162 results on '"Crispino JD"'
Search Results
2. Molecular insights into Down syndrome-associated leukemia.
- Author
-
Vyas P, Crispino JD, Vyas, Paresh, and Crispino, John D
- Published
- 2007
- Full Text
- View/download PDF
3. Control of Megakaryocyte Expansion and Bone Marrow Fibrosis by Lysyl Oxidase*
- Author
-
Alexia Eliades, Hector A. Lucero, Katya Ravid, Kelley A. Burridge, Hillary Johnston-Cox, Ajoy Bhupatiraju, Philip C. Trackman, Anna Rita Migliaccio, John D. Crispino, Nikolaos Papadantonakis, Eliades, A, Papadantonakis, N, Bhupatiraju, A, Burridge, Ka, Johnston-Cox, Ha, FRANCO MIGLIACCIO, ANNA RITA, Crispino, Jd, Lucero, Ha, Trackman, Pc, and Ravid, K
- Subjects
Male ,Platelet-derived growth factor ,Blotting, Western ,Fluorescent Antibody Technique ,Lysyl oxidase ,Biochemistry ,Polyploidy ,Protein-Lysine 6-Oxidase ,Receptor, Platelet-Derived Growth Factor beta ,chemistry.chemical_compound ,Mice ,Megakaryocyte ,Bone Marrow ,medicine ,Animals ,RNA, Messenger ,Molecular Biology ,Mitosis ,biology ,integumentary system ,urogenital system ,food and beverages ,megakaryocyte bone marrow fibrosis lysyl oxidase ,Cell Biology ,Flow Cytometry ,Molecular biology ,Haematopoiesis ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,chemistry ,Primary Myelofibrosis ,biology.protein ,Bone marrow ,Signal transduction ,Megakaryocytes ,Platelet-derived growth factor receptor ,Cell Division - Abstract
Lysyl oxidase (LOX), a matrix cross-linking protein, is known to be selectively expressed and to enhance a fibrotic phenotype. A recent study of ours showed that LOX oxidizes the PDGF receptor-β (PDGFR-β), leading to amplified downstream signaling. Here, we examined the expression and functions of LOX in megakaryocytes (MKs), the platelet precursors. Cells committed to the MK lineage undergo mitotic proliferation to yield diploid cells, followed by endomitosis and acquisition of polyploidy. Intriguingly, LOX expression is detected in diploid-tetraploid MKs, but scarce in polyploid MKs. PDGFR-BB is an inducer of mitotic proliferation in MKs. LOX inhibition with β-aminopropionitrile reduces PDGFR-BB binding to cells and downstream signaling, as well as its proliferative effect on the MK lineage. Inhibition of LOX activity has no influence on MK polyploidy. We next rationalized that, in a system with an abundance of low ploidy MKs, LOX could be highly expressed and with functional significance. Thus, we resorted to GATA-1(low) mice, where there is an increase in low ploidy MKs, augmented levels of PDGF-BB, and an extensive matrix of fibers. MKs from these mice display high expression of LOX, compared with control mice. Importantly, treatment of GATA-1(low) mice with β-aminopropionitrile significantly improves the bone marrow fibrotic phenotype, and MK number in the spleen. Thus, our in vitro and in vivo data support a novel role for LOX in regulating MK expansion by PDGF-BB and suggest LOX as a new potential therapeutic target for myelofibrosis.
- Published
- 2011
4. Disruption of cotranscriptional splicing suggests that RBM39 is a therapeutic target in acute lymphoblastic leukemia.
- Author
-
Jin Q, Harris E, Myers JA, Mehmood R, Cotton A, Shirnekhi HK, Baggett DW, Wen JQ, Schild AB, Bhansali RS, Klein J, Narina S, Pieters T, Yoshimi A, Pruett-Miller SM, Kriwacki R, Abdel-Wahab O, Malinge S, Ntziachristos P, Obeng EA, and Crispino JD
- Abstract
There are few options for patients with relapse/refractory B-cell acute lymphoblastic leukemia (B-ALL), thus this is a major area of unmet medical need. Here, we reveal that inclusion of a poison exon in RBM39, which could be induced both by CDK9 or CDK9 independent CMGC (cyclin-dependent kinases, mitogen-activated protein kinases, glycogen synthase kinases, CDC-like kinases) kinase inhibition, is recognized by the nonsense-mediated mRNA decay (NMD) pathway for degradation. Targeting this poison exon in RBM39 with CMGC inhibitors lead to protein downregulation and inhibition of ALL growth, particularly in relapse/refractory B-ALL. Mechanistically, disruption of co-transcriptional splicing by inhibition of CMGC kinases including DYRK1A, or inhibition of CDK9, which phosphorylate the C-terminal domain of RNA polymerase II (Pol II), results in alteration of SF3B1 and Pol II association. Disruption of SF3B1 and transcriptional elongation complex alters Pol II pausing, which promotes the inclusion of a poison exon in RBM39. Moreover, RBM39 ablation suppresses the growth of human B-ALL, and targeting RBM39 with sulfonamides, which degrade RBM39 protein, showed strong anti-tumor activity in preclinical models. Our data reveal that relapse/refractory B-ALL is susceptible to pharmacologic and genetic inhibition of RBM39 and provide two potential strategies to target this axis., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
5. Efficacy of DYRK1A inhibitors in novel models of Down syndrome acute lymphoblastic leukemia.
- Author
-
Carey-Smith SL, Simad MH, Panchal K, Aya-Bonilla C, Smolders H, Lin S, Armitage JD, Nguyen VT, Bentley K, Ford J, Singh S, Oommen J, Laurent AP, Mercher T, Crispino JD, Montgomery AP, Kassiou M, Besson T, Deau E, Meijer L, Cheung LC, Kotecha RS, and Malinge S
- Subjects
- Humans, Animals, Mice, Disease Models, Animal, Down Syndrome complications, Down Syndrome drug therapy, Protein-Tyrosine Kinases antagonists & inhibitors, Dyrk Kinases, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases genetics
- Published
- 2024
- Full Text
- View/download PDF
6. Malignant progression of preleukemic disorders.
- Author
-
Hall T, Gurbuxani S, and Crispino JD
- Subjects
- Humans, Preleukemia pathology, Preleukemia genetics, Myelodysplastic Syndromes pathology, Myelodysplastic Syndromes genetics, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute genetics, Animals, Precancerous Conditions pathology, Precancerous Conditions genetics, Mutation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Cell Transformation, Neoplastic metabolism, Disease Progression
- Abstract
Abstract: The spectrum of myeloid disorders ranges from aplastic bone marrow failure characterized by an empty bone marrow completely lacking in hematopoiesis to acute myeloid leukemia in which the marrow space is replaced by undifferentiated leukemic blasts. Recent advances in the capacity to sequence bulk tumor population as well as at a single-cell level has provided significant insight into the stepwise process of transformation to acute myeloid leukemia. Using models of progression in the context of germ line predisposition (trisomy 21, GATA2 deficiency, and SAMD9/9L syndrome), premalignant states (clonal hematopoiesis and clonal cytopenia of unknown significance), and myelodysplastic syndrome, we review the mechanisms of progression focusing on the hierarchy of clonal mutation and potential roles of transcription factor alterations, splicing factor mutations, and the bone marrow environment in progression to acute myeloid leukemia. Despite major advances in our understanding, preventing the progression of these disorders or treating them at the acute leukemia phase remains a major area of unmet medical need., (© 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
7. Building Synthetic Biosensors Using Red Blood Cell Proteins.
- Author
-
Dolberg TB, Gunnels TF, Ling T, Sarnese KA, Crispino JD, and Leonard JN
- Subjects
- Ligands, Membrane Proteins metabolism, Erythrocytes metabolism, Biosensing Techniques
- Abstract
As the use of engineered cell therapies expands from pioneering efforts in cancer immunotherapy to other applications, an attractive but less explored approach is the use of engineered red blood cells (RBCs). Compared to other cells, RBCs have a very long circulation time and reside in the blood compartment, so they could be ideally suited for applications as sentinel cells that enable in situ sensing and diagnostics. However, we largely lack tools for converting RBCs into biosensors. A unique challenge is that RBCs remodel their membranes during maturation, shedding many membrane components, suggesting that an RBC-specific approach may be needed. Toward addressing this need, here we develop a biosensing architecture built on RBC membrane proteins that are retained through erythropoiesis. This biosensor employs a mechanism in which extracellular ligand binding is transduced into intracellular reconstitution of a split output protein (including either a fluorophore or an enzyme). By comparatively evaluating a range of biosensor architectures, linker types, scaffold choices, and output signals, we identify biosensor designs and design features that confer substantial ligand-induced signal in vitro . Finally, we demonstrate that erythroid precursor cells engineered with our RBC-protein biosensors function in vivo. This study establishes a foundation for developing RBC-based biosensors that could ultimately address unmet needs including noninvasive monitoring of physiological signals for a range of diagnostic applications.
- Published
- 2024
- Full Text
- View/download PDF
8. Correction: A positive feedback loop regulation between NOTCH1 and USP11 in T-cell leukemia.
- Author
-
Fijalkowski I, Wang J, Jin Q, Van Laere J, Serafin V, Crispino JD, and Ntziachristos P
- Published
- 2024
- Full Text
- View/download PDF
9. A positive feedback loop regulation between NOTCH1 and USP11 in T-cell leukemia.
- Author
-
Fijalkowski I, Wang J, Jin Q, Van Laere J, Serafin V, Crispino JD, and Ntziachristos P
- Subjects
- Humans, Feedback, Basic Helix-Loop-Helix Transcription Factors, Receptor, Notch1 genetics, Thiolester Hydrolases, Leukemia, T-Cell
- Published
- 2024
- Full Text
- View/download PDF
10. Building synthetic biosensors using red blood cell proteins.
- Author
-
Dolberg TB, Gunnels TF, Ling T, Sarnese KA, Crispino JD, and Leonard JN
- Abstract
As the use of engineered cell therapies expands from pioneering efforts in cancer immunotherapy to other applications, an attractive but less explored approach is the use of engineered red blood cells (RBCs). Compared to other cells, RBCs have a very long circulation time and reside in the blood compartment, so they could be ideally suited for applications as sentinel cells that enable in situ sensing and diagnostics. However, we largely lack tools for converting RBCs into biosensors. A unique challenge is that RBCs remodel their membranes during maturation, shedding many membrane components, suggesting that an RBC-specific approach may be needed. Towards addressing this need, here we develop a biosensing architecture built on RBC membrane proteins that are retained through erythropoiesis. This biosensor employs a mechanism in which extracellular ligand binding is transduced into intracellular reconstitution of a split output protein (including either a fluorophore or an enzyme). By comparatively evaluating a range of biosensor architectures, linker types, scaffold choices, and output signals, we identify biosensor designs and design features that confer substantial ligand-induced signal in vitro . Finally, we demonstrate that erythroid precursor cells engineered with our RBC protein biosensors function in vivo . This study establishes a foundation for developing RBC-based biosensors that could ultimately address unmet needs including non-invasive monitoring of physiological signals for a range of diagnostic applications.
- Published
- 2023
- Full Text
- View/download PDF
11. Chromothripsis orchestrates leukemic transformation in blast phase MPN through targetable amplification of DYRK1A .
- Author
-
Brierley CK, Yip BH, Orlando G, Goyal H, Wen S, Wen J, Levine MF, Jakobsdottir GM, Rodriguez-Meira A, Adamo A, Bashton M, Hamblin A, Clark SA, O'Sullivan J, Murphy L, Olijnik AA, Cotton A, Narina S, Pruett-Miller SM, Enshaei A, Harrison C, Drummond M, Knapper S, Tefferi A, Antony-Debré I, Thongjuea S, Wedge DC, Constantinescu S, Papaemmanuil E, Psaila B, Crispino JD, and Mead AJ
- Abstract
Chromothripsis, the process of catastrophic shattering and haphazard repair of chromosomes, is a common event in cancer. Whether chromothripsis might constitute an actionable molecular event amenable to therapeutic targeting remains an open question. We describe recurrent chromothripsis of chromosome 21 in a subset of patients in blast phase of a myeloproliferative neoplasm (BP-MPN), which alongside other structural variants leads to amplification of a region of chromosome 21 in ∼25% of patients ('chr21amp'). We report that chr21amp BP-MPN has a particularly aggressive and treatment-resistant phenotype. The chr21amp event is highly clonal and present throughout the hematopoietic hierarchy. DYRK1A , a serine threonine kinase and transcription factor, is the only gene in the 2.7Mb minimally amplified region which showed both increased expression and chromatin accessibility compared to non-chr21amp BP-MPN controls. We demonstrate that DYRK1A is a central node at the nexus of multiple cellular functions critical for BP-MPN development, including DNA repair, STAT signalling and BCL2 overexpression. DYRK1A is essential for BP-MPN cell proliferation in vitro and in vivo , and DYRK1A inhibition synergises with BCL2 targeting to induce BP-MPN cell apoptosis. Collectively, these findings define the chr21amp event as a prognostic biomarker in BP-MPN and link chromothripsis to a druggable target.
- Published
- 2023
- Full Text
- View/download PDF
12. Introduction to a review series on hematopoietic stem cells.
- Author
-
Crispino JD
- Subjects
- Hematopoietic Stem Cells, Hematopoietic Stem Cell Transplantation
- Published
- 2023
- Full Text
- View/download PDF
13. Gata1s mutant mice display persistent defects in the erythroid lineage.
- Author
-
Ling T, Zhang K, Yang J, Gurbuxani S, and Crispino JD
- Subjects
- Animals, Mice, Cell Lineage, Protein Isoforms, Thrombopoiesis, Down Syndrome complications, Erythropoiesis genetics
- Abstract
GATA1 mutations that result in loss of the N-terminal 83 amino acids are a feature of myeloid leukemia in children with Down syndrome, rare familial cases of dyserythropoietic anemia, and a subset of cases of Diamond-Blackfan anemia. The Gata1s mouse model, which expresses only the short GATA1 isoform that begins at methionine 84, has been shown to have a defect in hematopoiesis, especially impaired erythropoiesis with expanded megakaryopoiesis, during gestation. However, these mice reportedly did not show any postnatal phenotype. Here, we demonstrate that Gata1s mutant mice display macrocytic anemia and features of aberrant megakaryopoiesis throughout life, culminating in profound splenomegaly and bone marrow fibrosis. These data support the use of this animal model for studies of GATA1 deficiencies., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. B cell class switch recombination is regulated by DYRK1A through MSH6 phosphorylation.
- Author
-
Stoler-Barak L, Harris E, Peres A, Hezroni H, Kuka M, Di Lucia P, Grenov A, Gurwicz N, Kupervaser M, Yip BH, Iannacone M, Yaari G, Crispino JD, and Shulman Z
- Subjects
- Phosphorylation, Germinal Center, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, B-Lymphocytes, Immunoglobulin Class Switching genetics
- Abstract
Protection from viral infections depends on immunoglobulin isotype switching, which endows antibodies with effector functions. Here, we find that the protein kinase DYRK1A is essential for B cell-mediated protection from viral infection and effective vaccination through regulation of class switch recombination (CSR). Dyrk1a-deficient B cells are impaired in CSR activity in vivo and in vitro. Phosphoproteomic screens and kinase-activity assays identify MSH6, a DNA mismatch repair protein, as a direct substrate for DYRK1A, and deletion of a single phosphorylation site impaired CSR. After CSR and germinal center (GC) seeding, DYRK1A is required for attenuation of B cell proliferation. These findings demonstrate DYRK1A-mediated biological mechanisms of B cell immune responses that may be used for therapeutic manipulation in antibody-mediated autoimmunity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. CALR goes rogue.
- Author
-
Melo-Cardenas J and Crispino JD
- Subjects
- Humans, Calreticulin, Computer Simulation, Biological Transport, Neoplasms, Myeloproliferative Disorders
- Published
- 2023
- Full Text
- View/download PDF
16. Bone Marrow Avatars: Mimicking Hematopoiesis in a Dish.
- Author
-
Derecka M and Crispino JD
- Subjects
- Humans, Hematopoiesis, Cell Differentiation, Organoids, Bone Marrow, Hematologic Neoplasms
- Abstract
Summary: Faithful recapitulation of human bone marrow complexity has been a major challenge for the sci-entific community for many years. In this issue of Cancer Discovery, Khan and colleagues present an improved induced pluripotent stem cell differentiation protocol that generates bone marrow organoids re-creating key characteristics of human marrow. See related article by Khan et al., p. 364 (8)., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
17. IL-13/IL-4 signaling contributes to fibrotic progression of the myeloproliferative neoplasms.
- Author
-
Melo-Cardenas J, Bezavada L, Crawford JC, Gurbuxani S, Cotton A, Kang G, Gossett J, Marinaccio C, Weinberg R, Hoffman R, Migliaccio AR, Zheng Y, Derecka M, Rinaldi CR, and Crispino JD
- Subjects
- Mice, Animals, Interleukin-13 therapeutic use, Interleukin-4, Signal Transduction genetics, Fibrosis, Disease Progression, Neoplasms complications, Myeloproliferative Disorders complications, Primary Myelofibrosis genetics
- Abstract
Myelofibrosis (MF) is a disease associated with high unmet medical needs because allogeneic stem cell transplantation is not an option for most patients, and JAK inhibitors are generally effective for only 2 to 3 years and do not delay disease progression. MF is characterized by dysplastic megakaryocytic hyperplasia and progression to fulminant disease, which is associated with progressively increasing marrow fibrosis. Despite evidence that the inflammatory milieu in MF contributes to disease progression, the specific factors that promote megakaryocyte growth are poorly understood. Here, we analyzed changes in the cytokine profiles of MF mouse models before and after the development of fibrosis, coupled with the analysis of bone marrow populations using single-cell RNA sequencing. We found high interleukin 13 (IL-13) levels in the bone marrow of MF mice. IL-13 promoted the growth of mutant megakaryocytes and induced surface expression of transforming growth factor β and collagen biosynthesis. Similarly, analysis of samples from patients with MF revealed elevated levels of IL-13 in the plasma and increased IL-13 receptor expression in marrow megakaryocytes. In vivo, IL-13 overexpression promoted disease progression, whereas reducing IL-13/IL-4 signaling reduced several features of the disease, including fibrosis. Finally, we observed an increase in the number of marrow T cells and mast cells, which are known sources of IL-13. Together, our data demonstrate that IL-13 is involved in disease progression in MF and that inhibition of the IL-13/IL-4 signaling pathway might serve as a novel therapeutic target to treat MF., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
18. Oncogenic deubiquitination controls tyrosine kinase signaling and therapy response in acute lymphoblastic leukemia.
- Author
-
Jin Q, Gutierrez Diaz B, Pieters T, Zhou Y, Narang S, Fijalkwoski I, Borin C, Van Laere J, Payton M, Cho BK, Han C, Sun L, Serafin V, Yacu G, Von Loocke W, Basso G, Veltri G, Dreveny I, Ben-Sahra I, Goo YA, Safgren SL, Tsai YC, Bornhauser B, Suraneni PK, Gaspar-Maia A, Kandela I, Van Vlierberghe P, Crispino JD, Tsirigos A, and Ntziachristos P
- Subjects
- Humans, Cell Line, Tumor, Glucocorticoids pharmacology, Glucocorticoids therapeutic use, Lymphocyte Specific Protein Tyrosine Kinase p56(lck) metabolism, Receptors, Glucocorticoid metabolism, Signal Transduction, Thiolester Hydrolases metabolism, Thiolester Hydrolases therapeutic use, Ubiquitin-Specific Peptidase 7 metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics
- Abstract
Dysregulation of kinase signaling pathways favors tumor cell survival and therapy resistance in cancer. Here, we reveal a posttranslational regulation of kinase signaling and nuclear receptor activity via deubiquitination in T cell acute lymphoblastic leukemia (T-ALL). We observed that the ubiquitin-specific protease 11 (USP11) is highly expressed and associates with poor prognosis in T-ALL. USP11 ablation inhibits leukemia progression in vivo, sparing normal hematopoiesis. USP11 forms a complex with USP7 to deubiquitinate the oncogenic lymphocyte cell-specific protein-tyrosine kinase (LCK) and enhance its activity. Impairment of LCK activity leads to increased glucocorticoid receptor (GR) expression and glucocorticoids sensitivity. Genetic knockout of USP7 improved the antileukemic efficacy of glucocorticoids in vivo. The transcriptional activation of GR target genes is orchestrated by the deubiquitinase activity and mediated via an increase in enhancer-promoter interaction intensity. Our data unveil how dysregulated deubiquitination controls leukemia survival and drug resistance, suggesting previously unidentified therapeutic combinations toward targeting leukemia.
- Published
- 2022
- Full Text
- View/download PDF
19. PU.1-c-Jun interaction is crucial for PU.1 function in myeloid development.
- Author
-
Zhao X, Bartholdy B, Yamamoto Y, Evans EK, Alberich-Jordà M, Staber PB, Benoukraf T, Zhang P, Zhang J, Trinh BQ, Crispino JD, Hoang T, Bassal MA, and Tenen DG
- Subjects
- Animals, Binding Sites, Cell Differentiation genetics, Mice, Promoter Regions, Genetic, Proto-Oncogene Proteins c-jun, Hematopoiesis genetics, Transcription Factor AP-1 genetics
- Abstract
The Ets transcription factor PU.1 is essential for inducing the differentiation of monocytes, macrophages, and B cells in fetal liver and adult bone marrow. PU.1 controls hematopoietic differentiation through physical interactions with other transcription factors, such as C/EBPα and the AP-1 family member c-Jun. We found that PU.1 recruits c-Jun to promoters without the AP-1 binding sites. To address the functional importance of this interaction, we generated PU.1 point mutants that do not bind c-Jun while maintaining normal DNA binding affinity. These mutants lost the ability to transactivate a target reporter that requires a physical PU.1-c-Jun interaction, and did not induce monocyte/macrophage differentiation of PU.1-deficient cells. Knock-in mice carrying these point mutations displayed an almost complete block in hematopoiesis and perinatal lethality. While the PU.1 mutants were expressed in hematopoietic stem and early progenitor cells, myeloid differentiation was severely blocked, leading to an almost complete loss of mature hematopoietic cells. Differentiation into mature macrophages could be restored by expressing PU.1 mutant fused to c-Jun, demonstrating that a physical PU.1-c-Jun interaction is crucial for the transactivation of PU.1 target genes required for myeloid commitment and normal PU.1 function in vivo during macrophage differentiation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
20. Introduction to a review series on megakaryopoiesis and platelet production.
- Author
-
Crispino JD
- Subjects
- Blood Platelets, Megakaryocytes, Thrombopoiesis
- Published
- 2022
- Full Text
- View/download PDF
21. Discovery of a signaling feedback circuit that defines interferon responses in myeloproliferative neoplasms.
- Author
-
Saleiro D, Wen JQ, Kosciuczuk EM, Eckerdt F, Beauchamp EM, Oku CV, Blyth GT, Fischietti M, Ilut L, Colamonici M, Palivos W, Atsaves PA, Tan D, Kocherginsky M, Weinberg RS, Fish EN, Crispino JD, Hoffman R, and Platanias LC
- Subjects
- Antiviral Agents therapeutic use, Feedback, Humans, Interferon-alpha pharmacology, Interferon-alpha therapeutic use, Signal Transduction, rho-Associated Kinases metabolism, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Neoplasms drug therapy
- Abstract
Interferons (IFNs) are key initiators and effectors of the immune response against malignant cells and also directly inhibit tumor growth. IFNα is highly effective in the treatment of myeloproliferative neoplasms (MPNs), but the mechanisms of action are unclear and it remains unknown why some patients respond to IFNα and others do not. Here, we identify and characterize a pathway involving PKCδ-dependent phosphorylation of ULK1 on serine residues 341 and 495, required for subsequent activation of p38 MAPK. We show that this pathway is essential for IFN-suppressive effects on primary malignant erythroid precursors from MPN patients, and that increased levels of ULK1 and p38 MAPK correlate with clinical response to IFNα therapy in these patients. We also demonstrate that IFNα treatment induces cleavage/activation of the ULK1-interacting ROCK1/2 proteins in vitro and in vivo, triggering a negative feedback loop that suppresses IFN responses. Overexpression of ROCK1/2 is seen in MPN patients and their genetic or pharmacological inhibition enhances IFN-anti-neoplastic responses in malignant erythroid precursors from MPN patients. These findings suggest the clinical potential of pharmacological inhibition of ROCK1/2 in combination with IFN-therapy for the treatment of MPNs., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
22. The chromosome 21 kinase DYRK1A: emerging roles in cancer biology and potential as a therapeutic target.
- Author
-
Rammohan M, Harris E, Bhansali RS, Zhao E, Li LS, and Crispino JD
- Subjects
- Chromosomes, Human, Pair 21 metabolism, Humans, Oncogenes, Phosphorylation, Dyrk Kinases, Neoplasms genetics, Protein Serine-Threonine Kinases genetics, Protein-Tyrosine Kinases genetics
- Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1 A (DYRK1A) is a serine/threonine kinase that belongs to the DYRK family of proteins, a subgroup of the evolutionarily conserved CMGC protein kinase superfamily. Due to its localization on chromosome 21, the biological significance of DYRK1A was initially characterized in the pathogenesis of Down syndrome (DS) and related neurodegenerative diseases. However, increasing evidence has demonstrated a prominent role in cancer through its ability to regulate biologic processes including cell cycle progression, DNA damage repair, transcription, ubiquitination, tyrosine kinase activity, and cancer stem cell maintenance. DYRK1A has been identified as both an oncogene and tumor suppressor in different models, underscoring the importance of cellular context in its function. Here, we review mechanistic contributions of DYRK1A to cancer biology and its role as a potential therapeutic target., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
23. Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape.
- Author
-
Tefferi A, Gangat N, Pardanani A, and Crispino JD
- Subjects
- Humans, Janus Kinase 2 genetics, Janus Kinase 2 metabolism, Mutation, Myeloproliferative Disorders genetics, Polycythemia Vera genetics, Primary Myelofibrosis diagnosis, Primary Myelofibrosis genetics, Primary Myelofibrosis therapy, Thrombocythemia, Essential diagnosis, Thrombocythemia, Essential genetics
- Abstract
Primary myelofibrosis (PMF) is one of three myeloproliferative neoplasms (MPN) that are morphologically and molecularly inter-related, the other two being polycythemia vera (PV) and essential thrombocythemia (ET). MPNs are characterized by JAK-STAT-activating JAK2, CALR, or MPL mutations that give rise to stem cell-derived clonal myeloproliferation, which is prone to leukemic and, in case of PV and ET, fibrotic transformation. Abnormal megakaryocyte proliferation is accompanied by bone marrow fibrosis and characterizes PMF, while the clinical phenotype is pathogenetically linked to ineffective hematopoiesis and aberrant cytokine expression. Among MPN-associated driver mutations, type 1-like CALR mutation has been associated with favorable prognosis in PMF, while ASXL1, SRSF2, U2AF1-Q157, EZH2, CBL, and K/NRAS mutations have been shown to be prognostically detrimental. Such information has enabled development of exclusively genetic (GIPSS) and clinically integrated (MIPSSv2) prognostic models that facilitate individualized treatment decisions. Allogeneic stem cell transplantation remains the only treatment modality in MF with the potential to prolong survival, whereas drug therapy, including JAK2 inhibitors, is directed mostly at the inflammatory component of the disease and is therefore palliative in nature. Similarly, disease-modifying activity remains elusive for currently available investigational drugs, while their additional value in symptom management awaits controlled confirmation. There is a need for genetic characterization of clinical observations followed by in vitro and in vivo preclinical studies that will hopefully identify therapies that target the malignant clone in MF to improve patient outcomes., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
24. Resident Self-Tissue of Proinflammatory Cytokines Rather Than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1 low Mice.
- Author
-
Zingariello M, Verachi P, Gobbo F, Martelli F, Falchi M, Mazzarini M, Valeri M, Sarli G, Marinaccio C, Melo-Cardenas J, Crispino JD, and Migliaccio AR
- Subjects
- Animals, Bone Marrow metabolism, Bone Marrow pathology, Endothelial Cells metabolism, Endothelial Cells pathology, Megakaryocytes metabolism, Megakaryocytes pathology, Mice, Cytokines metabolism, GATA1 Transcription Factor metabolism, Primary Myelofibrosis metabolism, Primary Myelofibrosis pathology
- Abstract
Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1
low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-β1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.- Published
- 2022
- Full Text
- View/download PDF
25. Clinical and biological aspects of myeloid leukemia in Down syndrome.
- Author
-
Boucher AC, Caldwell KJ, Crispino JD, and Flerlage JE
- Subjects
- Humans, Leukemia, Myeloid etiology, Leukemia, Myeloid metabolism, Down Syndrome complications, GATA1 Transcription Factor genetics, Leukemia, Myeloid pathology, Mutation
- Abstract
Children with Down syndrome are at an elevated risk of leukemia, especially myeloid leukemia (ML-DS). This malignancy is frequently preceded by transient abnormal myelopoiesis (TAM), which is self-limited expansion of fetal liver-derived megakaryocyte progenitors. An array of international studies has led to consensus in treating ML-DS with reduced-intensity chemotherapy, leading to excellent outcomes. In addition, studies performed in the past 20 years have revealed many of the genetic and epigenetic features of the tumors, including GATA1 mutations that are arguably associated with all cases of both TAM and ML-DS. Despite these advances in understanding the clinical and biological aspects of ML-DS, little is known about the mechanisms of relapse. Upon relapse, patients face a poor outcome, and there is no consensus on treatment. Future studies need to be focused on this challenging aspect of leukemia in children with DS., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
26. Introduction to a review series on molecular mechanisms of hematologic malignancies.
- Author
-
Crispino JD
- Subjects
- Humans, Hematologic Neoplasms
- Published
- 2021
- Full Text
- View/download PDF
27. LKB1/ STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms.
- Author
-
Marinaccio C, Suraneni P, Celik H, Volk A, Wen QJ, Ling T, Bulic M, Lasho T, Koche RP, Famulare CA, Farnoud N, Stein B, Schieber M, Gurbuxani S, Root DE, Younger ST, Hoffman R, Gangat N, Ntziachristos P, Chandel NS, Levine RL, Rampal RK, Challen GA, Tefferi A, and Crispino JD
- Subjects
- Animals, Disease Models, Animal, Disease Progression, Mice, Mice, Inbred C57BL, Mutation, Myeloproliferative Disorders genetics, AMP-Activated Protein Kinases genetics, Genes, Tumor Suppressor, Leukemia, Myeloid, Acute genetics
- Abstract
The myeloproliferative neoplasms (MPN) frequently progress to blast phase disease, an aggressive form of acute myeloid leukemia. To identify genes that suppress disease progression, we performed a focused CRISPR/Cas9 screen and discovered that depletion of LKB1/ Stk11 led to enhanced in vitro self-renewal of murine MPN cells. Deletion of Stk11 in a mouse MPN model caused rapid lethality with enhanced fibrosis, osteosclerosis, and an accumulation of immature cells in the bone marrow, as well as enhanced engraftment of primary human MPN cells in vivo . LKB1 loss was associated with increased mitochondrial reactive oxygen species and stabilization of HIF1α, and downregulation of LKB1 and increased levels of HIF1α were observed in human blast phase MPN specimens. Of note, we observed strong concordance of pathways that were enriched in murine MPN cells with LKB1 loss with those enriched in blast phase MPN patient specimens, supporting the conclusion that STK11 is a tumor suppressor in the MPNs. SIGNIFICANCE: Progression of the myeloproliferative neoplasms to acute myeloid leukemia occurs in a substantial number of cases, but the genetic basis has been unclear. We discovered that loss of LKB1/ STK11 leads to stabilization of HIF1a and promotes disease progression. This observation provides a potential therapeutic avenue for targeting progression. This article is highlighted in the In This Issue feature, p. 1307 ., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
28. Posttranslational regulation of FOXA1 by Polycomb and BUB3/USP7 deubiquitin complex in prostate cancer.
- Author
-
Park SH, Fong KW, Kim J, Wang F, Lu X, Lee Y, Brea LT, Wadosky K, Guo C, Abdulkadir SA, Crispino JD, Fang D, Ntziachristos P, Liu X, Li X, Wan Y, Goodrich DW, Zhao JC, and Yu J
- Abstract
Forkhead box protein A1 (FOXA1) is essential for androgen-dependent prostate cancer (PCa) growth. However, how FOXA1 levels are regulated remains elusive and its therapeutic targeting proven challenging. Here, we report FOXA1 as a nonhistone substrate of enhancer of zeste homolog 2 (EZH2), which methylates FOXA1 at lysine-295. This methylation is recognized by WD40 repeat protein BUB3, which subsequently recruits ubiquitin-specific protease 7 (USP7) to remove ubiquitination and enhance FOXA1 protein stability. They functionally converge in regulating cell cycle genes and promoting PCa growth. FOXA1 is a major therapeutic target of the inhibitors of EZH2 methyltransferase activities in PCa. FOXA1-driven PCa growth can be effectively mitigated by EZH2 enzymatic inhibitors, either alone or in combination with USP7 inhibitors. Together, our study reports EZH2-catalyzed methylation as a key mechanism to FOXA1 protein stability, which may be leveraged to enhance therapeutic targeting of PCa using enzymatic EZH2 inhibitors., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2021
- Full Text
- View/download PDF
29. The Role of Megakaryocytes in Myelofibrosis.
- Author
-
Melo-Cardenas J, Migliaccio AR, and Crispino JD
- Subjects
- Calreticulin genetics, Humans, Janus Kinase 2 genetics, Mutation, Receptors, Thrombopoietin genetics, Megakaryocytes, Myeloproliferative Disorders genetics, Neoplasms genetics, Primary Myelofibrosis genetics
- Abstract
Megakaryocytes give rise to platelets, which have a wide variety of functions in coagulation, immune response, inflammation, and tissue repair. Dysregulation of megakaryocytes is a key feature of in the myeloproliferative neoplasms, especially myelofibrosis. Megakaryocytes are among the main drivers of myelofibrosis by promoting myeloproliferation and bone marrow fibrosis. In vivo targeting of megakaryocytes by genetic and pharmacologic approaches ameliorates the disease, underscoring the important role of megakaryocytes in myeloproliferative neoplasms. Here we review the current knowledge of the function of megakaryocytes in the JAK2, CALR, and MPL-mutant myeloproliferative neoplasms., Competing Interests: Disclosure The authors have nothing to disclose., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
30. DYRK1A regulates B cell acute lymphoblastic leukemia through phosphorylation of FOXO1 and STAT3.
- Author
-
Bhansali RS, Rammohan M, Lee P, Laurent AP, Wen Q, Suraneni P, Yip BH, Tsai YC, Jenni S, Bornhauser B, Siret A, Fruit C, Pacheco-Benichou A, Harris E, Besson T, Thompson BJ, Goo YA, Hijiya N, Vilenchik M, Izraeli S, Bourquin JP, Malinge S, and Crispino JD
- Subjects
- Animals, Female, Forkhead Box Protein O1 genetics, Male, Mice, Mice, Transgenic, Neoplasm Proteins genetics, Phosphorylation genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Protein Serine-Threonine Kinases genetics, Protein-Tyrosine Kinases genetics, STAT3 Transcription Factor genetics, Dyrk Kinases, Forkhead Box Protein O1 metabolism, Neoplasm Proteins metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, STAT3 Transcription Factor metabolism
- Abstract
DYRK1A is a serine/threonine kinase encoded on human chromosome 21 (HSA21) that has been implicated in several pathologies of Down syndrome (DS), including cognitive deficits and Alzheimer's disease. Although children with DS are predisposed to developing leukemia, especially B cell acute lymphoblastic leukemia (B-ALL), the HSA21 genes that contribute to malignancies remain largely undefined. Here, we report that DYRK1A is overexpressed and required for B-ALL. Genetic and pharmacologic inhibition of DYRK1A decreased leukemic cell expansion and suppressed B-ALL development in vitro and in vivo. Furthermore, we found that FOXO1 and STAT3, transcription factors that are indispensable for B cell development, are critical substrates of DYRK1A. Loss of DYRK1A-mediated FOXO1 and STAT3 signaling disrupted DNA damage and ROS regulation, respectively, leading to preferential cell death in leukemic B cells. Thus, we reveal a DYRK1A/FOXO1/STAT3 axis that facilitates the development and maintenance of B-ALL.
- Published
- 2021
- Full Text
- View/download PDF
31. FBXO11 is a candidate tumor suppressor in the leukemic transformation of myelodysplastic syndrome.
- Author
-
Schieber M, Marinaccio C, Bolanos LC, Haffey WD, Greis KD, Starczynowski DT, and Crispino JD
- Subjects
- CRISPR-Cas Systems, Cell Line, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, F-Box Proteins genetics, Gene Deletion, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology, Protein-Arginine N-Methyltransferases genetics, Tumor Suppressor Proteins genetics, Cell Transformation, Neoplastic metabolism, F-Box Proteins metabolism, Leukemia, Myeloid, Acute metabolism, Myelodysplastic Syndromes metabolism, Protein-Arginine N-Methyltransferases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous myeloid malignancy characterized by blood cell morphological dysplasia, ineffective clonal hematopoiesis, and risk of transformation to secondary acute myeloid leukemia (sAML). A number of genetic abnormalities have been identified in MDS and sAML, but sensitive sequencing methods can detect these mutations in nearly all healthy individuals by 60 years of age. To discover novel cellular pathways that accelerate MDS and sAML, we performed a CRISPR/Cas9 screen in the human MDS-L cell line. We report here that loss of the F-Box protein FBXO11, a component of the SCF ubiquitin ligase complex, confers cytokine independent growth to MDS-L cells, suggesting a tumor suppressor role for FBXO11 in myeloid malignancies. Putative FBXO11 substrates are enriched for proteins with functions in RNA metabolism and, of note, spliceosome mutations that are commonly found in MDS/sAML are rare in patients with low FBXO11 expression. We also reveal that loss of FBXO11 leads to significant changes in transcriptional pathways influencing leukocyte proliferation, differentiation, and apoptosis. Last, we find that FBXO11 expression is reduced in patients with secondary AML. We conclude that loss of FBXO11 is a mechanism for disease transformation of MDS into AML, and may represent a future therapeutic target.
- Published
- 2020
- Full Text
- View/download PDF
32. Determinants and role of chromatin organization in acute leukemia.
- Author
-
Fang C, Rao S, Crispino JD, and Ntziachristos P
- Subjects
- Acute Disease, CCCTC-Binding Factor genetics, Gene Expression Regulation, Neoplastic, Humans, Leukemia genetics, Mutation, Chromatin metabolism, Leukemia metabolism
- Abstract
DNA is compacted into higher order structures that have major implications in gene regulation. These structures allow for long-range interactions of DNA elements, such as the association of promoters with their cognate enhancers. In recent years, mutations in genes that control these structures, including the cohesin-complex and the insulator-binding protein CTCF, have been found in a spectrum of hematologic disorders, and especially in acute leukemias. Cohesin and CTCF are critical for mediating looping and establishing boundaries within chromatin. Cells that harbor mutations in these genes display aberrant chromatin architecture and resulting differences in gene expression that contribute to leukemia initiation and progression. Here, we provide detailed discussion of the nature of 3D interactions and the way that they are disrupted in acute leukemia. Continued research in this area will provide new insights into the mechanisms of leukemogenesis and may shed light on novel treatment strategies.
- Published
- 2020
- Full Text
- View/download PDF
33. Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia.
- Author
-
Laurent AP, Siret A, Ignacimouttou C, Panchal K, Diop M, Jenni S, Tsai YC, Roos-Weil D, Aid Z, Prade N, Lagarde S, Plassard D, Pierron G, Daudigeos E, Lecluse Y, Droin N, Bornhauser BC, Cheung LC, Crispino JD, Gaudry M, Bernard OA, Macintyre E, Barin Bonnigal C, Kotecha RS, Geoerger B, Ballerini P, Bourquin JP, Delabesse E, Mercher T, and Malinge S
- Subjects
- Animals, Computational Biology methods, Disease Models, Animal, Disease Susceptibility, Gene Expression Profiling, Humans, Immunophenotyping, Leukemia, B-Cell therapy, Mice, Mice, Transgenic, Oncogenes, Protein Kinase Inhibitors pharmacology, Pyridones pharmacology, Pyrimidinones pharmacology, Down Syndrome complications, Down Syndrome genetics, Down Syndrome metabolism, Leukemia, B-Cell diagnosis, Leukemia, B-Cell etiology, Mitogen-Activated Protein Kinases metabolism, Signal Transduction drug effects, ras Proteins metabolism
- Abstract
Purpose: Children with Down syndrome (constitutive trisomy 21) that develop acute lymphoblastic leukemia (DS-ALL) have a 3-fold increased likelihood of treatment-related mortality coupled with a higher cumulative incidence of relapse, compared with other children with B-cell acute lymphoblastic leukemia (B-ALL). This highlights the lack of suitable treatment for Down syndrome children with B-ALL., Experimental Design: To facilitate the translation of new therapeutic agents into clinical trials, we built the first preclinical cohort of patient-derived xenograft (PDX) models of DS-ALL, comprehensively characterized at the genetic and transcriptomic levels, and have proven its suitability for preclinical studies by assessing the efficacy of drug combination between the MEK inhibitor trametinib and conventional chemotherapy agents., Results: Whole-exome and RNA-sequencing experiments revealed a high incidence of somatic alterations leading to RAS/MAPK pathway activation in our cohort of DS-ALL, as well as in other pediatric B-ALL presenting somatic gain of the chromosome 21 (B-ALL+21). In murine and human B-cell precursors, activated KRAS
G12D functionally cooperates with trisomy 21 to deregulate transcriptional networks that promote increased proliferation and self renewal, as well as B-cell differentiation blockade. Moreover, we revealed that inhibition of RAS/MAPK pathway activation using the MEK1/2 inhibitor trametinib decreased leukemia burden in several PDX models of B-ALL+21, and enhanced survival of DS-ALL PDX in combination with conventional chemotherapy agents such as vincristine., Conclusions: Altogether, using novel and suitable PDX models, this study indicates that RAS/MAPK pathway inhibition represents a promising strategy to improve the outcome of Down syndrome children with B-cell precursor leukemia., (©2020 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
34. Acute Megakaryocytic Leukemia.
- Author
-
McNulty M and Crispino JD
- Subjects
- Animals, Child, GATA1 Transcription Factor genetics, Humans, Mutation, Down Syndrome complications, Down Syndrome genetics, Leukemia, Megakaryoblastic, Acute etiology, Leukemia, Megakaryoblastic, Acute genetics
- Abstract
Acute megakaryoblastic leukemia (AMKL) is a rare malignancy affecting megakaryocytes, platelet-producing cells that reside in the bone marrow. Children with Down syndrome (DS) are particularly prone to developing the disease and have a different age of onset, distinct genetic mutations, and better prognosis as compared with individuals without DS who develop the disease. Here, we discuss the contributions of chromosome 21 genes and other genetic mutations to AMKL, the clinical features of the disease, and the differing features of DS- and non-DS-AMKL. Further studies elucidating the role of chromosome 21 genes in this disease may aid our understanding of how they function in other types of leukemia, in which they are frequently mutated or differentially expressed. Although researchers have made many insights into understanding AMKL, much more remains to be learned about its underlying molecular mechanisms., (Copyright © 2020 Cold Spring Harbor Laboratory Press; all rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
35. GATA1 mutations in red cell disorders.
- Author
-
Ling T and Crispino JD
- Subjects
- Hematologic Diseases genetics, Humans, Red-Cell Aplasia, Pure genetics, GATA1 Transcription Factor genetics, Hematologic Diseases pathology, Mutation, Red-Cell Aplasia, Pure pathology
- Abstract
GATA1 is an essential regulator of erythroid cell gene expression and maturation. In its absence, erythroid progenitors are arrested in differentiation and undergo apoptosis. Much has been learned about GATA1 function through animal models, which include genetic knockouts as well as ones with decreased levels of expression. However, even greater insights have come from the finding that a number of rare red cell disorders, including Diamond-Blackfan anemia, are associated with GATA1 mutations. These mutations affect the amino-terminal zinc finger (N-ZF) and the amino-terminus of the protein, and in both cases can alter the DNA-binding activity, which is primarily conferred by the third functional domain, the carboxyl-terminal zinc finger (C-ZF). Here we discuss the role of GATA1 in erythropoiesis with an emphasis on the mutations found in human patients with red cell disorders., (© 2019 International Union of Biochemistry and Molecular Biology.)
- Published
- 2020
- Full Text
- View/download PDF
36. Preface to IUBMB life special issue on GATA transcription factors in development, differentiation, and disease.
- Author
-
Strouboulis J and Crispino JD
- Subjects
- Animals, GATA Transcription Factors genetics, Humans, Leukemia genetics, Leukemia metabolism, Cell Differentiation, GATA Transcription Factors metabolism, Leukemia pathology, Molecular Biology, Periodicals as Topic
- Published
- 2020
- Full Text
- View/download PDF
37. Biological Characterization of 8-Cyclopropyl-2-(pyridin-3-yl)thiazolo[5,4- f ]quinazolin-9(8 H )-one, a Promising Inhibitor of DYRK1A.
- Author
-
Fruit C, Couly F, Bhansali R, Rammohan M, Lindberg MF, Crispino JD, Meijer L, and Besson T
- Abstract
Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) hyperactivity has been linked to the development of a number of human malignancies. DYRK1A is the most studied family member, and the discovery of novel specific inhibitors is attracting considerable interest. The 8-cyclopropyl-2(pyridin-3-yl)thiazolo[5,4- f ]quinazolin-9(8 H )-one (also called FC162 ) was found to be a promising inhibitor of DYRK1A and was characterized in biological experiments, by western transfer and flow cytometry on SH-SY5Y and pre-B cells. Here, the results obtained with FC162 are compared to well-characterized known DYRK1A inhibitors (e.g., Leucettine L41 and EHT1610 ).
- Published
- 2019
- Full Text
- View/download PDF
38. Chromatin occupancy and epigenetic analysis reveal new insights into the function of the GATA1 N terminus in erythropoiesis.
- Author
-
Ling T, Birger Y, Stankiewicz MJ, Ben-Haim N, Kalisky T, Rein A, Kugler E, Chen W, Fu C, Zhang K, Patel H, Sikora JW, Goo YA, Kelleher N, Zou L, Izraeli S, and Crispino JD
- Subjects
- Anemia, Diamond-Blackfan genetics, Anemia, Diamond-Blackfan physiopathology, Animals, Chromatin genetics, Epigenesis, Genetic genetics, Mice, Mice, Mutant Strains, Protein Isoforms, Erythropoiesis genetics, GATA1 Transcription Factor genetics
- Abstract
Mutations in GATA1, which lead to expression of the GATA1s isoform that lacks the GATA1 N terminus, are seen in patients with Diamond-Blackfan anemia (DBA). In our efforts to better understand the connection between GATA1s and DBA, we comprehensively studied erythropoiesis in Gata1s mice. Defects in yolks sac and fetal liver hematopoiesis included impaired terminal maturation and reduced numbers of erythroid progenitors. RNA-sequencing revealed that both erythroid and megakaryocytic gene expression patterns were altered by the loss of the N terminus, including aberrant upregulation of Gata2 and Runx1. Dysregulation of global H3K27 methylation was found in the erythroid progenitors upon loss of N terminus of GATA1. Chromatin-binding assays revealed that, despite similar occupancy of GATA1 and GATA1s, there was a striking reduction of H3K27me3 at regulatory elements of the Gata2 and Runx1 genes. Consistent with the observation that overexpression of GATA2 has been reported to impair erythropoiesis, we found that haploinsufficiency of Gata2 rescued the erythroid defects of Gata1s fetuses. Together, our integrated genomic analysis of transcriptomic and epigenetic signatures reveals that, Gata1 mice provide novel insights into the role of the N terminus of GATA1 in transcriptional regulation and red blood cell maturation which may potentially be useful for DBA patients., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
39. Activation of JAK/STAT Signaling in Megakaryocytes Sustains Myeloproliferation In Vivo .
- Author
-
Woods B, Chen W, Chiu S, Marinaccio C, Fu C, Gu L, Bulic M, Yang Q, Zouak A, Jia S, Suraneni PK, Xu K, Levine RL, Crispino JD, and Wen QJ
- Subjects
- Animals, Bone Marrow metabolism, Bone Marrow pathology, Cell Proliferation physiology, Female, Humans, Janus Kinase 2 genetics, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Myeloproliferative Disorders genetics, Point Mutation, STAT5 Transcription Factor genetics, Signal Transduction, Janus Kinase 2 metabolism, Megakaryocytes metabolism, Megakaryocytes pathology, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, STAT5 Transcription Factor metabolism
- Abstract
Purpose: The myeloproliferative neoplasms (MPN), including polycythemia vera, essential thrombocythemia, and primary myelofibrosis, are characterized by the expansion of the erythroid, megakaryocytic, and granulocytic lineages. A common feature of these disorders is the presence of abnormal megakaryocytes, which have been implicated as causative agents in the development of bone marrow fibrosis. However, the specific contributions of megakaryocytes to MPN pathogenesis remain unclear., Experimental Design: We used Pf4-Cre transgenic mice to drive expression of JAK2
V617F in megakaryocyte lineage-committed hematopoietic cells. We also assessed the critical role of mutant megakaryocytes in MPN maintenance through cell ablation studies in JAK2V617F and MPLW515L BMT models of MPN., Results: JAK2V617F -mutant presence in megakaryocytes was sufficient to induce enhanced erythropoiesis and promote fibrosis, which leads to a myeloproliferative state with expansion of mutant and nonmutant hematopoietic cells. The increased erythropoiesis was associated with elevated IL6 level, which was also required for aberrant erythropoiesis in vivo . Furthermore, depletion of megakaryocytes in the JAK2V617F and MPLW515L BMT models ameliorated polycythemia and leukocytosis in addition to expected effects on megakaryopoiesis., Conclusions: Our observations reveal that JAK/STAT pathway activation in megakaryocytes induces myeloproliferation and is necessary for MPN maintenance in vivo . These observations indicate that MPN clone can influence the behavior of the wild-type hematopoietic milieu, at least, in part, via altered production of proinflammatory cytokines and chemokines. Our findings resonate with patients who present with a clinical MPN and a low JAK2V617F allele burden, and support the development of MPN therapies aimed at targeting megakaryocytes., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
40. Mechanisms of Progression of Myeloid Preleukemia to Transformed Myeloid Leukemia in Children with Down Syndrome.
- Author
-
Labuhn M, Perkins K, Matzk S, Varghese L, Garnett C, Papaemmanuil E, Metzner M, Kennedy A, Amstislavskiy V, Risch T, Bhayadia R, Samulowski D, Hernandez DC, Stoilova B, Iotchkova V, Oppermann U, Scheer C, Yoshida K, Schwarzer A, Taub JW, Crispino JD, Weiss MJ, Hayashi Y, Taga T, Ito E, Ogawa S, Reinhardt D, Yaspo ML, Campbell PJ, Roberts I, Constantinescu SN, Vyas P, Heckl D, and Klusmann JH
- Published
- 2019
- Full Text
- View/download PDF
41. Myelofibrosis in 2019: moving beyond JAK2 inhibition.
- Author
-
Schieber M, Crispino JD, and Stein B
- Subjects
- Female, History, 21st Century, Humans, Male, Primary Myelofibrosis pathology, Protein Kinase Inhibitors pharmacology, Gene Amplification genetics, Janus Kinase 2 antagonists & inhibitors, Primary Myelofibrosis genetics, Protein Kinase Inhibitors therapeutic use
- Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by ineffective clonal hematopoiesis, splenomegaly, bone marrow fibrosis, and the propensity for transformation to acute myeloid leukemia. The discovery of mutations in JAK2, CALR, and MPL have uncovered activated JAK-STAT signaling as a primary driver of MF, supporting a rationale for JAK inhibition. However, JAK inhibition alone is insufficient for long-term remission and offers modest, if any, disease-modifying effects. Given this, there is great interest in identifying mechanisms that cooperate with JAK-STAT signaling to predict disease progression and rationally guide the development of novel therapies. This review outlines the latest discoveries in the biology of MF, discusses current clinical management of patients with MF, and summarizes the ongoing clinical trials that hope to change the landscape of MF treatment.
- Published
- 2019
- Full Text
- View/download PDF
42. Aurora Kinase A Inhibition Provides Clinical Benefit, Normalizes Megakaryocytes, and Reduces Bone Marrow Fibrosis in Patients with Myelofibrosis: A Phase I Trial.
- Author
-
Gangat N, Marinaccio C, Swords R, Watts JM, Gurbuxani S, Rademaker A, Fought AJ, Frankfurt O, Altman JK, Wen QJ, Farnoud N, Famulare CA, Patel A, Tapia R, Vallapureddy RR, Barath S, Graf A, Handlogten A, Zblewski D, Patnaik MM, Al-Kali A, Dinh YT, Englund Prahl K, Patel S, Nobrega JC, Tejera D, Thomassen A, Gao J, Ji P, Rampal RK, Giles FJ, Tefferi A, Stein B, and Crispino JD
- Subjects
- Aurora Kinase A, Fibrosis, Humans, Janus Kinase 2, Megakaryocytes, Primary Myelofibrosis
- Abstract
Purpose: Myelofibrosis is characterized by bone marrow fibrosis, atypical megakaryocytes, splenomegaly, constitutional symptoms, thrombotic and hemorrhagic complications, and a risk of evolution to acute leukemia. The JAK kinase inhibitor ruxolitinib provides therapeutic benefit, but the effects are limited. The purpose of this study was to determine whether targeting AURKA, which has been shown to increase maturation of atypical megakaryocytes, has potential benefit for patients with myelofibrosis., Patients and Methods: Twenty-four patients with myelofibrosis were enrolled in a phase I study at three centers. The objective of the study was to evaluate the safety and preliminary efficacy of alisertib. Correlative studies involved assessment of the effect of alisertib on the megakaryocyte lineage, allele burden, and fibrosis., Results: In addition to being well tolerated, alisertib reduced splenomegaly and symptom burden in 29% and 32% of patients, respectively, despite not consistently reducing the degree of inflammatory cytokines. Moreover, alisertib normalized megakaryocytes and reduced fibrosis in 5 of 7 patients for whom sequential marrows were available. Alisertib also decreased the mutant allele burden in a subset of patients., Conclusions: Given the limitations of ruxolitinib, novel therapies are needed for myelofibrosis. In this study, alisertib provided clinical benefit and exhibited the expected on-target effect on the megakaryocyte lineage, resulting in normalization of these cells and reduced fibrosis in the majority of patients for which sequential marrows were available. Thus, AURKA inhibition should be further developed as a therapeutic option in myelofibrosis. See related commentary by Piszczatowski and Steidl, p. 4868 ., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
43. GATA-1: A potential novel biomarker for the differentiation of essential thrombocythemia and myelofibrosis.
- Author
-
Lally J, Boasman K, Brown L, Martinelli V, Cappuccio I, Sovani V, Marinaccio C, Crispino JD, Graham C, and Rinaldi C
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers metabolism, Bone Marrow metabolism, Case-Control Studies, Diagnosis, Differential, Female, GATA1 Transcription Factor genetics, Gene Expression, Humans, Male, Middle Aged, NF-E2 Transcription Factor, p45 Subunit genetics, Primary Myelofibrosis genetics, Primary Myelofibrosis metabolism, Proto-Oncogene Protein c-fli-1 genetics, RNA, Messenger blood, RNA, Messenger genetics, Thrombocythemia, Essential genetics, Thrombocythemia, Essential metabolism, GATA1 Transcription Factor metabolism, Primary Myelofibrosis diagnosis, Thrombocythemia, Essential diagnosis
- Abstract
Essentials The BCR-ABL negative myeloproliferative neoplasms are subjected to unknown phenotypic modifiers. GATA-1 is upregulated in ET patients, regardless of treatment regimen or mutational status. Myelofibrosis (MF) megakaryocytes displayed decreased GATA-1 staining. GATA-1 may have utility as a diagnostic marker in ET and in its differential diagnosis from MF. ABSTRACT: Background The BCR-ABL-negative myeloproliferative neoplasms, i.e., polycythemia vera, essential thrombocythemia (ET), and myelofibrosis (MF), are characterized by mutations in JAK2, CALR, or MPL. However, an as yet unknown factor drives the precise disease phenotype. The hematopoietic transcription factor GATA-1 and its downstream targets NFE2 and FLI1 are responsible for determining erythroid and megakaryocyte lineages during hematopoietic stem cell differentiation. Previous studies have demonstrated a low level of GATA-1 expression in megakaryocytes from patients with MF. Objectives and methods The expression of GATA-1, NFE2 and FLI1 was studied for changes in the peripheral blood (PB) of ET patients. Peripheral blood samples were obtained from 36 ET patients, 14 MF patients, and seven healthy control donors. Total RNA from PB mononuclear cells (PBMCs) was extracted, and quantitative polymerase chain reaction was used to determine relative changes in gene expression. Protein levels of GATA-1 were also determined in bone marrow sections from ET and MF patients. Results GATA-1 mRNA was upregulated in ET patients, regardless of treatment regimen or mutational status. FLI1 expression was significantly downregulated, whereas NFE2 expression was unaffected by changes in GATA-1 mRNA levels. Megakaryocytes from ET patients showed increased protein levels of GATA-1 as compared with those from MF patients. Conclusions Our results confirmed, in PB, our previous data demonstrating elevated levels of GATA-1 mRNA in total bone marrow of ET patients. GATA-1 mRNA levels are independent of cytoreductive therapies, and may have utility as a diagnostic marker in ET and in its differential diagnosis from MF., (© 2019 International Society on Thrombosis and Haemostasis.)
- Published
- 2019
- Full Text
- View/download PDF
44. Regulation of MLL/COMPASS stability through its proteolytic cleavage by taspase1 as a possible approach for clinical therapy of leukemia.
- Author
-
Zhao Z, Wang L, Volk AG, Birch NW, Stoltz KL, Bartom ET, Marshall SA, Rendleman EJ, Nestler CM, Shilati J, Schiltz GE, Crispino JD, and Shilatifard A
- Subjects
- Animals, Chromatin metabolism, Disease Models, Animal, Disease Progression, Endopeptidases genetics, Enzyme Inhibitors pharmacology, Gene Knockout Techniques, HCT116 Cells, HEK293 Cells, Humans, Leukemia enzymology, Leukemia genetics, MCF-7 Cells, Mice, Myeloid-Lymphoid Leukemia Protein metabolism, Protein Stability, Survival Analysis, Endopeptidases metabolism, Leukemia therapy, Myeloid-Lymphoid Leukemia Protein genetics
- Abstract
Chromosomal translocations of the Mixed-lineage leukemia 1 ( MLL1 ) gene generate MLL chimeras that drive the pathogenesis of acute myeloid and lymphoid leukemia. The untranslocated MLL1 is a substrate for proteolytic cleavage by the endopeptidase threonine aspartase 1 (taspase1); however, the biological significance of MLL1 cleavage by this endopeptidase remains unclear. Here, we demonstrate that taspase1-dependent cleavage of MLL1 results in the destabilization of MLL. Upon loss of taspase1, MLL1 association with chromatin is markedly increased due to the stabilization of its unprocessed version, and this stabilization of the uncleaved MLL1 can result in the displacement of MLL chimeras from chromatin in leukemic cells. Casein kinase II (CKII) phosphorylates MLL1 proximal to the taspase1 cleavage site, facilitating its cleavage, and pharmacological inhibition of CKII blocks taspase1-dependent MLL1 processing, increases MLL1 stability, and results in the displacement of the MLL chimeras from chromatin. Accordingly, inhibition of CKII in a MLL-AF9 mouse model of leukemia delayed leukemic progression in vivo. This study provides insights into the direct regulation of the stability of MLL1 through its cleavage by taspase1, which can be harnessed for targeted therapeutic approaches for the treatment of aggressive leukemia as the result of MLL translocations., (© 2019 Zhao et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
45. AKT activation is a feature of CALR mutant myeloproliferative neoplasms.
- Author
-
Fu C, Wen QJ, Marinaccio C, Ling T, Chen W, Bulic M, Lasho T, Tefferi A, Crispino JD, and Xu K
- Subjects
- Cells, Cultured, Hematopoietic Stem Cells metabolism, Humans, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Proto-Oncogene Proteins c-akt genetics, Calreticulin genetics, Hematopoietic Stem Cells pathology, Mutation, Myeloproliferative Disorders pathology, Proto-Oncogene Proteins c-akt metabolism
- Published
- 2019
- Full Text
- View/download PDF
46. An activating mutation of the NSD2 histone methyltransferase drives oncogenic reprogramming in acute lymphocytic leukemia.
- Author
-
Swaroop A, Oyer JA, Will CM, Huang X, Yu W, Troche C, Bulic M, Durham BH, Wen QJ, Crispino JD, MacKerell AD Jr, Bennett RL, Kelleher NL, and Licht JD
- Subjects
- Amino Acid Substitution, HeLa Cells, Heterografts, Humans, Neoplasm Invasiveness, Neoplasm Transplantation, Cellular Reprogramming, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Mutation, Missense, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma enzymology, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Repressor Proteins genetics, Repressor Proteins metabolism
- Abstract
NSD2, a histone methyltransferase specific for methylation of histone 3 lysine 36 (H3K36), exhibits a glutamic acid to lysine mutation at residue 1099 (E1099K) in childhood acute lymphocytic leukemia (ALL), and cells harboring this mutation can become the predominant clone in relapsing disease. We studied the effects of this mutant enzyme in silico, in vitro, and in vivo using gene edited cell lines. The E1099K mutation altered enzyme/substrate binding and enhanced the rate of H3K36 methylation. As a result, cell lines harboring E1099K exhibit increased H3K36 dimethylation and reduced H3K27 trimethylation, particularly on nucleosomes containing histone H3.1. Mutant NSD2 cells exhibit reduced apoptosis and enhanced proliferation, clonogenicity, adhesion, and migration. In mouse xenografts, mutant NSD2 cells are more lethal and brain invasive than wildtype cells. Transcriptional profiling demonstrates that mutant NSD2 aberrantly activates factors commonly associated with neural and stromal lineages in addition to signaling and adhesion genes. Identification of these pathways provides new avenues for therapeutic interventions in NSD2 dysregulated malignancies.
- Published
- 2019
- Full Text
- View/download PDF
47. USP7 Cooperates with NOTCH1 to Drive the Oncogenic Transcriptional Program in T-Cell Leukemia.
- Author
-
Jin Q, Martinez CA, Arcipowski KM, Zhu Y, Gutierrez-Diaz BT, Wang KK, Johnson MR, Volk AG, Wang F, Wu J, Grove C, Wang H, Sokirniy I, Thomas PM, Goo YA, Abshiru NA, Hijiya N, Peirs S, Vandamme N, Berx G, Goosens S, Marshall SA, Rendleman EJ, Takahashi YH, Wang L, Rawat R, Bartom ET, Collings CK, Van Vlierberghe P, Strikoudis A, Kelly S, Ueberheide B, Mantis C, Kandela I, Bourquin JP, Bornhauser B, Serafin V, Bresolin S, Paganin M, Accordi B, Basso G, Kelleher NL, Weinstock J, Kumar S, Crispino JD, Shilatifard A, and Ntziachristos P
- Subjects
- Animals, Carcinogenesis genetics, Cell Proliferation genetics, Gene Expression Regulation, Neoplastic genetics, Genetic Therapy, Humans, Jurkat Cells, Leukemia, T-Cell pathology, Leukemia, T-Cell therapy, Mice, Signal Transduction genetics, Xenograft Model Antitumor Assays, Jumonji Domain-Containing Histone Demethylases genetics, Leukemia, T-Cell genetics, Receptor, Notch1 genetics, Ubiquitin-Specific Peptidase 7 genetics
- Abstract
Purpose: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease, affecting children and adults. Chemotherapy treatments show high response rates but have debilitating effects and carry risk of relapse. Previous work implicated NOTCH1 and other oncogenes. However, direct inhibition of these pathways affects healthy tissues and cancer alike. Our goal in this work has been to identify enzymes active in T-ALL whose activity could be targeted for therapeutic purposes., Experimental Design: To identify and characterize new NOTCH1 druggable partners in T-ALL, we coupled studies of the NOTCH1 interactome to expression analysis and a series of functional analyses in cell lines, patient samples, and xenograft models., Results: We demonstrate that ubiquitin-specific protease 7 (USP7) interacts with NOTCH1 and controls leukemia growth by stabilizing the levels of NOTCH1 and JMJD3 histone demethylase. USP7 is highly expressed in T-ALL and is transcriptionally regulated by NOTCH1. In turn, USP7 controls NOTCH1 levels through deubiquitination. USP7 binds oncogenic targets and controls gene expression through stabilization of NOTCH1 and JMJD3 and ultimately H3K27me3 changes. We also show that USP7 and NOTCH1 bind T-ALL superenhancers, and inhibition of USP7 leads to a decrease of the transcriptional levels of NOTCH1 targets and significantly blocks T-ALL cell growth in vitro and in vivo ., Conclusions: These results provide a new model for USP7 deubiquitinase activity through recruitment to oncogenic chromatin loci and regulation of both oncogenic transcription factors and chromatin marks to promote leukemia. Our studies also show that targeting USP7 inhibition could be a therapeutic strategy in aggressive leukemia., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
48. Dynamins 2 and 3 control the migration of human megakaryocytes by regulating CXCR4 surface expression and ITGB1 activity.
- Author
-
Suraneni PK, Corey SJ, Hession MJ, Ishaq R, Awomolo A, Hasan S, Shah C, Liu H, Wickrema A, Debili N, Crispino JD, Eklund EA, and Chen Y
- Subjects
- Actin Cytoskeleton, Cell Line, Cell Membrane metabolism, Cell Movement, Dynamin II antagonists & inhibitors, Dynamin II genetics, Dynamin III antagonists & inhibitors, Dynamin III genetics, Humans, Megakaryocytes cytology, Megakaryocytes metabolism, RNA Interference, RNA, Small Interfering metabolism, rab GTP-Binding Proteins metabolism, rhoA GTP-Binding Protein metabolism, Dynamin II metabolism, Dynamin III metabolism, Integrin beta1 metabolism, Receptors, CXCR4 metabolism
- Abstract
Megakaryocyte (MK) migration from the bone marrow periosteal niche toward the vascular niche is a prerequisite for proplatelet extension and release into the circulation. The mechanism for this highly coordinated process is poorly understood. Here we show that dynasore (DNSR), a small-molecule inhibitor of dynamins (DNMs), or short hairpin RNA knockdown of DNM2 and DNM3 impairs directional migration in a human MK cell line or MKs derived from cultured CD34
+ cells. Because cell migration requires actin cytoskeletal rearrangements, we measured actin polymerization and the activity of cytoskeleton regulator RhoA and found them to be decreased after inhibition of DNM2 and DNM3. Because SDF-1α is important for hematopoiesis, we studied the expression of its receptor CXCR4 in DNSR-treated cells. CXCR4 expression on the cell surface was increased, at least partially because of slower endocytosis and internalization after SDF-1α treatment. Combined inhibition of DNM2 and DNM3 or forced expression of dominant-negative Dnm2-K44A or GTPase-defective DNM3 diminished β1 integrin (ITGB1) activity. DNSR-treated MKs showed an abnormally clustered staining pattern of Rab11, a marker of recycling endosomes. This suggests decreased recruitment of the recycling pathway in DNSR-treated cells. Altogether, we show that the GTPase activity of DNMs, which governs endocytosis and regulates cell receptor trafficking, exerts control on MK migration toward SDF-1α gradients, such as those originating from the vascular niche. DNMs play a critical role in MKs by triggering membrane-cytoskeleton rearrangements downstream of CXCR4 and integrins., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
49. Dual Targeting of Aurora Kinases with AMG 900 Exhibits Potent Preclinical Activity Against Acute Myeloid Leukemia with Distinct Post-Mitotic Outcomes.
- Author
-
Payton M, Cheung HK, Ninniri MSS, Marinaccio C, Wayne WC, Hanestad K, Crispino JD, Juan G, and Coxon A
- Subjects
- Animals, Apoptosis drug effects, Aurora Kinases metabolism, Cell Cycle Checkpoints drug effects, Cell Differentiation drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Female, G1 Phase drug effects, Humans, Megakaryocytes drug effects, Megakaryocytes metabolism, Megakaryocytes pathology, Mice, Organophosphates pharmacology, Polyploidy, Protein Isoforms metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Quinazolines pharmacology, Tumor Burden, Tumor Suppressor Protein p53 metabolism, Xenograft Model Antitumor Assays, Aurora Kinases antagonists & inhibitors, Leukemia, Myeloid, Acute pathology, Mitosis drug effects, Phthalazines pharmacology
- Abstract
Aurora kinase A and B have essential and non-overlapping roles in mitosis, with elevated expression in a subset of human cancers, including acute myeloid leukemia (AML). In this study, pan-aurora kinase inhibitor (AKI) AMG 900 distinguishes itself as an anti-leukemic agent that is more uniformly potent against a panel of AML cell lines than are isoform-selective AKIs and classic AML drugs. AMG 900 inhibited AML cell growth by inducing polyploidization and/or apoptosis. AMG 900 and aurora-B-selective inhibitor AZD1152-hQPA showed comparable cellular effects on AML lines that do not harbor a FLT3 -ITD mutation. AMG 900 was active against P-glycoprotein-expressing AML cells resistant to AZD1152-hQPA and was effective at inducing expression of megakaryocyte-lineage markers (CD41, CD42) on human CHRF-288-11 cells and mouse Jak2
V617F cells. In MOLM-13 cells, inhibition of p-histone H3 by AMG 900 was associated with polyploidy, extra centrosomes, accumulation of p53 protein, apoptosis, and cleavage of Bcl-2 protein. Co-administration of cytarabine (Ara-C) with AMG 900 potentiated cell killing in a subset of AML lines, with evidence of attenuated polyploidization. AMG 900 inhibited the proliferation of primary human bone marrow cells in culture, with a better proliferation recovery profile relative to classic antimitotic drug docetaxel. In vivo , AMG 900 significantly reduced tumor burden in a systemic MOLM-13 xenograft model where we demonstrate the utility of 3'-deoxy-3'-18 F-fluorothymidine [18 F]FLT positron emission tomographic (PET)-CT imaging to measure the antiproliferative effects of AMG 900 in skeletal tissues in mice., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
50. A CHAF1B-Dependent Molecular Switch in Hematopoiesis and Leukemia Pathogenesis.
- Author
-
Volk A, Liang K, Suraneni P, Li X, Zhao J, Bulic M, Marshall S, Pulakanti K, Malinge S, Taub J, Ge Y, Rao S, Bartom E, Shilatifard A, and Crispino JD
- Subjects
- Adult, Animals, Binding Sites physiology, Cell Differentiation physiology, Cell Line, Tumor, Cell Proliferation genetics, Chromatin Assembly Factor-1 genetics, Exoribonucleases, Female, Hematopoiesis genetics, Humans, Jurkat Cells, Leukemia, Myeloid, Acute genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Binding physiology, Proteins genetics, Repressor Proteins, Ribonucleases, CCAAT-Enhancer-Binding Proteins metabolism, Chromatin metabolism, Chromatin Assembly Factor-1 metabolism, Hematopoiesis physiology, Leukemia, Myeloid, Acute pathology, Nucleosomes metabolism, Proteins metabolism
- Abstract
CHAF1B is the p60 subunit of the chromatin assembly factor (CAF1) complex, which is responsible for assembly of histones H3.1/H4 heterodimers at the replication fork during S phase. Here we report that CHAF1B is required for normal hematopoiesis while its overexpression promotes leukemia. CHAF1B has a pro-leukemia effect by binding chromatin at discrete sites and interfering with occupancy of transcription factors that promote myeloid differentiation, such as CEBPA. Reducing Chaf1b activity by either heterozygous deletion or overexpression of a CAF1 dominant negative allele is sufficient to suppress leukemogenesis in vivo without impairing normal hematopoiesis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.