2b (gata2b
+/- ), an orthologue of GATA2. Morphological analysis revealed myeloid and erythroid dysplasia in gata2b+/- kidney marrow. Because Gata2b could affect both transcription and chromatin accessibility during lineage differentiation, this was assessed by single-cell (sc) RNA-seq and single-nucleus (sn) ATAC-seq. Sn-ATAC-seq showed that the co-accessibility between the transcription start site (TSS) and a -3.5-4.1 kb putative enhancer was more robust in gata2b+/- zebrafish HSPCs compared to wild type, increasing gata2b expression and resulting in higher genome-wide Gata2b motif use in HSPCs. As a result of increased accessibility of the gata2b locus, gata2b+/- chromatin was also more accessible during lineage differentiation. scRNA-seq data revealed myeloid differentiation defects, that is, impaired cell cycle progression, reduced expression of cebpa and cebpb and increased signatures of ribosome biogenesis. These data also revealed a differentiation delay in erythroid progenitors, aberrant proliferative signatures and down-regulation of Gata1a, a master regulator of erythropoiesis, which worsened with age. These findings suggest that cell-intrinsic compensatory mechanisms, needed to obtain normal levels of Gata2b in heterozygous HSPCs to maintain their integrity, result in aberrant lineage differentiation, thereby representing a critical step in the predisposition to MDS., (© 2024 The Author(s). British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
9. Oncogene EVI1 drives acute myeloid leukemia via a targetable interaction with CTBP2.
- Author
-
Pastoors D, Havermans M, Mulet-Lazaro R, Brian D, Noort W, Grasel J, Hoogenboezem R, Smeenk L, Demmers JAA, Milsom MD, Enver T, Groen RWJ, Bindels E, and Delwel R
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Co-Repressor Proteins metabolism, Co-Repressor Proteins genetics, Protein Binding, Transcription Factors metabolism, Transcription Factors genetics, Alcohol Oxidoreductases metabolism, Alcohol Oxidoreductases genetics, DNA-Binding Proteins metabolism, DNA-Binding Proteins genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, MDS1 and EVI1 Complex Locus Protein metabolism, MDS1 and EVI1 Complex Locus Protein genetics
- Abstract
Acute myeloid leukemia (AML) driven by the activation of EVI1 due to chromosome 3q26/ MECOM rearrangements is incurable. Because transcription factors such as EVI1 are notoriously hard to target, insight into the mechanism by which EVI1 drives myeloid transformation could provide alternative avenues for therapy. Applying protein folding predictions combined with proteomics technologies, we demonstrate that interaction of EVI1 with CTBP1 and CTBP2 via a single PLDLS motif is indispensable for leukemic transformation. A 4× PLDLS repeat construct outcompetes binding of EVI1 to CTBP1 and CTBP2 and inhibits proliferation of 3q26/ MECOM rearranged AML in vitro and in xenotransplant models. This proof-of-concept study opens the possibility to target one of the most incurable forms of AML with specific EVI1-CTBP inhibitors. This has important implications for other tumor types with aberrant expression of EVI1 and for cancers transformed by different CTBP-dependent oncogenic transcription factors.
- Published
- 2024
- Full Text
- View/download PDF
10. SRSF2-P95H decreases JAK/STAT signaling in hematopoietic cells and delays myelofibrosis development in mice.
- Author
-
Willekens C, Laplane L, Dagher T, Benlabiod C, Papadopoulos N, Lacout C, Rameau P, Catelain C, Alfaro A, Edmond V, Signolle N, Marchand V, Droin N, Hoogenboezem R, Schneider RK, Penson A, Abdel-Wahab O, Giraudier S, Pasquier F, Marty C, Plo I, Villeval JL, Constantinescu SN, Porteu F, Vainchenker W, and Solary E
- Subjects
- Animals, Mice, Janus Kinase 2 genetics, Janus Kinases genetics, Mutation, RNA-Binding Proteins genetics, Signal Transduction, STAT Transcription Factors genetics, Hematopoietic Stem Cell Transplantation, Myeloproliferative Disorders genetics, Primary Myelofibrosis genetics
- Abstract
Heterozygous mutation targeting proline 95 in Serine/Arginine-rich Splicing Factor 2 (SRSF2) is associated with V617F mutation in Janus Activated Kinase 2 (JAK2) in some myeloproliferative neoplasms (MPNs), most commonly primary myelofibrosis. To explore the interaction of Srsf2
P95H with Jak2V617F , we generated Cre-inducible knock-in mice expressing these mutants under control of the stem cell leukemia (Scl) gene promoter. In transplantation experiments, Srsf2P95H unexpectedly delayed myelofibrosis induced by Jak2V617F and decreased TGFβ1 serum level. Srsf2P95H reduced the competitiveness of transplanted Jak2V617F hematopoietic stem cells while preventing their exhaustion. RNA sequencing of sorted megakaryocytes identified an increased number of splicing events when the two mutations were combined. Focusing on JAK/STAT pathway, Jak2 exon 14 skipping was promoted by Srsf2P95H , an event detected in patients with JAK2V617F and SRSF2P95 co-mutation. The skipping event generates a truncated inactive JAK2 protein. Accordingly, Srsf2P95H delays myelofibrosis induced by the thrombopoietin receptor agonist Romiplostim in Jak2 wild-type animals. These results unveil JAK2 exon 14 skipping promotion as a strategy to reduce JAK/STAT signaling in pathological conditions., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
11. Gata2-regulated Gfi1b expression controls endothelial programming during endothelial-to-hematopoietic transition.
- Author
-
Koyunlar C, Gioacchino E, Vadgama D, de Looper H, Zink J, Ter Borg MND, Hoogenboezem R, Havermans M, Sanders MA, Bindels E, Dzierzak E, Touw IP, and de Pater E
- Subjects
- Pregnancy, Female, Animals, Mice, Cell Differentiation, Hematopoietic Stem Cells metabolism, Transcription Factors genetics, Proto-Oncogene Proteins genetics, Repressor Proteins genetics, GATA2 Transcription Factor genetics, GATA2 Transcription Factor metabolism, Zebrafish metabolism, GATA2 Deficiency
- Abstract
The first hematopoietic stem cells (HSCs) are formed through endothelial-to-hematopoietic transition (EHT) during embryonic development. The transcription factor GATA2 is a crucial regulator of EHT and HSC function throughout life. Because patients with GATA2 haploinsufficiency have inborn mutations, prenatal defects are likely to influence disease development. In mice, Gata2 haploinsufficiency (Gata2+/-) reduces the number and functionality of embryonic hematopoietic stem and progenitor cells (HSPCs) generated through EHT. However, the embryonic HSPC pool is heterogeneous and the mechanisms underlying this defect in Gata2+/- embryos remain unclear. Here, we investigated whether Gata2 haploinsufficiency selectively affects a cellular subset undergoing EHT. We showed that Gata2+/- HSPCs initiate, but cannot fully activate, hematopoietic programming during EHT. In addition, due to the reduced activity of the endothelial repressor Gfi1b, Gata2+/- HSPCs cannot repress endothelial identity to complete maturation. Finally, we showed that hematopoietic-specific induction of gfi1b could restore HSC production in gata2b-null (gata2b-/-) zebrafish embryos. This study illustrates the pivotal role of Gata2 in the regulation of the transcriptional network governing HSPC identity throughout the EHT., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Adult human kidney organoids originate from CD24 + cells and represent an advanced model for adult polycystic kidney disease.
- Author
-
Xu Y, Kuppe C, Perales-Patón J, Hayat S, Kranz J, Abdallah AT, Nagai J, Li Z, Peisker F, Saritas T, Halder M, Menzel S, Hoeft K, Kenter A, Kim H, van Roeyen CRC, Lehrke M, Moellmann J, Speer T, Buhl EM, Hoogenboezem R, Boor P, Jansen J, Knopp C, Kurth I, Smeets B, Bindels E, Reinders MEJ, Baan C, Gribnau J, Hoorn EJ, Steffens J, Huber TB, Costa I, Floege J, Schneider RK, Saez-Rodriguez J, Freedman BS, and Kramann R
- Subjects
- Adult, Humans, TRPP Cation Channels genetics, Organoids, Kidney, CD24 Antigen genetics, Polycystic Kidney, Autosomal Dominant genetics, Cysts
- Abstract
Adult kidney organoids have been described as strictly tubular epithelia and termed tubuloids. While the cellular origin of tubuloids has remained elusive, here we report that they originate from a distinct CD24
+ epithelial subpopulation. Long-term-cultured CD24+ cell-derived tubuloids represent a functional human kidney tubule. We show that kidney tubuloids can be used to model the most common inherited kidney disease, namely autosomal dominant polycystic kidney disease (ADPKD), reconstituting the phenotypic hallmark of this disease with cyst formation. Single-cell RNA sequencing of CRISPR-Cas9 gene-edited PKD1- and PKD2-knockout tubuloids and human ADPKD and control tissue shows similarities in upregulation of disease-driving genes. Furthermore, in a proof of concept, we demonstrate that tolvaptan, the only approved drug for ADPKD, has a significant effect on cyst size in tubuloids but no effect on a pluripotent stem cell-derived model. Thus, tubuloids are derived from a tubular epithelial subpopulation and represent an advanced system for ADPKD disease modeling., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
13. Identification of High-Risk Multiple Myeloma With a Plasma Cell Leukemia-Like Transcriptomic Profile.
- Author
-
Hofste Op Bruinink D, Kuiper R, van Duin M, Cupedo T, van der Velden VHJ, Hoogenboezem R, van der Holt B, Beverloo HB, Valent ET, Vermeulen M, Gay F, Broijl A, Avet-Loiseau H, Munshi NC, Musto P, Moreau P, Zweegman S, van de Donk NWCJ, and Sonneveld P
- Subjects
- Humans, Prognosis, Transcriptome, Treatment Outcome, Leukemia, Plasma Cell genetics, Multiple Myeloma drug therapy
- Abstract
Purpose: Primary plasma cell leukemia (pPCL) is an aggressive subtype of multiple myeloma, which is distinguished from newly diagnosed multiple myeloma (NDMM) on the basis of the presence of ≥ 20% circulating tumor cells (CTCs). A molecular marker for pPCL is currently lacking, which could help identify NDMM patients with high-risk PCL-like disease, despite not having been recognized as such clinically., Methods: A transcriptomic classifier for PCL-like disease was bioinformatically constructed and validated by leveraging information on baseline CTC levels, tumor burden, and tumor transcriptomics from 154 patients with NDMM included in the Cassiopeia or HO143 trials and 29 patients with pPCL from the EMN12/HO129 trial. Its prognostic value was assessed in an independent cohort of 2,139 patients with NDMM from the HOVON-65/GMMG-HD4, HOVON-87/NMSG-18, EMN02/HO95, MRC-IX, Total Therapy 2, Total Therapy 3, and MMRF CoMMpass studies., Results: High CTC levels were associated with the expression of 1,700 genes, independent of tumor burden (false discovery rate < 0.05). Of these, 54 genes were selected by leave-one-out cross-validation to construct a transcriptomic classifier representing PCL-like disease. This not only demonstrated a sensitivity of 93% to identify pPCL in the validation cohort but also classified 10% of NDMM tumors as PCL-like. PCL-like MM transcriptionally and cytogenetically resembled pPCL, but presented with significantly lower CTC levels and tumor burden. Multivariate analyses in NDMM confirmed the significant prognostic value of PCL-like status in the context of Revised International Staging System stage, age, and treatment, regarding both progression-free (hazard ratio, 1.64; 95% CI, 1.30 to 2.07) and overall survival (hazard ratio, 1.89; 95% CI, 1.42 to 2.50)., Conclusion: pPCL was identified on the basis of a specific tumor transcriptome, which was also present in patients with high-risk NDMM, despite not being clinically leukemic. Incorporating PCL-like status into current risk models in NDMM may improve prognostic accuracy.
- Published
- 2022
- Full Text
- View/download PDF
14. Induced cell-autonomous neutropenia systemically perturbs hematopoiesis in Cebpa enhancer-null mice.
- Author
-
Avellino R, Mulet-Lazaro R, Havermans M, Hoogenboezem R, Smeenk L, Salomonis N, Schneider RK, Rombouts E, Bindels E, Grimes L, and Delwel R
- Subjects
- Animals, CCAAT-Enhancer-Binding Proteins genetics, CCAAT-Enhancer-Binding Proteins metabolism, Hematopoietic Stem Cells metabolism, Mice, Mice, Knockout, Transcription Factors metabolism, Hematopoiesis, Neutropenia genetics
- Abstract
The transcription factor C/EBPa initiates the neutrophil gene expression program in the bone marrow (BM). Knockouts of the Cebpa gene or its +37kb enhancer in mice show 2 major findings: (1) neutropenia in BM and blood; (2) decrease in long-term hematopoietic stem cell (LT-HSC) numbers. Whether the latter finding is cell-autonomous (intrinsic) to the LT-HSCs or an extrinsic event exerted on the stem cell compartment remained an open question. Flow cytometric analysis of the Cebpa +37kb enhancer knockout model revealed that the reduction in LT-HSC numbers observed was proportional to the degree of neutropenia. Single-cell transcriptomics of wild-type (WT) mouse BM showed that Cebpa is predominantly expressed in early myeloid-biased progenitors but not in LT-HSCs. These observations suggest that the negative effect on LT-HSCs is an extrinsic event caused by neutropenia. We transplanted whole BMs from +37kb enhancer-deleted mice and found that 40% of the recipient mice acquired full-blown neutropenia with severe dysplasia and a significant reduction in the total LT-HSC population. The other 60% showed initial signs of myeloid differentiation defects and dysplasia when they were sacrificed, suggesting they were in an early stage of the same pathological process. This phenotype was not seen in mice transplanted with WT BM. Altogether, these results indicate that Cebpa enhancer deletion causes cell-autonomous neutropenia, which reprograms and disturbs the quiescence of HSCs, leading to a systemic impairment of the hematopoietic process., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
15. Essential role for Gata2 in modulating lineage output from hematopoietic stem cells in zebrafish.
- Author
-
Gioacchino E, Koyunlar C, Zink J, de Looper H, de Jong M, Dobrzycki T, Mahony CB, Hoogenboezem R, Bosch D, van Strien PMH, van Royen ME, French PJ, Bindels E, Gussinklo KJ, Monteiro R, Touw IP, and de Pater E
- Subjects
- Animals, Cell Differentiation, GATA2 Transcription Factor genetics, Hematopoiesis, Mice, Monocytes, Zebrafish Proteins, Hematopoietic Stem Cells, Zebrafish
- Abstract
The differentiation of hematopoietic stem cells (HSCs) is tightly controlled to ensure a proper balance between myeloid and lymphoid cell output. GATA2 is a pivotal hematopoietic transcription factor required for generation and maintenance of HSCs. GATA2 is expressed throughout development, but because of early embryonic lethality in mice, its role during adult hematopoiesis is incompletely understood. Zebrafish contains 2 orthologs of GATA2: Gata2a and Gata2b, which are expressed in different cell types. We show that the mammalian functions of GATA2 are split between these orthologs. Gata2b-deficient zebrafish have a reduction in embryonic definitive hematopoietic stem and progenitor cell (HSPC) numbers, but are viable. This allows us to uniquely study the role of GATA2 in adult hematopoiesis. gata2b mutants have impaired myeloid lineage differentiation. Interestingly, this defect arises not in granulocyte-monocyte progenitors, but in HSPCs. Gata2b-deficient HSPCs showed impaired progression of the myeloid transcriptional program, concomitant with increased coexpression of lymphoid genes. This resulted in a decrease in myeloid-programmed progenitors and a relative increase in lymphoid-programmed progenitors. This shift in the lineage output could function as an escape mechanism to avoid a block in lineage differentiation. Our study helps to deconstruct the functions of GATA2 during hematopoiesis and shows that lineage differentiation flows toward a lymphoid lineage in the absence of Gata2b., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
16. The multiple myeloma microenvironment is defined by an inflammatory stromal cell landscape.
- Author
-
de Jong MME, Kellermayer Z, Papazian N, Tahri S, Hofste Op Bruinink D, Hoogenboezem R, Sanders MA, van de Woestijne PC, Bos PK, Khandanpour C, Vermeulen J, Moreau P, van Duin M, Broijl A, Sonneveld P, and Cupedo T
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols pharmacology, Bone Marrow drug effects, Bone Marrow immunology, Bone Marrow pathology, Cell Line, Tumor, Disease Progression, Female, Gene Expression Regulation, Neoplastic immunology, Humans, Male, Mesenchymal Stem Cells pathology, Middle Aged, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local prevention & control, Primary Cell Culture, Prospective Studies, RNA-Seq, Single-Cell Analysis, Tumor Microenvironment drug effects, Tumor Microenvironment genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Mesenchymal Stem Cells immunology, Multiple Myeloma immunology, Neoplasm Recurrence, Local immunology, Tumor Microenvironment immunology
- Abstract
Progression and persistence of malignancies are influenced by the local tumor microenvironment, and future eradication of currently incurable tumors will, in part, hinge on our understanding of malignant cell biology in the context of their nourishing surroundings. Here, we generated paired single-cell transcriptomic datasets of tumor cells and the bone marrow immune and stromal microenvironment in multiple myeloma. These analyses identified myeloma-specific inflammatory mesenchymal stromal cells, which spatially colocalized with tumor cells and immune cells and transcribed genes involved in tumor survival and immune modulation. Inflammatory stromal cell signatures were driven by stimulation with proinflammatory cytokines, and analyses of immune cell subsets suggested interferon-responsive effector T cell and CD8
+ stem cell memory T cell populations as potential sources of stromal cell-activating cytokines. Tracking stromal inflammation in individuals over time revealed that successful antitumor induction therapy is unable to revert bone marrow inflammation, predicting a role for mesenchymal stromal cells in disease persistence.- Published
- 2021
- Full Text
- View/download PDF
17. Extracellular Vesicles Derived From Adult and Fetal Bone Marrow Mesenchymal Stromal Cells Differentially Promote ex vivo Expansion of Hematopoietic Stem and Progenitor Cells.
- Author
-
Ghebes CA, Morhayim J, Kleijer M, Koroglu M, Erkeland SJ, Hoogenboezem R, Bindels E, van Alphen FPJ, van den Biggelaar M, Nolte MA, van der Eerden BCJ, Braakman E, Voermans C, and van de Peppel J
- Abstract
Recently, we and others have illustrated that extracellular vesicles (EVs) have the potential to support hematopoietic stem and progenitor cell (HSPC) expansion; however, the mechanism and processes responsible for the intercellular communication by EVs are still unknown. In the current study, we investigate whether primary human bone marrow derived mesenchymal stromal cells (BMSC) EVs isolated from two different origins, fetal (fEV) and adult (aEV) tissue, can increase the relative low number of HSPCs found in umbilical cord blood (UCB) and which EV-derived components are responsible for ex vivo HSPC expansion. Interestingly, aEVs and to a lesser extent fEVs, showed supportive ex vivo expansion capacity of UCB-HSPCs. Taking advantage of the two BMSC sources with different supportive effects, we analyzed the EV cargo and investigated how gene expression is modulated in HSPCs after incubation with aEVs and fEVs. Proteomics analyses of the protein cargo composition of the supportive aEV vs. the less-supportive fEV identified 90% of the Top100 exosome proteins present in the ExoCarta database. Gene Ontology (GO) analyses illustrated that the proteins overrepresented in aEVs were annotated to oxidation-reduction process, mitochondrial ATP synthesis coupled proton transport, or protein folding. In contrast, the proteins overrepresented in fEVs were annotated to extracellular matrix organization positive regulation of cell migration or transforming growth factor beta receptor (TGFBR) signaling pathway. Small RNA sequencing identified different molecular signatures between aEVs and fEVs. Interestingly, the microRNA cluster miR-99b/let-7e/miR-125a, previously identified to increase the number of HSPCs by targeting multiple pro-apoptotic genes, was highly and significantly enriched in aEVs. Although we identified significant differences in the supportive effects of aEVs and fEVs, RNAseq analyses of the 24 h treated HSPCs indicated that a limited set of genes was differentially regulated when compared to cells that were treated with cytokines only. Together, our study provides novel insights into the complex biological role of EVs and illustrates that aEVs and fEVs differentially support ex vivo expansion capacity of UCB-HSPCs. Together opening new means for the application of EVs in the discovery of therapeutics for more efficient ex vivo HSPC expansion., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Ghebes, Morhayim, Kleijer, Koroglu, Erkeland, Hoogenboezem, Bindels, van Alphen, van den Biggelaar, Nolte, van der Eerden, Braakman, Voermans and van de Peppel.)
- Published
- 2021
- Full Text
- View/download PDF
18. Increased CXCL4 expression in hematopoietic cells links inflammation and progression of bone marrow fibrosis in MPN.
- Author
-
Gleitz HFE, Dugourd AJF, Leimkühler NB, Snoeren IAM, Fuchs SNR, Menzel S, Ziegler S, Kröger N, Triviai I, Büsche G, Kreipe H, Banjanin B, Pritchard JE, Hoogenboezem R, Bindels EM, Schumacher N, Rose-John S, Elf S, Saez-Rodriguez J, Kramann R, and Schneider RK
- Subjects
- Animals, Bone Marrow immunology, Bone Marrow metabolism, Cell Proliferation, Disease Progression, Fibrosis etiology, Fibrosis immunology, Fibrosis metabolism, Humans, Inflammation etiology, Inflammation immunology, Inflammation metabolism, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Male, Megakaryocytes, Mice, Mice, Knockout, Mutation, Platelet Factor 4 genetics, Primary Myelofibrosis etiology, Primary Myelofibrosis immunology, Primary Myelofibrosis metabolism, Bone Marrow pathology, Fibrosis pathology, Inflammation pathology, Myeloproliferative Disorders complications, Platelet Factor 4 metabolism, Primary Myelofibrosis pathology
- Abstract
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm (MPN) that leads to progressive bone marrow (BM) fibrosis. Although the cellular mutations involved in the pathogenesis of PMF have been extensively investigated, the sequential events that drive stromal activation and fibrosis by hematopoietic-stromal cross-talk remain elusive. Using an unbiased approach and validation in patients with MPN, we determined that the differential spatial expression of the chemokine CXCL4/platelet factor-4 marks the progression of fibrosis. We show that the absence of hematopoietic CXCL4 ameliorates the MPN phenotype, reduces stromal cell activation and BM fibrosis, and decreases the activation of profibrotic pathways in megakaryocytes, inflammation in fibrosis-driving cells, and JAK/STAT activation in both megakaryocytes and stromal cells in 3 murine PMF models. Our data indicate that higher CXCL4 expression in MPN has profibrotic effects and is a mediator of the characteristic inflammation. Therefore, targeting CXCL4 might be a promising strategy to reduce inflammation in PMF., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
19. Gli1 + Mesenchymal Stromal Cells Are a Key Driver of Bone Marrow Fibrosis and an Important Cellular Therapeutic Target.
- Author
-
Schneider RK, Mullally A, Dugourd A, Peisker F, Hoogenboezem R, Van Strien PMH, Bindels EM, Heckl D, Büsche G, Fleck D, Müller-Newen G, Wongboonsin J, Ventura Ferreira M, Puelles VG, Saez-Rodriguez J, Ebert BL, Humphreys BD, and Kramann R
- Published
- 2018
- Full Text
- View/download PDF
20. Gli1 + Mesenchymal Stromal Cells Are a Key Driver of Bone Marrow Fibrosis and an Important Cellular Therapeutic Target.
- Author
-
Schneider RK, Mullally A, Dugourd A, Peisker F, Hoogenboezem R, Van Strien PMH, Bindels EM, Heckl D, Büsche G, Fleck D, Müller-Newen G, Wongboonsin J, Ventura Ferreira M, Puelles VG, Saez-Rodriguez J, Ebert BL, Humphreys BD, and Kramann R
- Subjects
- Animals, Cell Differentiation genetics, Humans, Mesenchymal Stem Cells pathology, Mice, Mice, Transgenic, Myofibroblasts pathology, Primary Myelofibrosis genetics, Primary Myelofibrosis metabolism, Primary Myelofibrosis pathology, Zinc Finger Protein GLI1 genetics, Zinc Finger Protein GLI1 metabolism, Cell Differentiation drug effects, Cell Proliferation drug effects, Mesenchymal Stem Cells metabolism, Myofibroblasts metabolism, Primary Myelofibrosis drug therapy, Pyridines pharmacology, Pyrimidines pharmacology, Zinc Finger Protein GLI1 antagonists & inhibitors
- Abstract
Bone marrow fibrosis (BMF) develops in various hematological and non-hematological conditions and is a central pathological feature of myelofibrosis. Effective cell-targeted therapeutics are needed, but the cellular origin of BMF remains elusive. Here, we show using genetic fate tracing in two murine models of BMF that Gli1
+ mesenchymal stromal cells (MSCs) are recruited from the endosteal and perivascular niche to become fibrosis-driving myofibroblasts in the bone marrow. Genetic ablation of Gli1+ cells abolished BMF and rescued bone marrow failure. Pharmacological targeting of Gli proteins with GANT61 inhibited Gli1+ cell expansion and myofibroblast differentiation and attenuated fibrosis severity. The same pathway is also active in human BMF, and Gli1 expression in BMF significantly correlates with the severity of the disease. In addition, GANT61 treatment reduced the myofibroblastic phenotype of human MSCs isolated from patients with BMF, suggesting that targeting of Gli proteins could be a relevant therapeutic strategy., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
21. Source tracking of Enterococcus moraviensis and E. haemoperoxidus.
- Author
-
Taučer-Kapteijn M, Hoogenboezem W, Hoogenboezem R, de Haas S, and Medema G
- Subjects
- Animals, Enterococcus classification, Feces microbiology, Humans, Netherlands, Species Specificity, Water Microbiology, Enterococcus isolation & purification, Geese microbiology, Groundwater microbiology, Mammals microbiology, Water Purification
- Abstract
Enterococci were detected occasionally in 100 L samples of water abstracted from a shallow aquifer in a natural dune infiltration area for drinking water production. Enterococcus moraviensis was the species most frequently identified in these samples. Because there are no existing reports of faecal sources of E. moraviensis and the closely related E. hemoperoxidus, this study aimed to find such sources of these two species in the dunes. Faecal samples from various animal species living in the vicinity of abstraction wells, were analysed for enterococci on Slanetz and Bartley Agar. From these samples, enterococci isolates (1,386 in total) were subsequently identified using matrix assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry. E. moraviensis was found in the faeces of geese, foxes and rabbits. Also, E. haemoperoxidus was isolated from goose faeces. Using hierarchical clustering, the species composition of Enterococcus spp. isolated from abstracted water formed one cluster with the species composition found in geese droppings. A sanitary survey supported the indication that feral geese may provide a substantial faecal load in particular parts of this dune infiltration area, close to the water abstraction system. This study confirms the faecal origin of E. moraviensis and E. haemoperoxidus from specific animals, which strengthens their significance as faecal indicators.
- Published
- 2017
- Full Text
- View/download PDF
22. MicroRNA-155 induces AML in combination with the loss of C/EBPA in mice.
- Author
-
Alemdehy MF, de Looper HW, Kavelaars FG, Sanders MA, Hoogenboezem R, Löwenberg B, Valk PJ, Touw IP, and Erkeland SJ
- Subjects
- Animals, Mice, CCAAT-Enhancer-Binding Protein-alpha genetics, Leukemia, Myeloid, Acute genetics, MicroRNAs genetics
- Published
- 2016
- Full Text
- View/download PDF
23. An autonomous CEBPA enhancer specific for myeloid-lineage priming and neutrophilic differentiation.
- Author
-
Avellino R, Havermans M, Erpelinck C, Sanders MA, Hoogenboezem R, van de Werken HJ, Rombouts E, van Lom K, van Strien PM, Gebhard C, Rehli M, Pimanda J, Beck D, Erkeland S, Kuiken T, de Looper H, Gröschel S, Touw I, Bindels E, and Delwel R
- Subjects
- Animals, CCAAT-Enhancer-Binding Protein-alpha genetics, Cell Line, Tumor, Gene Expression Regulation, Developmental, HEK293 Cells, HL-60 Cells, HeLa Cells, Hep G2 Cells, Humans, Jurkat Cells, K562 Cells, Mice, Mice, Knockout, U937 Cells, CCAAT-Enhancer-Binding Protein-alpha metabolism, Cell Differentiation genetics, Cell Lineage genetics, Enhancer Elements, Genetic, Myeloid Cells physiology, Myelopoiesis genetics, Neutrophils physiology
- Abstract
Neutrophilic differentiation is dependent on CCAAT enhancer-binding protein α (C/EBPα), a transcription factor expressed in multiple organs including the bone marrow. Using functional genomic technologies in combination with clustered regularly-interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 genome editing and in vivo mouse modeling, we show that CEBPA is located in a 170-kb topological-associated domain that contains 14 potential enhancers. Of these, 1 enhancer located +42 kb from CEBPA is active and engages with the CEBPA promoter in myeloid cells only. Germ line deletion of the homologous enhancer in mice in vivo reduces Cebpa levels exclusively in hematopoietic stem cells (HSCs) and myeloid-primed progenitor cells leading to severe defects in the granulocytic lineage, without affecting any other Cebpa-expressing organ studied. The enhancer-deleted progenitor cells lose their myeloid transcription program and are blocked in differentiation. Deletion of the enhancer also causes loss of HSC maintenance. We conclude that a single +42-kb enhancer is essential for CEBPA expression in myeloid cells only., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
24. ICL-induced miR139-3p and miR199a-3p have opposite roles in hematopoietic cell expansion and leukemic transformation.
- Author
-
Alemdehy MF, Haanstra JR, de Looper HW, van Strien PM, Verhagen-Oldenampsen J, Caljouw Y, Sanders MA, Hoogenboezem R, de Ru AH, Janssen GM, Smetsers SE, Bierings MB, van Veelen PA, von Lindern M, Touw IP, and Erkeland SJ
- Subjects
- Animals, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, DNA Repair genetics, DNA-Binding Proteins deficiency, Disease Models, Animal, Endonucleases deficiency, Hematopoietic Stem Cells metabolism, Leukemia metabolism, Leukemia pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cell Transformation, Neoplastic genetics, Hematopoietic Stem Cells pathology, Leukemia genetics, MicroRNAs genetics
- Abstract
Interstrand crosslinks (ICLs) are toxic DNA lesions that cause severe genomic damage during replication, especially in Fanconi anemia pathway-deficient cells. This results in progressive bone marrow failure and predisposes to acute myeloid leukemia (AML). The molecular mechanisms responsible for these defects are largely unknown. Using Ercc1-deficient mice, we show that Trp53 is responsible for ICL-induced bone marrow failure and that loss of Trp53 is leukemogenic in this model. In addition, Ercc1-deficient myeloid progenitors gain elevated levels of miR-139-3p and miR-199a-3p with age. These microRNAs exert opposite effects on hematopoiesis. Ectopic expression of miR-139-3p strongly inhibited proliferation of myeloid progenitors, whereas inhibition of miR-139-3p activity restored defective proliferation of Ercc1-deficient progenitors. Conversely, the inhibition of miR-199a-3p functions aggravated the myeloid proliferation defect in the Ercc1-deficient model, whereas its enforced expression enhanced proliferation of progenitors. Importantly, miR-199a-3p caused AML in a pre-leukemic mouse model, supporting its role as an onco-microRNA. Target genes include HuR for miR-139-3p and Prdx6, Runx1, and Suz12 for miR-199a-3p. The latter genes have previously been implicated as tumor suppressors in de novo and secondary AML. These findings show that, in addition to TRP53-controlled mechanisms, miR-139-3p and miR-199a-3p are involved in the defective hematopoietic function of ICL-repair deficient myeloid progenitors., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
25. Mutational spectrum of myeloid malignancies with inv(3)/t(3;3) reveals a predominant involvement of RAS/RTK signaling pathways.
- Author
-
Gröschel S, Sanders MA, Hoogenboezem R, Zeilemaker A, Havermans M, Erpelinck C, Bindels EM, Beverloo HB, Döhner H, Löwenberg B, Döhner K, Delwel R, and Valk PJ
- Subjects
- Alleles, Chromosome Banding, Chromosomes, Human, Pair 3, DNA Mutational Analysis, Epigenesis, Genetic, Exome, Gene Expression Profiling, Humans, Sequence Analysis, DNA, Sequence Analysis, RNA, Chromosome Inversion, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction, Translocation, Genetic, ras Proteins metabolism
- Abstract
Myeloid malignancies bearing chromosomal inv(3)/t(3;3) abnormalities are among the most therapy-resistant leukemias. Deregulated expression of EVI1 is the molecular hallmark of this disease; however, the genome-wide spectrum of cooperating mutations in this disease subset has not been systematically elucidated. Here, we show that 98% of inv(3)/t(3;3) myeloid malignancies harbor mutations in genes activating RAS/receptor tyrosine kinase (RTK) signaling pathways. In addition, hemizygous mutations in GATA2, as well as heterozygous alterations in RUNX1, SF3B1, and genes encoding epigenetic modifiers, frequently co-occur with the inv(3)/t(3;3) aberration. Notably, neither mutational patterns nor gene expression profiles differ across inv(3)/t(3;3) acute myeloid leukemia, chronic myeloid leukemia, and myelodysplastic syndrome cases, suggesting recognition of inv(3)/t(3;3) myeloid malignancies as a single disease entity irrespective of blast count. The high incidence of activating RAS/RTK signaling mutations may provide a target for a rational treatment strategy in this high-risk patient group., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
26. Two splice-factor mutant leukemia subgroups uncovered at the boundaries of MDS and AML using combined gene expression and DNA-methylation profiling.
- Author
-
Taskesen E, Havermans M, van Lom K, Sanders MA, van Norden Y, Bindels E, Hoogenboezem R, Reinders MJ, Figueroa ME, Valk PJ, Löwenberg B, Melnick A, and Delwel R
- Subjects
- Adult, Aged, Aged, 80 and over, Anemia, Refractory, with Excess of Blasts genetics, DNA Fingerprinting, Female, Humans, Male, Middle Aged, Nuclear Proteins genetics, Phosphoproteins genetics, RNA Splicing Factors, Ribonucleoprotein, U2 Small Nuclear genetics, Ribonucleoproteins genetics, Serine-Arginine Splicing Factors, Splicing Factor U2AF, DNA Methylation, Leukemia, Myeloid, Acute genetics, Mutation, Myelodysplastic Syndromes genetics
- Abstract
Mutations in splice factor (SF) genes occur more frequently in myelodysplastic syndromes (MDS) than in acute myeloid leukemias (AML). We sequenced complementary DNA from bone marrow of 47 refractory anemia with excess blasts (RAEB) patients, 29 AML cases with low marrow blast cell count, and 325 other AML patients and determined the presence of SF-hotspot mutations in SF3B1, U2AF35, and SRSF2. SF mutations were found in 10 RAEB, 12 AML cases with low marrow blast cell count, and 25 other AML cases. Our study provides evidence that SF-mutant RAEB and SF-mutant AML are clinically, cytologically, and molecularly highly similar. An integrated analysis of genomewide messenger RNA (mRNA) expression profiling and DNA-methylation profiling data revealed 2 unique patient clusters highly enriched for SF-mutant RAEB/AML. The combined genomewide mRNA expression profiling/DNA-methylation profiling signatures revealed 1 SF-mutant patient cluster with an erythroid signature. The other SF-mutant patient cluster was enriched for NRAS/KRAS mutations and showed an inferior survival. We conclude that SF-mutant RAEB/AML constitutes a related disorder overriding the artificial separation between AML and MDS, and that SF-mutant RAEB/AML is composed of 2 molecularly and clinically distinct subgroups. We conclude that SF-mutant disorders should be considered as myeloid malignancies that transcend the boundaries of AML and MDS., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
27. Integrated genome-wide genotyping and gene expression profiling reveals BCL11B as a putative oncogene in acute myeloid leukemia with 14q32 aberrations.
- Author
-
Abbas S, Sanders MA, Zeilemaker A, Geertsma-Kleinekoort WM, Koenders JE, Kavelaars FG, Abbas ZG, Mahamoud S, Chu IW, Hoogenboezem R, Peeters JK, van Drunen E, van Galen J, Beverloo HB, Löwenberg B, and Valk PJ
- Subjects
- Antigens, Surface metabolism, Case-Control Studies, Cell Proliferation, Gene Dosage, Gene Expression, Gene Expression Profiling, Genome-Wide Association Study, Genotype, Humans, Immunophenotyping, In Situ Hybridization, Fluorescence, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute metabolism, Polymorphism, Single Nucleotide, Sequence Analysis, DNA, T-Lymphocytes metabolism, Translocation, Genetic, Chromosome Aberrations, Chromosomes, Human, Pair 14, Leukemia, Myeloid, Acute genetics, Oncogenes, Repressor Proteins genetics, Tumor Suppressor Proteins genetics
- Abstract
Acute myeloid leukemia is a neoplasm characterized by recurrent molecular aberrations traditionally demonstrated by cytogenetic analyses. We used high density genome-wide genotyping and gene expression profiling to reveal acquired cryptic abnormalities in acute myeloid leukemia. By genome-wide genotyping of 137 cases of primary acute myeloid leukemia, we disclosed a recurrent focal amplification on chromosome 14q32, which included the genes BCL11B, CCNK, C14orf177 and SETD3, in two cases. In the affected cases, the BCL11B gene showed consistently high mRNA expression, whereas the expression of the other genes was unperturbed. Fluorescence in situ hybridization on 40 cases of acute myeloid leukemia with high BCL11B mRNA expression [2.5-fold above median; 40 out of 530 cases (7.5%)] revealed 14q32 abnormalities in two additional cases. In the four BCL11B-rearranged cases the 14q32 locus was fused to different partner chromosomes. In fact, in two cases, we demonstrated that the focal 14q32 amplifications were integrated into transcriptionally active loci. The translocations involving BCL11B result in increased expression of full-length BCL11B protein. The BCL11B-rearranged acute myeloid leukemias expressed both myeloid and T-cell markers. These biphenotypic acute leukemias all carried FLT3 internal tandem duplications, a characteristic marker of acute myeloid leukemia. BCL11B mRNA expression in acute myeloid leukemia appeared to be strongly associated with expression of other T-cell-specific genes. Myeloid 32D(GCSF-R) cells ectopically expressing Bcl11b showed decreased proliferation rate and less maturation. In conclusion, by an integrated approach involving high-throughput genome-wide genotyping and gene expression profiling we identified BCL11B as a candidate oncogene in acute myeloid leukemia.
- Published
- 2014
- Full Text
- View/download PDF
28. A single oncogenic enhancer rearrangement causes concomitant EVI1 and GATA2 deregulation in leukemia.
- Author
-
Gröschel S, Sanders MA, Hoogenboezem R, de Wit E, Bouwman BAM, Erpelinck C, van der Velden VHJ, Havermans M, Avellino R, van Lom K, Rombouts EJ, van Duin M, Döhner K, Beverloo HB, Bradner JE, Döhner H, Löwenberg B, Valk PJM, Bindels EMJ, de Laat W, and Delwel R
- Subjects
- Cell Line, Tumor, Chromosome Inversion, Humans, MDS1 and EVI1 Complex Locus Protein, Promoter Regions, Genetic, Transcriptional Activation, Translocation, Genetic, Chromosomes, Human, Pair 3, DNA-Binding Proteins genetics, Enhancer Elements, Genetic, GATA2 Transcription Factor genetics, Gene Expression Regulation, Neoplastic, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Proto-Oncogenes genetics, Transcription Factors genetics
- Abstract
Chromosomal rearrangements without gene fusions have been implicated in leukemogenesis by causing deregulation of proto-oncogenes via relocation of cryptic regulatory DNA elements. AML with inv(3)/t(3;3) is associated with aberrant expression of the stem-cell regulator EVI1. Applying functional genomics and genome-engineering, we demonstrate that both 3q rearrangements reposition a distal GATA2 enhancer to ectopically activate EVI1 and simultaneously confer GATA2 functional haploinsufficiency, previously identified as the cause of sporadic familial AML/MDS and MonoMac/Emberger syndromes. Genomic excision of the ectopic enhancer restored EVI1 silencing and led to growth inhibition and differentiation of AML cells, which could be replicated by pharmacologic BET inhibition. Our data show that structural rearrangements involving the chromosomal repositioning of a single enhancer can cause deregulation of two unrelated distal genes, with cancer as the outcome., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
29. Cooperativity of RUNX1 and CSF3R mutations in severe congenital neutropenia: a unique pathway in myeloid leukemogenesis.
- Author
-
Skokowa J, Steinemann D, Katsman-Kuipers JE, Zeidler C, Klimenkova O, Klimiankou M, Unalan M, Kandabarau S, Makaryan V, Beekman R, Behrens K, Stocking C, Obenauer J, Schnittger S, Kohlmann A, Valkhof MG, Hoogenboezem R, Göhring G, Reinhardt D, Schlegelberger B, Stanulla M, Vandenberghe P, Donadieu J, Zwaan CM, Touw IP, van den Heuvel-Eibrink MM, Dale DC, and Welte K
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Congenital Bone Marrow Failure Syndromes, Cytogenetic Analysis, Female, Humans, Male, Neutropenia genetics, Neutropenia pathology, Signal Transduction genetics, Young Adult, Cell Transformation, Neoplastic genetics, Core Binding Factor Alpha 2 Subunit genetics, Leukemia, Myeloid genetics, Mutation, Neutropenia congenital, Receptors, Colony-Stimulating Factor genetics
- Abstract
Severe congenital neutropenia (CN) is a preleukemic bone marrow failure syndrome with a 20% risk of evolving into leukemia or myelodysplastic syndrome (MDS). Patterns of acquisition of leukemia-associated mutations were investigated using next-generation deep-sequencing in 31 CN patients who developed leukemia or MDS. Twenty (64.5%) of the 31 patients had mutations in RUNX1. A majority of patients with RUNX1 mutations (80.5%) also had acquired CSF3R mutations. In contrast to their high frequency in CN patients who developed leukemia or MDS, RUNX1 mutations were found in only 9 of 307 (2.9%) patients with de novo pediatric acute myeloid leukemia. A sequential analysis at stages prior to overt leukemia revealed RUNX1 mutations to be late events in leukemic transformation. Single-cell analyses in 2 patients showed that RUNX1 and CSF3R mutations were present in the same malignant clone. Functional studies demonstrated elevated granulocyte colony-stimulating factor (G-CSF)-induced proliferation with diminished myeloid differentiation of hematopoietic CD34(+) cells coexpressing mutated forms of RUNX1 and CSF3R. The high frequency of cooperating RUNX1 and CSF3R mutations in CN patients suggests a novel molecular pathway of leukemogenesis: mutations in the hematopoietic cytokine receptor (G-CSFR) in combination with the second mutations in the downstream hematopoietic transcription fator (RUNX1). The detection of both RUNX1 and CSF3R mutations could be used as a marker for identifying CN patients with a high risk of progressing to leukemia or MDS.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.