273 results on '"John D. Crispino"'
Search Results
2. B cell class switch recombination is regulated by DYRK1A through MSH6 phosphorylation
- Author
-
Liat Stoler-Barak, Ethan Harris, Ayelet Peres, Hadas Hezroni, Mirela Kuka, Pietro Di Lucia, Amalie Grenov, Neta Gurwicz, Meital Kupervaser, Bon Ham Yip, Matteo Iannacone, Gur Yaari, John D. Crispino, and Ziv Shulman
- Subjects
Science - Abstract
Class switch recombination (CSR) is a process by which B cells switch their immunoglobulin isotype and develop pathogen-eliminating antibodies. Here, the authors show that a protein kinase DYRK1A is required for protection from viral infection through the regulation of CSR and effective clonal expansion.
- Published
- 2023
- Full Text
- View/download PDF
3. PU.1-c-Jun interaction is crucial for PU.1 function in myeloid development
- Author
-
Xinhui Zhao, Boris Bartholdy, Yukiya Yamamoto, Erica K. Evans, Meritxell Alberich-Jordà, Philipp B. Staber, Touati Benoukraf, Pu Zhang, Junyan Zhang, Bon Q. Trinh, John D. Crispino, Trang Hoang, Mahmoud A. Bassal, and Daniel G. Tenen
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The transcription factor PU.1 recruits c-Jun as a co-activator to promoters without AP-1 binding sites, and mice with point mutations in PU.1 that disrupts the interaction between PU1 and c-Jun have defects in PU.1 dependent blood lineages, including macrophages and B cells.
- Published
- 2022
- Full Text
- View/download PDF
4. Discovery of a signaling feedback circuit that defines interferon responses in myeloproliferative neoplasms
- Author
-
Diana Saleiro, Jeremy Q. Wen, Ewa M. Kosciuczuk, Frank Eckerdt, Elspeth M. Beauchamp, Chidera V. Oku, Gavin T. Blyth, Mariafausta Fischietti, Liliana Ilut, Marco Colamonici, William Palivos, Paula A. Atsaves, Dean Tan, Masha Kocherginsky, Rona Singer Weinberg, Eleanor N. Fish, John D. Crispino, Ronald Hoffman, and Leonidas C. Platanias
- Subjects
Science - Abstract
Interferon alpha (IFNalpha) therapy is showing promising results to treat myeloproliferative neoplasms (MPNs). Here, the authors show that IFNalpha response requires ULK1 phosphorylation to induce p38-MAPK signalling but it is counteracted by ROCK1-2 activation suggesting combination therapy of IFNalpha-ROCK1-2 inhibition may improve MPNs treatment.
- Published
- 2022
- Full Text
- View/download PDF
5. Resident Self-Tissue of Proinflammatory Cytokines Rather Than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1low Mice
- Author
-
Maria Zingariello, Paola Verachi, Francesca Gobbo, Fabrizio Martelli, Mario Falchi, Maria Mazzarini, Mauro Valeri, Giuseppe Sarli, Christian Marinaccio, Johanna Melo-Cardenas, John D. Crispino, and Anna Rita Migliaccio
- Subjects
myelofibrosis ,GATA1 ,proinflammatory cytokines ,TGF-β1 ,interleukin 8 ,megakaryocytes ,Microbiology ,QR1-502 - 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 Gata1low 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
6. Mediator Kinase Phosphorylation of STAT1 S727 Promotes Growth of Neoplasms With JAK-STAT Activation
- Author
-
Ioana I. Nitulescu, Sara C. Meyer, Qiang Jeremy Wen, John D. Crispino, Madeleine E. Lemieux, Ross L. Levine, Henry E. Pelish, and Matthew D. Shair
- Subjects
STAT1 ,MPN ,CDK8 ,Kinase inhibitor ,Leukemia ,Super-enhancer ,Cortistatin A ,Ruxolitinib ,Medicine ,Medicine (General) ,R5-920 - Abstract
Constitutive JAK-STAT signaling drives the proliferation of most myeloproliferative neoplasms (MPN) and a subset of acute myeloid leukemia (AML), but persistence emerges with chronic exposure to JAK inhibitors. MPN and post-MPN AML are dependent on tyrosine phosphorylation of STATs, but the role of serine STAT1 phosphorylation remains unclear. We previously demonstrated that Mediator kinase inhibitor cortistatin A (CA) reduced proliferation of JAK2-mutant AML in vitro and in vivo and also suppressed CDK8-dependent phosphorylation of STAT1 at serine 727. Here we report that phosphorylation of STAT1 S727 promotes the proliferation of AML cells with JAK-STAT pathway activation. Inhibition of serine phosphorylation by CA promotes growth arrest and differentiation, inhibits colony formation in MPN patient samples and reduces allele burden in MPN mouse models. These results reveal that STAT1 pS727 regulates growth and differentiation in JAK-STAT activated neoplasms and suggest that Mediator kinase inhibition represents a therapeutic strategy to regulate JAK-STAT signaling.
- Published
- 2017
- Full Text
- View/download PDF
7. The Hippo-p53 pathway in megakaryopoiesis
- Author
-
Praveen K Suraneni and John D. Crispino
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2016
- Full Text
- View/download PDF
8. Global transcriptome and chromatin occupancy analysis reveal the short isoform of GATA1 is deficient for erythroid specification and gene expression
- Author
-
Timothy M. Chlon, Maureen McNulty, Benjamin Goldenson, Alexander Rosinski, and John D. Crispino
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
GATA1 is a master transcriptional regulator of the differentiation of several related myeloid blood cell types, including erythrocytes and megakaryocytes. Germ-line mutations that cause loss of full length GATA1, but allow for expression of the short isoform (GATA1s), are associated with defective erythropoiesis in a subset of patients with Diamond Blackfan Anemia. Despite extensive studies of GATA1s in megakaryopoiesis, the mechanism by which GATA1s fails to support normal erythropoiesis is not understood. In this study, we used global gene expression and chromatin occupancy analysis to compare the transcriptional activity of GATA1s to GATA1. We discovered that compared to GATA1, GATA1s is less able to activate the erythroid gene expression program and terminal differentiation in cells with dual erythroid-megakaryocytic differentiation potential. Moreover, we found that GATA1s bound to many of its erythroid-specific target genes less efficiently than full length GATA1. These results suggest that the impaired ability of GATA1s to promote erythropoiesis in DBA may be caused by failure to occupy erythroid-specific gene regulatory elements.
- Published
- 2015
- Full Text
- View/download PDF
9. A novel role for survivin in erythroblast enucleation
- Author
-
Ganesan Keerthivasan, Hui Liu, Jacob M. Gump, Steven F. Dowdy, Amittha Wickrema, and John D. Crispino
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Background Nucleus free red blood cells are unique to mammals. During their terminal stage of differentiation, mammalian erythroblasts exit the cell cycle and enucleate. We previously found that survivin, a member of the chromosomal passenger complex that is required for cytokinesis, is highly expressed in late non-dividing cells. The role of survivin in enucleating erythroblasts is not known.Design and Methods In order to identify the role of survivin in these late erythroblasts, we performed proteomic analysis on survivin-bound protein complexes purified from murine erythroleukemia cells. Various molecular and cell biological techniques were used to confirm the presence and function of this novel complex. Furthermore, we used survivinfl/fl mice to study the effect of loss of survivin in enucleating erythroblasts.Results We found that survivin failed to co-localize with its known partners' inner centromere protein or Aurora-B in enucleating erythroblasts but rather exists in a multi-protein complex with epidermal growth factor receptor substrate15 and clathrin, two proteins that mediate endocytic vesicle trafficking. As evidence for a direct role of this latter complex in enucleation, we found that knockdown of the genes reduced the efficiency of enucleation of primary human erythroblasts. We also observed that loss of survivin in murine erythroblasts inhibited enucleation and that survivin-deficient cells harbored smaller cytoplasmic vacuoles. Interestingly, vacuolin-1, a small molecule that induces vacuole fusion, rescued the defective enucleation caused by survivin deficiency.Conclusions This study identified a novel role for survivin in erythroblast enucleation through previously unknown protein partners.
- Published
- 2012
- Full Text
- View/download PDF
10. Erythroblast Enucleation
- Author
-
Ganesan Keerthivasan, Amittha Wickrema, and John D. Crispino
- Subjects
Internal medicine ,RC31-1245 - Abstract
Even though the production of orthochromatic erythroblasts can be scaled up to fulfill clinical requirements, enucleation remains one of the critical rate-limiting steps in the production of transfusable red blood cells. Mammalian erythrocytes extrude their nucleus prior to entering circulation, likely to impart flexibility and improve the ability to traverse through capillaries that are half the size of erythrocytes. Recently, there have been many advances in our understanding of the mechanisms underlying mammalian erythrocyte enucleation. This review summarizes these advances, discusses the possible future directions in the field, and evaluates the prospects for improved ex vivo production of red blood cells.
- Published
- 2011
- Full Text
- View/download PDF
11. CALR goes rogue
- Author
-
Johanna Melo-Cardenas and John D. Crispino
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
12. Bone Marrow Avatars: Mimicking Hematopoiesis in a Dish
- Author
-
Marta Derecka and John D. Crispino
- Subjects
Oncology - Abstract
Summary: Faithful recapitulation of human bone marrow complexity has been a major challenge for the scientific 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).
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Dual Targeting of Aurora Kinases with AMG 900 Exhibits Potent Preclinical Activity Against Acute Myeloid Leukemia with Distinct Post-Mitotic Outcomes
- Author
-
Angela Coxon, Gloria Juan, John D. Crispino, Kelly Hanestad, William C. Wayne, Christian Marinaccio, Maria Stefania S. Ninniri, Hung-Kam Cheung, and Marc Payton
- 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 Jak2V617F 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′-18F-fluorothymidine [18F]FLT positron emission tomographic (PET)–CT imaging to measure the antiproliferative effects of AMG 900 in skeletal tissues in mice.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure from LKB1/STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms
- Author
-
John D. Crispino, Ayalew Tefferi, Grant A. Challen, Raajit K. Rampal, Ross L. Levine, Navdeep S. Chandel, Panagiotis Ntziachristos, Naseema Gangat, Ronald Hoffman, Scott T. Younger, David E. Root, Sandeep Gurbuxani, Michael Schieber, Brady Stein, Noushin Farnoud, Christopher A. Famulare, Richard P. Koche, Terra Lasho, Marinka Bulic, Te Ling, Qiang Jeremy Wen, Andrew Volk, Hamza Celik, Praveen Suraneni, and Christian Marinaccio
- Abstract
Supplementary Figure from LKB1/STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Materials and methods, Supplementary Figures S1-S8 from Dual Targeting of Aurora Kinases with AMG 900 Exhibits Potent Preclinical Activity Against Acute Myeloid Leukemia with Distinct Post-Mitotic Outcomes
- Author
-
Angela Coxon, Gloria Juan, John D. Crispino, Kelly Hanestad, William C. Wayne, Christian Marinaccio, Maria Stefania S. Ninniri, Hung-Kam Cheung, and Marc Payton
- Abstract
Supplementary Figure S1 shows the cellular effects of AKIs and anti-leukemic agents across a panel of human AML cell lines. Supplementary Figure S2 shows expression of aurora-A and aurora-B protein levels in four AML cell lines. Supplementary Figure S3 shows AMG 900 induces a dose-dependent increase in polyploidy, apoptosis, and p53 protein levels in MOLM-13 cells. Supplementary Figure S4 shows AMG 900 plus Ara-C combination matrix and CI determination in MOLM-13 cells. Supplementary Figure S5 shows AMG 900 induced apoptosis is attenuated by peptide inhibitors of caspases in MOLM-13 cells. Supplementary Figure S6 shows FC gating strategy for annexin-V coupled JC-1 assay. Supplementary Figure S7 shows the anti-proliferative effects of AMG 900 and AZD1152-hQPA on primary human bone marrow mononuclear cells in culture. Supplementary Figure S8 shows a moderate reduction in mouse body weight after AMG 900 treatment.
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Table from LKB1/STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms
- Author
-
John D. Crispino, Ayalew Tefferi, Grant A. Challen, Raajit K. Rampal, Ross L. Levine, Navdeep S. Chandel, Panagiotis Ntziachristos, Naseema Gangat, Ronald Hoffman, Scott T. Younger, David E. Root, Sandeep Gurbuxani, Michael Schieber, Brady Stein, Noushin Farnoud, Christopher A. Famulare, Richard P. Koche, Terra Lasho, Marinka Bulic, Te Ling, Qiang Jeremy Wen, Andrew Volk, Hamza Celik, Praveen Suraneni, and Christian Marinaccio
- Abstract
Supplementary Table from LKB1/STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
17. Data from LKB1/STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms
- Author
-
John D. Crispino, Ayalew Tefferi, Grant A. Challen, Raajit K. Rampal, Ross L. Levine, Navdeep S. Chandel, Panagiotis Ntziachristos, Naseema Gangat, Ronald Hoffman, Scott T. Younger, David E. Root, Sandeep Gurbuxani, Michael Schieber, Brady Stein, Noushin Farnoud, Christopher A. Famulare, Richard P. Koche, Terra Lasho, Marinka Bulic, Te Ling, Qiang Jeremy Wen, Andrew Volk, Hamza Celik, Praveen Suraneni, and Christian Marinaccio
- 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
- Published
- 2023
- Full Text
- View/download PDF
18. The chromosome 21 kinase DYRK1A: emerging roles in cancer biology and potential as a therapeutic target
- Author
-
Malini Rammohan, Ethan Harris, Rahul S. Bhansali, Emily Zhao, Loretta S. Li, and John D. Crispino
- Subjects
Cancer Research ,Genetics ,Molecular Biology - Published
- 2022
- Full Text
- View/download PDF
19. Figure S7 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
Efficacy of conventional chemotherapeutic agents alone or in combination with Trametinib in PDX B-ALL cells in vitro
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Data from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
Material and Methods, Figure legends
- Published
- 2023
- Full Text
- View/download PDF
21. Data from Activation of JAK/STAT Signaling in Megakaryocytes Sustains Myeloproliferation In Vivo
- Author
-
Qiang Jeremy Wen, John D. Crispino, Ross L. Levine, Kailin Xu, Praveen Kumar Suraneni, Shengxian Jia, Anouar Zouak, Qiong Yang, Marinka Bulic, Lilly Gu, Chunling Fu, Christian Marinaccio, Sophia Chiu, Wei Chen, and Brittany Woods
- 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 JAK2V617F 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.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Table 1 from USP7 Cooperates with NOTCH1 to Drive the Oncogenic Transcriptional Program in T-Cell Leukemia
- Author
-
Panagiotis Ntziachristos, Ali Shilatifard, John D. Crispino, Suresh Kumar, Joseph Weinstock, Neil L. Kelleher, Giuseppe Basso, Benedetta Accordi, Maddalena Paganin, Silvia Bresolin, Valentina Serafin, Beat Bornhauser, Jean-Pierre Bourquin, Irawati Kandela, Christine Mantis, Beatrix Ueberheide, Stephen Kelly, Alexandros Strikoudis, Pieter Van Vlierberghe, Clayton K. Collings, Elizabeth T. Bartom, Radhika Rawat, Lu Wang, Yoh-hei Takahashi, Emily J. Rendleman, Stacy A. Marshall, Steven Goosens, Geert Berx, Niels Vandamme, Sofie Peirs, Nobuko Hijiya, Nebiyu A. Abshiru, Young Ah Goo, Paul M. Thomas, Ivan Sokirniy, Hui Wang, Charles Grove, Jian Wu, Feng Wang, Andrew G. Volk, Megan R. Johnson, Kenneth K. Wang, Blanca T. Gutierrez-Diaz, Yixing Zhu, Kelly M. Arcipowski, Carlos A. Martinez, and Qi Jin
- Abstract
Supplementary Table 1
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Table 2 from USP7 Cooperates with NOTCH1 to Drive the Oncogenic Transcriptional Program in T-Cell Leukemia
- Author
-
Panagiotis Ntziachristos, Ali Shilatifard, John D. Crispino, Suresh Kumar, Joseph Weinstock, Neil L. Kelleher, Giuseppe Basso, Benedetta Accordi, Maddalena Paganin, Silvia Bresolin, Valentina Serafin, Beat Bornhauser, Jean-Pierre Bourquin, Irawati Kandela, Christine Mantis, Beatrix Ueberheide, Stephen Kelly, Alexandros Strikoudis, Pieter Van Vlierberghe, Clayton K. Collings, Elizabeth T. Bartom, Radhika Rawat, Lu Wang, Yoh-hei Takahashi, Emily J. Rendleman, Stacy A. Marshall, Steven Goosens, Geert Berx, Niels Vandamme, Sofie Peirs, Nobuko Hijiya, Nebiyu A. Abshiru, Young Ah Goo, Paul M. Thomas, Ivan Sokirniy, Hui Wang, Charles Grove, Jian Wu, Feng Wang, Andrew G. Volk, Megan R. Johnson, Kenneth K. Wang, Blanca T. Gutierrez-Diaz, Yixing Zhu, Kelly M. Arcipowski, Carlos A. Martinez, and Qi Jin
- Abstract
Supplementary Table 2
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Table 3 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
List of the SNVs identified through RNA-sequencing
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Table 7 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
Differentially expressed genes in the human B-ALL cohort
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Table 1 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
Characteristics of the B-ALL cohort
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Data from Activation of JAK/STAT Signaling in Megakaryocytes Sustains Myeloproliferation In Vivo
- Author
-
Qiang Jeremy Wen, John D. Crispino, Ross L. Levine, Kailin Xu, Praveen Kumar Suraneni, Shengxian Jia, Anouar Zouak, Qiong Yang, Marinka Bulic, Lilly Gu, Chunling Fu, Christian Marinaccio, Sophia Chiu, Wei Chen, and Brittany Woods
- Abstract
Supplementary figures and figure legends
- Published
- 2023
- Full Text
- View/download PDF
28. Supplementary Table 8 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
Reagents and Resources
- Published
- 2023
- Full Text
- View/download PDF
29. Figures S1-17 plus legends from USP7 Cooperates with NOTCH1 to Drive the Oncogenic Transcriptional Program in T-Cell Leukemia
- Author
-
Panagiotis Ntziachristos, Ali Shilatifard, John D. Crispino, Suresh Kumar, Joseph Weinstock, Neil L. Kelleher, Giuseppe Basso, Benedetta Accordi, Maddalena Paganin, Silvia Bresolin, Valentina Serafin, Beat Bornhauser, Jean-Pierre Bourquin, Irawati Kandela, Christine Mantis, Beatrix Ueberheide, Stephen Kelly, Alexandros Strikoudis, Pieter Van Vlierberghe, Clayton K. Collings, Elizabeth T. Bartom, Radhika Rawat, Lu Wang, Yoh-hei Takahashi, Emily J. Rendleman, Stacy A. Marshall, Steven Goosens, Geert Berx, Niels Vandamme, Sofie Peirs, Nobuko Hijiya, Nebiyu A. Abshiru, Young Ah Goo, Paul M. Thomas, Ivan Sokirniy, Hui Wang, Charles Grove, Jian Wu, Feng Wang, Andrew G. Volk, Megan R. Johnson, Kenneth K. Wang, Blanca T. Gutierrez-Diaz, Yixing Zhu, Kelly M. Arcipowski, Carlos A. Martinez, and Qi Jin
- Abstract
Supplementary Figs. 1-17 with legends
- Published
- 2023
- Full Text
- View/download PDF
30. Data from Aurora Kinase A Inhibition Provides Clinical Benefit, Normalizes Megakaryocytes, and Reduces Bone Marrow Fibrosis in Patients with Myelofibrosis: A Phase I Trial
- Author
-
John D. Crispino, Brady Stein, Ayalew Tefferi, Francis J. Giles, Raajit K. Rampal, Peng Ji, Juehua Gao, Amber Thomassen, Dalissa Tejera, Juan Carlos Nobrega, Shradha Patel, Kristen Englund Prahl, Yvonne Trang Dinh, Aref Al-kali, Mrinal M. Patnaik, Darci Zblewski, Amy Handlogten, Amy Graf, Stephanie Barath, Rangit R. Vallapureddy, Roberto Tapia, Akshar Patel, Christopher A. Famulare, Noushin Farnoud, Qiang Jeremy Wen, Jessica K. Altman, Olga Frankfurt, Angela J. Fought, Alfred Rademaker, Sandeep Gurbuxani, Justin M. Watts, Ronan Swords, Christian Marinaccio, and Naseema Gangat
- 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
- Published
- 2023
- Full Text
- View/download PDF
31. Supplementary Data from Aurora Kinase A Inhibition Provides Clinical Benefit, Normalizes Megakaryocytes, and Reduces Bone Marrow Fibrosis in Patients with Myelofibrosis: A Phase I Trial
- Author
-
John D. Crispino, Brady Stein, Ayalew Tefferi, Francis J. Giles, Raajit K. Rampal, Peng Ji, Juehua Gao, Amber Thomassen, Dalissa Tejera, Juan Carlos Nobrega, Shradha Patel, Kristen Englund Prahl, Yvonne Trang Dinh, Aref Al-kali, Mrinal M. Patnaik, Darci Zblewski, Amy Handlogten, Amy Graf, Stephanie Barath, Rangit R. Vallapureddy, Roberto Tapia, Akshar Patel, Christopher A. Famulare, Noushin Farnoud, Qiang Jeremy Wen, Jessica K. Altman, Olga Frankfurt, Angela J. Fought, Alfred Rademaker, Sandeep Gurbuxani, Justin M. Watts, Ronan Swords, Christian Marinaccio, and Naseema Gangat
- Abstract
Figure S1: Alisertib treatment did not result in a consistent cytokine response. Figure S2: Alisertib did not have a consistent effect on TGF-b levels. Figure S3: Alisertib induced GATA1 expression in the SET2 megakaryocytic cell line. Figure S4: AURKA inhibition induces megakaryocytic gene expression. Figure S5: Alisertib increases GATA1 expression in the bone marrow of patients on therapy. Table S1: Correlation of Genotype and prior JAK inhibitor therapy with response and adverse events Table S2: Summary of Treatment Related Adverse Events Table S3: Association between GATA1 staining, degree of fibrosis and clinical response
- Published
- 2023
- Full Text
- View/download PDF
32. Data from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- 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 KRASG12D 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.
- Published
- 2023
- Full Text
- View/download PDF
33. Supplementary Table 2 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
List of the SNVs identified through whole exome sequencing
- Published
- 2023
- Full Text
- View/download PDF
34. Supplementary Table 4 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
List of fusion transcripts identified through RNA-sequencing
- Published
- 2023
- Full Text
- View/download PDF
35. Data from USP7 Cooperates with NOTCH1 to Drive the Oncogenic Transcriptional Program in T-Cell Leukemia
- Author
-
Panagiotis Ntziachristos, Ali Shilatifard, John D. Crispino, Suresh Kumar, Joseph Weinstock, Neil L. Kelleher, Giuseppe Basso, Benedetta Accordi, Maddalena Paganin, Silvia Bresolin, Valentina Serafin, Beat Bornhauser, Jean-Pierre Bourquin, Irawati Kandela, Christine Mantis, Beatrix Ueberheide, Stephen Kelly, Alexandros Strikoudis, Pieter Van Vlierberghe, Clayton K. Collings, Elizabeth T. Bartom, Radhika Rawat, Lu Wang, Yoh-hei Takahashi, Emily J. Rendleman, Stacy A. Marshall, Steven Goosens, Geert Berx, Niels Vandamme, Sofie Peirs, Nobuko Hijiya, Nebiyu A. Abshiru, Young Ah Goo, Paul M. Thomas, Ivan Sokirniy, Hui Wang, Charles Grove, Jian Wu, Feng Wang, Andrew G. Volk, Megan R. Johnson, Kenneth K. Wang, Blanca T. Gutierrez-Diaz, Yixing Zhu, Kelly M. Arcipowski, Carlos A. Martinez, and Qi Jin
- 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.
- Published
- 2023
- Full Text
- View/download PDF
36. Supplementary Table 6 from Constitutive Activation of RAS/MAPK Pathway Cooperates with Trisomy 21 and Is Therapeutically Exploitable in Down Syndrome B-cell Leukemia
- Author
-
Sebastien Malinge, Thomas Mercher, Eric Delabesse, Jean-Pierre Bourquin, Paola Ballerini, Birgit Geoerger, Rishi S. Kotecha, Carole Barin Bonnigal, Elizabeth Macintyre, Olivier A. Bernard, Muriel Gaudry, John D. Crispino, Laurence C. Cheung, Beat C. Bornhauser, Nathalie Droin, Yann Lecluse, Estelle Daudigeos, Gaelle Pierron, Damien Plassard, Stephanie Lagarde, Nais Prade, Zakia Aid, Damien Roos-Weil, Yi-Chien Tsai, Silvia Jenni, M'Boyba Diop, Kunjal Panchal, Cathy Ignacimouttou, Aurélie Siret, and Anouchka P. Laurent
- Abstract
List of datasets enriched in murine B cell precursors (GSEA analyses)
- Published
- 2023
- Full Text
- View/download PDF
37. IL-13/IL-4 signaling contributes to fibrotic progression of the myeloproliferative neoplasms
- Author
-
Johanna Melo-Cardenas, Lavanya Bezavada, Jeremy Chase Crawford, Sandeep Gurbuxani, Anitria Cotton, Guolian Kang, Jeffrey Gossett, Christian Marinaccio, Rona Weinberg, Ronald Hoffman, Anna Rita Migliaccio, Yan Zheng, Marta Derecka, Ciro R. Rinaldi, and John D. Crispino
- Subjects
Interleukin-13 ,Myeloproliferative Disorders ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Fibrosis ,Mice ,Primary Myelofibrosis ,Neoplasms ,Disease Progression ,Animals ,Interleukin-4 ,Signal Transduction - 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.
- Published
- 2022
38. Gata1s mutant mice display persistent defects in the erythroid lineage
- Author
-
Te Ling, Kevin Zhang, Jiayue Yang, Sandeep Gurbuxani, and John D Crispino
- Subjects
Hematology - 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 (ML-DS), rare familial cases of dyserythropoietic anemia, and a subset of cases of Diamond-Blackfan Anemia (DBA). 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, specially impaired erythropoiesis with expanded megakaryopoiesis, during gestation. However, the mice were reported to not show any post-natal 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.
- Published
- 2022
39. Pharmacologic Inhibition of DYRK1A Results in MYC Hyperactivation and ERK Hyperphosphorylation rendering KMT2A-R ALL Cells Sensitive to BCL2 Inhibition
- Author
-
Christian Hurtz, V. S. S. Abhinav Ayyadevara, Gerald Wertheim, John A Chukinas, Joseph P Loftus, Sung June Lee, Anil Kumar, Rahul S Bhansali, Srividya Swaminathan, Huimin Geng, Thomas Milne, Xianxin Hua, Kathrin M Bernt, Thierry Besson, Junwei Shi, John D. Crispino, Martin Carroll, and Sarah K Tasian
- Abstract
KMT2A-rearranged (KMT2A-R) B cell acute lymphoblastic leukemia (ALL) is a high-risk disease in children and adults that is often chemotherapy resistant. To identify non-cytotoxic approaches to therapy, we performed a domain-specific kinome-wide CRISPR screen in KMT2A-R cell lines and patient derived xenograft samples (PDX) and identified dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) as a potential target. Pharmacologic inhibition of the KMT2A-fusion transcriptional co-regulator Menin released the KMT2A-fusion complex from the DYRK1A promoter thereby lowering DYRK1A expression levels confirming DYRK1A as a direct target of the KMT2A fusion oncogene. Direct pharmacologic inhibition of DYRK1A decreased cell proliferation of KMT2A-R ALL, thereby confirming the requirement of DYRK1A in this ALL subtype. To further understand the biologic function of DYRK1A in KMT2A-R ALL, we leveraged pharmacologic DYRK1A inhibitors in KMT2A-R PDX and cell line models. DYRK1A inhibition consistently led to upregulation of MYC protein levels, and hyperphosphorylation of ERK, which we confirmed via in vivo treatment experiments. Furthermore, DYRK1A inhibition decreased ALL burden in mice. Our results further demonstrate that DYRK1A inhibition induces the proapoptotic factor BIM, but ERK hyperphosphorylation is the driving event that induces cell cycle arrest. In contrast, combined treatment of KMT2A-R ALL cells in vitro and in vivo with DYRK1A inhibitors and the BCL2 inhibitor, venetoclax, synergistically decreases cell survival and reduced the leukemic burden in mice. Taken together these results demonstrate a unique function of DYRK1A specially in KMT2A-R ALL. Synergistic inhibition of DRYK1A and BCL2 may provide a low-toxic approach to treat this high risk ALL subtype.
- Published
- 2022
- Full Text
- View/download PDF
40. The Role of Megakaryocytes in Myelofibrosis
- Author
-
Anna Rita Migliaccio, John D. Crispino, and Johanna Melo-Cardenas
- Subjects
Inflammation ,Bone marrow fibrosis ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Megakaryocyte ,Neoplasms ,Myeloproliferation ,medicine ,Humans ,Platelet ,Myelofibrosis ,Myeloproliferative Disorders ,business.industry ,Hematology ,Janus Kinase 2 ,Tissue repair ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Primary Myelofibrosis ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,medicine.symptom ,Calreticulin ,business ,Megakaryocytes ,Receptors, Thrombopoietin ,030215 immunology - 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.
- Published
- 2021
- Full Text
- View/download PDF
41. LKB1/STK11 Is a Tumor Suppressor in the Progression of Myeloproliferative Neoplasms
- Author
-
Grant A. Challen, Ayalew Tefferi, Terra L. Lasho, Christian Marinaccio, Panagiotis Ntziachristos, Ronald Hoffman, Christopher Famulare, Hamza Celik, Marinka Bulic, Brady L. Stein, Naseema Gangat, Richard Koche, Te Ling, Praveen Suraneni, Noushin Farnoud, Ross L. Levine, David E. Root, Michael Schieber, Andrew Volk, Sandeep Gurbuxani, Raajit K. Rampal, Scott T. Younger, Navdeep S. Chandel, Qiang Jeremy Wen, and John D. Crispino
- Subjects
0301 basic medicine ,STK11 ,food and beverages ,Myeloid leukemia ,Biology ,medicine.disease ,law.invention ,03 medical and health sciences ,Osteosclerosis ,030104 developmental biology ,0302 clinical medicine ,HIF1A ,medicine.anatomical_structure ,Oncology ,Downregulation and upregulation ,law ,Fibrosis ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Suppressor ,Bone marrow - 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
- Published
- 2021
- Full Text
- View/download PDF
42. DDB1 and CUL4 Associated Factor 7 (DCAF7) Is Essential for Hematopoiesis
- Author
-
Johanna Melo-Cardenas, Lavanya Bezavada, Anitria Cotton, and John D. Crispino
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
43. FBXO11 is a candidate tumor suppressor in the leukemic transformation of myelodysplastic syndrome
- Author
-
John D. Crispino, Lyndsey Bolanos, Wendy D. Haffey, Michael Schieber, Daniel T. Starczynowski, Kenneth D. Greis, and Christian Marinaccio
- Subjects
Protein-Arginine N-Methyltransferases ,Spliceosome ,Myeloid ,medicine.medical_treatment ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,Article ,Cell Line ,law.invention ,law ,hemic and lymphatic diseases ,medicine ,Humans ,Secondary Acute Myeloid Leukemia ,Leukocyte proliferation ,Oncogenesis ,F-Box Proteins ,Tumor Suppressor Proteins ,Hematology ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Leukemia, Myeloid, Acute ,Leukemia ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Cytokine ,Oncology ,Myelodysplastic Syndromes ,Cancer research ,Suppressor ,CRISPR-Cas Systems ,Carcinogenesis ,Myelodysplastic syndrome ,Gene Deletion - 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
44. Determinants and role of chromatin organization in acute leukemia
- Author
-
Panagiotis Ntziachristos, John D. Crispino, Celestia Fang, and Sridhar Rao
- Subjects
0301 basic medicine ,CCCTC-Binding Factor ,Cancer Research ,Biology ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Enhancer ,Gene ,Regulation of gene expression ,Genetics ,Acute leukemia ,Leukemia ,Cohesin ,Hematology ,medicine.disease ,Chromatin ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,CTCF ,030220 oncology & carcinogenesis ,Acute Disease ,Mutation - 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
45. Introduction to a review series on megakaryopoiesis and platelet production
- Author
-
John D. Crispino
- Subjects
Blood Platelets ,Immunology ,Cell Biology ,Hematology ,Biochemistry ,Megakaryocytes ,Thrombopoiesis - Abstract
Hematologists have long been fascinated by how platelet production occurs, and our understanding continues to evolve as technology advances and as more and more genetic causes for thrombocytopenia are discovered. In this Review Series on megakaryopoiesis and platelet production edited by Associate Editor John Crispino, experts in the field discuss the latest insights into megakaryopoiesis in fetal and adult life, the varied roles that megakaryocytes are now recognized to play, the mechanics of platelet creation, and how all of this new knowledge underpins clinical disorders requiring diagnosis and management.
- Published
- 2022
46. The chromosome 21 kinase DYRK1A: emerging roles in cancer biology and potential as a therapeutic target
- Author
-
Malini, Rammohan, Ethan, Harris, Rahul S, Bhansali, Emily, Zhao, Loretta S, Li, and John D, Crispino
- Subjects
Chromosomes, Human, Pair 21 ,Neoplasms ,Humans ,Oncogenes ,Phosphorylation ,Protein Serine-Threonine Kinases ,Protein-Tyrosine Kinases - 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.
- Published
- 2021
47. Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape
- Author
-
Ayalew Tefferi, Naseema Gangat, Animesh Pardanani, and John D. Crispino
- Subjects
Cancer Research ,Myeloproliferative Disorders ,Oncology ,Primary Myelofibrosis ,Mutation ,Humans ,Janus Kinase 2 ,Polycythemia Vera ,Thrombocythemia, Essential - 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.
- Published
- 2021
48. Building the Future Therapies for Down Syndrome: The Third International Conference of the T21 Research Society
- Author
-
Anna Vazquez, Eric D. Hamlett, Sébastien Malinge, Yann Herault, Frances K. Wiseman, Bradley T. Christian, Laura del Hoyo Soriano, Patricia A Shaw, Floriana Costanzo, John D. Crispino, Eugenio Barone, Jean Maurice Delabar, Renata Bartesaghi, Alain D. Dekker, Juan Fortea Ormaechea, Véronique Brault, Marzia Perluigi, Peter Paul De Deyn, Mara Dierssen, Elizabeth M. C. Fisher, Tarik F. Haydar, Lisa M. Jacola, Marie-Claude Potier, Stephanie L. Sherman, Anita Bhattacharyya, Leonard J Abbeduto, William C. Mobley, Maria Florencia Iulita, Andre Strydom, Maria Carmona-Iragui, María Martínez de Lagrán, Jorge Busciglio, Pablo Helguera, Anna J. Esbensen, Barcelona Institute of Science and Technology (BIST), Universitat Pompeu Fabra [Barcelona] (UPF), CIBER de Enfermedades Raras (CIBERER), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Instituto de Investigación Médica Mercedes y Martín Ferreyra [Córdoba] (INIMEC), Universidad Nacional de Córdoba [Argentina]-Consejo Nacional de Investigaciones Científicas y Técnicas [Buenos Aires] (CONICET), University of Wisconsin-Madison, Universitat Autònoma de Barcelona (UAB), Fundació Catalana de Síndrome de Down [Barcelona], University College of London [London] (UCL), University of California [San Diego] (UC San Diego), University of California, Cincinnati Children's Hospital Medical Center, St Jude Children's Research Hospital, Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Medical University of South Carolina [Charleston] (MUSC), University of California [Davis] (UC Davis), University of California [Irvine] (UCI), Université de Montréal (UdeM), McGill University = Université McGill [Montréal, Canada], Northwestern University [Evanston], Telethon KIDS Institute, The University of Western Australia (UWA), Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome], Bambino Gesù Children’s Hospital [Rome, Italy], University of Bologna, University of Groningen [Groningen], University of Antwerp (UA), Boston University [Boston] (BU), Emory University [Atlanta, GA], King‘s College London, and Molecular Neuroscience and Ageing Research (MOLAR)
- Subjects
BIOMARKER ,Down syndrome ,media_common.quotation_subject ,Intellectual and Developmental Disabilities (IDD) ,[SDV]Life Sciences [q-bio] ,Clinical Sciences ,Intellectual disability ,Disease ,Mouse models ,s disease ,03 medical and health sciences ,Presentation ,Trisomy 21 Research Society ,medicine ,Genetics ,2.1 Biological and endogenous factors ,Language impairment ,Alzheimer’ ,Genetics (clinical) ,030304 developmental biology ,media_common ,Cancer ,0303 health sciences ,Scientific organization ,030305 genetics & heredity ,Neurodevelopmental disorders ,Neurosciences ,Cognition ,ADULTS ,Alzheimer's disease ,medicine.disease ,biomarkers ,down syndrome ,intellectual disability ,language impairment ,mouse models ,neurodevelopmental disorders ,trisomy 21 Research Society ,DYSFUNCTION ,3. Good health ,Brain Disorders ,Syndrome Conferences, Symposia or Workshops ,ALZHEIMERS-DISEASE ,GENE DOSAGE ,Engineering ethics ,Human medicine ,Trisomy ,Psychology ,Biomarkers - Abstract
International audience; Research focused on Down syndrome has increased in the last several years to advance understanding of the consequences of trisomy 21 (T21) on molecular and cellular processes and, ultimately, on individuals with Down syndrome. The Trisomy 21 Research Society (T21RS) is the premier scientific organization for researchers and clinicians studying Down syndrome. The Third International Conference of T21RS, held June 6–9, 2019, in Barcelona, Spain, brought together 429 scientists, families, and industry representatives to share the latest discoveries on underlying cellular and molecular mechanisms of T21, define cognitive and behavioral challenges and better understand comorbidities associated with Down syndrome, including Alzheimer’s disease and leukemia. Presentation of cutting-edge results in neuroscience, neurology, model systems, psychology, cancer, biomarkers and molecular and pharmacological therapeutic approaches demonstrate the compelling interest and continuing advancement in all aspects of understanding and ameliorating conditions associated with T21.
- Published
- 2021
- Full Text
- View/download PDF
49. GATA1 mutations in red cell disorders
- Author
-
John D. Crispino and Te Ling
- Subjects
0301 basic medicine ,Clinical Biochemistry ,Regulator ,Biology ,Red-Cell Aplasia, Pure ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Gene expression ,Genetics ,Humans ,GATA1 Transcription Factor ,Molecular Biology ,Gene knockout ,Zinc finger ,Red Cell ,GATA1 ,Cell Biology ,Hematologic Diseases ,Cell biology ,030104 developmental biology ,Apoptosis ,030220 oncology & carcinogenesis ,Mutation ,Erythropoiesis - 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.
- Published
- 2019
- Full Text
- View/download PDF
50. Activation of JAK/STAT Signaling in Megakaryocytes Sustains Myeloproliferation In Vivo
- Author
-
Chunling Fu, Praveen Suraneni, Sophia Chiu, Ross L. Levine, Lilly Gu, Shengxian Jia, Anouar Zouak, Wei Chen, Qiong Yang, John D. Crispino, Brittany Woods, Qiang Jeremy Wen, Marinka Bulic, Kailin Xu, and Christian Marinaccio
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Mice, Transgenic ,Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Polycythemia vera ,Megakaryocyte ,Bone Marrow ,hemic and lymphatic diseases ,Myeloproliferation ,STAT5 Transcription Factor ,medicine ,Animals ,Humans ,Point Mutation ,Myelofibrosis ,Cell Proliferation ,Megakaryopoiesis ,Myeloproliferative Disorders ,Essential thrombocythemia ,food and beverages ,Janus Kinase 2 ,medicine.disease ,Mice, Inbred C57BL ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Erythropoiesis ,Female ,Megakaryocytes ,Signal Transduction - 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 JAK2V617F 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.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.