26 results on '"A. DEMEYER"'
Search Results
2. Single-cell DNA amplicon sequencing reveals clonal heterogeneity and evolution in T-cell acute lymphoblastic leukemia
- Author
-
Albertí-Servera, Llucia, Demeyer, Sofie, Govaerts, Inge, Swings, Toon, De Bie, Jolien, Gielen, Olga, Brociner, Marco, Michaux, Lucienne, Maertens, Johan, Uyttebroeck, Anne, De Keersmaecker, Kim, Boeckx, Nancy, Segers, Heidi, and Cools, Jan
- Published
- 2021
- Full Text
- View/download PDF
3. Suz12 inactivation cooperates with JAK3 mutant signaling in the development of T-cell acute lymphoblastic leukemia
- Author
-
Broux, Michael, Prieto, Cristina, Demeyer, Sofie, Vanden Bempt, Marlies, Alberti-Servera, Llucia, Lodewijckx, Inge, Vandepoel, Roel, Mentens, Nicole, Gielen, Olga, Jacobs, Kris, Geerdens, Ellen, Vicente, Carmen, de Bock, Charles E., and Cools, Jan
- Published
- 2019
- Full Text
- View/download PDF
4. Hedgehog pathway activation in T-cell acute lymphoblastic leukemia predicts response to SMO and GLI1 inhibitors
- Author
-
Dagklis, Antonis, Demeyer, Sofie, De Bie, Jolien, Radaelli, Enrico, Pauwels, Daphnie, Degryse, Sandrine, Gielen, Olga, Vicente, Carmen, Vandepoel, Roel, Geerdens, Ellen, Uyttebroeck, Anne, Boeckx, Nancy, de Bock, Charles E., and Cools, Jan
- Published
- 2016
- Full Text
- View/download PDF
5. A Noncoding Germline Variant Creates a Gain-of-Function De Novo Enhancer That up-Regulates IL5 Transcription to Cause Familial Hypereosinophilia, Revealing a Previously Undescribed Mechanism for Human Disease
- Author
-
Kim, Jung-Hyun, primary, Bowman, Abbie, additional, Babu, Senbagavalli, additional, Cools, Jan, additional, Demeyer, Sofie, additional, Kazemian, Majid, additional, Leonard, Warren, additional, Lin, Jian-Xin, additional, Makiya, Michelle, additional, Perez, Mathieu, additional, Sweeney, Colin L., additional, Franco, Luis M, additional, Redekar, Neelam, additional, Lack, Justin, additional, Resar, Linda, additional, and Klion, Amy D, additional
- Published
- 2022
- Full Text
- View/download PDF
6. JAK3 mutants transform hematopoietic cells through JAK1 activation, causing T-cell acute lymphoblastic leukemia in a mouse model
- Author
-
Degryse, Sandrine, de Bock, Charles E., Cox, Luk, Demeyer, Sofie, Gielen, Olga, Mentens, Nicole, Jacobs, Kris, Geerdens, Ellen, Gianfelici, Valentina, Hulselmans, Gert, Fiers, Mark, Aerts, Stein, Meijerink, Jules P., Tousseyn, Thomas, and Cools, Jan
- Published
- 2014
- Full Text
- View/download PDF
7. Aberrant MYCN expression drives oncogenic hijacking of EZH2 as a transcriptional activator in peripheral T-cell lymphoma
- Author
-
Marlies Vanden Bempt, Koen Debackere, Sofie Demeyer, Quentin Van Thillo, Nienke Meeuws, Cristina Prieto, Sarah Provost, Nicole Mentens, Kris Jacobs, Olga Gielen, David Nittner, Seishi Ogawa, Keisuke Kataoka, Carlos Graux, Thomas Tousseyn, Jan Cools, Daan Dierickx, UCL - SSS/IREC/MONT - Pôle Mont Godinne, and UCL - (MGD) Service d'oncologie médicale
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of hematological cancers arising from the malignant transformation of mature T cells. In a cohort of 28 PTCL cases, we identified recurrent overexpression of MYCN, a member of the MYC family of oncogenic transcription factors. Approximately half of all PTCL cases was characterized by a MYC expression signature. Inducible expression of MYCN in lymphoid cells in a mouse model caused T-cell lymphoma that recapitulated human PTCL with an MYC expression signature. Integration of mouse and human expression data identified EZH2 as a key downstream target of MYCN. Remarkably, EZH2 was found to be an essential cofactor for the transcriptional activation of the MYCN-driven gene expression program, which was independent of methyltransferase activity but dependent on phosphorylation by CDK1. MYCN-driven T-cell lymphoma was sensitive to EZH2 degradation or CDK1 inhibition, which displayed synergy with US Food and Drug Administration-approved histone deacetylase (HDAC) inhibitors. ispartof: BLOOD vol:140 issue:23 pages:2463-2476 ispartof: location:United States status: published
- Published
- 2022
8. A Noncoding Germline Variant Creates a Gain-of-Function De Novo Enhancer That up-Regulates IL5 Transcription to Cause Familial Hypereosinophilia, Revealing a Previously Undescribed Mechanism for Human Disease
- Author
-
Jung-Hyun Kim, Abbie Bowman, Senbagavalli Babu, Jan Cools, Sofie Demeyer, Majid Kazemian, Warren Leonard, Jian-Xin Lin, Michelle Makiya, Mathieu Perez, Colin L. Sweeney, Luis M Franco, Neelam Redekar, Justin Lack, Linda Resar, and Amy D Klion
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
9. SOCS1 Haploinsufficiency Presenting As Incidental Refractory Thrombocytopenia in a Pediatric Patient with Inflammatory Symptoms and History of Sars Cov-2 Infection
- Author
-
Lapinski, Jillian, primary, Ngo, Sandra Hoang, additional, Lee, Pui Y, additional, Walkovich, Kelly J., additional, Hannibal, Mark, additional, DeMeyer, Lauren, additional, Mohan, Smriti, additional, and Michniacki, Thomas F., additional
- Published
- 2021
- Full Text
- View/download PDF
10. Clonal Heterogeneity and Evolution during Treatment in High Hyperdiploid B-Cell Acute Lymphoblastic Leukemia
- Author
-
Aertgeerts, Margo, Demeyer, Sofie, Lamote, Jochen, De Bie, Jolien, Michaux, Lucienne, Cools, Jan, and Segers, Heidi
- Published
- 2023
- Full Text
- View/download PDF
11. Overexpression of wild-type IL-7Rα promotes T-cell acute lymphoblastic leukemia/lymphoma
- Author
-
Silva, Ana, primary, Almeida, Afonso R. M., additional, Cachucho, Ana, additional, Neto, João L., additional, Demeyer, Sofie, additional, de Matos, Mafalda, additional, Hogan, Thea, additional, Li, Yunlei, additional, Meijerink, Jules, additional, Cools, Jan, additional, Grosso, Ana Rita, additional, Seddon, Benedict, additional, and Barata, João T., additional
- Published
- 2021
- Full Text
- View/download PDF
12. Mutant JAK3 signaling is increased by loss of wild-type JAK3 or by acquisition of secondary JAK3 mutations in T-ALL
- Author
-
Sandrine Degryse, Simon Bornschein, Jean Soulier, Emilie Leroy, Ellen Geerdens, Marlies Vanden Bempt, Sofie Demeyer, Charles E. de Bock, Jan Cools, Kris Jacobs, Stefan N. Constantinescu, Olga Gielen, Christine J. Harrison, and UCL - SSS/DDUV/SIGN - Cell signalling
- Subjects
Models, Molecular ,0301 basic medicine ,Immunology ,Mutant ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,Mutation Rate ,Cell Line, Tumor ,medicine ,Humans ,Point Mutation ,Alleles ,STAT5 ,Genetics ,Mutation ,Lymphoid Neoplasia ,Janus kinase 3 ,Point mutation ,Wild type ,Janus Kinase 3 ,Cell Biology ,Hematology ,medicine.disease ,Molecular biology ,Leukemia ,030104 developmental biology ,biology.protein ,Janus kinase ,Signal Transduction - Abstract
The Janus kinase 3 (JAK3) tyrosine kinase is mutated in 10% to 16% of T-cell acute lymphoblastic leukemia (T-ALL) cases. JAK3 mutants induce constitutive JAK/STAT signaling and cause leukemia when expressed in the bone marrow cells of mice. Surprisingly, we observed that one third of JAK3-mutant T-ALL cases harbor 2 JAK3 mutations, some of which are monoallelic and others that are biallelic. Our data suggest that wild-type JAK3 competes with mutant JAK3 (M511I) for binding to the common γ chain and thereby suppresses its oncogenic potential. We demonstrate that JAK3 (M511I) can increase its limited oncogenic potential through the acquisition of an additional mutation in the mutant JAK3 allele. These double JAK3 mutants show increased STAT5 activation and increased potential to transform primary mouse pro-T cells to interleukin-7-independent growth and were not affected by wild-type JAK3 expression. These data extend our insight into the oncogenic properties of JAK3 mutations and provide an explanation of why progression of JAK3-mutant T-ALL cases can be associated with the accumulation of additional JAK3 mutations.
- Published
- 2018
13. SOCS1 Haploinsufficiency Presenting As Incidental Refractory Thrombocytopenia in a Pediatric Patient with Inflammatory Symptoms and History of Sars Cov-2 Infection
- Author
-
Smriti Mohan, Sandra Hoang Ngo, Thomas F. Michniacki, Kelly Walkovich, Pui Y. Lee, Mark C. Hannibal, Lauren DeMeyer, and Jillian Lapinski
- Subjects
311.Disorders of Platelet Number or Function: Clinical and Epidemiological ,Pediatrics ,medicine.medical_specialty ,Suppressor of cytokine signaling 1 ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Pediatric patient ,Medicine ,Refractory Thrombocytopenia ,business ,Haploinsufficiency - Abstract
BACKGROUND: Immune thrombocytopenia purpura (ITP) has a complex pathogenesis and may be a primary diagnosis or secondary to an underlying condition 1. Evaluation for underlying diagnoses in patients presenting with atypical features of classic ITP is key, as this can impact treatment decisions, therapy response, and prognosis. Genetic variants that predispose patients to ITP are especially important to investigate as patients may be at risk for additional autoimmune phenomenon or malignancy. The SARS CoV-2 pandemic has added further complexity as reports suggest the infection can lead to autoimmunity in those with genetic predispositions 2,3. Loss of the suppressor of cytokine signaling 1 (SOCS1) function has been described to manifest with autoinflammatory syndrome, with or without immunodeficiency 4,5. Reports of autoimmunity developing in patients with SOCS1 haploinsufficiency after SARS CoV-2 infection are documented, including multi-system inflammatory syndrome (MIS-C) 2. A proposed mechanism of this virus-triggered autoimmunity includes a transient innate and adaptive immunodeficiency 3. This raises the question whether patients harboring genetic variants with risk of autoimmunity are placed at an even higher risk for ITP in the wake of SARS-CoV2 infection. CASE PRESENTATION: We present a 6-year-old female with isolated thrombocytopenia of 4,000/uL identified during evaluation for severe arthralgias unresponsive to corticosteroid treatment (maximum dose 1mg/kg/day) over a 6-month period. Laboratory results at presentation were consistent with ITP, including presence of platelet autoantibodies. Evaluation revealed hypocellularity for age (~50%) on bone marrow evaluation as well as elevated IgE (2080 kU/L), with IgA, IgM, and IgG levels within reference range. She had a remote history of SARS CoV-2-like illness and SARS CoV-2 antibodies were found present in serologic assay, without a history of vaccination. Genetic testing, including chromosomal microarray from peripheral blood and marrow, was included in the diagnostic workup given concern for a history of developmental delays with macrocephaly and necessity to rule-out malignancy with the patient noted to have a 5 mega-base deletion at 16p13.2p13.11, which includes the SOCS1 gene. Comprehensive next generation sequencing for additional immune dysregulation/primary immunodeficiency associated variants was unremarkable. Functional studies of surface expression of interferon-inducible genes (CD169 (SIGLEC-1)) and STAT1 phosphorylation via analysis of CD14+ monocytes revealed excess interferon signaling previously described in patients with SOCS1 haploinsufficiency (Figure 1). Measurements of B-cell-activating factor were also found to be extremely elevated at 6432 pg/mL. The patient's ITP course was complicated by hematuria, melena and refractory platelet response to first line therapy consisting of intravenous immunoglobulin 1 g/kg x2 doses and 2 mg/kg/day prednisolone. She required escalation to high dose methylprednisolone (30mg/kg), rituximab 375 mg/m 2/weekly x4 doses, and concurrent romiplostim (2 doses) for control of thrombocytopenia and bleeding manifestations. Her rheumatologic symptoms subsided with initiation of corticosteroids, and she has subsequently completed a prolonged corticosteroid taper. She currently has a normal platelet count with non-steroidal anti-inflammatory therapy utilized for arthralgia management with plan to transition to JAK inhibition for maintenance therapy. CONCLUSION: This case highlights the potential impact of investigating for susceptibility genes for ITP with consideration for broader testing including targeted next generation sequencing panels or microarray analysis in patients with atypical ITP presentations or response to therapy, as knowledge of this patient's underlying genetics led to earlier treatment and use of alternative agents. Additionally, the case adds the novel finding of bone marrow hypocellularity to the clinical phenotype of SOCS1 haploinsufficiency, as this has not yet been reported and contributes to the literature on the relationship of autoimmunity and SARS CoV-2 infections in patients with predisposing genetic variants. Figure 1 Figure 1. Disclosures Walkovich: Horizon Pharmaceuticals: Honoraria, Membership on an entity's Board of Directors or advisory committees; Pharming: Honoraria, Membership on an entity's Board of Directors or advisory committees; Swedish Orphan Biovitrum AB (Sobi): Consultancy, Honoraria; X4 Pharmaceuticals: Other: Local PI for clinical trial involving mavorixafor and patients with neutropenia.
- Published
- 2021
14. Overexpression of wild-type IL-7Rα promotes T-cell acute lymphoblastic leukemia/lymphoma
- Author
-
Thea Hogan, João Luís Neto, Benedict Seddon, Sofie Demeyer, Ana Patricia Silva, Yunlei Li, Mafalda Matos, Ana Rita Grosso, Afonso R. M. Almeida, Jules P.P. Meijerink, João T. Barata, Jan Cools, Ana Cachucho, Pathology, DCV - Departamento de Ciências da Vida, UCIBIO - Applied Molecular Biosciences Unit, and Repositório da Universidade de Lisboa
- Subjects
Oncology ,medicine.medical_specialty ,Carcinogenesis ,T cell ,Immunology ,Mice, Transgenic ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,Interleukin-7 Receptor alpha Subunit ,03 medical and health sciences ,Mice ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,media_common.cataloged_instance ,Animals ,Humans ,European union ,030304 developmental biology ,media_common ,0303 health sciences ,Acute lymphoblastic leukemia-lymphoma ,Lymphoid Neoplasia ,Receptors, Interleukin-7 ,Thymocytes ,business.industry ,Gene Expression Regulation, Leukemic ,European research ,Cell Biology ,Hematology ,Neoplasms, Experimental ,3. Good health ,Neoplasm Proteins ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,business ,Protein overexpression ,Signal Transduction - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is often not cured by frontline chemotherapy, and efforts to improve treatment by targeting oncogenes such as NOTCH1 have been hampered by toxicity. Silva and colleagues studied primary patient samples to show that high-level interleukin 7 receptor α (IL7Rα) gene expression correlates with ongoing, oncogenic IL7R-mediated signaling. Using new in vivo models, they characterized the impact of IL7Rα expression on the pathogenesis of T-ALL and its response to various targeted therapies that reduce IL7-related signaling., Key Points Mice overexpressing IL-7Rα develop leukemia with features of human T-ALL and sensitivity to ruxolitinib, dactolisib, and venetoclax.T-ALL patients with high levels of wild-type IL7R present with evidence of ongoing, oncogenic-like IL-7R–mediated activation of signaling., Visual Abstract, Tight regulation of IL-7Rα expression is essential for normal T-cell development. IL-7Rα gain-of-function mutations are known drivers of T-cell acute lymphoblastic leukemia (T-ALL). Although a subset of patients with T-ALL display high IL7R messenger RNA levels and cases with IL7R gains have been reported, the impact of IL-7Rα overexpression, rather than mutational activation, during leukemogenesis remains unclear. In this study, overexpressed IL-7Rα in tetracycline-inducible Il7r transgenic and Rosa26 IL7R knockin mice drove potential thymocyte self-renewal, and thymus hyperplasia related to increased proliferation of T-cell precursors, which subsequently infiltrated lymph nodes, spleen, and bone marrow, ultimately leading to fatal leukemia. The tumors mimicked key features of human T-ALL, including heterogeneity in immunophenotype and genetic subtype between cases, frequent hyperactivation of the PI3K/Akt pathway paralleled by downregulation of p27Kip1 and upregulation of Bcl-2, and gene expression signatures evidencing activation of JAK/STAT, PI3K/Akt/mTOR and Notch signaling. Notably, we also found that established tumors may no longer require high levels of IL-7R expression upon secondary transplantation and progressed in the absence of IL-7, but remain sensitive to inhibitors of IL-7R–mediated signaling ruxolitinib (Jak1), AZD1208 (Pim), dactolisib (PI3K/mTOR), palbociclib (Cdk4/6), and venetoclax (Bcl-2). The relevance of these findings for human disease are highlighted by the fact that samples from patients with T-ALL with high wild-type IL7R expression display a transcriptional signature resembling that of IL-7–stimulated pro-T cells and, critically, of IL7R-mutant cases of T-ALL. Overall, our study demonstrates that high expression of IL-7Rα can promote T-cell tumorigenesis, even in the absence of IL-7Rα mutational activation.
- Published
- 2019
15. Mutant JAK3 signaling is increased by loss of wild-type JAK3 or by acquisition of secondary JAK3 mutations in T-ALL
- Author
-
Degryse, Sandrine, Bornschein, Simon, de Bock, Charles E., Leroy, Emilie, Vanden Bempt, Marlies, Demeyer, Sofie, Jacobs, Kris, Geerdens, Ellen, Gielen, Olga, Soulier, Jean, Harrison, Christine J., Constantinescu, Stefan N., and Cools, Jan
- Published
- 2018
- Full Text
- View/download PDF
16. Evolution of Clinically Relevant Subclones during Chemotherapy Treatment of ALL As Determined By Single-Cell DNA and RNA Sequencing
- Author
-
Albertí Servera, Llucia, primary, Demeyer, Sofie, additional, Govaerts, Inge, additional, Gielen, Olga, additional, Segers, Heidi, additional, Uyttebroeck, Anne, additional, Boeckx, Nancy, additional, Maertens, Johan A., additional, De Keersmaecker, Kim, additional, and Cools, Jan, additional
- Published
- 2019
- Full Text
- View/download PDF
17. Evolution of Clinically Relevant Subclones during Chemotherapy Treatment of ALL As Determined By Single-Cell DNA and RNA Sequencing
- Author
-
Llucia Albertí Servera, Nancy Boeckx, Kim De Keersmaecker, Olga Gielen, Inge Govaerts, Anne Uyttebroeck, Sofie Demeyer, Jan Cools, Heidi Segers, and Johan Maertens
- Subjects
Immunology ,Cancer ,Genomics ,Cell Biology ,Hematology ,Computational biology ,Biology ,medicine.disease ,Biochemistry ,Chemotherapy regimen ,Genome ,Gene expression profiling ,Leukemia ,Acute lymphocytic leukemia ,medicine ,Gene - Abstract
Acute lymphoblastic leukemia (ALL), which is the most common cancer in children, shows extensive genetic intra-tumoral heterogeneity. This heterogeneity might be the underlying reason for an incomplete response to treatment and for the development of relapse. In order to envision the clinical implementation of a refined risk-category strategy based on ALL subclonal composition, it is essential to first generate a reference single-cell map and accumulate evidence on how the subclonal composition affects the response to treatment. For that, we performed large-scale and integrative single-cell genome and transcriptome profiling of pediatric samples at diagnosis, during drug treatment and in case of relapse. We used the 10x Genomics platform for single-cell RNA-sequencing analysis (around 4000 cells per sample) and the Tapestri Platform (Mission Bio) for targeted single-cell DNA-sequencing (around 5000 cells per sample) of the most mutated genomic regions in ALL. For the later, we developed a custom panel that covers 305 ALL mutational hotspots across 110 genes. We have determined a reference single-cell map of the cellular (based on the gene expression profile) and the clonal composition (based on the co-occurrence of mutations at each individual cell) for pediatric ALL at diagnosis (8 T-ALL and 10 B-ALL patients). We have also reconstructed the tumor phylogeny highlighting the order of mutational acquisition and the most likely pattern of evolution. Moreover, we have studied how T-ALL evolves during drug treatment at single-cell resolution in 4 patients, unraveling which are the most sensitive clones to the therapy, which are the most resistant ones and when relapse clones originated. Single-cell RNA-sequencing also provided information on how normal hematopoiesis recovers during chemotherapy treatment. The results show that ALL is typically composed by a major clone and 5-10 smaller clones that have different sensitivities to the therapy. We have been able to detect minor clones ( Disclosures Maertens: Gilead Sciences: Other: Grants, personal fees and non-financial support; Cidara: Other: Personal fees and non-financial support; Amplyx: Other: Personal fees and non-financial support; F2G: Other: Personal fees and non-financial support; Merck: Other: Personal fees and non-financial support; Pfizer: Other: Grant and personal fees; Astellas Pharma: Other: Personal fees and non-financial support.
- Published
- 2019
18. Hedgehog pathway activation in T-cell acute lymphoblastic leukemia predicts response to SMO and GLI1 inhibitors
- Author
-
Sofie Demeyer, Antonis Dagklis, Nancy Boeckx, Charles E. de Bock, Sandrine Degryse, Jan Cools, Ellen Geerdens, Anne Uyttebroeck, Jolien De Bie, Roel Vandepoel, Daphnie Pauwels, Carmen Vicente, Olga Gielen, and Enrico Radaelli
- Subjects
0301 basic medicine ,Male ,animal structures ,Indian hedgehog ,Pyridines ,Immunology ,Vismodegib ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,Zinc Finger Protein GLI1 ,03 medical and health sciences ,Mice ,GLI1 ,medicine ,Animals ,Humans ,Anilides ,Hedgehog Proteins ,Sonic hedgehog ,Hedgehog ,biology ,Gene Expression Regulation, Leukemic ,Cell Biology ,Hematology ,biology.organism_classification ,Smoothened Receptor ,Xenograft Model Antitumor Assays ,Hedgehog signaling pathway ,Neoplasm Proteins ,030104 developmental biology ,Pyrimidines ,embryonic structures ,Cancer research ,biology.protein ,Ectopic expression ,Female ,Smoothened ,medicine.drug ,Signal Transduction - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive childhood leukemia that is caused by the accumulation of multiple genomic lesions resulting in transcriptional deregulation and increased cell proliferation and survival. Through analysis of gene expression data, we provide evidence that the hedgehog pathway is activated in 20% of T-ALL samples. Hedgehog pathway activation is associated with ectopic expression of the hedgehog ligands Sonic hedgehog (SHH) or Indian hedgehog (IHH), and with upregulation of the transcription factor GLI1 Ectopic expression of SHH or IHH in mouse T cells in vivo caused hedgehog pathway activation in both lymphoid and epithelial cells in the thymus and resulted in increased expression of important T-cell stimulatory ligands (Dll4, Il7, and Vegf) by thymic epithelial cells. In T-ALL cell lines, pharmacological inhibition or short interfering RNA-mediated knockdown of SMO or GLI1 led to decreased cell proliferation. Moreover, primary T-ALL cases with high GLI1 messenger RNA levels, but not those with low or undetectable GLI1 expression, were sensitive to hedgehog pathway inhibition by GANT61 or GDC-0449 (vismodegib) using ex vivo cultures and in vivo xenograft models. We identify the hedgehog pathway as a novel therapeutic target in T-ALL and demonstrate that hedgehog inhibitors approved by the US Food and Drug Administration could be used for the treatment of this rare leukemia.
- Published
- 2016
19. Use of Crispr/Cas Genome Editing in Ba/F3 Cells to Generate the Fip1l1-Pdgfra and Nup214-Abl1 Fusion Genes
- Author
-
Vanden Bempt, Marlies, primary, de Bock, Charles E, additional, Mentens, Nicole, additional, Gielen, Olga, additional, Geerdens, Ellen, additional, Demeyer, Sofie, additional, and Cools, Jan, additional
- Published
- 2014
- Full Text
- View/download PDF
20. Synergism Between HOXA9 and Mutant JAK3 (M511I) Leads to Rapid Leukemia Development within an in Vivo Murine Bone Marrow Transplant Model
- Author
-
de Bock, Charles E, primary, Degryse, Sandrine, additional, Demeyer, Sofie, additional, Sweron, Bram, additional, Gielen, Olga, additional, Mentens, Nicole, additional, Geerdens, Ellen, additional, and Cools, Jan, additional
- Published
- 2014
- Full Text
- View/download PDF
21. Use of Crispr/Cas Genome Editing in Ba/F3 Cells to Generate the Fip1l1-Pdgfra and Nup214-Abl1 Fusion Genes
- Author
-
Jan Cools, Marlies Vanden Bempt, Ellen Geerdens, Olga Gielen, Charles E. de Bock, Nicole Mentens, and Sofie Demeyer
- Subjects
ABL ,Cas9 ,Immunology ,Cell Biology ,Hematology ,PDGFRA ,Biology ,Biochemistry ,Fusion protein ,Molecular biology ,Fusion gene ,Genome editing ,CRISPR ,Gene - Abstract
CRISPR/Cas genome editing is a powerful tool to precisely induce chromosomal breaks and to modify genes of interest. Cas9, an RNA-guided DNA endonuclease derived from Streptococcus pyogenes, is able to generate double stranded breaks (DSBs) in the genomic locus to where it is directed by its guide RNA (gRNA) component. The DSBs are subsequently repaired by one of the two main host repair mechanisms: the error-prone Non-homologous end joining (NHEJ) pathway or the very specific Homology-directed repair (HDR) pathway. We aimed to use CRISPR/Cas genome editing to generate the Fip1l1-Pdgfra and Nup214-Abl1 fusion genes by inducing chromosomal rearrangements in the interleukin-3 dependent Ba/F3 cell line. Prior to generating the chromosomal rearrangements, we optimized CRISPR/Cas genome editing in Ba/F3 cells, by targeting Cas9 to exon 24 of CD45, a cell surface transmembrane protein, of which inactivation can be easily detected by flow cytometry. Electroporation of Ba/F3 cells with plasmids expressing Cas9 and the specific guide RNA led to efficient inactivation of the CD45 gene, as measured by flow cytometry (30% of the cells showed loss of CD45 expression). The use of the Cas9 nickase variant led to an increased efficiency of CD45 inactivation with 58% of the cells showing loss of CD45 expression. We then extended these studies to assess the efficiency of homology-directed repair to introduce a specific mutation, using a single strand donor template to generate a premature stop codon in exon 24 of CD45. The successful introduction of the novel stop codon in CD45 was confirmed by PCR amplification of the targeted exon followed by massive parallel sequencing (MiSeq, Illumina) and we observed this endogenous mutation in 80% of the Ba/F3 clones. Having optimised the use and efficiency of CRISPR/Cas in Ba/F3 cells, we aimed to introduce double stranded breaks simultaneously in the genes Fip1l1 and Pdgfra to generate a cell based model for the FIP1L1-PDGFRA fusion gene as observed in chronic eosinophilic leukemia. Double strand breaks were introduced in Fip1l1 exon 23, 31, 32 or 34 together with simultaneous breaks in Pdgfra exon 12, both located on mouse chromosome 5. Upon IL3 removal, cells harbouring the deletion and fusion gene were able to survive, grow and form colonies in semi-solid medium, as was shown before for Ba/F3 cells transduced with retroviral vectors expressing FIP1L1-PDGFRA. The presence of the deletion was confirmed by PCR, and fusion protein expression was detected by Western blotting. A fusion between exon 1 of Fip1l1 and exon 12 of Pdgfra could also transform the cells, which confirmed earlier findings that the transforming capacities of the fusion protein are independent of Fip1l1 and dependent on the interruption of the juxtamembrane region of PDGFRA. The expression and phosphorylation levels of Fip1l1-Pdgfra were compared between the CRISPR/Cas generated Ba/F3 cells and retrovirally transduced cells overexpressing FIP1L1-PDGFRA. As expected, retrovirally transduced cells showed a much higher protein expression level of FIP1L1-PDGFRA, and much stronger phosphorylation compared to the CRISPR/Cas generated cells, in which the endogenous Fip1l1 promoter is used to drive the expression of the fusion protein. We also observed a difference in sensitivity to inhibition by imatinib, a kinase inhibitor with strong activity against PDGFRA. The same strategy was followed to generate a fusion between Nup214 and Abl1, as observed in a subset of T-cell acute lymphoblastic leukemia cases. Ba/F3 cells harbouring the Nup214-Abl1 fusion gene were able to survive and grow independent of IL3. The presence of the fusion gene was confirmed by PCR, and fusion protein expression was detected by Western blotting. Taken together, these data show that CRISPR/Cas induced chromosomal translocations in cells more faithfully recapitulate gene expression levels and sensitivity to chemotherapeutics when compared to retroviral transduction based expression of an oncogene. In conclusion, we have now designed and implemented an optimised platform to use CRISPR/Cas genome editing in Ba/F3 cells and measure gRNA efficacy by massive parallel sequencing. Our data confirm that the CRISPR/Cas genome editing system can be used to generate chromosomal rearrangements in Ba/F3 cells and provides a method to generate improved cell based models for the study of oncogenic tyrosine kinases. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
22. Synergism Between HOXA9 and Mutant JAK3 (M511I) Leads to Rapid Leukemia Development within an in Vivo Murine Bone Marrow Transplant Model
- Author
-
Sofie Demeyer, Nicole Mentens, Jan Cools, Charles E. de Bock, Sandrine Degryse, Bram Sweron, Olga Gielen, and Ellen Geerdens
- Subjects
Oncogene ,Immunology ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry ,Transplantation ,Leukemia ,Haematopoiesis ,medicine.anatomical_structure ,medicine ,Cancer research ,Bone marrow ,Stem cell ,Progenitor cell ,CD8 - Abstract
Activation mutations in JAK3 occur in 16% of T-cell acute lymphoblastic leukemia (T-ALL) cases, and co-occur frequently with HOXA cluster rearrangement. Genomic rearrangement of the HOXA cluster results in increased expression of HOXA9 and HOXA10. However it remains unclear if either HOXA9 or HOXA10 can cooperate with activating JAK3 mutations during oncogenic transformation and leukemogenesis. We have previously shown that JAK3 mutations lead to cell transformation and cause a long latency T-ALL in vivo using a mouse bone marrow transplant model. In this study we demonstrate that co-expression of the activating JAK3(M511I) protein with HOXA9 cooperate to develop leukemia within 30 days of transplant using an in vivo bone marrow transplant model. In our cooperative model, murine hematopoietic stem / progenitor cells were co-transduced with either both retroviral vectors encoding JAK3(M511I)/GFP and HOXA9/mCherry or each individually and then injected into sub-lethally irradiated recipient mice. Mice transplanted with bone marrow cells expressing JAK3(M511I) mutant alone developed T-ALL in 120 to 150 days. In sharp contrast, mice transplanted with cells expressing both JAK3(M511I) and HOXA9 showed rapid leukemia development within 30 days after transplant. Leukemia development was characterized by the rapid and specific increase in GFP-mCherry double positive cells. These animals showed high WBC, and splenomegaly and accumulation of immature CD8 single positive cells in the thymus. Similar experiments with HOXA10 did not show cooperation suggesting that HOXA9 is the more important oncogene in HOXA rearranged leukemias when a JAK3 activating mutation is present. To determine the underlying genetic mechanism for cooperation between HOXA9 and JAK3(M511I) the single positive JAK3 and double positive JAK3/HOXA9 expressing cells were isolated from thymi of leukemic mice for both epigenomic profiling using ATAC-seq and gene expression profiling. These analyses identified genetic pathways activated by the co-expression of HOXA9 and JAK3(M511I) mutation and provide mechanistic insight into the synergistic interaction between these two factors in driving leukemia development. Treatment of the animals with a JAK kinase inhibitor resulted in delayed leukemia development, confirming that the leukemia cells remain sensitive to the JAK inhibitor. This mouse model provides insight in the cooperation between oncogenes in leukemia development and provides a model for the study of targeted agents in this setting. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
23. A Noncoding Germline Variant Creates a Gain-of-Function De NovoEnhancer That up-Regulates IL5Transcription to Cause Familial Hypereosinophilia, Revealing a Previously Undescribed Mechanism for Human Disease
- Author
-
Kim, Jung-Hyun, Bowman, Abbie, Babu, Senbagavalli, Cools, Jan, Demeyer, Sofie, Kazemian, Majid, Leonard, Warren, Lin, Jian-Xin, Makiya, Michelle, Perez, Mathieu, Sweeney, Colin L., Franco, Luis M, Redekar, Neelam, Lack, Justin, Resar, Linda, and Klion, Amy D
- Published
- 2022
- Full Text
- View/download PDF
24. SOCS1Haploinsufficiency Presenting As Incidental Refractory Thrombocytopenia in a Pediatric Patient with Inflammatory Symptoms and History of Sars Cov-2 Infection
- Author
-
Lapinski, Jillian, Ngo, Sandra Hoang, Lee, Pui Y, Walkovich, Kelly J., Hannibal, Mark, DeMeyer, Lauren, Mohan, Smriti, and Michniacki, Thomas F.
- Abstract
Immune thrombocytopenia purpura (ITP) has a complex pathogenesis and may be a primary diagnosis or secondary to an underlying condition 1. Evaluation for underlying diagnoses in patients presenting with atypical features of classic ITP is key, as this can impact treatment decisions, therapy response, and prognosis. Genetic variants that predispose patients to ITP are especially important to investigate as patients may be at risk for additional autoimmune phenomenon or malignancy. The SARS CoV-2 pandemic has added further complexity as reports suggest the infection can lead to autoimmunity in those with genetic predispositions 2,3. Loss of the suppressor of cytokine signaling 1 (SOCS1) function has been described to manifest with autoinflammatory syndrome, with or without immunodeficiency 4,5. Reports of autoimmunity developing in patients with SOCS1haploinsufficiency after SARS CoV-2 infection are documented, including multi-system inflammatory syndrome (MIS-C) 2. A proposed mechanism of this virus-triggered autoimmunity includes a transient innate and adaptive immunodeficiency 3. This raises the question whether patients harboring genetic variants with risk of autoimmunity are placed at an even higher risk for ITP in the wake of SARS-CoV2 infection.
- Published
- 2021
- Full Text
- View/download PDF
25. Synergism Between HOXA9 and Mutant JAK3 (M511I) Leads to Rapid Leukemia Development within an in VivoMurine Bone Marrow Transplant Model
- Author
-
de Bock, Charles E, Degryse, Sandrine, Demeyer, Sofie, Sweron, Bram, Gielen, Olga, Mentens, Nicole, Geerdens, Ellen, and Cools, Jan
- Abstract
Activation mutations in JAK3 occur in 16% of T-cell acute lymphoblastic leukemia (T-ALL) cases, and co-occur frequently with HOXA cluster rearrangement. Genomic rearrangement of the HOXA cluster results in increased expression of HOXA9 and HOXA10. However it remains unclear if either HOXA9 or HOXA10 can cooperate with activating JAK3 mutations during oncogenic transformation and leukemogenesis. We have previously shown that JAK3 mutations lead to cell transformation and cause a long latency T-ALL in vivousing a mouse bone marrow transplant model. In this study we demonstrate that co-expression of the activating JAK3(M511I) protein with HOXA9 cooperate to develop leukemia within 30 days of transplant using an in vivo bone marrow transplant model. In our cooperative model, murine hematopoietic stem / progenitor cells were co-transduced with either both retroviral vectors encoding JAK3(M511I)/GFP and HOXA9/mCherry or each individually and then injected into sub-lethally irradiated recipient mice. Mice transplanted with bone marrow cells expressing JAK3(M511I) mutant alone developed T-ALL in 120 to 150 days. In sharp contrast, mice transplanted with cells expressing both JAK3(M511I) and HOXA9 showed rapid leukemia development within 30 days after transplant. Leukemia development was characterized by the rapid and specific increase in GFP-mCherry double positive cells. These animals showed high WBC, and splenomegaly and accumulation of immature CD8 single positive cells in the thymus. Similar experiments with HOXA10 did not show cooperation suggesting that HOXA9 is the more important oncogene in HOXA rearranged leukemias when a JAK3 activating mutation is present. To determine the underlying genetic mechanism for cooperation between HOXA9 and JAK3(M511I) the single positive JAK3 and double positive JAK3/HOXA9 expressing cells were isolated from thymi of leukemic mice for both epigenomic profiling using ATAC-seq and gene expression profiling. These analyses identified genetic pathways activated by the co-expression of HOXA9 and JAK3(M511I) mutation and provide mechanistic insight into the synergistic interaction between these two factors in driving leukemia development. Treatment of the animals with a JAK kinase inhibitor resulted in delayed leukemia development, confirming that the leukemia cells remain sensitive to the JAK inhibitor. This mouse model provides insight in the cooperation between oncogenes in leukemia development and provides a model for the study of targeted agents in this setting.
- Published
- 2014
- Full Text
- View/download PDF
26. Use of Crispr/Cas Genome Editing in Ba/F3 Cells to Generate the Fip1l1-Pdgfraand Nup214-Abl1Fusion Genes
- Author
-
Vanden Bempt, Marlies, de Bock, Charles E, Mentens, Nicole, Gielen, Olga, Geerdens, Ellen, Demeyer, Sofie, and Cools, Jan
- Abstract
CRISPR/Cas genome editing is a powerful tool to precisely induce chromosomal breaks and to modify genes of interest. Cas9, an RNA-guided DNA endonuclease derived from Streptococcus pyogenes, is able to generate double stranded breaks (DSBs) in the genomic locus to where it is directed by its guide RNA (gRNA) component. The DSBs are subsequently repaired by one of the two main host repair mechanisms: the error-prone Non-homologous end joining (NHEJ) pathway or the very specific Homology-directed repair (HDR) pathway. We aimed to use CRISPR/Cas genome editing to generate the Fip1l1-Pdgfra and Nup214-Abl1 fusion genes by inducing chromosomal rearrangements in the interleukin-3 dependent Ba/F3 cell line.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.