72 results on '"Martens JHA"'
Search Results
2. DNA methylation oscillation defines classes of enhancers
- Author
-
Henk Stunnenberg, Cutler A, Ivliev A, Lancashire L, Mattia Frontini, Simon Heath, Stephan Beck, Wallace C, Steven W. Wingett, Peter Fraser, Kate Downes, Biola-Maria Javierre, Emanuele Libertini, Garcia Ar, Martens Jha, Gut M, Luigi Grassi, Debjani Paul, Jonathan Cairns, Willem H. Ouwehand, Gut I, Mikhail Spivakov, and Rifat Hamoudi
- Subjects
0303 health sciences ,biology ,dNaM ,Genomics ,Computational biology ,Genome project ,Bisulfite ,03 medical and health sciences ,0302 clinical medicine ,Histone ,030220 oncology & carcinogenesis ,DNA methylation ,biology.protein ,Enhancer ,030304 developmental biology ,Epigenomics - Abstract
Understanding the regulatory landscape of human cells requires the integration of genomic and epigenomic maps, capturing combinatorial levels of cell type-specific and invariant activity states.Here, we segmented whole-genome bisulfite sequencing-derived methylomes into consecutive blocks of co-methylation (COMETs) to obtain spatial variation patterns of DNA methylation (DNAm oscillations) integrated with histone modifications and promoter-enhancer interactions derived from promoter capture Hi-C (PCHi-C) sequencing of the same purified blood cells.Mapping DNAm oscillations onto regulatory genome annotation revealed that enhancers are enriched for DNAm hyper-oscillations (>30-fold), where multiple machine learning models support DNAm as predictive of enhancer location. Based on this analysis, we report overall predictive power of 99% for DNAm oscillations, 77.3% for DNaseI, 41% for CGIs, 20% for UMRs and 0% for LMRs, demonstrating the power of DNAm oscillations over other methods for enhancer prediction. Methylomes of activated and non-activated CD4+ T cells indicate that DNAm oscillations exist in both states irrespective of activation; hence they can be used to determine the location of latent enhancers.Our approach advances the identification of tissue-specific regulatory elements and outperforms previous approaches defining enhancer classes based on DNA methylation.
- Published
- 2018
- Full Text
- View/download PDF
3. Genetic Drivers of Epigenetic and Transcriptional Variation in Human Immune Cells
- Author
-
Chen, L, Ge, B, Casale, FP, Vasquez, L, Kwan, T, Garrido-Martín, D, Watt, S, Yan, Y, Kundu, K, Ecker, S, Datta, A, Richardson, D, Burden, F, Mead, D, Mann, AL, Fernandez, JM, Rowlston, S, Wilder, SP, Farrow, S, Shao, X, Lambourne, JJ, Redensek, A, Albers, CA, Amstislavskiy, V, Ashford, S, Berentsen, K, Bomba, L, Bourque, G, Bujold, D, Busche, S, Caron, M, Chen, S-H, Cheung, W, Delaneau, O, Dermitzakis, ET, Elding, H, Colgiu, I, Bagger, FO, Flicek, P, Habibi, E, Iotchkova, V, Janssen-Megens, E, Kim, B, Lehrach, H, Lowy, E, Mandoli, A, Matarese, F, Maurano, MT, Morris, JA, Pancaldi, V, Pourfarzad, F, Rehnstrom, K, Rendon, A, Risch, T, Sharifi, N, Simon, M-M, Sultan, M, Valencia, A, Walter, K, Wang, S-Y, Frontini, M, Antonarakis, SE, Clarke, L, Yaspo, M-L, Beck, S, Guigo, R, Rico, D, Martens, JHA, Ouwehand, WH, Kuijpers, TW, Paul, DS, Stunnenberg, HG, Stegle, O, Downes, K, Pastinen, T, and Soranzo, N
- Subjects
transription ,DNA methylation ,QTL ,monocyte ,t-cell ,neutrophil ,histone modification ,immune ,3. Good health ,EWAS ,allele specific - Abstract
Characterizing the multifaceted contribution of genetic and epigenetic factors to disease phenotypes is a major challenge in human genetics and medicine. We carried out high-resolution genetic, epigenetic, and transcriptomic profiling in three major human immune cell types (CD14$^{+}$ monocytes, CD16$^{+}$ neutrophils, and naive CD4$^{+}$ T cells) from up to 197 individuals. We assess, quantitatively, the relative contribution of $\textit{cis}$-genetic and epigenetic factors to transcription and evaluate their impact as potential sources of confounding in epigenome-wide association studies. Further, we characterize highly coordinated genetic effects on gene expression, methylation, and histone variation through quantitative trait locus (QTL) mapping and allele-specific (AS) analyses. Finally, we demonstrate colocalization of molecular trait QTLs at 345 unique immune disease loci. This expansive, high-resolution atlas of multi-omics changes yields insights into cell-type-specific correlation between diverse genomic inputs, more generalizable correlations between these inputs, and defines molecular events that may underpin complex disease risk.
4. Trained immunity suppression determines kidney allograft survival.
- Author
-
Jonkman I, Jacobs MME, Negishi Y, Yanginlar C, Martens JHA, Baltissen M, Vermeulen M, van den Hoogen MWF, Baas M, van der Vlag J, Fayad ZA, Teunissen AJP, Madsen JC, Ochando J, Joosten LAB, Netea MG, Mulder WJM, Mhlanga MM, Hilbrands LB, Rother N, and Duivenvoorden R
- Subjects
- Humans, Male, Female, Middle Aged, Prognosis, Immunosuppressive Agents, Adult, Follow-Up Studies, Kidney Failure, Chronic surgery, Kidney Failure, Chronic immunology, Leukocytes, Mononuclear immunology, Allografts, Glomerular Filtration Rate, Kidney Function Tests, Trained Immunity, Kidney Transplantation, Graft Survival immunology, Immunity, Innate, Graft Rejection immunology
- Abstract
The innate immune system plays an essential role in regulating the immune responses to kidney transplantation, but the mechanisms through which innate immune cells influence long-term graft survival are unclear. The current study highlights the vital role of trained immunity in kidney allograft survival. Trained immunity describes the epigenetic and metabolic changes that innate immune cells undergo following an initial stimulus, allowing them have a stronger inflammatory response to subsequent stimuli. We stimulated healthy peripheral blood mononuclear cells with pretransplant and posttransplant serum of kidney transplant patients and immunosuppressive drugs in an in vitro trained immunity assay and measured tumor necrosis factor and interleukin 6 cytokine levels in the supernatant as a readout for trained immunity. We show that the serum of kidney transplant recipients collected 1 week after transplantation can suppress trained immunity. Importantly, we found that kidney transplant recipients whose serum most strongly suppressed trained immunity rarely experienced graft loss. This suppressive effect of posttransplant serum is likely mediated by previously unreported effects of immunosuppressive drugs. Our findings provide mechanistic insights into the role of innate immunity in kidney allograft survival, uncovering trained immunity as a potential therapeutic target for improving graft survival., Competing Interests: Declaration of competing interest The authors of this manuscript have conflicts of interest to disclose as described by the American Journal of Transplantation. L. A. B. Joosten is scientific founder of TTxD, LembaTX, and SalvinaTX. M. G. Netea is scientific founder of TTxD and Biotrip. W. J. M. Mulder is scientific founder of TTxD and Biotrip. Other authors of this manuscript have no conflicts of interest to disclose as described by the American Journal of Transplantation., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Seasonal variation in BCG-induced trained immunity.
- Author
-
Kilic G, Debisarun PA, Alaswad A, Baltissen MP, Lamers LA, de Bree LCJ, Benn CS, Aaby P, Dijkstra H, Lemmers H, Martens JHA, Domínguez-Andrés J, van Crevel R, Li Y, Xu CJ, and Netea MG
- Subjects
- Humans, Adult, Male, Female, Young Adult, Killer Cells, Natural immunology, Vaccination, Healthy Volunteers, Interferon-gamma immunology, Interferon-gamma metabolism, Trained Immunity, BCG Vaccine immunology, BCG Vaccine administration & dosage, Seasons, Cytokines immunology, Cytokines metabolism, Immunologic Memory, Leukocytes, Mononuclear immunology, Immunity, Innate
- Abstract
The Bacille Calmette-Guerin (BCG) vaccine is a well-established inducer of innate immune memory (also termed trained immunity), causing increased cytokine production upon heterologous secondary stimulation. Innate immune responses are known to be influenced by season, but whether seasons impact induction of trained immunity is not known. To explore the influence of season on innate immune memory induced by the BCG vaccine, we vaccinated healthy volunteers with BCG either during winter or spring. Three months later, we measured the ex vivo cytokine responses against heterologous stimuli, analyzed gene expressions and epigenetic signatures of the immune cells, and compared these with the baseline before vaccination. BCG vaccination during winter induced a stronger increase in the production of pro-inflammatory cytokines by peripheral blood mononuclear cells (PBMCs) upon stimulation with different bacterial and fungal stimuli, compared to BCG vaccination in spring. In contrast, winter BCG vaccination resulted in lower IFNγ release in PBMCs compared to spring BCG vaccination. Furthermore, NK cells of the winter-vaccinated people had a greater pro-inflammatory cytokine and IFNγ production capacity upon heterologous stimulation. BCG had only minor effects on the transcriptome of monocytes 3 months later. In contrast, we identified season-dependent epigenetic changes in monocytes and NK cells induced by vaccination, partly explaining the higher immune cell reactivity in the winter BCG vaccination group. These results suggest that BCG vaccination during winter is more prone to induce a robust trained immunity response by activating and reprogramming the immune cells, especially NK cells. (Dutch clinical trial registry no. NL58219.091.16)., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: MGN is a scientific founder of TTxD, Lemba and Biotrip. All other authors declare that they have no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Trained immunity is regulated by T cell-induced CD40-TRAF6 signaling.
- Author
-
Jacobs MME, Maas RJF, Jonkman I, Negishi Y, Tielemans Zamora W, Yanginlar C, van Heck J, Matzaraki V, Martens JHA, Baltissen M, Vermeulen M, Morla-Folch J, Ranzenigo A, Wang W, Umali M, Ochando J, van der Vlag J, Hilbrands LB, Joosten LAB, Netea MG, Mulder WJM, van Leent MMT, Mhlanga MM, Teunissen AJP, Rother N, and Duivenvoorden R
- Subjects
- Animals, Mice, T-Lymphocytes immunology, T-Lymphocytes metabolism, Humans, Male, Heart Transplantation, Trained Immunity, CD40 Antigens metabolism, TNF Receptor-Associated Factor 6 metabolism, Signal Transduction, Mice, Inbred C57BL
- Abstract
Trained immunity is characterized by histone modifications and metabolic changes in innate immune cells following exposure to inflammatory signals, leading to heightened responsiveness to secondary stimuli. Although our understanding of the molecular regulation of trained immunity has increased, the role of adaptive immune cells herein remains largely unknown. Here, we show that T cells modulate trained immunity via cluster of differentiation 40-tissue necrosis factor receptor-associated factor 6 (CD40-TRAF6) signaling. CD40-TRAF6 inhibition modulates functional, transcriptomic, and metabolic reprogramming and modifies histone 3 lysine 4 trimethylation associated with trained immunity. Besides in vitro studies, we reveal that single-nucleotide polymorphisms in the proximity of CD40 are linked to trained immunity responses in vivo and that combining CD40-TRAF6 inhibition with cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4-Ig)-mediated co-stimulatory blockade induces long-term graft acceptance in a murine heart transplantation model. Combined, our results reveal that trained immunity is modulated by CD40-TRAF6 signaling between myeloid and adaptive immune cells and that this can be leveraged for therapeutic purposes., Competing Interests: Declaration of interests J.O., L.A.B.J., M.G.N., and W.J.M.M. declare that they are scientific founders of Trained Therapeutics Discovery., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Susceptibility of pediatric acute lymphoblastic leukemia to STAT3 inhibition depends on p53 induction.
- Author
-
Gasparoli L, Virely C, Tsakaneli A, Che N, Edwards D, Bartram J, Hubank M, Pal D, Heidenreich O, Martens JHA, De Boer J, and Williams O
- Subjects
- Child, Humans, Cell Cycle Proteins metabolism, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-mdm2 genetics, Recurrence, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Advances in the clinical management of pediatric B-cell acute lymphoblastic leukemia (B-ALL) have dramatically improved outcomes for this disease. However, relapsed and high-risk disease still contribute to significant numbers of treatment failures. Development of new, broad range therapies is urgently needed for these cases. We previously reported the susceptibility of ETV6-RUNX1+ pediatric B-ALL to inhibition of signal transducer and activator of transcription 3 (STAT3) activity. In the present study, we demonstrate that pharmacological or genetic inhibition of STAT3 results in p53 induction and that CRISPR-mediated TP53 knockout substantially reverses susceptibility to STAT3 inhibition. Furthermore, we demonstrate that sensitivity to STAT3 inhibition in patient-derived xenograft (PDX) B-ALL samples is not restricted to any particular disease subtype, but rather depends on TP53 status, the only resistant samples being TP53 mutant. Induction of p53 following STAT3 inhibition is not directly dependent on MDM2 but correlates with degradation of MDM4. As such, STAT3 inhibition exhibits synergistic in vitro and in vivo anti-leukemia activity when combined with MDM2 inhibition. Taken together with the relatively low frequency of TP53 mutations in this disease, these data support the future development of combined STAT3/ MDM2 inhibition in the therapy of refractory and relapsed pediatric B-ALL.
- Published
- 2024
- Full Text
- View/download PDF
8. GFI1B and LSD1 repress myeloid traits during megakaryocyte differentiation.
- Author
-
Venhuizen J, van Bergen MGJM, Bergevoet SM, Gilissen D, Spruijt CG, Wingens L, van den Akker E, Vermeulen M, Jansen JH, Martens JHA, and van der Reijden BA
- Subjects
- Humans, Cell Differentiation genetics, Histone Demethylases genetics, Histone Demethylases metabolism, Gene Expression Regulation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism, Megakaryocytes metabolism, Hematopoiesis genetics
- Abstract
The transcription factor Growth Factor Independence 1B (GFI1B) recruits Lysine Specific Demethylase 1 A (LSD1/KDM1A) to stimulate gene programs relevant for megakaryocyte and platelet biology. Inherited pathogenic GFI1B variants result in thrombocytopenia and bleeding propensities with varying intensity. Whether these affect similar gene programs is unknow. Here we studied transcriptomic effects of four patient-derived GFI1B variants (GFI1B
T174N ,H181Y ,R184P ,Q287* ) in MEG01 megakaryoblasts. Compared to normal GFI1B, each variant affected different gene programs with GFI1BQ287* uniquely failing to repress myeloid traits. In line with this, single cell RNA-sequencing of induced pluripotent stem cell (iPSC)-derived megakaryocytes revealed a 4.5-fold decrease in the megakaryocyte/myeloid cell ratio in GFI1BQ287* versus normal conditions. Inhibiting the GFI1B-LSD1 interaction with small molecule GSK-LSD1 resulted in activation of myeloid genes in normal iPSC-derived megakaryocytes similar to what was observed for GFI1BQ287* iPSC-derived megakaryocytes. Thus, GFI1B and LSD1 facilitate gene programs relevant for megakaryopoiesis while simultaneously repressing programs that induce myeloid differentiation., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
9. Erythroid Predominance in Bone Marrow Biopsies of Acute Myeloid Leukemia Patients after Decitabine Treatment Correlates with Mutation Profile and Complete Remission.
- Author
-
Tiso F, Hebeda KM, Langemeijer SMC, de Graaf AO, Martens JHA, Koorenhof-Scheele TN, Knops R, Kroeze LI, van der Reijden BA, and Jansen JH
- Subjects
- Humans, Male, Female, Middle Aged, Retrospective Studies, Aged, Adult, Biopsy, Antimetabolites, Antineoplastic therapeutic use, Azacitidine therapeutic use, DNA Methylation, Aged, 80 and over, Treatment Outcome, Decitabine therapeutic use, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute pathology, Mutation, Bone Marrow pathology, Remission Induction
- Abstract
Introduction: Acute myeloid leukemia (AML) patients may receive hypomethylating agents such as decitabine (DAC) as part of their treatment. Not all patients respond to this therapy, and if they do, the clinical response may occur only after 3-6 courses of treatment. Hence, early biomarkers predicting response would be very useful., Methods: We retrospectively analyzed a cohort of 22 AML patients who were treated with DAC. Histology of the bone marrow biopsy, pathogenic mutations, and methylation status were related to the treatment response., Results: In 8/22 (36%) patients, an erythroid dominant response (EDR) pattern, defined as a ratio of myeloid cells/erythroid cells <1, was observed. In the remaining 14 cases, a myeloid predominance was preserved during treatment. No difference in the hypomethylating effect of DAC treatment was observed in patients with and without EDR, as global 5-methylcytosine levels dropped similarly in both groups. Mutational analysis by NGS using a panel of commonly mutated genes in AML showed that patients with an early EDR harbored on average less mutations, with U2AF1 mutations occurring more frequently, whereas RUNX1 mutations were underrepresented compared to non-EDR cases. Interestingly, the development of an EDR correlated with complete remission (7/8 cases with an EDR vs. only 2/14 cases without an EDR)., Conclusion: We conclude that early histological bone marrow examination for the development of an EDR may be helpful to predict response in AML patients during treatment with DAC., (© 2024 S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
10. Acid ceramidase regulates innate immune memory.
- Author
-
Rother N, Yanginlar C, Prévot G, Jonkman I, Jacobs M, van Leent MMT, van Heck J, Matzaraki V, Azzun A, Morla-Folch J, Ranzenigo A, Wang W, van der Meel R, Fayad ZA, Riksen NP, Hilbrands LB, Lindeboom RGH, Martens JHA, Vermeulen M, Joosten LAB, Netea MG, Mulder WJM, van der Vlag J, Teunissen AJP, and Duivenvoorden R
- Subjects
- Histones, Lysine, Sphingolipids genetics, Immunity, Innate, Acid Ceramidase genetics, Acid Ceramidase metabolism, Trained Immunity
- Abstract
Innate immune memory, also called "trained immunity," is a functional state of myeloid cells enabling enhanced immune responses. This phenomenon is important for host defense, but also plays a role in various immune-mediated conditions. We show that exogenously administered sphingolipids and inhibition of sphingolipid metabolizing enzymes modulate trained immunity. In particular, we reveal that acid ceramidase, an enzyme that converts ceramide to sphingosine, is a potent regulator of trained immunity. We show that acid ceramidase regulates the transcription of histone-modifying enzymes, resulting in profound changes in histone 3 lysine 27 acetylation and histone 3 lysine 4 trimethylation. We confirm our findings by identifying single-nucleotide polymorphisms in the region of ASAH1, the gene encoding acid ceramidase, that are associated with the trained immunity cytokine response. Our findings reveal an immunomodulatory effect of sphingolipids and identify acid ceramidase as a relevant therapeutic target to modulate trained immunity responses in innate immune-driven disorders., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. Understanding blood development and leukemia using sequencing-based technologies and human cell systems.
- Author
-
Heuts BMH and Martens JHA
- Abstract
Our current understanding of human hematopoiesis has undergone significant transformation throughout the years, challenging conventional views. The evolution of high-throughput technologies has enabled the accumulation of diverse data types, offering new avenues for investigating key regulatory processes in blood cell production and disease. In this review, we will explore the opportunities presented by these advancements for unraveling the molecular mechanisms underlying normal and abnormal hematopoiesis. Specifically, we will focus on the importance of enhancer-associated regulatory networks and highlight the crucial role of enhancer-derived transcription regulation. Additionally, we will discuss the unprecedented power of single-cell methods and the progression in using in vitro human blood differentiation system, in particular induced pluripotent stem cell models, in dissecting hematopoietic processes. Furthermore, we will explore the potential of ever more nuanced patient profiling to allow precision medicine approaches. Ultimately, we advocate for a multiparameter, regulatory network-based approach for providing a more holistic understanding of normal hematopoiesis and blood disorders., 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 © 2023 Heuts and Martens.)
- Published
- 2023
- Full Text
- View/download PDF
12. Saponin-based adjuvants enhance antigen cross-presentation in human CD11c + CD1c + CD5 - CD163 + conventional type 2 dendritic cells.
- Author
-
Ho NI, Huis In 't Veld LGM, van Eck van der Sluijs J, Heuts BMH, Looman MWG, Kers-Rebel ED, van den Dries K, Dolstra H, Martens JHA, Hobo W, and Adema GJ
- Subjects
- Humans, Animals, Mice, CD8-Positive T-Lymphocytes, Adjuvants, Immunologic pharmacology, Dendritic Cells, Cross-Priming, Saponins pharmacology
- Abstract
Background: Adjuvants are key for effective vaccination against cancer and chronic infectious diseases. Saponin-based adjuvants (SBAs) are unique among adjuvants in their ability to induce robust cell-mediated immune responses in addition to antibody responses. Recent preclinical studies revealed that SBAs induced cross-presentation and lipid bodies in otherwise poorly cross-presenting CD11b
+ murine dendritic cells (DCs)., Method: Here, we investigated the response of human DC subsets to SBAs with RNA sequencing and pathway analyses, lipid body induction visualized by laser scanning microscopy, antigen translocation to the cytosol, and antigen cross-presentation to CD8+ T cells., Results: RNA sequencing of SBA-treated conventional type 1 DC (cDC1) and type 2 DC (cDC2) subsets uncovered that SBAs upregulated lipid-related pathways in CD11c+ CD1c+ cDC2s, especially in the CD5- CD163+ CD14+ cDC2 subset. Moreover, SBAs induced lipid bodies and enhanced endosomal antigen translocation into the cytosol in this particular cDC2 subset. Finally, SBAs enhanced cross-presentation only in cDC2s, which requires the CD163+ CD14+ cDC2 subset., Conclusions: These data thus identify the CD163+ CD14+ cDC2 subset as the main SBA-responsive DC subset in humans and imply new strategies to optimize the application of saponin-based adjuvants in a potent cancer vaccine., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2023
- Full Text
- View/download PDF
13. Inducible MLL-AF9 Expression Drives an AML Program during Human Pluripotent Stem Cell-Derived Hematopoietic Differentiation.
- Author
-
Heuts BMH, Arza-Apalategi S, Alkema SG, Tijchon E, Jussen L, Bergevoet SM, van der Reijden BA, and Martens JHA
- Subjects
- Child, Humans, Myeloid-Lymphoid Leukemia Protein genetics, Myeloid-Lymphoid Leukemia Protein metabolism, Cell Differentiation genetics, Monocytes metabolism, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Pluripotent Stem Cells metabolism
- Abstract
A t(9;11)(p22;q23) translocation produces the MLL-AF9 fusion protein, which is found in up to 25% of de novo AML cases in children. Despite major advances, obtaining a comprehensive understanding of context-dependent MLL-AF9-mediated gene programs during early hematopoiesis is challenging. Here, we generated a human inducible pluripotent stem cell (hiPSC) model with a doxycycline dose-dependent MLL-AF9 expression. We exploited MLL-AF9 expression as an oncogenic hit to uncover epigenetic and transcriptomic effects on iPSC-derived hematopoietic development and the transformation into (pre-)leukemic states. In doing so, we observed a disruption in early myelomonocytic development. Accordingly, we identified gene profiles that were consistent with primary MLL-AF9 AML and uncovered high-confidence MLL-AF9-associated core genes that are faithfully represented in primary MLL-AF9 AML, including known and presently unknown factors. Using single-cell RNA-sequencing, we identified an increase of CD34 expressing early hematopoietic progenitor-like cell states as well as granulocyte-monocyte progenitor-like cells upon MLL-AF9 activation. Our system allows for careful chemically controlled and stepwise in vitro hiPSC-derived differentiation under serum-free and feeder-free conditions. For a disease that currently lacks effective precision medicine, our system provides a novel entry-point into exploring potential novel targets for personalized therapeutic strategies.
- Published
- 2023
- Full Text
- View/download PDF
14. High expression of an intragenic long noncoding RNA misinterpreted as high FTO oncogene expression in NPM1 mutant acute myeloid leukemia.
- Author
-
Arza-Apalategi S, Heuts BMH, Dooijes MTM, Gilissen D, van der Heijden AJP, Jansen JH, Martens JHA, and van der Reijden BA
- Subjects
- Humans, Alpha-Ketoglutarate-Dependent Dioxygenase FTO genetics, Mutation, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oncogenes, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, RNA, Long Noncoding genetics
- Published
- 2023
- Full Text
- View/download PDF
15. scANANSE gene regulatory network and motif analysis of single-cell clusters.
- Author
-
Smits JGA, Arts JA, Frölich S, Snabel RR, Heuts BMH, Martens JHA, van Heeringen SJ, and Zhou H
- Subjects
- Sequence Analysis, RNA methods, Software, Transcription Factors genetics, Gene Regulatory Networks, Leukocytes, Mononuclear
- Abstract
The recent development of single-cell techniques is essential to unravel complex biological systems. By measuring the transcriptome and the accessible genome on a single-cell level, cellular heterogeneity in a biological environment can be deciphered. Transcription factors act as key regulators activating and repressing downstream target genes, and together they constitute gene regulatory networks that govern cell morphology and identity. Dissecting these gene regulatory networks is crucial for understanding molecular mechanisms and disease, especially within highly complex biological systems. The gene regulatory network analysis software ANANSE and the motif enrichment software GimmeMotifs were both developed to analyse bulk datasets. We developed scANANSE, a software pipeline for gene regulatory network analysis and motif enrichment using single-cell RNA and ATAC datasets. The scANANSE pipeline can be run from either R or Python. First, it exports data from standard single-cell objects. Next, it automatically runs multiple comparisons of cell cluster data. Finally, it imports the results back to the single-cell object, where the result can be further visualised, integrated, and interpreted. Here, we demonstrate our scANANSE pipeline on a publicly available PBMC multi-omics dataset. It identifies well-known cell type-specific hematopoietic factors. Importantly, we also demonstrated that scANANSE combined with GimmeMotifs is able to predict transcription factors with both activating and repressing roles in gene regulation., Competing Interests: No competing interests were disclosed., (Copyright: © 2023 Smits JGA et al.)
- Published
- 2023
- Full Text
- View/download PDF
16. Differences in Immune Responses in Individuals of Indian and European Origin: Relevance for the COVID-19 Pandemic.
- Author
-
Geckin B, Zoodsma M, Kilic G, Debisarun PA, Rakshit S, Adiga V, Ahmed A, Parthiban C, Kumar NC, D'Souza G, Baltissen MP, Martens JHA, Domínguez-Andrés J, Li Y, Vyakarnam A, and Netea MG
- Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, large differences in susceptibility and mortality due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have been reported between populations in Europe and South Asia. While both host and environmental factors (including Mycobacterium bovis BCG vaccination) have been proposed to explain this, the potential biological substrate of these differences is unknown. We purified peripheral blood mononuclear cells from individuals living in India and the Netherlands at baseline and 10 to 12 weeks after BCG vaccination. We compared chromatin accessibility between the two populations at baseline, as well as gene transcription profiles and cytokine production capacities upon stimulation. The chromatin accessibility of genes important for adaptive immunity was higher in the Indians than in the Europeans, while the latter had more accessible chromatin regions in genes of the innate immune system. At the transcriptional level, we observed that the Indian volunteers displayed a more tolerant immune response to stimulation, in contrast to a more exaggerated response in the Europeans. BCG vaccination strengthened the tolerance program in the Indians but not in the Europeans. These differences may partly explain the different impact of COVID-19 on the two populations. IMPORTANCE In this study, we assessed the differences in immune responses in individuals from India and Europe. This aspect is of great relevance, because of the described differences in morbidity and mortality between India and Europe during the pandemic. We found a significant difference in chromatin accessibility in immune cells from the two populations, followed by a more balanced and effective response in individuals from India. These exciting findings represent a very important piece of the puzzle for understanding the COVID-19 pandemic at a global level.
- Published
- 2023
- Full Text
- View/download PDF
17. Transcription factor HNF1β controls a transcriptional network regulating kidney cell structure and tight junction integrity.
- Author
-
Tholen LE, Latta F, Martens JHA, Hoenderop JGJ, and de Baaij JHF
- Subjects
- Mice, Animals, Tight Junctions metabolism, Kidney metabolism, Epithelial Cells metabolism, Hepatocyte Nuclear Factors genetics, Hepatocyte Nuclear Factors metabolism, Electrolytes metabolism, Hepatocyte Nuclear Factor 1-beta genetics, Transcription Factors metabolism, Gene Regulatory Networks
- Abstract
Mutations in the hepatocyte nuclear factor (HNF)1β gene ( HNF1B ) cause autosomal dominant tubulointerstitial kidney disease, a rare and heterogeneous disease characterized by renal cysts and/or malformation, maturity-onset diabetes of the young, hypomagnesemia, and hypokalemia. The electrolyte disturbances may develop in the distal part of the nephron, which is important for fine-tuning of Mg
2+ and Ca2+ reabsorption. Therefore, we aimed to study the transcriptional network directed by HNF1β in the distal part of the nephron. We combined HNF1β chromatin immunoprecipitation-sequencing and mRNA expression data to identify direct targets of HNF1β in a renal distal convoluted tubule cell line (mpkDCT). Gene Ontology term pathway analysis demonstrated enrichment of cell polarity, cell-cell junction, and cytoskeleton pathways in the dataset. Genes directly and indirectly regulated by HNF1β within these pathways included members of the apical and basolateral polarity complexes including Crumbs protein homolog 3 ( Crb3 ), partitioning defective 6 homolog-β ( Pard6b ), and LLGL Scribble cell polarity complex component 2 ( Llgl2 ). In monolayers of mouse inner medullary collecting duct 3 cells expressing dominant negative Hnf1b, tight junction integrity was compromised, as observed by reduced transepithelial electrical resistance values and increased permeability for fluorescein (0.4 kDa) compared with wild-type cells. Expression of dominant negative Hnf1b also led to a decrease in height (30%) and an increase in surface (58.5%) of cells grown on membranes. Moreover, three-dimensional spheroids formed by cells expressing dominant negative Hnf1b were reduced in size compared with wild-type spheroids (30%). Together, these findings demonstrate that HNF1β directs a transcriptional network regulating tight junction integrity and cell structure in the distal part of the nephron. NEW & NOTEWORTHY Genetic defects in transcription factor hepatocyte nuclear factor (HNF)1β cause a heterogeneous disease characterized by electrolyte disturbances, kidney cysts, and diabetes. By combining RNA-sequencing and HNF1β chromatin immunoprecipitation-sequencing data, we identified new HNF1β targets that were enriched for cell polarity pathways. Newly discovered targets included members of polarity complexes Crb3, Pard6b, and Llgl2. Functional assays in kidney epithelial cells demonstrated decreased tight junction integrity and a loss of typical cuboidal morphology in mutant Hnf1b cells.- Published
- 2023
- Full Text
- View/download PDF
18. Regulome analysis in B-acute lymphoblastic leukemia exposes Core Binding Factor addiction as a therapeutic vulnerability.
- Author
-
Wray JP, Deltcheva EM, Boiers C, Richardson SЕ, Chhetri JB, Brown J, Gagrica S, Guo Y, Illendula A, Martens JHA, Stunnenberg HG, Bushweller JH, Nimmo R, and Enver T
- Subjects
- Adult, Child, Humans, Core Binding Factors, B-Lymphocytes, Gene Fusion, Core Binding Factor Alpha 2 Subunit genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy
- Abstract
The ETV6-RUNX1 onco-fusion arises in utero, initiating a clinically silent pre-leukemic state associated with the development of pediatric B-acute lymphoblastic leukemia (B-ALL). We characterize the ETV6-RUNX1 regulome by integrating chromatin immunoprecipitation- and RNA-sequencing and show that ETV6-RUNX1 functions primarily through competition for RUNX1 binding sites and transcriptional repression. In pre-leukemia, this results in ETV6-RUNX1 antagonization of cell cycle regulation by RUNX1 as evidenced by mass cytometry analysis of B-lineage cells derived from ETV6-RUNX1 knock-in human pluripotent stem cells. In frank leukemia, knockdown of RUNX1 or its co-factor CBFβ results in cell death suggesting sustained requirement for RUNX1 activity which is recapitulated by chemical perturbation using an allosteric CBFβ-inhibitor. Strikingly, we show that RUNX1 addiction extends to other genetic subtypes of pediatric B-ALL and also adult disease. Importantly, inhibition of RUNX1 activity spares normal hematopoiesis. Our results suggest that chemical intervention in the RUNX1 program may provide a therapeutic opportunity in ALL., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
19. Dietary methionine starvation impairs acute myeloid leukemia progression.
- Author
-
Cunningham A, Erdem A, Alshamleh I, Geugien M, Pruis M, Pereira-Martins DA, van den Heuvel FAJ, Wierenga ATJ, Ten Berge H, Dennebos R, van den Boom V, Hogeling SM, Weinhäuser I, Knops R, de Blaauw P, Heiner-Fokkema MR, Woolthuis C, Günther UL, Rego EM, Martens JHA, Jansen JH, Schwalbe H, Huls G, and Schuringa JJ
- Subjects
- Mice, Animals, S-Adenosylmethionine metabolism, S-Adenosylmethionine therapeutic use, Histones metabolism, Racemethionine, Methionine, Leukemia, Myeloid, Acute pathology
- Abstract
Targeting altered tumor cell metabolism might provide an attractive opportunity for patients with acute myeloid leukemia (AML). An amino acid dropout screen on primary leukemic stem cells and progenitor populations revealed a number of amino acid dependencies, of which methionine was one of the strongest. By using various metabolite rescue experiments, nuclear magnetic resonance-based metabolite quantifications and 13C-tracing, polysomal profiling, and chromatin immunoprecipitation sequencing, we identified that methionine is used predominantly for protein translation and to provide methyl groups to histones via S-adenosylmethionine for epigenetic marking. H3K36me3 was consistently the most heavily impacted mark following loss of methionine. Methionine depletion also reduced total RNA levels, enhanced apoptosis, and induced a cell cycle block. Reactive oxygen species levels were not increased following methionine depletion, and replacement of methionine with glutathione or N-acetylcysteine could not rescue phenotypes, excluding a role for methionine in controlling redox balance control in AML. Although considered to be an essential amino acid, methionine can be recycled from homocysteine. We uncovered that this is primarily performed by the enzyme methionine synthase and only when methionine availability becomes limiting. In vivo, dietary methionine starvation was not only tolerated by mice, but also significantly delayed both cell line and patient-derived AML progression. Finally, we show that inhibition of the H3K36-specific methyltransferase SETD2 phenocopies much of the cytotoxic effects of methionine depletion, providing a more targeted therapeutic approach. In conclusion, we show that methionine depletion is a vulnerability in AML that can be exploited therapeutically, and we provide mechanistic insight into how cells metabolize and recycle methionine., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
20. Identification of transcription factors dictating blood cell development using a bidirectional transcription network-based computational framework.
- Author
-
Heuts BMH, Arza-Apalategi S, Frölich S, Bergevoet SM, van den Oever SN, van Heeringen SJ, van der Reijden BA, and Martens JHA
- Subjects
- Humans, Transcription Factors genetics, Transcription Factors metabolism, Algorithms, Blood Cells metabolism, Computational Biology, Gene Regulatory Networks, Leukemia, Myeloid, Acute genetics
- Abstract
Advanced computational methods exploit gene expression and epigenetic datasets to predict gene regulatory networks controlled by transcription factors (TFs). These methods have identified cell fate determining TFs but require large amounts of reference data and experimental expertise. Here, we present an easy to use network-based computational framework that exploits enhancers defined by bidirectional transcription, using as sole input CAGE sequencing data to correctly predict TFs key to various human cell types. Next, we applied this Analysis Algorithm for Networks Specified by Enhancers based on CAGE (ANANSE-CAGE) to predict TFs driving red and white blood cell development, and THP-1 leukemia cell immortalization. Further, we predicted TFs that are differentially important to either cell line- or primary- associated MLL-AF9-driven gene programs, and in primary MLL-AF9 acute leukemia. Our approach identified experimentally validated as well as thus far unexplored TFs in these processes. ANANSE-CAGE will be useful to identify transcription factors that are key to any cell fate change using only CAGE-seq data as input., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. Dynamics of broad H3K4me3 domains uncover an epigenetic switch between cell identity and cancer-related genes.
- Author
-
Belhocine M, Simonin M, Abad Flores JD, Cieslak A, Manosalva I, Pradel L, Smith C, Mathieu EL, Charbonnier G, Martens JHA, Stunnenberg HG, Maqbool MA, Mikulasova A, Russell LJ, Rico D, Puthier D, Ferrier P, Asnafi V, and Spicuglia S
- Subjects
- Epigenesis, Genetic, Histones metabolism, Humans, Oncogenes, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Broad domains of H3K4 methylation have been associated with consistent expression of tissue-specific, cell identity, and tumor suppressor genes. Here, we identified broad domain-associated genes in healthy human thymic T cell populations and a collection of T cell acute lymphoblastic leukemia (T-ALL) primary samples and cell lines. We found that broad domains are highly dynamic throughout T cell differentiation, and their varying breadth allows the distinction between normal and neoplastic cells. Although broad domains preferentially associate with cell identity and tumor suppressor genes in normal thymocytes, they flag key oncogenes in T-ALL samples. Moreover, the expression of broad domain-associated genes, both coding and noncoding, is frequently deregulated in T-ALL. Using two distinct leukemic models, we showed that the ectopic expression of T-ALL oncogenic transcription factor preferentially impacts the expression of broad domain-associated genes in preleukemic cells. Finally, an H3K4me3 demethylase inhibitor differentially targets T-ALL cell lines depending on the extent and number of broad domains. Our results show that the regulation of broad H3K4me3 domains is associated with leukemogenesis, and suggest that the presence of these structures might be used for epigenetic prioritization of cancer-relevant genes, including long noncoding RNAs., (© 2022 Belhocine et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
22. CBX2 shapes chromatin accessibility promoting AML via p38 MAPK signaling pathway.
- Author
-
Del Gaudio N, Di Costanzo A, Liu NQ, Conte L, Dell'Aversana C, Bove G, Benedetti R, Montella L, Ciardiello F, Carafa V, Ambrosino C, Tucci V, Conte M, Martens JHA, Stunnenberg HG, Nebbioso A, and Altucci L
- Subjects
- Humans, Signal Transduction, p38 Mitogen-Activated Protein Kinases genetics, p38 Mitogen-Activated Protein Kinases metabolism, Chromatin genetics, Leukemia, Myeloid, Acute drug therapy, Polycomb Repressive Complex 1 genetics, Polycomb Repressive Complex 1 metabolism
- Abstract
Background: The dynamic epigenome and proteins specialized in the interpretation of epigenetic marks critically contribute to leukemic pathogenesis but also offer alternative therapeutic avenues. Targeting newly discovered chromatin readers involved in leukemogenesis may thus provide new anticancer strategies. Accumulating evidence suggests that the PRC1 complex member CBX2 is overexpressed in solid tumors and promotes cancer cell survival. However, its role in leukemia is still unclear., Methods: We exploited reverse genetic approaches to investigate the role of CBX2 in human leukemic cell lines and ex vivo samples. We also analyzed phenotypic effects following CBX2 silencing using cellular and molecular assays and related functional mechanisms by ATAC-seq and RNA-seq. We then performed bioinformatic analysis of ChIP-seq data to explore the influence of histone modifications in CBX2-mediated open chromatin sites. Lastly, we used molecular assays to determine the contribution of CBX2-regulated pathways to leukemic phenotype., Results: We found CBX2 overexpressed in leukemia both in vitro and ex vivo samples compared to CD34
+ cells. Decreased CBX2 RNA levels prompted a robust reduction in cell proliferation and induction of apoptosis. Similarly, sensitivity to CBX2 silencing was observed in primary acute myeloid leukemia samples. CBX2 suppression increased genome-wide chromatin accessibility followed by alteration of leukemic cell transcriptional programs, resulting in enrichment of cell death pathways and downregulation of survival genes. Intriguingly, CBX2 silencing induced epigenetic reprogramming at p38 MAPK-associated regulatory sites with consequent deregulation of gene expression., Conclusions: Our results identify CBX2 as a crucial player in leukemia progression and highlight a potential druggable CBX2-p38 MAPK network in AML., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
23. Direct targeted therapy for MLL-fusion-driven high-risk acute leukaemias.
- Author
-
Cantilena S, Gasparoli L, Pal D, Heidenreich O, Klusmann JH, Martens JHA, Faille A, Warren AJ, Karsa M, Pandher R, Somers K, Williams O, and de Boer J
- Subjects
- Acute Disease, Apoptosis, Cell Proliferation, Child, Epigenesis, Genetic, Humans, Infant, Leukemia genetics, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism
- Abstract
Background: Improving the poor prognosis of infant leukaemias remains an unmet clinical need. This disease is a prototypical fusion oncoprotein-driven paediatric cancer, with MLL (KMT2A)-fusions present in most cases. Direct targeting of these driving oncoproteins represents a unique therapeutic opportunity. This rationale led us to initiate a drug screening with the aim of discovering drugs that can block MLL-fusion oncoproteins., Methods: A screen for inhibition of MLL-fusion proteins was developed that overcomes the traditional limitations of targeting transcription factors. This luciferase reporter-based screen, together with a secondary western blot screen, was used to prioritize compounds. We characterized the lead compound, disulfiram (DSF), based on its efficient ablation of MLL-fusion proteins. The consequences of drug-induced MLL-fusion inhibition were confirmed by cell proliferation, colony formation, apoptosis assays, RT-qPCR, in vivo assays, RNA-seq and ChIP-qPCR and ChIP-seq analysis. All statistical tests were two-sided., Results: Drug-induced inhibition of MLL-fusion proteins by DSF resulted in a specific block of colony formation in MLL-rearranged cells in vitro, induced differentiation and impeded leukaemia progression in vivo. Mechanistically, DSF abrogates MLL-fusion protein binding to DNA, resulting in epigenetic changes and down-regulation of leukaemic programmes setup by the MLL-fusion protein., Conclusion: DSF can directly inhibit MLL-fusion proteins and demonstrate antitumour activity both in vitro and in vivo, providing, to our knowledge, the first evidence for a therapy that directly targets the initiating oncogenic MLL-fusion protein., (© 2022 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.)
- Published
- 2022
- Full Text
- View/download PDF
24. Presence of mutant p53 increases stem cell frequency and is associated with reduced binding to classic TP53 binding sites in cell lines and primary AMLs.
- Author
-
Gerritsen M, Hilgendorf S, Yi G, Wierenga ATJ, Schuringa JJ, Martens JHA, and Vellenga E
- Subjects
- Binding Sites, Cell Line, DNA, Hematopoietic Stem Cells metabolism, Humans, Mutation, Leukemia, Myeloid, Acute pathology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
With an overall 5%-10% incidence rate in acute myeloid leukemia (AML), the occurrence of TP53 mutations is low compared with that in solid tumors. However, when focusing on high-risk groups including secondary AML (sAML) and therapy-related AMLs, the frequency of mutations reaches up to 35%. Mutations may include loss of heterozygosity (LOH) or deletion of the 17p allele, but are mostly missense substitutions that are located in the DNA-binding domain. Despite elaborate research on the effects of TP53 mutations in solid tumors, in hematological malignancies, the effects of TP53 mutations versus loss of TP53 remain unclear and under debate. Here, we compared the cellular effects of a TP53 mutant and loss of TP53 in human hematopoietic stem and progenitor cells (HSPCs). We found that when expressing TP53 mutant or loss of TP53 using siRNA, CD34
+ /CD38- cells have a significantly enhanced replating potential, which could not be demonstrated for the CD34+ /CD38+ population. Using RNA-sequencing analysis, we found a loss of expression of p53 target genes in cells with TP53 knockdown. In contrast, an increased expression of a large number of genes was observed when expressing TP53 mutant, resulting in an increase in expression of genes involved in megakaryocytic differentiation, plasma membrane binding, and extracellular structure organization. When binding of p53 wild type and p53 mutant was compared in cell lines, we found that mutant p53 binds to a large number of binding sites genomewide, contrary to wild-type p53, for which binding is restricted to genes with a p53 binding motif. These findings were verified in primary AMLs with and without mutated TP53. In conclusion, in our models, we identified overlapping effects of TP53 mutant and loss of TP53 on in vitro stem cell properties but distinct effects on DNA binding and gene expression., Competing Interests: Conflict of Interest Disclosure The authors declare that there is no conflict of interest regarding the publication of this article., (Copyright © 2022 ISEH -- Society for Hematology and Stem Cells. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
25. Genetic diversity within leukemia-associated immunophenotype-defined subclones in AML.
- Author
-
Tiso F, Koorenhof-Scheele TN, Huys E, Martens JHA, de Graaf AO, van der Reijden BA, Langemeijer SMC, Preijers FWMB, Kroeze LI, and Jansen JH
- Subjects
- Clonal Evolution, Gene Expression Regulation, Leukemic, Genetic Variation, Humans, Immunophenotyping, Mutation, Leukemia, Myeloid, Acute genetics
- Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous disease showing dynamic clonal evolution patterns over time. Various subclones may be present simultaneously and subclones may show a different expansion pattern and respond differently to applied therapies. It is already clear that immunophenotyping and genetic analyses may yield overlapping, but also complementary information. Detailed information on the genetic make-up of immunophenotypically defined subclones is however scarce. We performed error-corrected sequencing for 27 myeloid leukemia driver genes in 86, FACS-sorted immunophenotypically characterized normal and aberrant subfractions in 10 AML patients. We identified three main scenarios. In the first group of patients, the two techniques were equally well characterizing the malignancy. In the second group, most of the isolated populations did not express aberrant immunophenotypes but still harbored several genetic aberrancies, indicating that the information obtained only by immunophenotyping would be incomplete. Vice versa, one patient was identified in which genetic mutations were found only in a small fraction of the immunophenotypically defined malignant populations, indicating that the genetic analysis gave an incomplete picture of the disease. We conclude that currently, characterization of leukemic cells in AML by molecular and immunophenotypic techniques is complementary, and infer that both techniques should be used in parallel in order to obtain the most complete view on the disease., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
26. Characterization of a genomic region 8 kb downstream of GFI1B associated with myeloproliferative neoplasms.
- Author
-
van Bergen MGJM, van Oorschot R, Bergevoet SM, de Graaf AO, Tönnissen ELRTM, Stevens-Linders E, Neveling K, Jansen PWTC, Baltissen MPA, Vermeulen M, Mandoli A, Martens JHA, Preijers F, Jansen JH, and van der Reijden BA
- Subjects
- Adult, Aged, Alleles, CRISPR-Cas Systems genetics, Female, Gene Expression Regulation genetics, Genome, Human genetics, Genotype, High-Throughput Nucleotide Sequencing, Humans, K562 Cells, Male, Middle Aged, Myeloid Cells metabolism, Myeloid Cells pathology, Myeloproliferative Disorders pathology, Neoplasms pathology, Phagocytosis genetics, Polymorphism, Single Nucleotide genetics, Young Adult, Genetic Predisposition to Disease, Myeloproliferative Disorders genetics, Neoplasms genetics, Proto-Oncogene Proteins genetics, Repressor Proteins genetics
- Abstract
A genomic locus 8 kb downstream of the transcription factor GFI1B (Growth Factor Independence 1B) predisposes to clonal hematopoiesis and myeloproliferative neoplasms. One of the most significantly associated polymorphisms in this region is rs621940-G. GFI1B auto-represses GFI1B, and altered GFI1B expression contributes to myeloid neoplasms. We studied whether rs621940-G affects GFI1B expression and growth of immature cells. GFI1B ChIP-seq showed clear binding to the rs621940 locus. Preferential binding of various hematopoietic transcription factors to either the rs621940-C or -G allele was observed, but GFI1B showed no preference. In gene reporter assays the rs621940 region inhibited GFI1B promoter activity with the G-allele having less suppressive effects compared to the C-allele. However, CRISPR-Cas9 mediated deletion of the locus in K562 cells did not alter GFI1B expression nor auto-repression. In healthy peripheral blood mononuclear cells GFI1B expression did not differ consistently between the rs621940 alleles. Long range and targeted deep sequencing did not detect consistent effects of rs621940-G on allelic GFI1B expression either. Finally, we observed that myeloid colony formation was not significantly affected by either rs621940 allele in 193 healthy donors. Together, these findings show no evidence that rs621940 or its locus affect GFI1B expression, auto-repression or growth of immature myeloid cells., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
27. The EHA Research Roadmap: Normal Hematopoiesis.
- Author
-
Jaffredo T, Balduini A, Bigas A, Bernardi R, Bonnet D, Canque B, Charbord P, Cumano A, Delwel R, Durand C, Fibbe W, Forrester L, de Franceschi L, Ghevaert C, Gjertsen B, Gottgens B, Graf T, Heidenreich O, Hermine O, Higgs D, Kleanthous M, Klump H, Kouskoff V, Krause D, Lacaud G, Celso CL, Martens JHA, Méndez-Ferrer S, Menendez P, Oostendorp R, Philipsen S, Porse B, Raaijmakers M, Robin C, Stunnenberg H, Theilgaard-Mönch K, Touw I, Vainchenker W, Corrons JV, Yvernogeau L, and Schuringa JJ
- Published
- 2021
- Full Text
- View/download PDF
28. Cell type-specific novel long non-coding RNA and circular RNA in the BLUEPRINT hematopoietic transcriptomes atlas.
- Author
-
Grassi L, Izuogu OG, Jorge NAN, Seyres D, Bustamante M, Burden F, Farrow S, Farahi N, Martin FJ, Frankish A, Mudge JM, Kostadima M, Petersen R, Lambourne JJ, Rowlston S, Martin-Rendon E, Clarke L, Downes K, Estivill X, Flicek P, Martens JHA, Yaspo ML, Stunnenberg HG, Ouwehand WH, Passetti F, Turro E, and Frontini M
- Subjects
- Gene Expression Profiling, High-Throughput Nucleotide Sequencing, RNA, Circular, Sequence Analysis, RNA, RNA, Long Noncoding genetics, Transcriptome
- Abstract
Transcriptional profiling of hematopoietic cell subpopulations has helped to characterize the developmental stages of the hematopoietic system and the molecular bases of malignant and non-malignant blood diseases. Previously, only the genes targeted by expression microarrays could be profiled genome-wide. High-throughput RNA sequencing, however, encompasses a broader repertoire of RNA molecules, without restriction to previously annotated genes. We analyzed the BLUEPRINT consortium RNA-sequencing data for mature hematopoietic cell types. The data comprised 90 total RNA-sequencing samples, each composed of one of 27 cell types, and 32 small RNA-sequencing samples, each composed of one of 11 cell types. We estimated gene and isoform expression levels for each cell type using existing annotations from Ensembl. We then used guided transcriptome assembly to discover unannotated transcripts. We identified hundreds of novel non-coding RNA genes and showed that the majority have cell type-dependent expression. We also characterized the expression of circular RNA and found that these are also cell type-specific. These analyses refine the active transcriptional landscape of mature hematopoietic cells, highlight abundant genes and transcriptional isoforms for each blood cell type, and provide a valuable resource for researchers of hematologic development and diseases. Finally, we made the data accessible via a web-based interface: https://blueprint.haem.cam.ac.uk/bloodatlas/.
- Published
- 2021
- Full Text
- View/download PDF
29. Recent insights into Histone Acetyltransferase-1 : biological function and involvement in pathogenesis.
- Author
-
Poziello A, Nebbioso A, Stunnenberg HG, Martens JHA, Carafa V, and Altucci L
- Subjects
- Acetylation, Histones metabolism, DNA Methylation, Histone Acetyltransferases genetics
- Abstract
Acetylation of histone and non-histone proteins is a post-translational modification mostly associated with activation of gene transcription. The first histone acetyltransferase (HAT) identified as modifying newly synthesized histone H4 in yeast was a type B HAT named HAT1 . Although it was the first HAT to be discovered, HAT1 remains one of the most poorly studied enzymes in its class. In addition to its well-established role in the cytoplasm, recent findings have revealed new and intriguing aspects of the function of HAT1 in the nucleus. Several studies have described its involvement in regulating different pathways associated with a wide range of diseases, including cancer. This review focuses on our current understanding of HAT1 , highlighting its importance in regulating chromatin replication and gene expression. This previously unknown role for HAT1 opens up novel scenarios in which further studies will be required to better understand its function.
- Published
- 2021
- Full Text
- View/download PDF
30. Specific proteome changes in platelets from individuals with GATA1-, GFI1B-, and RUNX1-linked bleeding disorders.
- Author
-
Van Bergen MGJM, Marneth AE, Hoogendijk AJ, Van Alphen FPJ, Van den Akker E, Laros-Van Gorkom BAP, Hoeks M, Simons A, De Munnik SA, Janssen JJWM, Martens JHA, Jansen JH, Meijer AB, and Van der Reijden BA
- Subjects
- Homeostasis, Humans, Mutation genetics, Signal Transduction, Transcription Factors genetics, Transcription Factors metabolism, Blood Platelet Disorders metabolism, Blood Platelets metabolism, Core Binding Factor Alpha 2 Subunit metabolism, GATA1 Transcription Factor metabolism, Proteome metabolism, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism
- Abstract
Mutations in the transcription factors GATA binding factor 1 (GATA1), growth factor independence 1B (GFI1B), and Runt-related transcription factor 1 (RUNX1) cause familial platelet and bleeding disorders. Mutant platelets exhibit common abnormalities including an α-granule reduction resulting in a grayish appearance in blood smears. This suggests that similar pathways are deregulated by different transcription factor mutations. To identify common factors, full platelet proteomes from 11 individuals with mutant GATA1R216Q, GFI1BQ287*, RUNX1Q154Rfs, or RUNX1TD2-6 and 28 healthy controls were examined by label-free quantitative mass spectrometry. In total, 2875 platelet proteins were reliably quantified. Clustering analysis of more than 300 differentially expressed proteins revealed profound differences between cases and controls. Among cases, 44 of 143 significantly downregulated proteins were assigned to platelet function, hemostasis, and granule biology, in line with platelet dysfunction and bleedings. Remarkably, none of these proteins were significantly diminished in all affected cases. Similarly, no proteins were commonly overrepresented in all affected cases compared with controls. These data indicate that the studied transcription factor mutations alter platelet proteomes in distinct largely nonoverlapping manners. This work provides the quantitative landscape of proteins that affect platelet function when deregulated by mutated transcription factors in inherited bleeding disorders., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
31. Mutational mechanisms shaping the coding and noncoding genome of germinal center derived B-cell lymphomas.
- Author
-
Hübschmann D, Kleinheinz K, Wagener R, Bernhart SH, López C, Toprak UH, Sungalee S, Ishaque N, Kretzmer H, Kreuz M, Waszak SM, Paramasivam N, Ammerpohl O, Aukema SM, Beekman R, Bergmann AK, Bieg M, Binder H, Borkhardt A, Borst C, Brors B, Bruns P, Carrillo de Santa Pau E, Claviez A, Doose G, Haake A, Karsch D, Haas S, Hansmann ML, Hoell JI, Hovestadt V, Huang B, Hummel M, Jäger-Schmidt C, Kerssemakers JNA, Korbel JO, Kube D, Lawerenz C, Lenze D, Martens JHA, Ott G, Radlwimmer B, Reisinger E, Richter J, Rico D, Rosenstiel P, Rosenwald A, Schillhabel M, Stilgenbauer S, Stadler PF, Martín-Subero JI, Szczepanowski M, Warsow G, Weniger MA, Zapatka M, Valencia A, Stunnenberg HG, Lichter P, Möller P, Loeffler M, Eils R, Klapper W, Hoffmann S, Trümper L, Küppers R, Schlesner M, and Siebert R
- Subjects
- Adult, B-Lymphocytes metabolism, Cell Line, Cell Line, Tumor, Genes, Immunoglobulin genetics, HeLa Cells, Hep G2 Cells, Human Umbilical Vein Endothelial Cells, Humans, Immunoglobulin Class Switching genetics, K562 Cells, MCF-7 Cells, Somatic Hypermutation, Immunoglobulin genetics, V(D)J Recombination genetics, Genome genetics, Germinal Center metabolism, Lymphoma, B-Cell genetics, Mutation genetics
- Abstract
B cells have the unique property to somatically alter their immunoglobulin (IG) genes by V(D)J recombination, somatic hypermutation (SHM) and class-switch recombination (CSR). Aberrant targeting of these mechanisms is implicated in lymphomagenesis, but the mutational processes are poorly understood. By performing whole genome and transcriptome sequencing of 181 germinal center derived B-cell lymphomas (gcBCL) we identified distinct mutational signatures linked to SHM and CSR. We show that not only SHM, but presumably also CSR causes off-target mutations in non-IG genes. Kataegis clusters with high mutational density mainly affected early replicating regions and were enriched for SHM- and CSR-mediated off-target mutations. Moreover, they often co-occurred in loci physically interacting in the nucleus, suggesting that mutation hotspots promote increased mutation targeting of spatially co-localized loci (termed hypermutation by proxy). Only around 1% of somatic small variants were in protein coding sequences, but in about half of the driver genes, a contribution of B-cell specific mutational processes to their mutations was found. The B-cell-specific mutational processes contribute to both lymphoma initiation and intratumoral heterogeneity. Overall, we demonstrate that mutational processes involved in the development of gcBCL are more complex than previously appreciated, and that B cell-specific mutational processes contribute via diverse mechanisms to lymphomagenesis.
- Published
- 2021
- Full Text
- View/download PDF
32. Potential impact of celiac disease genetic risk factors on T cell receptor signaling in gluten-specific CD4+ T cells.
- Author
-
Bakker OB, Ramírez-Sánchez AD, Borek ZA, de Klein N, Li Y, Modderman R, Kooy-Winkelaar Y, Johannesen MK, Matarese F, Martens JHA, Kumar V, van Bergen J, Qiao SW, Lundin KEA, Sollid LM, Koning F, Wijmenga C, Withoff S, and Jonkers IH
- Subjects
- Biomarkers metabolism, CD4-Positive T-Lymphocytes metabolism, Celiac Disease chemically induced, Celiac Disease pathology, Cytokines immunology, Cytokines metabolism, Gene Expression Profiling, Gene Expression Regulation, Glutens administration & dosage, Glutens immunology, Humans, Receptors, Antigen, T-Cell genetics, Transcriptome, CD4-Positive T-Lymphocytes immunology, Celiac Disease genetics, Celiac Disease immunology, Receptors, Antigen, T-Cell immunology
- Abstract
Celiac disease is an auto-immune disease in which an immune response to dietary gluten leads to inflammation and subsequent atrophy of small intestinal villi, causing severe bowel discomfort and malabsorption of nutrients. The major instigating factor for the immune response in celiac disease is the activation of gluten-specific CD4+ T cells expressing T cell receptors that recognize gluten peptides presented in the context of HLA-DQ2 and DQ8. Here we provide an in-depth characterization of 28 gluten-specific T cell clones. We assess their transcriptional and epigenetic response to T cell receptor stimulation and link this to genetic factors associated with celiac disease. Gluten-specific T cells have a distinct transcriptional profile that mostly resembles that of Th1 cells but also express cytokines characteristic of other types of T-helper cells. This transcriptional response appears not to be regulated by changes in chromatin state, but rather by early upregulation of transcription factors and non-coding RNAs that likely orchestrate the subsequent activation of genes that play a role in immune pathways. Finally, integration of chromatin and transcription factor binding profiles suggest that genes activated by T cell receptor stimulation of gluten‑specific T cells may be impacted by genetic variation at several genetic loci associated with celiac disease.
- Published
- 2021
- Full Text
- View/download PDF
33. The USP7-TRIM27 axis mediates non-canonical PRC1.1 function and is a druggable target in leukemia.
- Author
-
Maat H, Atsma TJ, Hogeling SM, Rodríguez López A, Jaques J, Olthuis M, de Vries MP, Gravesteijn C, Brouwers-Vos AZ, van der Meer N, Datema S, Salzbrunn J, Huls G, Baas R, Martens JHA, van den Boom V, and Schuringa JJ
- Abstract
In an attempt to unravel functionality of the non-canonical PRC1.1 Polycomb complex in human leukemogenesis, we show that USP7 and TRIM27 are integral components of PRC1.1. USP7 interactome analyses show that PRC1.1 is the predominant Polycomb complex co-precipitating with USP7. USP7 inhibition results in PRC1.1 disassembly and loss of chromatin binding, coinciding with reduced H2AK119ub and H3K27ac levels and diminished gene transcription of active PRC1.1-controlled loci, whereas H2AK119ub marks are also lost at PRC1 loci. TRIM27 and USP7 are reciprocally required for incorporation into PRC1.1, and TRIM27 knockdown partially rescues USP7 inhibitor sensitivity. USP7 inhibitors effectively impair proliferation in AML cells in vitro , also independent of the USP7-MDM2-TP53 axis, and MLL-AF9-induced leukemia is delayed in vivo in human leukemia xenografts. We propose a model where USP7 counteracts TRIM27 E3 ligase activity, thereby maintaining PRC1.1 integrity and function. Moreover, USP7 inhibition may be a promising new strategy to treat AML patients., Competing Interests: The authors declare no conflict of interests., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
34. The Role of Necroptosis: Biological Relevance and Its Involvement in Cancer.
- Author
-
Della Torre L, Nebbioso A, Stunnenberg HG, Martens JHA, Carafa V, and Altucci L
- Abstract
Regulated cell death mechanisms are essential for the maintenance of cellular homeostasis. Evasion of cell death is one of the most important hallmarks of cancer. Necroptosis is a caspase independent form of regulated cell death, investigated as a novel therapeutic strategy to eradicate apoptosis resistant cancer cells. The process can be triggered by a variety of stimuli and is controlled by the activation of RIP kinases family as well as MLKL. The well-studied executor, RIPK1, is able to modulate key cellular events through the interaction with several proteins, acting as strategic crossroads of several molecular pathways. Little evidence is reported about its involvement in tumorigenesis. In this review, we summarize current studies on the biological relevance of necroptosis, its contradictory role in cancer and its function in cell fate control. Targeting necroptosis might be a novel therapeutic intervention strategy in anticancer therapies as a pharmacologically controllable event.
- Published
- 2021
- Full Text
- View/download PDF
35. Stronger induction of trained immunity by mucosal BCG or MTBVAC vaccination compared to standard intradermal vaccination.
- Author
-
Vierboom MPM, Dijkman K, Sombroek CC, Hofman SO, Boot C, Vervenne RAW, Haanstra KG, van der Sande M, van Emst L, Domínguez-Andrés J, Moorlag SJCFM, Kocken CHM, Thole J, Rodríguez E, Puentes E, Martens JHA, van Crevel R, Netea MG, Aguilo N, Martin C, and Verreck FAW
- Subjects
- Acetylation, Administration, Intranasal, Animals, Bone Marrow drug effects, Bone Marrow immunology, Bone Marrow microbiology, Cellular Reprogramming genetics, Cellular Reprogramming immunology, Female, Gene Expression Regulation, Histones genetics, Histones immunology, Injections, Intravenous, Interleukin-1beta genetics, Interleukin-1beta immunology, Interleukin-6 genetics, Interleukin-6 immunology, Lung drug effects, Lung immunology, Lung microbiology, Macaca mulatta, Male, Monocytes drug effects, Monocytes immunology, Monocytes microbiology, Mycobacterium tuberculosis pathogenicity, Respiratory Mucosa microbiology, Tuberculosis, Pulmonary genetics, Tuberculosis, Pulmonary immunology, Tuberculosis, Pulmonary microbiology, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha immunology, BCG Vaccine administration & dosage, Immunity, Mucosal, Mycobacterium tuberculosis immunology, Respiratory Mucosa immunology, Tuberculosis Vaccines administration & dosage, Tuberculosis, Pulmonary prevention & control
- Abstract
BCG vaccination can strengthen protection against pathogens through the induction of epigenetic and metabolic reprogramming of innate immune cells, a process called trained immunity. We and others recently demonstrated that mucosal or intravenous BCG better protects rhesus macaques from Mycobacterium tuberculosis infection and TB disease than standard intradermal vaccination, correlating with local adaptive immune signatures. In line with prior mouse data, here, we show in rhesus macaques that intravenous BCG enhances innate cytokine production associated with changes in H3K27 acetylation typical of trained immunity. Alternative delivery of BCG does not alter the cytokine production of unfractionated bronchial lavage cells. However, mucosal but not intradermal vaccination, either with BCG or the M. tuberculosis -derived candidate MTBVAC, enhances innate cytokine production by blood- and bone marrow-derived monocytes associated with metabolic rewiring, typical of trained immunity. These results provide support to strategies for improving TB vaccination and, more broadly, modulating innate immunity via mucosal surfaces., Competing Interests: E.R., E.P., N.A., and C.M. are co-inventors on a patent on a tuberculosis vaccine held by the University of Zaragoza and Biofabri. E.R. and E.P. are employees of Biofabri. J.T. is an employee of the TuBerculosis Vaccine Initiative and an advisor to Biofabri. M.G.N. holds a patent on the inhibition of trained immunity with nanobiologics, and a patent on the stimulation of trained immunity with nanobiologics. M.G.N. is also a scientific founder of Trained Therapeutix and Discovery (TTxD). The remaining authors of the Radboud UMC and the authors from the BPRC declare no competing interests., (© 2020 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
36. Blueprint of human thymopoiesis reveals molecular mechanisms of stage-specific TCR enhancer activation.
- Author
-
Cieslak A, Charbonnier G, Tesio M, Mathieu EL, Belhocine M, Touzart A, Smith C, Hypolite G, Andrieu GP, Martens JHA, Janssen-Megens E, Gut M, Gut I, Boissel N, Petit A, Puthier D, Macintyre E, Stunnenberg HG, Spicuglia S, and Asnafi V
- Subjects
- Animals, Apoptosis Regulatory Proteins genetics, Cell Differentiation genetics, Cell Nucleus metabolism, Chromatin metabolism, DNA Demethylation, DNA Methylation genetics, Epigenome, Gene Expression Regulation, Gene Rearrangement, T-Lymphocyte, Histones metabolism, Homeodomain Proteins genetics, Humans, Lymphocyte Activation immunology, Mice, Protein Binding, Protein Processing, Post-Translational, Stem Cells cytology, T-Lymphocytes cytology, Thymocytes metabolism, Enhancer Elements, Genetic, Hematopoiesis genetics, Receptors, Antigen, T-Cell genetics, Thymus Gland cytology
- Abstract
Cell differentiation is accompanied by epigenetic changes leading to precise lineage definition and cell identity. Here we present a comprehensive resource of epigenomic data of human T cell precursors along with an integrative analysis of other hematopoietic populations. Although T cell commitment is accompanied by large scale epigenetic changes, we observed that the majority of distal regulatory elements are constitutively unmethylated throughout T cell differentiation, irrespective of their activation status. Among these, the TCRA gene enhancer (Eα) is in an open and unmethylated chromatin structure well before activation. Integrative analyses revealed that the HOXA5-9 transcription factors repress the Eα enhancer at early stages of T cell differentiation, while their decommission is required for TCRA locus activation and enforced αβ T lineage differentiation. Remarkably, the HOXA-mediated repression of Eα is paralleled by the ectopic expression of homeodomain-related oncogenes in T cell acute lymphoblastic leukemia. These results highlight an analogous enhancer repression mechanism at play in normal and cancer conditions, but imposing distinct developmental constraints., Competing Interests: Disclosures: The authors declare no competing interests exist., (© 2020 Cieslak et al.)
- Published
- 2020
- Full Text
- View/download PDF
37. Chromatin activation as a unifying principle underlying pathogenic mechanisms in multiple myeloma.
- Author
-
Ordoñez R, Kulis M, Russiñol N, Chapaprieta V, Carrasco-Leon A, García-Torre B, Charalampopoulou S, Clot G, Beekman R, Meydan C, Duran-Ferrer M, Verdaguer-Dot N, Vilarrasa-Blasi R, Soler-Vila P, Garate L, Miranda E, San José-Enériz E, Rodriguez-Madoz JR, Ezponda T, Martínez-Turrilas R, Vilas-Zornoza A, Lara-Astiaso D, Dupéré-Richer D, Martens JHA, El-Omri H, Taha RY, Calasanz MJ, Paiva B, San Miguel J, Flicek P, Gut I, Melnick A, Mitsiades CS, Licht JD, Campo E, Stunnenberg HG, Agirre X, Prosper F, and Martin-Subero JI
- Subjects
- Cell Line, DNA-Binding Proteins metabolism, Epigenesis, Genetic, Humans, NF-kappa B metabolism, Osteogenesis genetics, Receptors, Notch metabolism, Signal Transduction, TOR Serine-Threonine Kinases metabolism, Thioredoxins metabolism, Transcription Factors metabolism, Tumor Suppressor Protein p53 metabolism, Up-Regulation, Chromatin metabolism, Gene Expression Regulation, Neoplastic, Multiple Myeloma genetics, Plasma Cells metabolism
- Abstract
Multiple myeloma (MM) is a plasma cell neoplasm associated with a broad variety of genetic lesions. In spite of this genetic heterogeneity, MMs share a characteristic malignant phenotype whose underlying molecular basis remains poorly characterized. In the present study, we examined plasma cells from MM using a multi-epigenomics approach and demonstrated that, when compared to normal B cells, malignant plasma cells showed an extensive activation of regulatory elements, in part affecting coregulated adjacent genes. Among target genes up-regulated by this process, we found members of the NOTCH, NF-kB, MTOR signaling, and TP53 signaling pathways. Other activated genes included sets involved in osteoblast differentiation and response to oxidative stress, all of which have been shown to be associated with the MM phenotype and clinical behavior. We functionally characterized MM-specific active distant enhancers controlling the expression of thioredoxin ( TXN ), a major regulator of cellular redox status and, in addition, identified PRDM5 as a novel essential gene for MM. Collectively, our data indicate that aberrant chromatin activation is a unifying feature underlying the malignant plasma cell phenotype., (© 2020 Ordoñez et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2020
- Full Text
- View/download PDF
38. CD34 + acute myeloid leukemia cells with low levels of reactive oxygen species show increased expression of stemness genes and can be targeted by the BCL2 inhibitor venetoclax.
- Author
-
Mattes K, Gerritsen M, Folkerts H, Geugien M, van den Heuvel FA, Svendsen AF, Yi G, Martens JHA, and Vellenga E
- Subjects
- Humans, Proto-Oncogene Proteins c-bcl-2 genetics, Reactive Oxygen Species, Sulfonamides, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Published
- 2020
- Full Text
- View/download PDF
39. CBFβ-SMMHC Affects Genome-wide Polycomb Repressive Complex 1 Activity in Acute Myeloid Leukemia.
- Author
-
Cordonnier G, Mandoli A, Cagnard N, Hypolite G, Lhermitte L, Verhoeyen E, Asnafi V, Dillon N, Macintyre E, Martens JHA, and Bond J
- Subjects
- Animals, Epigenesis, Genetic, Female, HeLa Cells, Heterografts, Humans, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred NOD, Oncogene Proteins, Fusion genetics, Polycomb Repressive Complex 1 genetics, Polycomb Repressive Complex 2 genetics, Polycomb Repressive Complex 2 metabolism, Transcriptional Activation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Oncogene Proteins, Fusion metabolism, Polycomb Repressive Complex 1 metabolism
- Abstract
Mutations and deletions of polycomb repressive complex (PRC) components are increasingly recognized to affect tumor biology in a range of cancers. However, little is known about how genetic alterations of PRC-interacting molecules such as the core binding factor (CBF) complex influence polycomb activity. We report that the acute myeloid leukemia (AML)-associated CBFβ-SMMHC fusion oncoprotein physically interacts with the PRC1 complex and that these factors co-localize across the AML genome in an apparently PRC2-independent manner. Depletion of CBFβ-SMMHC caused substantial increases in genome-wide PRC1 binding and marked changes in the association between PRC1 and the CBF DNA-binding subunit RUNX1. PRC1 was more likely to be associated with actively transcribed genes in CBFβ-SMMHC-expressing cells. CBFβ-SMMHC depletion had heterogeneous effects on gene expression, including significant reductions in transcription of ribosomal loci occupied by PRC1. Our results provide evidence that CBFβ-SMMHC markedly and diversely affects polycomb recruitment and transcriptional regulation across the AML genome., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
40. HIF1/2-exerted control over glycolytic gene expression is not functionally relevant for glycolysis in human leukemic stem/progenitor cells.
- Author
-
Wierenga ATJ, Cunningham A, Erdem A, Lopera NV, Brouwers-Vos AZ, Pruis M, Mulder AB, Günther UL, Martens JHA, Vellenga E, and Schuringa JJ
- Abstract
Background: Hypoxia-inducible factors (HIF)1 and 2 are transcription factors that regulate the homeostatic response to low oxygen conditions. Since data related to the importance of HIF1 and 2 in hematopoietic stem and progenitors is conflicting, we investigated the chromatin binding profiles of HIF1 and HIF2 and linked that to transcriptional networks and the cellular metabolic state., Methods: Genome-wide ChIPseq and ChIP-PCR experiments were performed to identify HIF1 and HIF2 binding sites in human acute myeloid leukemia (AML) cells and healthy CD34
+ hematopoietic stem/progenitor cells. Transcriptome studies were performed to identify gene expression changes induced by hypoxia or by overexpression of oxygen-insensitive HIF1 and HIF2 mutants. Metabolism studies were performed by 1D-NMR, and glucose consumption and lactate production levels were determined by spectrophotometric enzyme assays. CRISPR-CAS9-mediated HIF1, HIF2, and ARNT-/- lines were generated to study the functional consequences upon loss of HIF signaling, in vitro and in vivo upon transplantation of knockout lines in xenograft mice., Results: Genome-wide ChIP-seq and transcriptome studies revealed that overlapping HIF1- and HIF2-controlled loci were highly enriched for various processes including metabolism, particularly glucose metabolism, but also for chromatin organization, cellular response to stress and G protein-coupled receptor signaling. ChIP-qPCR validation studies confirmed that glycolysis-related genes but not genes related to the TCA cycle or glutaminolysis were controlled by both HIF1 and HIF2 in leukemic cell lines and primary AMLs, while in healthy human CD34+ cells these loci were predominantly controlled by HIF1 and not HIF2. However, and in contrast to our initial hypotheses, CRISPR/Cas9-mediated knockout of HIF signaling did not affect growth, internal metabolite concentrations, glucose consumption or lactate production under hypoxia, not even in vivo upon transplantation of knockout cells into xenograft mice., Conclusion: These data indicate that, while HIFs exert control over glycolysis but not OxPHOS gene expression in human leukemic cells, this is not critically important for their metabolic state. In contrast, inhibition of BCR-ABL did impact on glucose consumption and lactate production regardless of the presence of HIFs. These data indicate that oncogene-mediated control over glycolysis can occur independently of hypoxic signaling modules., Competing Interests: Competing interestsThe authors declare that they have no competing interests., (© The Author(s). 2019.)- Published
- 2019
- Full Text
- View/download PDF
41. The acute myeloid leukemia associated AML1-ETO fusion protein alters the transcriptome and cellular progression in a single-oncogene expressing in vitro induced pluripotent stem cell based granulocyte differentiation model.
- Author
-
Tijchon E, Yi G, Mandoli A, Smits JGA, Ferrari F, Heuts BMH, Wijnen F, Kim B, Janssen-Megens EM, Schuringa JJ, and Martens JHA
- Subjects
- Cell Proliferation genetics, Cells, Cultured, Core Binding Factor Alpha 2 Subunit genetics, Granulocytes metabolism, Humans, Leukemia, Myeloid, Acute genetics, Leukopoiesis genetics, Monocytes physiology, Myelopoiesis genetics, Oncogene Proteins, Fusion genetics, Oncogenes physiology, RUNX1 Translocation Partner 1 Protein genetics, Transfection, Cell Differentiation genetics, Cell Transformation, Neoplastic genetics, Core Binding Factor Alpha 2 Subunit physiology, Granulocytes physiology, Induced Pluripotent Stem Cells physiology, Oncogene Proteins, Fusion physiology, RUNX1 Translocation Partner 1 Protein physiology, Transcriptome genetics
- Abstract
Acute myeloid leukemia (AML) is characterized by recurrent mutations that affect normal hematopoiesis. The analysis of human AMLs has mostly been performed using end-point materials, such as cell lines and patient derived AMLs that also carry additional contributing mutations. The molecular effects of a single oncogenic hit, such as expression of the AML associated oncoprotein AML1-ETO on hematopoietic development and transformation into a (pre-) leukemic state still needs further investigation. Here we describe the development and characterization of an induced pluripotent stem cell (iPSC) system that allows in vitro differentiation towards different mature myeloid cell types such as monocytes and granulocytes. During in vitro differentiation we expressed the AML1-ETO fusion protein and examined the effects of the oncoprotein on differentiation and the underlying alterations in the gene program at 8 different time points. Our analysis revealed that AML1-ETO as a single oncogenic hit in a non-mutated background blocks granulocytic differentiation, deregulates the gene program via altering the acetylome of the differentiating granulocytic cells, and induces t(8;21) AML associated leukemic characteristics. Together, these results reveal that inducible oncogene expression during in vitro differentiation of iPS cells provides a valuable platform for analysis of aberrant regulation in disease., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
42. The AAA+ATPase RUVBL2 is essential for the oncogenic function of c-MYB in acute myeloid leukemia.
- Author
-
Armenteros-Monterroso E, Zhao L, Gasparoli L, Brooks T, Pearce K, Mansour MR, Martens JHA, de Boer J, and Williams O
- Subjects
- ATPases Associated with Diverse Cellular Activities genetics, Animals, Cell Line, Tumor, DNA Helicases genetics, Disease Models, Animal, Disease Progression, Gene Knockdown Techniques, Hematopoiesis genetics, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Mice, Protein Binding, Proto-Oncogene Proteins c-myb metabolism, Xenograft Model Antitumor Assays, ATPases Associated with Diverse Cellular Activities metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, DNA Helicases metabolism, Gene Expression Regulation, Leukemic, Leukemia, Myeloid, Acute genetics, Proto-Oncogene Proteins c-myb genetics
- Abstract
Subtype-specific leukemia oncogenes drive aberrant gene expression profiles that converge on common essential mediators to ensure leukemia self-renewal and inhibition of differentiation. The transcription factor c-MYB functions as one such mediator in a diverse range of leukemias. Here we show for the first time that transcriptional repression of myeloid differentiation associated c-MYB target genes in AML is enforced by the AAA+ ATPase RUVBL2. Silencing RUVBL2 expression resulted in increased binding of c-MYB to these loci and their transcriptional activation. RUVBL2 inhibition resulted in AML cell apoptosis and severely impaired disease progression of established AML in engrafted mice. In contrast, such inhibition had little impact on normal hematopoietic progenitor differentiation. These data demonstrate that RUVBL2 is essential for the oncogenic function of c-MYB in AML by governing inhibition of myeloid differentiation. They also indicate that targeting the control of c-MYB function by RUVBL2 is a promising approach to developing future anti-AML therapies.
- Published
- 2019
- Full Text
- View/download PDF
43. Large-scale in vitro production of red blood cells from human peripheral blood mononuclear cells.
- Author
-
Heshusius S, Heideveld E, Burger P, Thiel-Valkhof M, Sellink E, Varga E, Ovchynnikova E, Visser A, Martens JHA, von Lindern M, and van den Akker E
- Subjects
- Biomarkers, Bioreactors, Cell Proliferation, Erythroblasts metabolism, Erythrocytes metabolism, Gene Expression Profiling, Humans, Immunophenotyping, Leukocytes, Mononuclear metabolism, Primary Cell Culture, Reticulocytes metabolism, Transcriptome, Batch Cell Culture Techniques standards, Cell Differentiation genetics, Erythroblasts cytology, Erythrocytes cytology, Erythropoiesis genetics, Leukocytes, Mononuclear cytology
- Abstract
Transfusion of donor-derived red blood cells (RBC) is the most common form of cellular therapy. Donor availability and the potential risk of alloimmunization and other transfusion-related complications may, however, limit the availability of transfusion units, especially for chronically transfused patients. In vitro cultured, customizable RBC would negate these concerns and further increase precision medicine. Large-scale, cost-effective production depends on optimization of culture conditions. We developed a defined medium and adapted our protocols to good manufacturing practice (GMP) culture requirements, which reproducibly provided pure erythroid cultures from peripheral blood mononuclear cells without prior CD34+ isolation, and a 3 × 107-fold increase in erythroblasts in 25 days (or from 100 million peripheral blood mononuclear cells, 2 to 4 mL packed red cells can be produced). Expanded erythroblast cultures could be differentiated to CD71dimCD235a+CD44+CD117-DRAQ5- RBC in 12 days. More than 90% of the cells enucleated and expressed adult hemoglobin as well as the correct blood group antigens. Deformability and oxygen-binding capacity of cultured RBC was comparable to in vivo reticulocytes. Daily RNA sampling during differentiation followed by RNA-sequencing provided a high-resolution map/resource of changes occurring during terminal erythropoiesis. The culture process was compatible with upscaling using a G-Rex bioreactor with a capacity of 1 L per reactor, allowing transition toward clinical studies and small-scale applications., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
44. Ring sideroblasts in AML are associated with adverse risk characteristics and have a distinct gene expression pattern.
- Author
-
Berger G, Gerritsen M, Yi G, Koorenhof-Scheele TN, Kroeze LI, Stevens-Kroef M, Yoshida K, Shiraishi Y, van den Berg E, Schepers H, Huls G, Mulder AB, Ogawa S, Martens JHA, Jansen JH, and Vellenga E
- Subjects
- Abnormal Karyotype, Adult, Aged, Aged, 80 and over, Biomarkers, Tumor, Cell Differentiation genetics, Female, Gene Expression Profiling, Genetic Association Studies, Genetic Predisposition to Disease, High-Throughput Nucleotide Sequencing, Humans, Immunophenotyping, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Mutation, RNA Splicing, Tumor Suppressor Protein p53 genetics, Erythroblasts metabolism, Erythroblasts pathology, Gene Expression Regulation, Leukemic, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Transcriptome
- Abstract
Ring sideroblasts (RS) emerge as result of aberrant erythroid differentiation leading to excessive mitochondrial iron accumulation, a characteristic feature for myelodysplastic syndromes (MDS) with mutations in the spliceosome gene SF3B1. However, RS can also be observed in patients diagnosed with acute myeloid leukemia (AML). The objective of this study was to characterize RS in patients with AML. Clinically, RS-AML is enriched for ELN adverse risk (55%). In line with this finding, 35% of all cases had complex cytogenetic aberrancies, and TP53 was most recurrently mutated in this cohort (37%), followed by DNMT3A (26%), RUNX1 (25%), TET2 (20%), and ASXL1 (19%). In contrast to RS-MDS, the incidence of SF3B1 mutations was low (8%). Whole-exome sequencing and SNP array analysis on a subset of patients did not uncover a single genetic defect underlying the RS phenotype. Shared genetic defects between erythroblasts and total mononuclear cell fraction indicate common ancestry for the erythroid lineage and the myeloid blast cells in patients with RS-AML. RNA sequencing analysis on CD34+ AML cells revealed differential gene expression between RS-AML and non RS-AML cases, including genes involved in megakaryocyte and erythroid differentiation. Furthermore, several heme metabolism-related genes were found to be upregulated in RS- CD34+ AML cells, as was observed in SF3B1mut MDS. These results demonstrate that although the genetic background of RS-AML differs from that of RS-MDS, they have certain downstream effector pathways in common., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
45. The alarmin S100A9 hampers osteoclast differentiation from human circulating precursors by reducing the expression of RANK.
- Author
-
Di Ceglie I, Blom AB, Davar R, Logie C, Martens JHA, Habibi E, Böttcher LM, Roth J, Vogl T, Goodyear CS, van der Kraan PM, van Lent PL, and van den Bosch MH
- Subjects
- Bone Resorption, Calgranulin B pharmacology, Cell Differentiation drug effects, Gene Expression Regulation drug effects, Histone Code, Humans, Inflammation chemically induced, Inflammation genetics, Interleukin-1 antagonists & inhibitors, Lipopolysaccharide Receptors analysis, Macrophage Colony-Stimulating Factor pharmacology, Monocytes cytology, RANK Ligand pharmacology, Receptor Activator of Nuclear Factor-kappa B genetics, Recombinant Proteins pharmacology, Tumor Necrosis Factor-alpha antagonists & inhibitors, Calgranulin B physiology, Monocytes drug effects, Osteoclasts cytology, Receptor Activator of Nuclear Factor-kappa B biosynthesis
- Abstract
The alarmin S100A8/A9 is implicated in sterile inflammation-induced bone resorption and has been shown to increase the bone-resorptive capacity of mature osteoclasts. Here, we investigated the effects of S100A9 on osteoclast differentiation from human CD14
+ circulating precursors. Hereto, human CD14+ monocytes were isolated and differentiated toward osteoclasts with M-CSF and receptor activator of NF-κB (RANK) ligand (RANKL) in the presence or absence of S100A9. Tartrate-resistant acid phosphatase staining showed that exposure to S100A9 during monocyte-to-osteoclast differentiation strongly decreased the numbers of multinucleated osteoclasts. This was underlined by a decreased resorption of a hydroxyapatite-like coating. The thus differentiated cells showed a high mRNA and protein production of proinflammatory factors after 16 h of exposure. In contrast, at d 4, the cells showed a decreased production of the osteoclast-promoting protein TNF-α. Interestingly, S100A9 exposure during the first 16 h of culture only was sufficient to reduce osteoclastogenesis. Using fluorescently labeled RANKL, we showed that, within this time frame, S100A9 inhibited the M-CSF-mediated induction of RANK. Chromatin immunoprecipitation showed that this was associated with changes in various histone marks at the epigenetic level. This S100A9-induced reduction in RANK was in part recovered by blocking TNF-α but not IL-1. Together, our data show that S100A9 impedes monocyte-to-osteoclast differentiation, probably via a reduction in RANK expression.-Di Ceglie, I., Blom, A. B., Davar, R., Logie, C., Martens, J. H. A., Habibi, E., Böttcher, L.-M., Roth, J., Vogl, T., Goodyear, C. S., van der Kraan, P. M., van Lent, P. L., van den Bosch, M. H. The alarmin S100A9 hampers osteoclast differentiation from human circulating precursors by reducing the expression of RANK.- Published
- 2019
- Full Text
- View/download PDF
46. Molecular mechanisms of bleeding disorderassociated GFI1B Q287* mutation and its affected pathways in megakaryocytes and platelets.
- Author
-
van Oorschot R, Hansen M, Koornneef JM, Marneth AE, Bergevoet SM, van Bergen MGJM, van Alphen FPJ, van der Zwaan C, Martens JHA, Vermeulen M, Jansen PWTC, Baltissen MPA, Gorkom BAPL, Janssen H, Jansen JH, von Lindern M, Meijer AB, van den Akker E, and van der Reijden BA
- Subjects
- Blood Coagulation Disorders genetics, Blood Coagulation Disorders metabolism, Blood Platelets metabolism, Cell Differentiation, Co-Repressor Proteins genetics, Co-Repressor Proteins metabolism, Histone Deacetylase 1 genetics, Histone Deacetylase 1 metabolism, Histone Deacetylase 2 genetics, Histone Deacetylase 2 metabolism, Histone Demethylases genetics, Histone Demethylases metabolism, Humans, Induced Pluripotent Stem Cells metabolism, Megakaryocytes metabolism, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Phenotype, Protein Interaction Maps, Proteome analysis, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism, Blood Coagulation Disorders pathology, Blood Platelets pathology, Gene Expression Regulation, Induced Pluripotent Stem Cells pathology, Megakaryocytes pathology, Mutation, Proto-Oncogene Proteins genetics, Repressor Proteins genetics
- Abstract
Dominant-negative mutations in the transcription factor Growth Factor Independence-1B (GFI1B), such as GFI1B
Q287* , cause a bleeding disorder characterized by a plethora of megakaryocyte and platelet abnormalities. The deregulated molecular mechanisms and pathways are unknown. Here we show that both normal and Q287* mutant GFI1B interacted most strongly with the lysine specific demethylase-1 - REST corepressor - histone deacetylase (LSD1-RCOR-HDAC) complex in megakaryoblasts. Sequestration of this complex by GFI1BQ287* and chemical separation of GFI1B from LSD1 induced abnormalities in normal megakaryocytes comparable to those seen in patients. Megakaryocytes derived from GFI1BQ287* -induced pluripotent stem cells also phenocopied abnormalities seen in patients. Proteome studies on normal and mutant-induced pluripotent stem cell-derived megakaryocytes identified a multitude of deregulated pathways downstream of GFI1BQ287* including cell division and interferon signaling. Proteome studies on platelets from GFI1BQ287* patients showed reduced expression of proteins implicated in platelet function, and elevated expression of proteins normally downregulated during megakaryocyte differentiation. Thus, GFI1B and LSD1 regulate a broad developmental program during megakaryopoiesis, and GFI1BQ287* deregulates this program through LSD1-RCOR-HDAC sequestering., (Copyright© 2019 Ferrata Storti Foundation.)- Published
- 2019
- Full Text
- View/download PDF
47. BRD9 binds cell type-specific chromatin regions regulating leukemic cell survival via STAT5 inhibition.
- Author
-
Del Gaudio N, Di Costanzo A, Liu NQ, Conte L, Migliaccio A, Vermeulen M, Martens JHA, Stunnenberg HG, Nebbioso A, and Altucci L
- Subjects
- Antigens, CD34 metabolism, Cell Proliferation, Cell Survival, HeLa Cells, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, RNA Interference, RNA, Small Interfering metabolism, STAT5 Transcription Factor antagonists & inhibitors, Suppressor of Cytokine Signaling 3 Protein metabolism, Transcription Factors antagonists & inhibitors, Transcription Factors genetics, Transcriptome, Tumor Cells, Cultured, Tumor Suppressor Proteins antagonists & inhibitors, Chromatin metabolism, Leukemia, Myeloid, Acute pathology, STAT5 Transcription Factor metabolism, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Leukemia is characterized by genetic and epigenetic mutations resulting in selection of cancer cells, which are unable to differentiate. Although genetic alterations are difficult to target, the epigenome is intrinsically dynamic and readily offers new therapeutic strategies. Thus, identifying cancer-specific context-dependent targets and unraveling their biological function may open up new therapeutic perspectives. Here we identify bromodomain-containing protein 9 (BRD9) as a critical target required in acute myeloid leukemia (AML). We show that BRD9 is overexpressed in AML cells including ex vivo primary blasts compared with CD34
+ cells. By targeting BRD9 expression in AML, we observed an alteration in proliferation and survival, ultimately resulting in the induction of apoptosis. Intriguingly, genome-wide profiling revealed that BRD9 binds enhancer regions in a cell type-specific manner, regulating cell type-related processes. We unveil a novel BRD9-sustained STAT5 pathway activation via regulation of SOCS3 expression levels. Our findings identify a previously undescribed BRD9-STAT5 axis as critical for leukemia maintenance, suggesting BRD9 as a potential therapeutic target.- Published
- 2019
- Full Text
- View/download PDF
48. CBFβ-MYH11 interferes with megakaryocyte differentiation via modulating a gene program that includes GATA2 and KLF1.
- Author
-
Yi G, Mandoli A, Jussen L, Tijchon E, van Bergen MGJM, Cordonnier G, Hansen M, Kim B, Nguyen LN, Jansen PWTC, Vermeulen M, van der Reijden B, van den Akker E, Bond J, and Martens JHA
- Subjects
- Binding Sites, Cell Differentiation, Cell Line, Tumor, Cell Proliferation, Epigenesis, Genetic, Erythroid Cells cytology, Erythroid Cells metabolism, Erythropoiesis genetics, Gene Expression Profiling, Gene Knockdown Techniques, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Megakaryocytes cytology, Oncogene Proteins, Fusion metabolism, Protein Binding, Thrombopoiesis, Transcription, Genetic, GATA2 Transcription Factor metabolism, Gene Expression Regulation, Leukemic, Kruppel-Like Transcription Factors metabolism, Megakaryocytes metabolism, Oncogene Proteins, Fusion genetics
- Abstract
The inv(16) acute myeloid leukemia-associated CBFβ-MYH11 fusion is proposed to block normal myeloid differentiation, but whether this subtype of leukemia cells is poised for a unique cell lineage remains unclear. Here, we surveyed the functional consequences of CBFβ-MYH11 in primary inv(16) patient blasts, upon expression during hematopoietic differentiation in vitro and upon knockdown in cell lines by multi-omics profiling. Our results reveal that primary inv(16) AML cells share common transcriptomic signatures and epigenetic determiners with megakaryocytes and erythrocytes. Using in vitro differentiation systems, we reveal that CBFβ-MYH11 knockdown interferes with normal megakaryocyte maturation. Two pivotal regulators, GATA2 and KLF1, are identified to complementally occupy RUNX1-binding sites upon fusion protein knockdown, and overexpression of GATA2 partly induces a gene program involved in megakaryocyte-directed differentiation. Together, our findings suggest that in inv(16) leukemia, the CBFβ-MYH11 fusion inhibits primed megakaryopoiesis by attenuating expression of GATA2/KLF1 and interfering with a balanced transcriptional program involving these two factors.
- Published
- 2019
- Full Text
- View/download PDF
49. RUNX1 mutations enhance self-renewal and block granulocytic differentiation in human in vitro models and primary AMLs.
- Author
-
Gerritsen M, Yi G, Tijchon E, Kuster J, Schuringa JJ, Martens JHA, and Vellenga E
- Subjects
- CCAAT-Enhancer-Binding Proteins biosynthesis, CCAAT-Enhancer-Binding Proteins genetics, Cell Differentiation genetics, Cell Transformation, Neoplastic genetics, Chromatin Immunoprecipitation, Core Binding Factor Alpha 2 Subunit biosynthesis, Core Binding Factor Alpha 2 Subunit blood, Core Binding Factor Alpha 2 Subunit metabolism, Fetal Blood metabolism, Granulocytes metabolism, Humans, Leukemia, Myeloid, Acute metabolism, Protein Binding, Core Binding Factor Alpha 2 Subunit genetics, Granulocytes pathology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mutation
- Abstract
To unravel molecular mechanisms by which Runt-related transcription factor 1 (RUNX1) mutations contribute to leukemic transformation, we introduced the RUNX1-S291fs300X mutation in human CD34
+ stem/progenitor cells and in human induced pluripotent stem cells (iPSCs). In both models, RUNX1mut overexpression strongly impaired myeloid commitment. Instead, self-renewal was enhanced, as shown, by increased long-term culture-initiating cell frequencies and enhanced colony-forming cell replating capacity. Long-term suspension cultures with RUNX1mut-transduced cord blood (CB) CD34+ cells continued for more than 100 days, during which the cells displayed an immature granulocyte-macrophage progenitor-like CD34+ /CD123+ /CD45RA+ phenotype. The CD34+ /CD38- hematopoietic stem cell (HSC) population most likely acted as cell of origin, as HSCs provided the best long-term proliferative potential on overexpression of RUNX1mut. CEBPA expression was reduced in RUNX1mut cells, and reexpression of CEBPA partly restored differentiation. RNA-seq analysis on CB/iPSC systems and on primary patient samples confirmed that RUNX1 mutations induce a myeloid differentiation block, and that a common set of RUNX1mut-upregulated target genes was strongly enriched for gene ontology terms associated with nucleosome assembly and chromatin structure. Interestingly, in comparison with AML1-ETO binding in acute myeloid leukemias (AMLs), we found significantly distinct genomic distribution and differential expression for RUNX1mut of genes such as TCF4 , MEIS1 , and HMGA2 that may potentially contribute to the underlying difference in clinical outcomes between RUNX1mut and AML1-ETO patients. In conclusion, RUNX1mut appears to induce a specific transcriptional program that contributes to leukemic transformation., (© 2019 by The American Society of Hematology.)- Published
- 2019
- Full Text
- View/download PDF
50. Chromatin-Based Classification of Genetically Heterogeneous AMLs into Two Distinct Subtypes with Diverse Stemness Phenotypes.
- Author
-
Yi G, Wierenga ATJ, Petraglia F, Narang P, Janssen-Megens EM, Mandoli A, Merkel A, Berentsen K, Kim B, Matarese F, Singh AA, Habibi E, Prange KHM, Mulder AB, Jansen JH, Clarke L, Heath S, van der Reijden BA, Flicek P, Yaspo ML, Gut I, Bock C, Schuringa JJ, Altucci L, Vellenga E, Stunnenberg HG, and Martens JHA
- Subjects
- Humans, Nucleophosmin, Chromatin genetics, Chromatin metabolism, Chromatin pathology, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit metabolism, Leukemia, Myeloid, Acute classification, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Mutation, Nuclear Proteins genetics, Nuclear Proteins metabolism, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism
- Abstract
Global investigation of histone marks in acute myeloid leukemia (AML) remains limited. Analyses of 38 AML samples through integrated transcriptional and chromatin mark analysis exposes 2 major subtypes. One subtype is dominated by patients with NPM1 mutations or MLL-fusion genes, shows activation of the regulatory pathways involving HOX-family genes as targets, and displays high self-renewal capacity and stemness. The second subtype is enriched for RUNX1 or spliceosome mutations, suggesting potential interplay between the 2 aberrations, and mainly depends on IRF family regulators. Cellular consequences in prognosis predict a relatively worse outcome for the first subtype. Our integrated profiling establishes a rich resource to probe AML subtypes on the basis of expression and chromatin data., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.