43 results on '"Chagraoui J"'
Search Results
2. Immunotherapeutic targeting of surfaceome heterogeneity in AML.
- Author
-
Bordeleau ME, Audemard É, Métois A, Theret L, Lisi V, Farah A, Spinella JF, Chagraoui J, Moujaber O, Aubert L, Khakipoor B, Mallinger L, Boivin I, Mayotte N, Hajmirza A, Bonneil É, Béliveau F, Pfammatter S, Feghaly A, Boucher G, Gendron P, Thibault P, Barabé F, Lemieux S, Richard-Carpentier G, Hébert J, Lavallée VP, Roux PP, and Sauvageau G
- Subjects
- Humans, Membrane Proteins metabolism, Leukemia, Myeloid, Acute immunology, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Acute pathology, Immunotherapy methods
- Abstract
Immunotherapy remains underexploited in acute myeloid leukemia (AML) compared to other hematological malignancies. Currently, gemtuzumab ozogamicin is the only therapeutic antibody approved for this disease. Here, to identify potential targets for immunotherapeutic intervention, we analyze the surface proteome of 100 genetically diverse primary human AML specimens for the identification of cell surface proteins and conduct single-cell transcriptome analyses on a subset of these specimens to assess antigen expression at the sub-population level. Through this comprehensive effort, we successfully identify numerous antigens and markers preferentially expressed by primitive AML cells. Many identified antigens are targeted by therapeutic antibodies currently under clinical evaluation for various cancer types, highlighting the potential therapeutic value of the approach. Importantly, this initiative uncovers AML heterogeneity at the surfaceome level, identifies several antigens and potential primitive cell markers characterizing AML subgroups, and positions immunotherapy as a promising approach to target AML subgroup specificities., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. KBTBD4-mediated reduction of MYC is critical for hematopoietic stem cell expansion upon UM171 treatment.
- Author
-
Chagraoui J, Girard S, Mallinger L, Mayotte N, Tellechea MF, and Sauvageau G
- Subjects
- Hematopoiesis, Cell Cycle, Cell Differentiation, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cell Transplantation methods
- Abstract
Abstract: Ex vivo expansion of hematopoietic stem cells (HSCs) is gaining importance for cell and gene therapy, and requires a shift from dormancy state to activation and cycling. However, abnormal or excessive HSC activation results in reduced self-renewal ability and increased propensity for myeloid-biased differentiation. We now report that activation of the E3 ligase complex CRL3KBTBD4 by UM171 not only induces epigenetic changes through CoREST1 degradation but also controls chromatin-bound master regulator of cell cycle entry and proliferative metabolism (MYC) levels to prevent excessive activation and maintain lympho-myeloid potential of expanded populations. Furthermore, reconstitution activity and multipotency of UM171-treated HSCs are specifically compromised when MYC levels are experimentally increased despite degradation of CoREST1., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
4. DELE1 haploinsufficiency causes resistance to mitochondrial stress-induced apoptosis in monosomy 5/del(5q) AML.
- Author
-
Spinella JF, Chagraoui J, Moison C, Lavallée VP, Boivin I, Gracias D, Lavallée S, Carpentier GR, Beliveau F, Hébert J, and Sauvageau G
- Subjects
- Adult, Humans, Apoptosis genetics, Chromosome Deletion, Chromosomes, Human, Pair 5 genetics, Haploinsufficiency, Leukemia, Myeloid, Acute genetics, Monosomy, Mitochondrial Proteins genetics
- Abstract
Monosomy 5 and deletions of the chromosome 5q (-5/del(5q)) are recurrent events in de novo adult acute myeloid leukemia (AML), reaching up to 40% of cases in secondary AML. These chromosome anomalies are associated with TP53 mutations and with very poor prognosis. Using the large Leucegene genomic and transcriptomic dataset composed of 48 -5/del(5q) patient specimens and 367 control AML, we identified DELE1 - located in the common deleted region - as the most consistently downregulated gene in these leukemias. DELE1 encodes a mitochondrial protein recently characterized as the relay of mitochondrial stress to the cytosol through a newly defined OMA1-DELE1-HRI pathway which ultimately leads to the activation of ATF4, the master transcription factor of the integrated stress response. Here, we showed that the partial loss of DELE1 expression observed in -5/del(5q) patients was sufficient to significantly reduce the sensitivity to mitochondrial stress in AML cells. Overall, our results suggest that DELE1 haploinsufficiency could represent a new driver mechanism in -5/del(5q) AML., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. CBFA2T3::GLIS2 pediatric acute megakaryoblastic leukemia is sensitive to BCL-XL inhibition by navitoclax and DT2216.
- Author
-
Gress V, Roussy M, Boulianne L, Bilodeau M, Cardin S, El-Hachem N, Lisi V, Khakipoor B, Rouette A, Farah A, Théret L, Aubert L, Fatima F, Audemard É, Thibault P, Bonneil É, Chagraoui J, Laramée L, Gendron P, Jouan L, Jammali S, Paré B, Simpson SM, Tran TH, Duval M, Teira P, Bittencourt H, Santiago R, Barabé F, Sauvageau G, Smith MA, Hébert J, Roux PP, Gruber TA, Lavallée VP, Wilhelm BT, and Cellot S
- Subjects
- Humans, Child, Child, Preschool, Animals, Mice, Proteomics, Transcription Factors, Proto-Oncogene Proteins c-bcl-2, Repressor Proteins, Leukemia, Megakaryoblastic, Acute drug therapy, Leukemia, Megakaryoblastic, Acute genetics, Leukemia, Megakaryoblastic, Acute pathology, Antineoplastic Agents
- Abstract
Abstract: Acute megakaryoblastic leukemia (AMKL) is a rare, developmentally restricted, and highly lethal cancer of early childhood. The paucity and hypocellularity (due to myelofibrosis) of primary patient samples hamper the discovery of cell- and genotype-specific treatments. AMKL is driven by mutually exclusive chimeric fusion oncogenes in two-thirds of the cases, with CBFA2T3::GLIS2 (CG2) and NUP98 fusions (NUP98r) representing the highest-fatality subgroups. We established CD34+ cord blood-derived CG2 models (n = 6) that sustain serial transplantation and recapitulate human leukemia regarding immunophenotype, leukemia-initiating cell frequencies, comutational landscape, and gene expression signature, with distinct upregulation of the prosurvival factor B-cell lymphoma 2 (BCL2). Cell membrane proteomic analyses highlighted CG2 surface markers preferentially expressed on leukemic cells compared with CD34+ cells (eg, NCAM1 and CD151). AMKL differentiation block in the mega-erythroid progenitor space was confirmed by single-cell profiling. Although CG2 cells were rather resistant to BCL2 genetic knockdown or selective pharmacological inhibition with venetoclax, they were vulnerable to strategies that target the megakaryocytic prosurvival factor BCL-XL (BCL2L1), including in vitro and in vivo treatment with BCL2/BCL-XL/BCL-W inhibitor navitoclax and DT2216, a selective BCL-XL proteolysis-targeting chimera degrader developed to limit thrombocytopenia in patients. NUP98r AMKL were also sensitive to BCL-XL inhibition but not the NUP98r monocytic leukemia, pointing to a lineage-specific dependency. Navitoclax or DT2216 treatment in combination with low-dose cytarabine further reduced leukemic burden in mice. This work extends the cellular and molecular diversity set of human AMKL models and uncovers BCL-XL as a therapeutic vulnerability in CG2 and NUP98r AMKL., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
6. Towards clinically meaningful expansion of human HSCs.
- Author
-
Tellechea MF, Chagraoui J, Cohen S, and Sauvageau G
- Published
- 2023
- Full Text
- View/download PDF
7. HMGA2 expression defines a subset of human AML with immature transcriptional signature and vulnerability to G2/M inhibition.
- Author
-
Moison C, Spinella JF, Chagraoui J, Lavallée VP, Lehnertz B, Thiollier C, Boivin I, Mayotte N, MacRae T, Marinier A, Hébert J, and Sauvageau G
- Subjects
- Antigens, CD34, Cell Cycle Checkpoints, Humans, Up-Regulation, Leukemia, Myeloid, Acute pathology
- Abstract
High-mobility group AT-hook 2 (HMGA2) is a nonhistone chromatin-binding protein that is normally expressed in stem cells of various tissues and aberrantly detected in several tumor types. We recently observed that one-fourth of human acute myeloid leukemia (AML) specimens express HMGA2, which associates with a very poor prognosis. We present results indicating that HMGA2+ AMLs share a distinct transcriptional signature representing an immature phenotype. Using single-cell analyses, we showed that HMGA2 is expressed in CD34+ subsets of stem cells and early progenitors, whether normal or derived from AML specimens. Of interest, we found that one of the strongest gene expression signatures associated with HMGA2 in AML is the upregulation of G2/M checkpoint genes. Whole-genome CRISPR/Cas9 screening in HMGA2 overexpressing cells further revealed a synthetic lethal interaction with several G2/M checkpoint genes. Accordingly, small molecules that target G2/M proteins were preferentially active in vitro and in vivo on HMGA2+ AML specimens. Together, our findings suggest that HMGA2 is a key functional determinant in AML and is associated with stem cell features, G2/M status, and related drug sensitivity., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. CEACAM1 is a novel culture-compatible surface marker of expanded long-term reconstituting hematopoietic stem cells.
- Author
-
Ansari U, Tomellini E, Chagraoui J, Lehnertz B, Mayotte N, Bordeleau ME, Roux PP, and Sauvageau G
- Subjects
- Biomarkers, Cell Adhesion Molecules, Antigens, CD, Hematopoietic Stem Cells
- Published
- 2022
- Full Text
- View/download PDF
9. HLF expression defines the human hematopoietic stem cell state.
- Author
-
Lehnertz B, Chagraoui J, MacRae T, Tomellini E, Corneau S, Mayotte N, Boivin I, Durand A, Gracias D, and Sauvageau G
- Subjects
- Hematopoiesis, Hematopoietic Stem Cells cytology, Humans, Single-Cell Analysis, Basic-Leucine Zipper Transcription Factors genetics, Gene Expression, Hematopoietic Stem Cells metabolism, Transcriptome
- Abstract
Hematopoietic stem cells (HSCs) sustain blood cell homeostasis throughout life and can regenerate all blood lineages after transplantation. Despite this clear functional definition, highly enriched isolation of human HSCs can currently only be achieved through combinatorial assessment of multiple surface antigens. Although several transgenic HSC reporter mouse strains have been described, no analogous approach to prospectively isolate human HSCs has been reported. To identify genes with the most selective expression in human HSCs, we profiled population and single-cell transcriptomes of unexpanded and ex vivo cultured cord blood-derived hematopoietic stem and progenitor cells as well as peripheral blood, adult bone marrow, and fetal liver. On the basis of these analyses, we propose the master transcription factor HLF (hepatic leukemia factor) as one of the most specific HSC marker genes. To directly track its expression in human hematopoietic cells, we developed a genomic HLF reporter strategy, capable of selectively labeling the most immature blood cells on the basis of a single engineered parameter. Most importantly, HLF-expressing cells comprise all stem cell activity in culture and in vivo during serial transplantation. Taken together, these results experimentally establish HLF as a defining gene of the human HSC state and outline a new approach to continuously mark these cells with high fidelity., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
10. Zinc finger protein E4F1 cooperates with PARP-1 and BRG1 to promote DNA double-strand break repair.
- Author
-
Moison C, Chagraoui J, Caron MC, Gagné JP, Coulombe Y, Poirier GG, Masson JY, and Sauvageau G
- Subjects
- Breast Neoplasms genetics, Cell Proliferation, Cell Survival, Chromatin Assembly and Disassembly, DNA Damage, Gene Expression Regulation, Neoplastic, Gene Silencing, Homologous Recombination, Humans, Protein Binding, Repressor Proteins deficiency, Signal Transduction, Ubiquitin-Protein Ligases deficiency, DNA Breaks, Double-Stranded, DNA Helicases metabolism, DNA Repair, Nuclear Proteins metabolism, Poly (ADP-Ribose) Polymerase-1 metabolism, Repressor Proteins metabolism, Transcription Factors metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Zinc finger (ZnF) proteins represent one of the largest families of human proteins, although most remain uncharacterized. Given that numerous ZnF proteins are able to interact with DNA and poly(ADP ribose), there is growing interest in understanding their mechanism of action in the maintenance of genome integrity. We now report that the ZnF protein E4F transcription factor 1 (E4F1) is an actor in DNA repair. Indeed, E4F1 is rapidly recruited, in a poly(ADP ribose) polymerase (PARP)-dependent manner, to DNA breaks and promotes ATR/CHK1 signaling, DNA-end resection, and subsequent homologous recombination. Moreover, we identify E4F1 as a regulator of the ATP-dependent chromatin remodeling SWI/SNF complex in DNA repair. E4F1 binds to the catalytic subunit BRG1/SMARCA4 and together with PARP-1 mediates its recruitment to DNA lesions. We also report that a proportion of human breast cancers show amplification and overexpression of E4F1 or BRG1 that are mutually exclusive with BRCA1 / 2 alterations. Together, these results reveal a function of E4F1 in the DNA damage response that orchestrates proper signaling and repair of double-strand breaks and document a molecular mechanism for its essential role in maintaining genome integrity and cell survival., Competing Interests: The authors declare no competing interest.
- Published
- 2021
- Full Text
- View/download PDF
11. UM171 Preserves Epigenetic Marks that Are Reduced in Ex Vivo Culture of Human HSCs via Potentiation of the CLR3-KBTBD4 Complex.
- Author
-
Chagraoui J, Girard S, Spinella JF, Simon L, Bonneil E, Mayotte N, MacRae T, Coulombe-Huntington J, Bertomeu T, Moison C, Tomellini E, Thibault P, Tyers M, Marinier A, and Sauvageau G
- Subjects
- Histone Deacetylases metabolism, Humans, Co-Repressor Proteins genetics, Co-Repressor Proteins metabolism, Epigenesis, Genetic, Hematopoietic Stem Cells metabolism
- Abstract
Human hematopoietic stem cells (HSCs) exhibit attrition of their self-renewal capacity when cultured ex vivo, a process that is partially reversed upon treatment with epigenetic modifiers, most notably inhibitors of histone deacetylases (HDACs) or lysine-specific demethylase LSD1. A recent study showed that the human HSC self-renewal agonist UM171 modulates the CoREST complex, leading to LSD1 degradation, whose inhibition mimics the activity of UM171. The mechanism underlying the UM171-mediated loss of CoREST function remains undetermined. We now report that UM171 potentiates the activity of a CULLIN3-E3 ubiquitin ligase (CRL3) complex whose target specificity is dictated by the poorly characterized Kelch/BTB domain protein KBTBD4. CRL3
KBTBD4 targets components of the LSD1/RCOR1 corepressor complex for proteasomal degradation, hence re-establishing H3K4me2 and H3K27ac epigenetic marks, which are rapidly decreased upon ex vivo culture of human HSCs., Competing Interests: Declaration of interests Except for G.S., the authors declare no competing interests. G.S. is founder and CEO of ExCellThera, a small biotech that owns an exclusive license to UM171., (Copyright © 2020 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
12. UM171-Expanded Cord Blood Transplants Support Robust T Cell Reconstitution with Low Rates of Severe Infections.
- Author
-
Dumont-Lagacé M, Li Q, Tanguay M, Chagraoui J, Kientega T, Cardin GB, Brasey A, Trofimov A, Carli C, Ahmad I, Bambace NM, Bernard L, Kiss TL, Roy J, Roy DC, Lemieux S, Perreault C, Rodier F, Dufresne SF, Busque L, Lachance S, Sauvageau G, Cohen S, and Delisle JS
- Subjects
- Fetal Blood, Humans, Retrospective Studies, T-Lymphocytes, Cord Blood Stem Cell Transplantation adverse effects, Graft vs Host Disease
- Abstract
Rapid T cell reconstitution following hematopoietic stem cell transplantation (HSCT) is essential for protection against infections and has been associated with lower incidence of chronic graft-versus-host disease (cGVHD), relapse, and transplant-related mortality (TRM). While cord blood (CB) transplants are associated with lower rates of cGVHD and relapse, their low stem cell content results in slower immune reconstitution and higher risk of graft failure, severe infections, and TRM. Recently, results of a phase I/II trial revealed that single UM171-expanded CB transplant allowed the use of smaller CB units without compromising engraftment (www.clinicaltrials.gov, NCT02668315). We assessed T cell reconstitution in patients who underwent transplantation with UM171-expanded CB grafts and retrospectively compared it to that of patients receiving unmanipulated CB transplants. While median T cell dose infused was at least 2 to 3 times lower than that of unmanipulated CB, numbers and phenotype of T cells at 3, 6, and 12 months post-transplant were similar between the 2 cohorts. T cell receptor sequencing analyses revealed that UM171 patients had greater T cell diversity and higher numbers of clonotypes at 12 months post-transplant. This was associated with higher counts of naive T cells and recent thymic emigrants, suggesting active thymopoiesis and correlating with the demonstration that UM171 expands common lymphoid progenitors in vitro. UM171 patients also showed rapid virus-specific T cell reactivity and significantly reduced incidence of severe infections. These results suggest that UM171 patients benefit from rapid T cell reconstitution, which likely contributes to the absence of moderate/severe cGVHD, infection-related mortality, and late TRM observed in this cohort., (Copyright © 2020 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Hematopoietic stem cell transplantation using single UM171-expanded cord blood: a single-arm, phase 1-2 safety and feasibility study.
- Author
-
Cohen S, Roy J, Lachance S, Delisle JS, Marinier A, Busque L, Roy DC, Barabé F, Ahmad I, Bambace N, Bernard L, Kiss T, Bouchard P, Caudrelier P, Landais S, Larochelle F, Chagraoui J, Lehnertz B, Corneau S, Tomellini E, van Kampen JJA, Cornelissen JJ, Dumont-Lagacé M, Tanguay M, Li Q, Lemieux S, Zandstra PW, and Sauvageau G
- Subjects
- Adolescent, Adult, Cell Self Renewal drug effects, Cells, Cultured drug effects, Cells, Cultured transplantation, Cord Blood Stem Cell Transplantation adverse effects, Disease-Free Survival, Feasibility Studies, Febrile Neutropenia etiology, Female, Graft Survival, Graft vs Host Disease etiology, Hematologic Neoplasms therapy, Hematopoietic Stem Cell Transplantation adverse effects, Hematopoietic Stem Cells cytology, Humans, Infant, Newborn, Male, Middle Aged, Proportional Hazards Models, Treatment Outcome, Young Adult, Cord Blood Stem Cell Transplantation methods, Hematopoietic Stem Cell Transplantation methods, Hematopoietic Stem Cells drug effects, Indoles pharmacology, Pyrimidines pharmacology
- Abstract
Background: Benefits of cord blood transplantation include low rates of relapse and chronic graft-versus-host disease (GVHD). However, the use of cord blood is rapidly declining because of the high incidence of infections, severe acute GVHD, and transplant-related mortality. UM171, a haematopoietic stem cell self-renewal agonist, has been shown to expand cord blood stem cells and enhance multilineage blood cell reconstitution in mice. We aimed to investigate the safety and feasibility of single UM171-expanded cord blood transplantation in patients with haematological malignancies who do not have a suitable HLA-matched donor., Methods: This single-arm, open-label, phase 1-2 safety and feasibility study was done at two hospitals in Canada. The study had two parts. In part 1, patients received two cord blood units (one expanded with UM171 and one unmanipulated cord blood) until UM171-expanded cord blood demonstrated engraftment. Once engraftment was documented we initiated part 2, reported here, in which patients received a single UM171-expanded cord blood unit with a dose de-escalation design to determine the minimal cord blood unit cell dose that achieved prompt engraftment. Eligible patients were aged 3-64 years, weighed 12 kg or more, had a haematological malignancy with an indication for allogeneic hematopoietic stem cell transplant and did not have a suitable HLA-matched donor, and a had a Karnofsky performance status score of 70% or more. Five clinical sites were planned to participate in the study; however, only two study sites opened, both of which only treated adult patients, thus no paediatric patients (aged <18 years) were recruited. Patients aged younger than 50 years without comorbidities received a myeloablative conditioning regimen (cyclophosphamide 120 mg/kg, fludarabine 75 mg/m
2 , and 12 Gy total body irradiation) and patients aged older than 50 years and those with comorbidities received a less myeloablative conditioning regimen (cyclophosphamide 50 mg/kg, thiotepa 10 mg/kg, fludarabine 150 mg/m2 , and 4 Gy total body irradiation). Patients were infused with the 7-day UM171-expanded CD34-positive cells and the lymphocyte-containing CD34-negative fraction. The primary endpoints were feasibility of UM171 expansion, safety of the transplant, kinetics of hematopoietic reconstitution (time to neutrophil and platelet engraftment) of UM171-expanded cord blood, and minimal pre-expansion cord blood unit cell dose that achieved prompt engraftment. We analysed feasibility in all enrolled patients and all other primary outcomes were analysed per protocol, in all patients who received single UM171-expanded cord blood transplantation. This trial has been completed and was registered with ClinicalTrials.gov, NCT02668315., Findings: Between Feb 17, 2016, and Nov 11, 2018, we enrolled 27 patients, four of whom received two cord blood units for safety purposes in part 1 of the study. 23 patients were subsequently enrolled in part 2 to receive a single UM171-expanded cord blood transplant and 22 patients received a single UM171-expanded cord blood transplantation. At data cutoff (Dec 31, 2018), median follow-up was 18 months (IQR 12-22). The minimal cord blood unit cell dose at thaw that achieved prompt engraftment as a single cord transplant after UM171 expansion was 0·52 × 105 CD34-positive cells. We successfully expanded 26 (96%) of 27 cord blood units with UM171. Among the 22 patients who received single UM171-expanded cord blood transplantation, median time to engraftment of 100 neutrophils per μL was 9·5 days (IQR 8-12), median time to engraftment of 500 neutrophils per μL was 18 days (12·5-20·0), and no graft failure occurred. Median time to platelet recovery was 42 days (IQR 35-47). The most common non-haematological adverse events were grade 3 febrile neutropenia (16 [73%] of 22 patients) and bacteraemia (nine [41%]). No unexpected adverse events were observed. One (5%) of 22 patients died due to treatment-related diffuse alveolar haemorrhage., Interpretation: Our preliminary findings suggest that UM171 cord blood stem cell expansion is feasible, safe, and allows for the use of small single cords without compromising engraftment. UM171-expanded cord blood might have the potential to overcome the disadvantages of other cord blood transplants while maintaining the benefits of low risk of chronic GVHD and relapse, and warrants further investigation in randomised trials., Funding: Canadian Institutes of Health Research, Canadian Cancer Society and Stem Cell Network., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
14. UM171 induces a homeostatic inflammatory-detoxification response supporting human HSC self-renewal.
- Author
-
Chagraoui J, Lehnertz B, Girard S, Spinella JF, Fares I, Tomellini E, Mayotte N, Corneau S, MacRae T, Simon L, and Sauvageau G
- Subjects
- Biomarkers, Cell Differentiation, Cell Proliferation, Gene Expression Profiling, Humans, Metabolic Detoxication, Phase I, Reactive Oxygen Species, Signal Transduction drug effects, Transcriptome, Cell Self Renewal drug effects, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells drug effects, Hematopoietic Stem Cells metabolism, Homeostasis drug effects, Indoles pharmacology, Pyrimidines pharmacology
- Abstract
Elucidation of the molecular cues required to balance adult stem cell self-renewal and differentiation is critical for advancing cellular therapies. Herein, we report that the hematopoietic stem cell (HSC) self-renewal agonist UM171 triggers a balanced pro- and anti-inflammatory/detoxification network that relies on NFKB activation and protein C receptor-dependent ROS detoxification, respectively. We demonstrate that within this network, EPCR serves as a critical protective component as its deletion hypersensitizes primitive hematopoietic cells to pro-inflammatory signals and ROS accumulation resulting in compromised stem cell function. Conversely, abrogation of the pro-inflammatory activity of UM171 through treatment with dexamethasone, cAMP elevating agents or NFkB inhibitors abolishes EPCR upregulation and HSC expansion. Together, these results show that UM171 stimulates ex vivo HSC expansion by establishing a critical balance between key pro- and anti-inflammatory mediators of self-renewal., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
15. A Fanci knockout mouse model reveals common and distinct functions for FANCI and FANCD2.
- Author
-
Dubois EL, Guitton-Sert L, Béliveau M, Parmar K, Chagraoui J, Vignard J, Pauty J, Caron MC, Coulombe Y, Buisson R, Jacquet K, Gamblin C, Gao Y, Laprise P, Lebel M, Sauvageau G, D d'Andrea A, and Masson JY
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Fanconi Anemia metabolism, Fanconi Anemia pathology, Fanconi Anemia Complementation Group D2 Protein metabolism, Fanconi Anemia Complementation Group Proteins metabolism, Female, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, Oocytes metabolism, Rad51 Recombinase genetics, Rad51 Recombinase metabolism, Spermatocytes metabolism, DNA Repair, Fanconi Anemia genetics, Fanconi Anemia Complementation Group D2 Protein genetics, Fanconi Anemia Complementation Group Proteins genetics
- Abstract
Fanconi Anemia (FA) clinical phenotypes are heterogenous and rely on a mutation in one of the 22 FANC genes (FANCA-W) involved in a common interstrand DNA crosslink-repair pathway. A critical step in the activation of FA pathway is the monoubiquitination of FANCD2 and its binding partner FANCI. To better address the clinical phenotype associated with FANCI and the epistatic relationship with FANCD2, we created the first conditional inactivation model for FANCI in mouse. Fanci -/- mice displayed typical FA features such as delayed development in utero, microphtalmia, cellular sensitivity to mitomycin C, occasional limb abnormalities and hematological deficiencies. Interestingly, the deletion of Fanci leads to a strong meiotic phenotype and severe hypogonadism. FANCI was localized in spermatocytes and spermatids and in the nucleus of oocytes. Both FANCI and FANCD2 proteins co-localized with RPA along meiotic chromosomes, albeit at different levels. Consistent with a role in meiotic recombination, FANCI interacted with RAD51 and stimulated D-loop formation, unlike FANCD2. The double knockout Fanci-/- Fancd2-/- also showed epistatic relationship for hematological defects while being not epistatic with respect to generating viable mice in crosses of double heterozygotes. Collectively, this study highlights common and distinct functions of FANCI and FANCD2 during mouse development, meiotic recombination and hematopoiesis., (© The Author(s) 2019. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2019
- Full Text
- View/download PDF
16. Integrin-α3 Is a Functional Marker of Ex Vivo Expanded Human Long-Term Hematopoietic Stem Cells.
- Author
-
Tomellini E, Fares I, Lehnertz B, Chagraoui J, Mayotte N, MacRae T, Bordeleau MÈ, Corneau S, Bisaillon R, and Sauvageau G
- Subjects
- Animals, Antigens, CD34 metabolism, Cell Proliferation, Cell Self Renewal, Down-Regulation, Fetal Blood cytology, Gene Expression Profiling, Humans, Mice, Phenotype, Proto-Oncogene Proteins c-myc metabolism, Biomarkers metabolism, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Integrin alpha3 metabolism
- Abstract
Transplantation of expanded hematopoietic stem cells (HSCs) and gene therapy based on HSC engineering have emerged as promising approaches for the treatment of hematological diseases. Nevertheless, the immunophenotype of cultured HSCs remains poorly defined. Here, we identify Integrin-α3 (ITGA3) as a marker of cultured human HSCs. Exploiting the pyrimidoindole derivative UM171 to expand cord blood (CB) cells, we show that ITGA3 expression is sufficient to separate the primitive EPCR
+ CD90+ CD133+ CD34+ CD45RA- HSC population into two functionally distinct fractions presenting mostly short-term (ITGA3- ) and both short-term and long-term (ITGA3+ ) repopulating potential. ITGA3+ cells exhibit robust multilineage differentiation potential, serial reconstitution ability in immunocompromised mice, and an HSC-specific transcriptomic signature. Moreover, ITGA3 expression is functionally required for the long-term engraftment of CB cells. Altogether, our results indicate that ITGA3 is a reliable marker of cultured human long-term repopulating HSCs (LT-HSCs) and represents an important tool to improve the accuracy of prospective HSC identification in culture., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
17. UM171 Enhances Lentiviral Gene Transfer and Recovery of Primitive Human Hematopoietic Cells.
- Author
-
Ngom M, Imren S, Maetzig T, Adair JE, Knapp DJHF, Chagraoui J, Fares I, Bordeleau ME, Sauvageau G, Leboulch P, Eaves C, and Humphries RK
- Abstract
Enhanced gene transfer efficiencies and higher yields of transplantable transduced human hematopoietic stem cells are continuing goals for improving clinical protocols that use stemcell-based gene therapies. Here, we examined the effect of the HSC agonist UM171 on these endpoints in both in vitro and in vivo systems. Using a 22-hr transduction protocol, we found that UM171 significantly enhances both the lentivirus-mediated transduction and yield of CD34
+ and CD34+ CD45RA- hematopoietic cells from human cord blood to give a 6-fold overall higher recovery of transduced hematopoietic stem cells, including cells with long-term lympho-myeloid repopulating activity in immunodeficient mice. The ability of UM171 to enhance gene transfer to primitive cord blood hematopoietic cells extended to multiple lentiviral pseudotypes, gamma retroviruses, and non-integrating lentiviruses and to adult bone marrow cells. UM171, thus, provides an interesting reagent for improving the ex vivo production of gene-modified cells and for reducing requirements of virus for a broad range of applications.- Published
- 2018
- Full Text
- View/download PDF
18. Transcriptomic landscape of acute promyelocytic leukemia reveals aberrant surface expression of the platelet aggregation agonist Podoplanin.
- Author
-
Lavallée VP, Chagraoui J, MacRae T, Marquis M, Bonnefoy A, Krosl J, Lemieux S, Marinier A, Pabst C, Rivard GÉ, Hébert J, and Sauvageau G
- Subjects
- Adult, Aged, Animals, Female, Flow Cytometry, Humans, Leukemia, Promyelocytic, Acute genetics, Male, Membrane Glycoproteins genetics, Mice, Middle Aged, Platelet Aggregation, Thrombocytopenia etiology, Tretinoin pharmacology, Hemorrhage etiology, Leukemia, Promyelocytic, Acute complications, Membrane Glycoproteins physiology, Transcriptome
- Abstract
Acute promyelocytic leukemia (APL) is a medical emergency because of associated lethal early bleeding, a condition preventable by prompt diagnosis and therapeutic intervention. The mechanisms underlying the hemostatic anomalies of APL are not completely elucidated. RNA-sequencing-based characterization of APL (n = 30) was performed and compared to that of other acute myeloid leukemia (n = 400) samples and normal promyelocytes. Perturbations in the transcriptome of coagulation and fibrinolysis-related genes in APL extend beyond known culprits and now include Thrombin, Factor X and Urokinase Receptor. Most intriguingly, the Podoplanin (PDPN) gene, involved in platelet aggregation, is aberrantly expressed in APL promyelocytes and is the most distinctive transcript for this disease. Using an antibody panel optimized for AML diagnosis by flow cytometry, we also found that PDPN was the most specific surface marker for APL, and that all-trans retinoic acid therapy rapidly decreases its expression. Functional studies showed that engineered overexpression of this gene in human leukemic cells causes aberrant platelet binding, activation and aggregation. PDPN-expressing primary APL cells, but not PDPN-negative primary leukemias, specifically induce platelet binding, activation and aggregation. Finally, PDPN expression on leukemia cells in a xenograft model was associated with thrombocytopenia and prolonged bleeding time in vivo. Together our results suggest that PDPN may contribute to the hemostatic perturbations found in APL.
- Published
- 2018
- Full Text
- View/download PDF
19. H3 K27M/I mutations promote context-dependent transformation in acute myeloid leukemia with RUNX1 alterations.
- Author
-
Lehnertz B, Zhang YW, Boivin I, Mayotte N, Tomellini E, Chagraoui J, Lavallée VP, Hébert J, and Sauvageau G
- Subjects
- Adolescent, Aged, 80 and over, Animals, DNA Methylation, Female, Humans, Lysine metabolism, Male, Mice, Middle Aged, Oncogene Proteins, Fusion genetics, RUNX1 Translocation Partner 1 Protein genetics, Sequence Analysis, DNA, Core Binding Factor Alpha 2 Subunit genetics, Histones genetics, Leukemia, Myeloid, Acute genetics, Mutation, Missense, Transformation, Genetic
- Abstract
Neomorphic missense mutations affecting crucial lysine residues in histone H3 genes significantly contribute to a variety of solid cancers. Despite the high prevalence of H3
K27M mutations in pediatric glioblastoma and their well-established impact on global histone H3 lysine 27 di- and trimethylation (H3K27me2/3), the relevance of these mutations has not been studied in acute myeloid leukemia (AML). Here, we report the first identification of H3K27M and H3K27I mutations in patients with AML. We find that these lesions are major determinants of reduced H3K27me2/3 in these patients and that they are associated with common aberrations in the RUNX1 gene. We demonstrate that H3K27I/M mutations are strong disease accelerators in a RUNX1-RUNX1T1 AML mouse model, suggesting that H3K27me2/3 has an important and selective leukemia-suppressive activity in this genetic context., (© 2017 by The American Society of Hematology.)- Published
- 2017
- Full Text
- View/download PDF
20. Chemogenomic Landscape of RUNX1 -mutated AML Reveals Importance of RUNX1 Allele Dosage in Genetics and Glucocorticoid Sensitivity.
- Author
-
Simon L, Lavallée VP, Bordeleau ME, Krosl J, Baccelli I, Boucher G, Lehnertz B, Chagraoui J, MacRae T, Ruel R, Chantigny Y, Lemieux S, Marinier A, Hébert J, and Sauvageau G
- Subjects
- Adult, Aged, Aged, 80 and over, Cell Line, Tumor, Female, Gene Expression Regulation, Leukemic, Gene Silencing, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Alleles, Core Binding Factor Alpha 2 Subunit genetics, Drug Resistance, Neoplasm genetics, Gene Dosage, Glucocorticoids pharmacology, Leukemia, Myeloid, Acute genetics, Mutation
- Abstract
Purpose: RUNX1 -mutated ( RUNX1
mut ) acute myeloid leukemia (AML) is associated with adverse outcome, highlighting the urgent need for a better genetic characterization of this AML subgroup and for the design of efficient therapeutic strategies for this disease. Toward this goal, we further dissected the mutational spectrum and gene expression profile of RUNX1mut AML and correlated these results to drug sensitivity to identify novel compounds targeting this AML subgroup. Experimental Design: RNA-sequencing of 47 RUNX1mut primary AML specimens was performed and sequencing results were compared to those of RUNX1 wild-type samples. Chemical screens were also conducted using RUNX1mut specimens to identify compounds selectively affecting the viability of RUNX1mut AML. Results: We show that samples with no remaining RUNX1 wild-type allele are clinically and genetically distinct and display a more homogeneous gene expression profile. Chemical screening revealed that most RUNX1mut specimens are sensitive to glucocorticoids (GCs) and we confirmed that GCs inhibit AML cell proliferation through their interaction with the glucocorticoid receptor (GR). We observed that specimens harboring RUNX1 mutations expected to result in low residual RUNX1 activity are most sensitive to GCs, and that coassociating mutations as well as GR levels contribute to GC sensitivity. Accordingly, acquired glucocorticoid sensitivity was achieved by negatively regulating RUNX1 expression in human AML cells. Conclusions: Our findings show the profound impact of RUNX1 allele dosage on gene expression profile and glucocorticoid sensitivity in AML, thereby opening opportunities for preclinical testing which may lead to drug repurposing and improved disease characterization. Clin Cancer Res; 23(22); 6969-81. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
21. UBAP2L is amplified in a large subset of human lung adenocarcinoma and is critical for epithelial lung cell identity and tumor metastasis.
- Author
-
Aucagne R, Girard S, Mayotte N, Lehnertz B, Lopes-Paciencia S, Gendron P, Boucher G, Chagraoui J, and Sauvageau G
- Subjects
- A549 Cells, Adenocarcinoma genetics, Adenocarcinoma pathology, Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Carrier Proteins genetics, Epithelial Cells pathology, Epithelial-Mesenchymal Transition genetics, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Mice, Neoplasm Metastasis, Neoplasm Proteins genetics, Respiratory Mucosa pathology, Adenocarcinoma metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Carrier Proteins biosynthesis, Epithelial Cells metabolism, Gene Expression Regulation, Neoplastic, Lung Neoplasms metabolism, Neoplasm Proteins biosynthesis, Respiratory Mucosa metabolism
- Abstract
The ubiquitin-associated protein 2-like ( UBAP2L ) gene remains poorly studied in human and mouse development. UBAP2L interacts with the Polycomb group protein B lymphoma Mo-MLV insertion region 1 homolog (BMI1) and determines the activity of mouse hematopoietic stem cells in vivo Here we show that loss of Ubap2l leads to disorganized respiratory epithelium of mutant neonates, which die of respiratory failure. We also show that UBAP2L overexpression leads to epithelial-mesenchymal transition-like phenotype in a non-small cell lung carcinoma (NSCLC) cell line. UBAP2L is amplified in 15% of human primary lung adenocarcinoma specimens. Such patients express higher levels of UBAP2L and show a reduction in survival when compared with those who do not have this gene amplification. Supporting a possible role for UBAP2L in lung tumor progression, NSCLC cells engineered to express low levels of this gene produce much smaller tumors in vivo than wild-type control cells. Together, these results suggest that UBAP2L contributes to epithelial lung cell identity in mice and that it plays an important role in human lung adenocarcinoma.-Aucagne, R., Girard, S., Mayotte, N., Lehnertz, B., Lopes-Paciencia, S., Gendron, P., Boucher, G., Chagraoui, J., Sauvageau, G. UBAP2L is amplified in a large subset of human lung adenocarcinoma and is critical for epithelial lung cell identity and tumor metastasis., (© FASEB.)
- Published
- 2017
- Full Text
- View/download PDF
22. EPCR expression marks UM171-expanded CD34 + cord blood stem cells.
- Author
-
Fares I, Chagraoui J, Lehnertz B, MacRae T, Mayotte N, Tomellini E, Aubert L, Roux PP, and Sauvageau G
- Subjects
- Animals, Cell Culture Techniques, Cell Proliferation drug effects, Endothelial Protein C Receptor, Female, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology, Humans, Mice, Inbred NOD, Mice, SCID, Antigens, CD analysis, Antigens, CD34 analysis, Fetal Blood cytology, Hematopoietic Stem Cells drug effects, Indoles pharmacology, Pyrimidines pharmacology, Receptors, Cell Surface analysis
- Abstract
A small subset of human cord blood CD34
+ cells express endothelial protein C receptor (EPCR/CD201/PROCR) when exposed to the hematopoietic stem cell (HSC) self-renewal agonist UM171. In this article, we show that EPCR-positive UM171-treated cells, as opposed to EPCR-negative cells, exhibit robust multilineage repopulation and serial reconstitution ability in immunocompromised mice. In contrast to other stem cell markers, such as CD38, EPCR expression is maintained when cells are introduced in culture, irrespective of UM171 treatment. Although engineered overexpression of EPCR fails to reproduce the effects of UM171 on HSC activity, its expression is required for the repopulating activity of human HSCs. Altogether, our results indicate that EPCR is a reliable and cell culture-compatible marker of UM171-expanded human cord blood HSCs., (© 2017 by The American Society of Hematology.)- Published
- 2017
- Full Text
- View/download PDF
23. High-throughput screening in niche-based assay identifies compounds to target preleukemic stem cells.
- Author
-
Gerby B, Veiga DF, Krosl J, Nourreddine S, Ouellette J, Haman A, Lavoie G, Fares I, Tremblay M, Litalien V, Ottoni E, Kosic M, Geoffrion D, Ryan J, Maddox PS, Chagraoui J, Marinier A, Hébert J, Sauvageau G, Kwok BH, Roux PP, and Hoang T
- Subjects
- 2-Methoxyestradiol, Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Estradiol pharmacology, Humans, Jurkat Cells, LIM Domain Proteins genetics, LIM Domain Proteins metabolism, Mice, Neoplastic Stem Cells pathology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, T-Cell Acute Lymphocytic Leukemia Protein 1, Transcription Factors genetics, Transcription Factors metabolism, Tumor Microenvironment genetics, Tumor Stem Cell Assay, Xenograft Model Antitumor Assays, Estradiol analogs & derivatives, Neoplastic Stem Cells metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Tumor Microenvironment drug effects
- Abstract
Current chemotherapies for T cell acute lymphoblastic leukemia (T-ALL) efficiently reduce tumor mass. Nonetheless, disease relapse attributed to survival of preleukemic stem cells (pre-LSCs) is associated with poor prognosis. Herein, we provide direct evidence that pre-LSCs are much less chemosensitive to existing chemotherapy drugs than leukemic blasts because of a distinctive lower proliferative state. Improving therapies for T-ALL requires the development of strategies to target pre-LSCs that are absolutely dependent on their microenvironment. Therefore, we designed a robust protocol for high-throughput screening of compounds that target primary pre-LSCs maintained in a niche-like environment, on stromal cells that were engineered for optimal NOTCH1 activation. The multiparametric readout takes into account the intrinsic complexity of primary cells in order to specifically monitor pre-LSCs, which were induced here by the SCL/TAL1 and LMO1 oncogenes. We screened a targeted library of compounds and determined that the estrogen derivative 2-methoxyestradiol (2-ME2) disrupted both cell-autonomous and non-cell-autonomous pathways. Specifically, 2-ME2 abrogated pre-LSC viability and self-renewal activity in vivo by inhibiting translation of MYC, a downstream effector of NOTCH1, and preventing SCL/TAL1 activity. In contrast, normal hematopoietic stem/progenitor cells remained functional. These results illustrate how recapitulating tissue-like properties of primary cells in high-throughput screening is a promising avenue for innovation in cancer chemotherapy., Competing Interests: The authors have declared that no conflict of interest exists.
- Published
- 2016
- Full Text
- View/download PDF
24. E4F1 is a master regulator of CHK1-mediated functions.
- Author
-
Grote D, Moison C, Duhamel S, Chagraoui J, Girard S, Yang J, Mayotte N, Coulombe Y, Masson JY, Brown GW, Meloche S, and Sauvageau G
- Subjects
- Animals, Apoptosis genetics, Bone Marrow metabolism, Bone Marrow pathology, Checkpoint Kinase 1, DNA Damage genetics, DNA Replication genetics, DNA-Binding Proteins biosynthesis, Gene Expression Regulation, Developmental, HEK293 Cells, Hematopoietic Stem Cells metabolism, Humans, Mice, Mouse Embryonic Stem Cells metabolism, Polycomb Repressive Complex 1 metabolism, Protein Kinases genetics, Proteolysis, Proto-Oncogene Proteins metabolism, Repressor Proteins, Transcription Factors biosynthesis, Ubiquitin-Protein Ligases, DNA-Binding Proteins genetics, Genomic Instability, Polycomb Repressive Complex 1 genetics, Protein Kinases biosynthesis, Proto-Oncogene Proteins genetics, Transcription Factors genetics
- Abstract
It has been previously shown that the polycomb protein BMI1 and E4F1 interact physically and genetically in the hematopoietic system. Here, we report that E4f1 is essential for hematopoietic cell function and survival. E4f1 deletion induces acute bone marrow failure characterized by apoptosis of progenitors while stem cells are preserved. E4f1-deficient cells accumulate DNA damage and show defects in progression through S phase and mitosis, revealing a role for E4F1 in cell-cycle progression and genome integrity. Importantly, we showed that E4F1 interacts with and protects the checkpoint kinase 1 (CHK1) protein from degradation. Finally, defects observed in E4f1-deficient cells were fully reversed by ectopic expression of Chek1. Altogether, our results classify E4F1 as a master regulator of CHK1 activity that ensures high fidelity of DNA replication, thus safeguarding genome stability., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. UBAP2L is a novel BMI1-interacting protein essential for hematopoietic stem cell activity.
- Author
-
Bordeleau ME, Aucagne R, Chagraoui J, Girard S, Mayotte N, Bonneil E, Thibault P, Pabst C, Bergeron A, Barabé F, Hébert J, Sauvageau M, Boutonnet C, Meloche S, and Sauvageau G
- Subjects
- Animals, Bone Marrow Cells metabolism, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Down-Regulation, Gene Deletion, Gene Knockdown Techniques, HEK293 Cells, Humans, Leukemia, Myeloid, Acute metabolism, Mice, Inbred C57BL, Polycomb-Group Proteins metabolism, Protein Binding, RNA, Small Interfering metabolism, Ubiquitin-Protein Ligases metabolism, Carrier Proteins metabolism, Hematopoietic Stem Cells metabolism, Polycomb Repressive Complex 1 metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Multipotent long-term repopulating hematopoietic stem cells (LT-HSCs) can self-renew or differentiate into the less primitive short-term repopulating stem cells (ST-HSCs), which themselves produce progenitors that ensure the daily supply of all essential blood components. The Polycomb group (PcG) protein BMI1 is essential for the activity of both HSCs and progenitor cells. Although BMI1 operates by suppressing the Ink4a/Arf locus in progenitors and ST-HSCs, the mechanisms through which this gene regulates the activity of LT-HSCs remain poorly understood. Toward this goal, we isolated BMI1-containing protein complexes and identified UBAP2L as a novel BMI1-interacting protein. We also showed that UBAP2L is preferentially expressed in mouse and human HSC-enriched populations when compared with more mature cell types, and that this gene is essential for the activity of LT-HSCs. In contrast to what is observed for Bmi1 knockdown, we found that UBAP2L depletion does not affect the Ink4a/Arf locus. Given that we demonstrated that BMI1 overexpression is able to rescue the deleterious effects of Ubap2l downregulation on LT-HSC activity and that UBAP2L is part of a PcG subcomplex comprising BMI1, we propose a model in which at least 2 different BMI1-containing PcG complexes regulate HSC activity, which are distinguishable by the presence of UBAP2L., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
26. Cord blood expansion. Pyrimidoindole derivatives are agonists of human hematopoietic stem cell self-renewal.
- Author
-
Fares I, Chagraoui J, Gareau Y, Gingras S, Ruel R, Mayotte N, Csaszar E, Knapp DJ, Miller P, Ngom M, Imren S, Roy DC, Watts KL, Kiem HP, Herrington R, Iscove NN, Humphries RK, Eaves CJ, Cohen S, Marinier A, Zandstra PW, and Sauvageau G
- Subjects
- Animals, Cell Culture Techniques, Fetal Blood cytology, Fetal Blood physiology, Genetic Therapy methods, Hematopoiesis physiology, Hematopoietic Stem Cell Transplantation methods, Hematopoietic Stem Cells physiology, Humans, Immunocompromised Host, Indoles chemistry, Mice, Pyrimidines chemistry, Small Molecule Libraries chemistry, Small Molecule Libraries pharmacology, Fetal Blood drug effects, Hematopoiesis drug effects, Hematopoietic Stem Cells drug effects, Indoles pharmacology, Pyrimidines pharmacology, Receptors, Aryl Hydrocarbon antagonists & inhibitors, Regeneration drug effects
- Abstract
The small number of hematopoietic stem and progenitor cells in cord blood units limits their widespread use in human transplant protocols. We identified a family of chemically related small molecules that stimulates the expansion ex vivo of human cord blood cells capable of reconstituting human hematopoiesis for at least 6 months in immunocompromised mice. The potent activity of these newly identified compounds, UM171 being the prototype, is independent of suppression of the aryl hydrocarbon receptor, which targets cells with more-limited regenerative potential. The properties of UM171 make it a potential candidate for hematopoietic stem cell transplantation and gene therapy., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
27. RNAi screen identifies Jarid1b as a major regulator of mouse HSC activity.
- Author
-
Cellot S, Hope KJ, Chagraoui J, Sauvageau M, Deneault É, MacRae T, Mayotte N, Wilhelm BT, Landry JR, Ting SB, Krosl J, Humphries K, Thompson A, and Sauvageau G
- Subjects
- Animals, Cells, Cultured, DNA-Binding Proteins genetics, Gene Expression Regulation drug effects, Gene Knockdown Techniques, Hematopoiesis drug effects, Hematopoietic Stem Cells metabolism, Histone Demethylases genetics, Histone Demethylases physiology, Jumonji Domain-Containing Histone Demethylases genetics, Mice, Mice, Congenic, Mice, Inbred C57BL, RNA, Small Interfering genetics, RNA, Small Interfering pharmacology, Transcription Factors genetics, Transcription Factors physiology, Validation Studies as Topic, DNA-Binding Proteins physiology, Hematopoiesis genetics, Hematopoietic Stem Cells physiology, High-Throughput Screening Assays methods, Jumonji Domain-Containing Histone Demethylases physiology, RNA Interference physiology
- Abstract
Histone methylation is a dynamic and reversible process proposed to directly impact on stem cell fate. The Jumonji (JmjC) domain-containing family of demethylases comprises 27 members that target mono-, di-, and trimethylated lysine residues of histone (or nonhistone) proteins. To evaluate their role in regulation of hematopoietic stem cell (HSC) behavior, we performed an in vivo RNAi-based functional screen and demonstrated that Jarid1b and Jhdm1f play opposing roles in regulation of HSC activity. Decrease in Jarid1b levels correlated with an in vitro expansion of HSCs with preserved long-term in vivo lymphomyeloid differentiation potential. Through RNA sequencing analysis, Jarid1b knockdown was associated with increased expression levels of several HSC regulators (Hoxa7, Hoxa9, Hoxa10, Hes1, Gata2) and reduced levels of differentiation-associated genes. shRNA against Jhdmlf, in contrast, impaired hematopoietic reconstitution of bone marrow cells. Together, our studies identified Jarid1b as a negative regulator of HSC activity and Jhdmlf as a positive regulator of HSC activity., Competing Interests: Conflict-of-interest disclosure: The authors declare no competing financial interests.
- Published
- 2013
- Full Text
- View/download PDF
28. Posttranslational regulation of self-renewal capacity: insights from proteome and phosphoproteome analyses of stem cell leukemia.
- Author
-
Trost M, Sauvageau M, Hérault O, Deleris P, Pomiès C, Chagraoui J, Mayotte N, Meloche S, Sauvageau G, and Thibault P
- Subjects
- Amino Acid Sequence, Animals, DNA-Binding Proteins analysis, DNA-Binding Proteins metabolism, Enhancer of Zeste Homolog 2 Protein, Enzyme Activation, Histone-Lysine N-Methyltransferase analysis, Histone-Lysine N-Methyltransferase metabolism, Humans, Mice, Molecular Sequence Data, Phosphorylation, Polycomb Repressive Complex 2, Polycomb-Group Proteins, Protein Interaction Maps, Protein Processing, Post-Translational, Repressor Proteins analysis, Repressor Proteins metabolism, Transcription Factors analysis, Transcription Factors metabolism, Tumor Cells, Cultured, p38 Mitogen-Activated Protein Kinases analysis, p38 Mitogen-Activated Protein Kinases metabolism, Leukemia, Myeloid, Acute metabolism, Neoplastic Stem Cells metabolism, Proteome analysis, Proteome metabolism
- Abstract
We recently generated 2 phenotypically similar Hoxa9+Meis1 overexpressing acute myeloid leukemias that differ by their in vivo biologic behavior. The first leukemia, named FLA2, shows a high frequency of leukemia stem cells (LSCs; 1 in 1.4 cells), whereas the second, FLB1, is more typical with a frequency of LSCs in the range of 1 per several hundred cells. To gain insights into possible mechanisms that determine LSC self-renewal, we profiled and compared the abundance of nuclear and cytoplasmic proteins and phosphoproteins from these leukemias using quantitative proteomics. These analyses revealed differences in proteins associated with stem cell fate, including a hyperactive p38 MAP kinase in FLB1 and a differentially localized Polycomb group protein Ezh2, which is mostly nuclear in FLA2 and predominantly cytoplasmic in FLB1. Together, these newly documented proteomes and phosphoproteomes represent a unique resource with more than 440 differentially expressed proteins and 11 543 unique phosphopeptides, of which 80% are novel and 7% preferentially phosphorylated in the stem cell-enriched leukemia., Competing Interests: Conflict-of-interest disclosure: The authors declare no competing financial interests.
- Published
- 2012
- Full Text
- View/download PDF
29. A role for GPx3 in activity of normal and leukemia stem cells.
- Author
-
Herault O, Hope KJ, Deneault E, Mayotte N, Chagraoui J, Wilhelm BT, Cellot S, Sauvageau M, Andrade-Navarro MA, Hébert J, and Sauvageau G
- Subjects
- Animals, Base Sequence, Blotting, Southern, Cell Line, DNA-Binding Proteins metabolism, Flow Cytometry, Fluorescence, Gene Expression Profiling, Genetic Vectors genetics, Homeodomain Proteins metabolism, Humans, Mice, Microscopy, Confocal, Molecular Sequence Data, Myeloid Ecotropic Viral Integration Site 1 Protein, Neoplasm Proteins metabolism, Reactive Oxygen Species metabolism, Real-Time Polymerase Chain Reaction, Sequence Analysis, DNA, Transcription Factors metabolism, Gene Expression Regulation, Neoplastic physiology, Glutathione Peroxidase metabolism, Leukemia metabolism, Neoplastic Stem Cells metabolism, Stem Cells metabolism
- Abstract
The determinants of normal and leukemic stem cell self-renewal remain poorly characterized. We report that expression of the reactive oxygen species (ROS) scavenger glutathione peroxidase 3 (GPx3) positively correlates with the frequency of leukemia stem cells (LSCs) in Hoxa9+Meis1-induced leukemias. Compared with a leukemia with a low frequency of LSCs, a leukemia with a high frequency of LSCs showed hypomethylation of the Gpx3 promoter region, and expressed high levels of Gpx3 and low levels of ROS. LSCs and normal hematopoietic stem cells (HSCs) engineered to express Gpx3 short hairpin RNA (shRNA) were much less competitive in vivo than control cells. However, progenitor cell proliferation and differentiation was not affected by Gpx3 shRNA. Consistent with this, HSCs overexpressing Gpx3 were significantly more competitive than control cells in long-term repopulation experiments, and overexpression of the self-renewal genes Prdm16 or Hoxb4 boosted Gpx3 expression. In human primary acute myeloid leukemia samples, GPX3 expression level directly correlated with adverse prognostic outcome, revealing a potential novel target for the eradication of LSCs.
- Published
- 2012
- Full Text
- View/download PDF
30. A key role for EZH2 and associated genes in mouse and human adult T-cell acute leukemia.
- Author
-
Simon C, Chagraoui J, Krosl J, Gendron P, Wilhelm B, Lemieux S, Boucher G, Chagnon P, Drouin S, Lambert R, Rondeau C, Bilodeau A, Lavallée S, Sauvageau M, Hébert J, and Sauvageau G
- Subjects
- Acute Disease, Animals, DNA-Binding Proteins genetics, Enhancer of Zeste Homolog 2 Protein, Histone-Lysine N-Methyltransferase genetics, Humans, Leukemia-Lymphoma, Adult T-Cell genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Polycomb Repressive Complex 2, Polycomb-Group Proteins, Protein Binding, Repressor Proteins genetics, Repressor Proteins metabolism, Transcription Factors genetics, DNA-Binding Proteins metabolism, Histone-Lysine N-Methyltransferase metabolism, Leukemia-Lymphoma, Adult T-Cell metabolism, Transcription Factors metabolism
- Abstract
In this study, we show the high frequency of spontaneous γδ T-cell leukemia (T-ALL) occurrence in mice with biallelic deletion of enhancer of zeste homolog 2 (Ezh2). Tumor cells show little residual H3K27 trimethylation marks compared with controls. EZH2 is a component of the PRC2 Polycomb group protein complex, which is associated with DNA methyltransferases. Using next-generation sequencing, we identify alteration in gene expression levels of EZH2 and acquired mutations in PRC2-associated genes (DNMT3A and JARID2) in human adult T-ALL. Together, these studies document that deregulation of EZH2 and associated genes leads to the development of mouse, and likely human, T-ALL.
- Published
- 2012
- Full Text
- View/download PDF
31. Asymmetric segregation and self-renewal of hematopoietic stem and progenitor cells with endocytic Ap2a2.
- Author
-
Ting SB, Deneault E, Hope K, Cellot S, Chagraoui J, Mayotte N, Dorn JF, Laverdure JP, Harvey M, Hawkins ED, Russell SM, Maddox PS, Iscove NN, and Sauvageau G
- Subjects
- Adaptor Protein Complex 2 antagonists & inhibitors, Adaptor Protein Complex 2 genetics, Adaptor Protein Complex alpha Subunits antagonists & inhibitors, Adaptor Protein Complex alpha Subunits genetics, Animals, Biomarkers metabolism, Blotting, Western, Cell Differentiation, Cell Lineage, Cell Proliferation, Flow Cytometry, Gene Expression Profiling, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells physiology, Leukemia metabolism, Leukemia pathology, Mice, Neoplastic Stem Cells metabolism, Oligonucleotide Array Sequence Analysis, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells physiology, Adaptor Protein Complex 2 metabolism, Adaptor Protein Complex alpha Subunits metabolism, Asymmetric Cell Division physiology, Cell Polarity genetics, Endocytosis genetics, Hematopoietic Stem Cells cytology, Neoplastic Stem Cells pathology, Stem Cells cytology
- Abstract
The stem cell-intrinsic model of self-renewal via asymmetric cell division (ACD) posits that fate determinants be partitioned unequally between daughter cells to either activate or suppress the stemness state. ACD is a purported mechanism by which hematopoietic stem cells (HSCs) self-renew, but definitive evidence for this cellular process remains open to conjecture. To address this issue, we chose 73 candidate genes that function within the cell polarity network to identify potential determinants that may concomitantly alter HSC fate while also exhibiting asymmetric segregation at cell division. Initial gene-expression profiles of polarity candidates showed high and differential expression in both HSCs and leukemia stem cells. Altered HSC fate was assessed by our established in vitro to in vivo screen on a subcohort of candidate polarity genes, which revealed 6 novel positive regulators of HSC function: Ap2a2, Gpsm2, Tmod1, Kif3a, Racgap1, and Ccnb1. Interestingly, live-cell videomicroscopy of the endocytic protein AP2A2 shows instances of asymmetric segregation during HSC/progenitor cell cytokinesis. These results contribute further evidence that ACD is functional in HSC self-renewal, suggest a role for Ap2a2 in HSC activity, and provide a unique opportunity to prospectively analyze progeny from HSC asymmetric divisions.
- Published
- 2012
- Full Text
- View/download PDF
32. An anticlastogenic function for the Polycomb Group gene Bmi1.
- Author
-
Chagraoui J, Hébert J, Girard S, and Sauvageau G
- Subjects
- Animals, Cell Cycle physiology, Cell Line, Chromosomes metabolism, DNA Damage, DNA Repair, Fibroblasts cytology, Fibroblasts physiology, Gene Expression Regulation, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Histones genetics, Histones metabolism, Humans, Mice, Mice, Knockout, Nuclear Proteins genetics, Polycomb Repressive Complex 1, Polycomb-Group Proteins, Proto-Oncogene Proteins genetics, Recombination, Genetic, Repressor Proteins genetics, Nuclear Proteins metabolism, Proto-Oncogene Proteins metabolism, Repressor Proteins metabolism
- Abstract
BMI1 is a key component of multiprotein Polycomb repression complex 1 (PRC1), and its disruption in mice induces severe aplastic anemia by early adulthood. The contributing mechanisms responsible for this phenotype remain elusive. Here we show that transformed human cell lines as well as primitive hematopoietic cells exhibit a high frequency of spontaneous chromosome breaks upon BMI1 depletion and are hypersensitive to genotoxic agents. Consistent with these observations, we found that BMI1 is recruited rapidly to DNA damage foci where it blocks transcriptional elongation. We also show that BMI1 contributes to homologous recombination DNA repair and is required for checkpoint recovery. Taken together, our results suggest that BMI1 is critical for the maintenance of chromosome integrity in both normal and transformed cells.
- Published
- 2011
- Full Text
- View/download PDF
33. Genome-wide interrogation of Mammalian stem cell fate determinants by nested chromosome deletions.
- Author
-
Fortier S, Bilodeau M, Macrae T, Laverdure JP, Azcoitia V, Girard S, Chagraoui J, Ringuette N, Hébert J, Krosl J, Mayotte N, and Sauvageau G
- Subjects
- Animals, Cell Line, Chromosome Mapping, Female, Humans, Male, Mice, Cell Differentiation, Chromosome Deletion, Embryonic Stem Cells cytology, Genome, Mammals genetics
- Abstract
Understanding the function of important DNA elements in mammalian stem cell genomes would be enhanced by the availability of deletion collections in which segmental haploidies are precisely characterized. Using a modified Cre-loxP-based system, we now report the creation and characterization of a collection of ∼1,300 independent embryonic stem cell (ESC) clones enriched for nested chromosomal deletions. Mapping experiments indicate that this collection spans over 25% of the mouse genome with good representative coverage of protein-coding genes, regulatory RNAs, and other non-coding sequences. This collection of clones was screened for in vitro defects in differentiation of ESC into embryoid bodies (EB). Several putative novel haploinsufficient regions, critical for EB development, were identified. Functional characterization of one of these regions, through BAC complementation, identified the ribosomal gene Rps14 as a novel haploinsufficient determinant of embryoid body formation. This new library of chromosomal deletions in ESC (DelES: http://bioinfo.iric.ca/deles) will serve as a unique resource for elucidation of novel protein-coding and non-coding regulators of ESC activity., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2010
- Full Text
- View/download PDF
34. A mutant allele of the Swi/Snf member BAF250a determines the pool size of fetal liver hemopoietic stem cell populations.
- Author
-
Krosl J, Mamo A, Chagraoui J, Wilhelm BT, Girard S, Louis I, Lessard J, Perreault C, and Sauvageau G
- Subjects
- Alleles, Animals, Blotting, Western, Cell Count, Cell Proliferation, Colony-Forming Units Assay, DNA-Binding Proteins metabolism, Female, Flow Cytometry, Gene Expression Profiling, Hematopoiesis, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology, Liver embryology, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, Nuclear Proteins metabolism, Oligonucleotide Array Sequence Analysis, Time Factors, Transcription Factors, DNA-Binding Proteins genetics, Hematopoietic Stem Cells metabolism, Liver metabolism, Mutation, Nuclear Proteins genetics
- Abstract
It is believed that hemopoietic stem cells (HSC), which colonize the fetal liver (FL) rapidly, expand to establish a supply of HSCs adequate for maintenance of hemopoiesis throughout life. Accordingly, FL HSCs are actively cycling as opposed to their predominantly quiescent bone marrow counterparts, suggesting that the FL microenvironment provides unique signals that support HSC proliferation and self-renewal. We now report the generation and characterization of mice with a mutant allele of Baf250a lacking exons 2 and 3. Baf250a(E2E3/E2E3) mice are viable until E19.5, but do not survive beyond birth. Most interestingly, FL HSC numbers are markedly higher in these mice than in control littermates, thus raising the possibility that Baf250a determines the HSC pool size in vivo. Limit dilution experiments indicate that the activity of Baf250a(E2E3/E2E3) HSC is equivalent to that of the wild-type counterparts. The Baf250a(E2E3/E2E3) FL-derived stroma, in contrast, exhibits a hemopoiesis-supporting potential superior to the developmentally matched controls. To our knowledge, this demonstration is the first that a mechanism operating in a cell nonautonomous manner canexpand the pool size of the fetal HSC populations.
- Published
- 2010
- Full Text
- View/download PDF
35. A functional screen to identify novel effectors of hematopoietic stem cell activity.
- Author
-
Deneault E, Cellot S, Faubert A, Laverdure JP, Fréchette M, Chagraoui J, Mayotte N, Sauvageau M, Ting SB, and Sauvageau G
- Subjects
- Adult Stem Cells metabolism, Animals, Bone Marrow Cells, Cell Differentiation, Hematopoietic Stem Cells metabolism, Humans, Mice, Mice, Inbred C57BL, NIH 3T3 Cells, Specific Pathogen-Free Organisms, Adult Stem Cells cytology, Gene Regulatory Networks, Hematopoietic Stem Cells cytology, Nuclear Proteins analysis
- Abstract
Despite tremendous progress made toward the identification of the molecular circuitry that governs cell fate in embryonic stem cells, genes controlling this process in the adult hematopoietic stem cell have proven to be more difficult to unmask. We now report the results of a novel gain-of-function screening approach, which identified a series of 18 nuclear factors that affect hematopoietic stem cell activity. Overexpression of ten of these factors resulted in an increased repopulating activity compared to unmanipulated cells. Interestingly, at least four of the 18 factors, Fos, Tcfec, Hmgb1, and Sfpi1, show non-cell-autonomous functions. The utilization of this screening method together with the creation of a database enriched for potential determinants of hematopoietic stem cell self-renewal will serve as a resource to uncover regulatory networks in these cells.
- Published
- 2009
- Full Text
- View/download PDF
36. The signaling protein Wnt4 enhances thymopoiesis and expands multipotent hematopoietic progenitors through beta-catenin-independent signaling.
- Author
-
Louis I, Heinonen KM, Chagraoui J, Vainio S, Sauvageau G, and Perreault C
- Subjects
- Animals, Cell Differentiation immunology, Flow Cytometry, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells metabolism, Immunoblotting, Mice, Multipotent Stem Cells immunology, Multipotent Stem Cells metabolism, Reverse Transcriptase Polymerase Chain Reaction, Thymus Gland cytology, Thymus Gland immunology, Wnt Proteins immunology, Wnt4 Protein, beta Catenin immunology, Hematopoietic Stem Cells cytology, Multipotent Stem Cells cytology, Signal Transduction immunology, Thymus Gland growth & development, Wnt Proteins metabolism, beta Catenin metabolism
- Abstract
Despite studies based on deletion or activation of intracellular components of the canonical Wingless related (Wnt) pathway, the role of Wnts in hematolymphopoiesis remains controversial. Using gain-of-function and loss-of-function models, we found that Wnt4 differentially affected diverse subsets of hematopoietic stem and progenitor cells. Bone-marrow and thymic Lin(-)Sca1(+)Kit(hi) cells (LSKs) were the key targets of Wnt4. In adult mice, Wnt4-induced expansion of Flt3(+) bone-marrow LSKs (lymphoid-primed multipotent progenitors) led to a sizeable accumulation of the most immature thymocyte subsets (upstream of beta-selection) and a major increase in thymopoiesis. Conversely, Wnt4(-/-) neonates showed low frequencies of bone-marrow LSKs and thymic hypocellularity. We provide compelling evidence that Wnt4 activates noncanonical (beta-catenin-independent) signaling and that its effects on hematopoietic cells are mainly non-cell-autonomous. Our work shows that Wnt4 overexpression has a unique ability to expand Flt3(+) LSKs in adults and demonstrates that noncanonical Wnt signaling regulates thymopoiesis.
- Published
- 2008
- Full Text
- View/download PDF
37. Sustained in vitro trigger of self-renewal divisions in Hoxb4hiPbx1(10) hematopoietic stem cells.
- Author
-
Cellot S, Krosl J, Chagraoui J, Meloche S, Humphries RK, and Sauvageau G
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Cell Division physiology, Cells, Cultured, Cyclin D3, Cyclin-Dependent Kinase Inhibitor p27 genetics, Cyclins genetics, Down-Regulation genetics, Gene Expression Profiling, Hematopoietic Stem Cells cytology, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Pre-B-Cell Leukemia Transcription Factor 1, Proto-Oncogene Proteins c-myc genetics, Repressor Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction methods, Transcription, Genetic genetics, Hematopoietic Stem Cells physiology, Homeodomain Proteins physiology, Transcription Factors physiology
- Abstract
Factors that trigger and sustain self-renewal divisions in tissue stem cells remain poorly characterized. By modulating the levels of Hoxb4 and its co-factor Pbxl in primary hematopoietic cells (Hoxb4hiPbxl(10) cells), we report an in vitro expansion of mouse hematopoietic stem cells (HSCs) by 105-fold over 2 weeks, with subsequent preservation of HSC properties. Clonal analyses of the hematopoietic system in recipients of expanded HSCs indicate that up to 70% of Hoxb4hiPbxl(10) stem cells present at initiation of culture underwent self-renewal in vitro. In this setting, Hoxb4 and its co-factor did not promote an increase in DNA synthesis, or a decrease in doubling time of Scal+Lin- cells when compared to controls. Q-PCR analyses further revealed a downregulation of Cdknlb (p27Kipl) and Mxdl (MadI) transcript levels in Hoxb4hiPbxl(l0) primitive cells, accompanied by a more subtle increase in c-myc and reduction in Ccnd3 (Cyclin D3). We thus put forward this strategy as an efficient in vitro HSC expansion tool, enabling a further step into the avenue of self-renewal molecular effectors.
- Published
- 2007
- Full Text
- View/download PDF
38. E4F1: a novel candidate factor for mediating BMI1 function in primitive hematopoietic cells.
- Author
-
Chagraoui J, Niessen SL, Lessard J, Girard S, Coulombe P, Sauvageau M, Meloche S, and Sauvageau G
- Subjects
- Animals, Apoptosis, Cell Cycle, Colony-Forming Units Assay, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p16 metabolism, DNA-Binding Proteins genetics, Humans, Immunoprecipitation, Liver metabolism, Mice, Mice, Knockout, Nuclear Proteins genetics, Polycomb Repressive Complex 1, Proto-Oncogene Proteins genetics, Repressor Proteins genetics, Saccharomyces cerevisiae, Transcription Factors genetics, Tumor Suppressor Protein p14ARF genetics, Tumor Suppressor Protein p14ARF metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Two-Hybrid System Techniques, Ubiquitin-Protein Ligases, Cell Proliferation, DNA-Binding Proteins metabolism, Hematopoietic Stem Cells metabolism, Nuclear Proteins physiology, Proto-Oncogene Proteins physiology, Repressor Proteins physiology, Transcription Factors metabolism
- Abstract
The Polycomb group gene Bmi1 is essential for the proliferation of neural and hematopoietic stem cells. Much remains to be learned about the pathways involved in the severe hematopoietic phenotype observed in Bmi1 homozygous mutant mice except for the fact that loss of p53 or concomitant loss of p16(Ink4a) and p19(Arf) functions achieves only a partial rescue. Here we report the identification of E4F1, an inhibitor of cellular proliferation, as a novel BMI1-interacting partner in hematopoietic cells. We provide evidence that Bmi1 and E4f1 genetically interact in the hematopoietic compartment to regulate cellular proliferation. Most importantly, we demonstrate that reduction of E4f1 levels through RNA interference mediated knockdown is sufficient to rescue the clonogenic and repopulating ability of Bmi1(-/-) hematopoietic cells up to 3 mo post-transplantation. Using cell lines and MEF, we also demonstrate that INK4A/ARF and p53 are not essential for functional interaction between Bmi1 and E4f1. Together, these findings identify E4F1 as a key modulator of BMI1 activity in primitive hematopoietic cells.
- Published
- 2006
- Full Text
- View/download PDF
39. Expression of Pitx2 in stromal cells is required for normal hematopoiesis.
- Author
-
Kieusseian A, Chagraoui J, Kerdudo C, Mangeot PE, Gage PJ, Navarro N, Izac B, Uzan G, Forget BG, and Dubart-Kupperschmitt A
- Subjects
- Animals, Cells, Cultured, Colony-Forming Units Assay, Fetus, Homeodomain Proteins genetics, Homozygote, Lentivirus genetics, Liver metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Nuclear Proteins genetics, RNA, Small Interfering pharmacology, Stem Cells cytology, Stem Cells metabolism, Stromal Cells cytology, Transcription Factors, Transfection, Homeobox Protein PITX2, Gene Expression Regulation, Developmental, Hematopoiesis physiology, Homeodomain Proteins physiology, Liver cytology, Nuclear Proteins physiology, Stromal Cells metabolism
- Abstract
Although the expression of Pitx2, a bicoid family homeodomain transcription factor, is highly regulated during hematopoiesis, its function during this process was not documented; we thus studied hematopoiesis in Pitx2-null mice. We found that Pitx2(-/-) embryos display hypoplastic livers with reduced numbers of hematopoietic cells, but these cells had normal hematopoietic potential, as evidenced by colony-forming assays, immature progenitor cell assays, and long-term repopulation assays. Because the microenvironment is also crucial to the development of normal hematopoiesis, we established Pitx2(-/-) and Pitx2(+/+) stromas from fetal liver and studied their hematopoietic supportive capacity. We showed that the frequency of cobblestone area-forming cells was 4-fold decreased when using Pitx2(-/-) stromal cells compared with Pitx2(+/+) stromal cells, whatever the Pitx2 genotype of hematopoietic cells tested in this assay. This defect was rescued by expression of Pitx2 into Pitx2(-/-) fetal liver stromal cells, demonstrating a major and direct role of Pitx2 in the hematopoietic supportive capacity of fetal liver stroma. Finally, we showed a reduced capacity of MS5 stromal cells expressing Pitx2 RNAi to support human hematopoiesis. Altogether these data showed that Pitx2 has major functions in the hematopoietic supportive capacity of fetal liver and adult bone marrow stromal cells.
- Published
- 2006
- Full Text
- View/download PDF
40. Human endothelial cells derived from circulating progenitors display specific functional properties compared with mature vessel wall endothelial cells.
- Author
-
Bompais H, Chagraoui J, Canron X, Crisan M, Liu XH, Anjo A, Tolla-Le Port C, Leboeuf M, Charbord P, Bikfalvi A, and Uzan G
- Subjects
- Animals, Base Sequence, Cell Culture Techniques, Cell Division, Colony-Forming Units Assay, DNA Primers, Endothelium, Vascular ultrastructure, Flow Cytometry, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Microscopy, Electron, Neovascularization, Physiologic, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells ultrastructure, Umbilical Veins, Cell Differentiation physiology, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Stem Cells cytology
- Abstract
Endothelial progenitor cells (EPCs) were shown to be present in systemic circulation and cord blood. We investigated whether EPCs display specific properties compared with mature endothelial cells. Human cord blood CD34+ cells were isolated and adherent cells were amplified under endothelial conditions. Expression of specific markers identified them as endothelial cells, also called endothelial progenitor-derived cells (EPDCs). When compared to mature endothelial cells, human umbilical vein endothelial cells (HUVECs) and human bone marrow endothelial cells (HBMECs), endothelial markers, were expressed to the same extent except for KDR, which is expressed more in EPDCs. They display a higher proliferation potential. Functional studies demonstrated that EPDCs were more sensitive to angiogenic factors, which afford these cells greater protection against cell death compared with HUVECs. Moreover, EPDCs exhibit more hematopoietic supportive activity than HUVECs. Finally, studies in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice demonstrated that human circulating EPCs are able to colonize a Matrigel plug. EPDCs display the morphology and phenotype of endothelial cells. Their functional features indicate, however, that although these cells have undergone some differentiation steps, they still have the properties of immature cells, suggesting greater tissue repair capabilities. Future use of in vitro amplified peripheral blood EPDCs may constitute a challenging strategy for cell therapy.
- Published
- 2004
- Full Text
- View/download PDF
41. Fetal liver stroma consists of cells in epithelial-to-mesenchymal transition.
- Author
-
Chagraoui J, Lepage-Noll A, Anjo A, Uzan G, and Charbord P
- Subjects
- Animals, Biomarkers, Blotting, Western, Cell Differentiation, Cell Line drug effects, Epithelial Cells cytology, Gestational Age, Hematopoiesis, Extramedullary, Hepatocytes cytology, Humans, Liver cytology, Mesoderm cytology, Mice, Mice, Inbred C57BL, Microscopy, Electron, Microscopy, Fluorescence, Oncostatin M, Peptides pharmacology, Phenotype, Reverse Transcriptase Polymerase Chain Reaction, Stromal Cells cytology, Stromal Cells drug effects, Liver embryology
- Abstract
Liver becomes the predominant site of hematopoiesis by 11.5 dpc (days after coitus) in the mouse and 15 gestational weeks in humans and stays so until the end of gestation. The reason the liver is the major hematopoietic site during fetal life is not clear. In this work, we tried to define which of the fetal liver microenvironmental cell populations would be associated with the development of hematopoiesis and found that a population of cells with mixed endodermal and mesodermal features corresponded to hematopoietic-supportive fetal liver stroma. Stromal cells generated from primary cultures or stromal lines from mouse or human fetal liver in the hematopoietic florid phase expressed both mesenchymal markers (vimentin, osteopontin, collagen I, alpha smooth muscle actin, thrombospondin-1, EDa fibronectin, calponin, Stro-1 antigens, myocyte-enhancer factor 2C) and epithelial (alpha-fetoprotein, cytokeratins 8 and 18, albumin, E-cadherin, hepatocyte nuclear factor 3 alpha) markers. Such a cell population fits with the description of cells in epithelial-to-mesenchymal transition (EMT), often observed during development, including that of the liver. The hematopoietic supportive capacity of EMT cells was lost after hepatocytic maturation, induced by oncostatin M in the cell line AFT024. EMT cells were observed in the fetal liver microenvironment during the hematopoietic phase but not in nonhematopoietic liver by the end of gestation and in the adult. EMT cells represent a novel stromal cell type that may be generated from hepatic endodermal or mesenchymal stem cells or even from circulating hematopoietic stem cells (HSCs) seeding the liver rudiment.
- Published
- 2003
- Full Text
- View/download PDF
42. Forced expression of p21 in GPIIb-p21 transgenic mice induces abnormalities in the proliferation of erythroid and megakaryocyte progenitors and primitive hematopoietic cells.
- Author
-
Albanese P, Chagraoui J, Charon M, Cocault L, Dusanter-Fourt I, Romeo PH, and Uzan G
- Subjects
- Animals, Blood Cell Count, Cell Differentiation, Cell Division, Cell Lineage, Colony-Forming Units Assay, Cyclin-Dependent Kinase Inhibitor p21, Cyclins biosynthesis, Cyclins genetics, DNA, Complementary genetics, Erythroid Precursor Cells cytology, Erythroid Precursor Cells metabolism, Gene Expression Regulation, Genes, Synthetic, Hematopoiesis genetics, Hematopoietic Stem Cells metabolism, Homeostasis, Humans, Megakaryocytes cytology, Megakaryocytes metabolism, Mice, Mice, Transgenic, Recombinant Fusion Proteins physiology, Cyclins physiology, Hematopoiesis physiology, Hematopoietic Stem Cells cytology, Platelet Membrane Glycoprotein IIb genetics, Promoter Regions, Genetic
- Abstract
Objective: p21(WAF1/Cip/kip) and p27(Kip1) are cyclin-dependant kinase inhibitors controlling cell-cycle exit and differentiation of numerous cell types. Among hematopoietic cells, megakaryocytes express high levels of p21, while in erythroid cells, p27(Kip1) is predominant. As p21 and p27 could display overlapping functions and as megakaryocytes and erythroid cells derive from a bipotent progenitor, we developed an in vivo model to determine the specific role of p21 in controlling the proliferation/differentiation balance of erythroid and megakaryocytic progenitors., Methods: Transgenic mice that overexpressed p21 under the control of the human GPIIb promoter in early progenitors and along megakaryocytic differentiation were generated. Different subsets of hematopoietic progenitors (BFU and CFU) and primitive cells (CAFC, LTC-IC) were analyzed by methylcellulose assay. Phenotypic evolution and clonogenic properties of the lin(-) population were analyzed along erythroid and megakaryocytic differentiation., Results: We observed p21 ectopic expression in early hematopoietic progenitors (lin(-)Sca(+)), megakaryocytes, and, to a lesser extent, erythroid cells. This expression induced an important decrease in the number of CFU-MK, BFU-E, CFU-E, primitive progenitors (CAFC day 35), and LTC-IC, but did not affect the maturation process of these cells and the blood cell count., Conclusions: We show that variation of p21 expression level changes the fate of hematopoietic cells by favoring either proliferation or differentiation pathways. This effect of p21 is exerted not only at the level of primitive progenitors but also in more mature progenitors. However, in vivo, a systemic compensation mechanism is most likely activated in response to variations of the flow of progenitor production.
- Published
- 2002
- Full Text
- View/download PDF
43. Ontogenic emergence of the hematon, a morphogenetic stromal unit that supports multipotential hematopoietic progenitors in mouse bone marrow.
- Author
-
Blazsek I, Chagraoui J, and Péault B
- Subjects
- Animals, B-Lymphocytes cytology, Bone Development physiology, Bone Marrow embryology, Bone Marrow growth & development, Bone Marrow physiology, Cell Adhesion physiology, Cell Aggregation physiology, Cell Communication physiology, Cell Culture Techniques methods, Cell Differentiation physiology, Embryo, Mammalian, Femur anatomy & histology, Hematopoiesis physiology, Hematopoietic Stem Cells physiology, Immunohistochemistry, Immunophenotyping, Mice, Mice, Inbred C57BL, Morphogenesis, Stromal Cells physiology, Bone Marrow Cells cytology, Hematopoietic Stem Cells cytology, Stromal Cells cytology
- Abstract
Development of the full repertoire of hematopoietic-lymphopoietic cells from a single stem cell requires specific contacts with stromal cells. The spatio-temporal organization of these cell associations in the bone marrow in ontogeny is, however, not well understood. In the adult, 10% of marrow cells form a cohort of compact aggregates, the hematon. In the hematon mesenchymal cells (Stro-1(+)), perivascular lipocytes (desmin(+)), endothelial cells (CD34(+), Flk-1(+), Sca-1(+)), and macrophages amalgamate with the hematopoietic progenitors long-term culture-initiating cells (LTC-IC), cobblestone area-forming cell (CAFC), high-proliferative-potential colony-forming unit (HPP-CFU), granulocyte-macrophage (GM)-CFU, and burst-forming unit-erythroid (BFU-E). During endochondral ossification of the femur, GM-CFU and day 7 CAFC numbers increased progressively from day 17 of gestation, but primitive, day 35 LTC-IC appeared from postnatal day 2. Unexpectedly, bone marrow (BM) taken between embryonic day 17 and day 5 was unable to support myeloid cell production in long-term cultures or to support day 35 LTC-IC growth. However, a gain in stromal cell competence occurred between days 7 and 10, which was correlated with the emergence of hematon in the BM. Thus, acquisition of hematopoietic competence by BM lags behind for approximately 10 days after the initial hematopoietic cell influx. In the adult, the hematon fraction was 3.7-fold enriched in day 35 LTC-IC over the buffy coat. It produced more GM-CFU and HPP-CFU in myeloid culture and more B cells in lymphopoietic "switch" cultures. It is reported that stromal hematopoietic units named hematons are specific morphogenetic structures that emerge at a well-defined postnatal stage of development in long bones, delineate discrete territories for hematopoietic stem cell seeding and development, embody the most productive hematogenous compartment in the BM, and probably enclose a morphogenetic organizer. (Blood. 2000;96:3763-3771)
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.