27 results on '"Muschen, M"'
Search Results
2. Developmental partitioning of SYK and ZAP70 prevents autoimmunity and cancer
- Author
-
Sadras, T, Martin, M, Kume, K, Robinson, ME, Saravanakumar, S, Lenz, G, Chen, Z, Song, JY, Siddiqi, T, Oksa, L, Knapp, AM, Cutler, J, Cosgun, KN, Klemm, L, Ecker, V, Winchester, J, Ghergus, D, Soulas-Sprauel, P, Kiefer, F, Heisterkamp, N, Pandey, A, Ngo, V, Wang, L, Jumaa, H, Buchner, M, Ruland, J, Chan, W-C, Meffre, E, Martin, T, Muschen, M, Sadras, T, Martin, M, Kume, K, Robinson, ME, Saravanakumar, S, Lenz, G, Chen, Z, Song, JY, Siddiqi, T, Oksa, L, Knapp, AM, Cutler, J, Cosgun, KN, Klemm, L, Ecker, V, Winchester, J, Ghergus, D, Soulas-Sprauel, P, Kiefer, F, Heisterkamp, N, Pandey, A, Ngo, V, Wang, L, Jumaa, H, Buchner, M, Ruland, J, Chan, W-C, Meffre, E, Martin, T, and Muschen, M
- Abstract
Even though SYK and ZAP70 kinases share high sequence homology and serve analogous functions, their expression in B and T cells is strictly segregated throughout evolution. Here, we identified aberrant ZAP70 expression as a common feature in a broad range of B cell malignancies. We validated SYK as the kinase that sets the thresholds for negative selection of autoreactive and premalignant clones. When aberrantly expressed in B cells, ZAP70 competes with SYK at the BCR signalosome and redirects SYK from negative selection to tonic PI3K signaling, thereby promoting B cell survival. In genetic mouse models for B-ALL and B-CLL, conditional expression of Zap70 accelerated disease onset, while genetic deletion impaired malignant transformation. Inducible activation of Zap70 during B cell development compromised negative selection of autoreactive B cells, resulting in pervasive autoantibody production. Strict segregation of the two kinases is critical for normal B cell selection and represents a central safeguard against the development of autoimmune disease and B cell malignancies.
- Published
- 2021
3. Author Correction: Metabolic gatekeeper function of B-lymphoid transcription factors (Nature (2017) DOI: 10.1038/nature21076)
- Author
-
Chan L. N., Chan, L, Chen, Z, Braas, D, Lee, J, Xiao, G, Geng, H, Cosgun, K, Hurtz, C, Shojaee, S, Cazzaniga, V, Schjerven, H, Ernst, T, Hochhaus, A, Kornblau, S, Konopleva, M, Pufall, M, Cazzaniga, G, Liu, G, Milne, T, Koeffler, H, Ross, T, Sanchez-Garcia, I, Borkhardt, A, Yamamoto, K, Dickins, R, Graeber, T, Muschen, M, Chan L. N., Chen Z., Braas D., Lee J. -W., Xiao G., Geng H., Cosgun K. N., Hurtz C., Shojaee S., Cazzaniga V., Schjerven H., Ernst T., Hochhaus A., Kornblau S. M., Konopleva M., Pufall M. A., Cazzaniga G., Liu G. J., Milne T. A., Koeffler H. P., Ross T. S., Sanchez-Garcia I., Borkhardt A., Yamamoto K. R., Dickins R. A., Graeber T. G., Muschen M., Chan L. N., Chan, L, Chen, Z, Braas, D, Lee, J, Xiao, G, Geng, H, Cosgun, K, Hurtz, C, Shojaee, S, Cazzaniga, V, Schjerven, H, Ernst, T, Hochhaus, A, Kornblau, S, Konopleva, M, Pufall, M, Cazzaniga, G, Liu, G, Milne, T, Koeffler, H, Ross, T, Sanchez-Garcia, I, Borkhardt, A, Yamamoto, K, Dickins, R, Graeber, T, Muschen, M, Chan L. N., Chen Z., Braas D., Lee J. -W., Xiao G., Geng H., Cosgun K. N., Hurtz C., Shojaee S., Cazzaniga V., Schjerven H., Ernst T., Hochhaus A., Kornblau S. M., Konopleva M., Pufall M. A., Cazzaniga G., Liu G. J., Milne T. A., Koeffler H. P., Ross T. S., Sanchez-Garcia I., Borkhardt A., Yamamoto K. R., Dickins R. A., Graeber T. G., and Muschen M.
- Abstract
In Fig. 3c of this Letter, the the effects of CRISPR-Cas9-mediated deletion of NR3C1, TXNIP and CNR2 in patient-derived B-lineage leukaemia cells were shown. For curves depicting NR3C1 (left graph), data s for TXNIP (middle graph) were inadvertently plotted. This figure has been corrected online, and the original Fig. 3c is shown as Supplementary Information to this Amendment for transparency. The error does not affect the conclusions of the Letter. In addition, Source Data files have been added for the Figs. 1-4 and Extended Data Figs. 1-10 of the original Letter.
- Published
- 2018
4. Coactivation of NF-kappa B and Notch signaling is sufficient to induce B-cell transformation and enables B-myeloid conversion
- Author
-
Xiu, Y, Dong, QZ, Fu, L, Bossler, A, Tang, XB, Boyce, B, Borcherding, N, Leidinger, M, Sardina, JL, Xue, HH, Li, QC, Feldman, A, Aifantis, I, Boccalatte, F, Wang, LL, Jin, ML, Khoury, J, Wang, W, Hu, SM, Yuan, YZ, Wang, ED, Yuan, J, Janz, S, Colgan, J, Habelhah, H, Waldschmidt, T, Muschen, M, Bagg, A, Darbro, B, and Zhao, C
- Abstract
NF-kappa B and Notch signaling can be simultaneously activated in a variety of B-cell lymphomas. Patients with B-cell lymphoma occasionally develop clonally related myeloid tumors with poor prognosis. Whether concurrent activation of both pathways is sufficient to induce B-cell transformation and whether the signaling initiates B-myebid conversion in a pathological context are largely unknown. Here, we provide genetic evidence that concurrent activation of NF-kappa B and Notch signaling in committed B cells is sufficient to induce B-cell lymphomatous transformation and primes common progenitor cells to convert to myeloid lineage through dedifferentiation, not transdifferentiation. Intriguingly, the converted myeloid cells can further transform, albeit at low frequency, into myeloid leukemia. Mechanistically, coactivation of NF-kappa B and Notch signaling endows committed B cells with the ability to self renew. Downregulation of BACH2, a lymphoma and myeloid gene suppressor, but not upregulation of CEBP/alpha and/or downregulation of B-cell transcription factors, is an early event in both B-cell transformation and myeloid conversion. Interestingly, a DNA hypomethylating drug not only effectively eliminated the converted myeloid leukemia cells, but also restored the expression of green fluorescent protein, which had been lost in converted myeloid leukemia cells. Collectively, our results suggest that targeting NF-kappa B and Notch signaling will not only improve lymphoma treatment, but also prevent the lymphomato-myeloid tumor conversion. Importantly, DNA hypomethylating drugs might efficiently treat these converted myeloid neoplasms.
- Published
- 2020
5. Metabolic gatekeeper function of B-lymphoid transcription factors (vol 542, pg 479, 2017)
- Author
-
Chan, LN, Chen, Z, Braas, D, Lee, J-W, Xiao, G, Geng, H, Cosgun, KN, Hurtz, C, Shojaee, S, Cazzaniga, V, Schjerven, H, Ernst, T, Hochhaus, A, Kornblau, SM, Konopleva, M, Pufall, MA, Cazzaniga, G, Liu, GJ, Milne, TA, Koeffler, HP, Ross, TS, Sanchez-Garcia, I, Borkhardt, A, Yamamoto, KR, Dickins, RA, Graeber, TG, and Muschen, M
- Abstract
In Fig. 3c of this Letter, the the effects of CRISPR-Cas9-mediated deletion of NR3C1, TXNIP and CNR2 in patient-derived B-lineage leukaemia cells were shown. For curves depicting NR3C1 (left graph), data s for TXNIP (middle graph) were inadvertently plotted. This figure has been corrected online, and the original Fig. 3c is shown as Supplementary Information to this Amendment for transparency. The error does not affect the conclusions of the Letter. In addition, Source Data files have been added for the Figs. 1-4 and Extended Data Figs. 1-10 of the original Letter.
- Published
- 2018
6. Novel BAFF-R CAR T-cell Therapy for CD19 Antigen-loss Relapsed B Cell Tumors
- Author
-
Kwak, L.W., primary, Qin, H., additional, Dong, Z., additional, Wang, X., additional, Cheng, W.A., additional, Smith, D., additional, Song, J.Y., additional, Aldoss, I., additional, Muschen, M., additional, and Forman, S.J., additional
- Published
- 2019
- Full Text
- View/download PDF
7. Antagonism of B cell enhancer networks by STAT5 drives leukemia and poor patient survival
- Author
-
Katerndahl, C.D.S., Heltemes-Harris, L.M., Willette, M.J.L., Henzler, C.M., Frietze, S., Yang, R., Schjerven, H., Silverstein, K.A.T., Ramsey, L.B., Hubbard, G., Wells, A.D., Kuiper, R.P, Scheijen, B., Leeuwen, F.N. van, Muschen, M., Kornblau, S.M., Farrar, M.A., Katerndahl, C.D.S., Heltemes-Harris, L.M., Willette, M.J.L., Henzler, C.M., Frietze, S., Yang, R., Schjerven, H., Silverstein, K.A.T., Ramsey, L.B., Hubbard, G., Wells, A.D., Kuiper, R.P, Scheijen, B., Leeuwen, F.N. van, Muschen, M., Kornblau, S.M., and Farrar, M.A.
- Abstract
Item does not contain fulltext, The transcription factor STAT5 has a critical role in B cell acute lymphoblastic leukemia (B-ALL). How STAT5 mediates this effect is unclear. Here we found that activation of STAT5 worked together with defects in signaling components of the precursor to the B cell antigen receptor (pre-BCR), including defects in BLNK, BTK, PKCbeta, NF-kappaB1 and IKAROS, to initiate B-ALL. STAT5 antagonized the transcription factors NF-kappaB and IKAROS by opposing regulation of shared target genes. Super-enhancers showed enrichment for STAT5 binding and were associated with an opposing network of transcription factors, including PAX5, EBF1, PU.1, IRF4 and IKAROS. Patients with a high ratio of active STAT5 to NF-kappaB or IKAROS had more-aggressive disease. Our studies indicate that an imbalance of two opposing transcriptional programs drives B-ALL and suggest that restoring the balance of these pathways might inhibit B-ALL.
- Published
- 2017
8. The Public Repository of Xenografts enables discovery and randomized phase II-like trials in mice
- Author
-
Townsend, E.C. (Elizabeth), Murakami, M.A. (Mark), Christodoulou, A. (Alexandra), Christie, A.L. (Amanda), Köster, J. (Johannes), DeSouza, T.A. (Tiffany), Morgan, E.A. (Elizabeth), Kallgren, S.P. (Scott), Liu, H. (Huiyun), Wu, S.-C. (Shuo-Chieh), Plana, O. (Olivia), Montero, J. (Joan), Stevenson, K.E. (Kristen), Rao, P. (Prakash), Vadhi, R. (Raga), Andreeff, M. (Michael), Armand, P. (Philippe), Ballen, K.K. (Karen), Barzaghi-Rinaudo, P. (Patrizia), Cahill, S. (Sarah), Clark, R.A. (Rachael), Cooke, V.G. (Vesselina), Davids, M.S. (Matthew), DeAngelo, D.J. (Daniel), Dorfman, D.M., Eaton, H. (Hilary), Ebert, B.L. (Benjamin), Etchin, J. (Julia), Firestone, B. (Brant), Fisher, D.C. (David), Freedman, A.S. (Arnold), Galinsky, (), I.A. (Ilene), Gao, H. (Hui), Garcia, (), J.S. (Jacqueline), Gamache-Ottou, F. (Francine), Graubert, T.A. (Timothy), Gutierrez, A. (Alejandro), Halilovic, E. (Ensar), Harris, M.H. (Marian), Herbert, Z.T. (Zachary), Horwitz, S.M. (Steven), Inghirami, G. (Giorgio), Intlekofer, A.M. (Andrew), Ito, M. (Moriko), Izraeli, S. (Shai), Jacobsen, E.D. (Eric), Jacobson, C.A. (Caron), Jeay, S. (Sébastien), Jeremias, I. (Irmela), Kelliher, M.A. (Michelle), Koch, R. (Raphael), Konopleva, M. (Marina), Kopp, N. (Nadja), Kornblau, S.M. (Steven), Kung, A.L. (Andrew), Kupper, T.S. (Thomas), LeBoeuf, N.R. (Nicole), LaCasce, A.S. (Ann), Lees, E. (Emma), Li, L.S. (Loretta), Look, A.T. (Thomas), Murakami, M. (Masato), Muschen, M. (Markus), Neuberg, D. (Donna), Ng, S.Y. (Samuel), Odejde, O.O. (Oreofe), Orkin, S.H. (Stuart), Paquette, R.R. (Rachel), Place, A.A. (Andrew), Roderick, J.E. (Justine), Ryan, J.A. (Jeremy), Sallan, S.E. (Stephen), Shoji, B. (Brent), Silverman, L.B. (Lewis), Soiffer, R.J. (Robert), Steensma, D.P. (David), Stegmaier, K. (Kimberley), Stone, R.M. (Richard), Tamburini, J. (Jerome), Thorner, A.R. (Aaron), Hummelen, P. (Paul) van, Wadleigh, M. (Martha), Wiesmann, M. (Marion), Weng, A.P. (Andrew), Wuerthner, J.U. (Jens), Williams, D.A. (David), Wollison, B.M. (Bruce), Lane, A.A. (Andrew), Letai, A. (Anthony), Bertagnolli, M.M. (Monica), Ritz, J. (Jerome), Brown, M. (Myles), Long, H. (Henry), Aster, J.C. (Jon), Shipp, M.A. (Margaret), Griffin, J.D. (James), Weinstock, D.M. (David), Townsend, E.C. (Elizabeth), Murakami, M.A. (Mark), Christodoulou, A. (Alexandra), Christie, A.L. (Amanda), Köster, J. (Johannes), DeSouza, T.A. (Tiffany), Morgan, E.A. (Elizabeth), Kallgren, S.P. (Scott), Liu, H. (Huiyun), Wu, S.-C. (Shuo-Chieh), Plana, O. (Olivia), Montero, J. (Joan), Stevenson, K.E. (Kristen), Rao, P. (Prakash), Vadhi, R. (Raga), Andreeff, M. (Michael), Armand, P. (Philippe), Ballen, K.K. (Karen), Barzaghi-Rinaudo, P. (Patrizia), Cahill, S. (Sarah), Clark, R.A. (Rachael), Cooke, V.G. (Vesselina), Davids, M.S. (Matthew), DeAngelo, D.J. (Daniel), Dorfman, D.M., Eaton, H. (Hilary), Ebert, B.L. (Benjamin), Etchin, J. (Julia), Firestone, B. (Brant), Fisher, D.C. (David), Freedman, A.S. (Arnold), Galinsky, (), I.A. (Ilene), Gao, H. (Hui), Garcia, (), J.S. (Jacqueline), Gamache-Ottou, F. (Francine), Graubert, T.A. (Timothy), Gutierrez, A. (Alejandro), Halilovic, E. (Ensar), Harris, M.H. (Marian), Herbert, Z.T. (Zachary), Horwitz, S.M. (Steven), Inghirami, G. (Giorgio), Intlekofer, A.M. (Andrew), Ito, M. (Moriko), Izraeli, S. (Shai), Jacobsen, E.D. (Eric), Jacobson, C.A. (Caron), Jeay, S. (Sébastien), Jeremias, I. (Irmela), Kelliher, M.A. (Michelle), Koch, R. (Raphael), Konopleva, M. (Marina), Kopp, N. (Nadja), Kornblau, S.M. (Steven), Kung, A.L. (Andrew), Kupper, T.S. (Thomas), LeBoeuf, N.R. (Nicole), LaCasce, A.S. (Ann), Lees, E. (Emma), Li, L.S. (Loretta), Look, A.T. (Thomas), Murakami, M. (Masato), Muschen, M. (Markus), Neuberg, D. (Donna), Ng, S.Y. (Samuel), Odejde, O.O. (Oreofe), Orkin, S.H. (Stuart), Paquette, R.R. (Rachel), Place, A.A. (Andrew), Roderick, J.E. (Justine), Ryan, J.A. (Jeremy), Sallan, S.E. (Stephen), Shoji, B. (Brent), Silverman, L.B. (Lewis), Soiffer, R.J. (Robert), Steensma, D.P. (David), Stegmaier, K. (Kimberley), Stone, R.M. (Richard), Tamburini, J. (Jerome), Thorner, A.R. (Aaron), Hummelen, P. (Paul) van, Wadleigh, M. (Martha), Wiesmann, M. (Marion), Weng, A.P. (Andrew), Wuerthner, J.U. (Jens), Williams, D.A. (David), Wollison, B.M. (Bruce), Lane, A.A. (Andrew), Letai, A. (Anthony), Bertagnolli, M.M. (Monica), Ritz, J. (Jerome), Brown, M. (Myles), Long, H. (Henry), Aster, J.C. (Jon), Shipp, M.A. (Margaret), Griffin, J.D. (James), and Weinstock, D.M. (David)
- Abstract
More than 90% of drugs with preclinical activity fail in human trials, largely due to insufficient efficacy. We hypothesized that adequately powered trials of patient-derived xenografts (PDX) in mice could efficiently define therapeutic activity across heterogeneous tumors. To address this hypothesis, we established a large, publicly available repository of well-characterized leukemia and lymphoma PDXs that undergo orthotopic engraftment, called the Public Repository of Xenografts (PRoXe). PRoXe includes all de-identified information relevant to the primary specimens and the PDXs derived from them. Using this repository, we demonstrate that large studies of acute leukemia PDXs that mimic human randomized clinical trials can characterize drug efficacy and generate transcriptional, functional, and proteomic biomarkers in both treatment-naive and relapsed/refractory disease.
- Published
- 2016
- Full Text
- View/download PDF
9. Epigenetic regulation of gene expression by Ikaros, HDAC1 and Casein Kinase II in leukemia
- Author
-
Song, C, primary, Pan, X, additional, Ge, Z, additional, Gowda, C, additional, Ding, Y, additional, Li, H, additional, Li, Z, additional, Yochum, G, additional, Muschen, M, additional, Li, Q, additional, Payne, K J, additional, and Dovat, S, additional
- Published
- 2015
- Full Text
- View/download PDF
10. RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia
- Author
-
Papaemmanuil, E, Rapado, I, Li, Y, Potter, N, Wedge, D, Tubio, J, Alexandrov, L, Van Loo, P, Cooke, S, Marshall, J, Martincorena, I, Hinton, J, Gundem, G, Van Delft, F, Nik Zainal, S, Jones, D, Ramakrishna, M, Titley, I, Stebbings, L, Leroy, C, Menzies, A, Gamble, J, Robinson, B, Mudie, L, Raine, K, O'Meara, S, Teague, J, Butler, A, Cazzaniga, G, Biondi, A, Zuna, J, Kempski, H, Muschen, M, Ford, A, Stratton, M, Greaves, M, Campbell, P, CAZZANIGA, GIOVANNI ITALO, BIONDI, ANDREA, Campbell, P., Papaemmanuil, E, Rapado, I, Li, Y, Potter, N, Wedge, D, Tubio, J, Alexandrov, L, Van Loo, P, Cooke, S, Marshall, J, Martincorena, I, Hinton, J, Gundem, G, Van Delft, F, Nik Zainal, S, Jones, D, Ramakrishna, M, Titley, I, Stebbings, L, Leroy, C, Menzies, A, Gamble, J, Robinson, B, Mudie, L, Raine, K, O'Meara, S, Teague, J, Butler, A, Cazzaniga, G, Biondi, A, Zuna, J, Kempski, H, Muschen, M, Ford, A, Stratton, M, Greaves, M, Campbell, P, CAZZANIGA, GIOVANNI ITALO, BIONDI, ANDREA, and Campbell, P.
- Abstract
The ETV6-RUNX1 fusion gene, found in 25% of childhood acute lymphoblastic leukemia (ALL) cases, is acquired in utero but requires additional somatic mutations for overt leukemia. We used exome and low-coverage whole-genome sequencing to characterize secondary events associated with leukemic transformation. RAG-mediated deletions emerge as the dominant mutational process, characterized by recombination signal sequence motifs near breakpoints, incorporation of non-templated sequence at junctions, ∼30-fold enrichment at promoters and enhancers of genes actively transcribed in B cell development and an unexpectedly high ratio of recurrent to non-recurrent structural variants. Single-cell tracking shows that this mechanism is active throughout leukemic evolution, with evidence of localized clustering and reiterated deletions. Integration of data on point mutations and rearrangements identifies ATF7IP and MGA as two new tumor-suppressor genes in ALL. Thus, a remarkably parsimonious mutational process transforms ETV6-RUNX1-positive lymphoblasts, targeting the promoters, enhancers and first exons of genes that normally regulate B cell differentiation. © 2014 Nature America, Inc
- Published
- 2014
11. Biology and targeting options for kinase signaling in acute lymphoblastic leukemia
- Author
-
Shojaee, S., primary and Muschen, M., additional
- Published
- 2014
- Full Text
- View/download PDF
12. Identification of different Ikaros cDNA transcripts in Philadelphia-positive adult acute lymphoblastic leukemia by a high-throughput capillary electrophoresis sizing method
- Author
-
Iacobucci, I., primary, Lonetti, A., additional, Cilloni, D., additional, Messa, F., additional, Ferrari, A., additional, Zuntini, R., additional, Ferrari, S., additional, Ottaviani, E., additional, Arruga, F., additional, Paolini, S., additional, Papayannidis, C., additional, Piccaluga, P. P., additional, Soverini, S., additional, Saglio, G., additional, Pane, F., additional, Baruzzi, A., additional, Vignetti, M., additional, Berton, G., additional, Vitale, A., additional, Chiaretti, S., additional, Muschen, M., additional, Foa, R., additional, Baccarani, M., additional, and Martinelli, G., additional
- Published
- 2008
- Full Text
- View/download PDF
13. Fas ligand, tumor necrosis factor-d expression, and apoptosis during allograft rejection and tolerance
- Author
-
Josien, R., Muschen, M., Gilbert, E., Douiliard, P., Heslan, J.-M., Souliliou, J.-P., and Cuturi, M.-C.
- Published
- 1998
- Full Text
- View/download PDF
14. Deranged CD95 system in a case of Churg-Strauss vasculitis
- Author
-
Muschen, M., Warskulat, U., Haussinger, D., Moers, C., Simon, D., and Even, J.
- Abstract
GASTROENTEROLOGY 1998;114:1351-1352
- Published
- 1998
- Full Text
- View/download PDF
15. RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia
- Author
-
Elli Papaemmanuil, Nicola E. Potter, Adam Butler, Anthony M. Ford, Giovanni Cazzaniga, Ludmil B. Alexandrov, Jon W. Teague, Jose M. C. Tubio, Peter Van Loo, Gunes Gundem, Sarah O’Meara, Catherine Leroy, Inmaculada Rapado, Yang Li, Lucy Stebbings, Susanna L. Cooke, Helena Kempski, Mel Greaves, Ian Titley, David Jones, Inigo Martincorena, Manasa Ramakrishna, Ben Robinson, Jonathan Hinton, John Marshall, Michael R. Stratton, Markus Müschen, Serena Nik-Zainal, Andrew Menzies, David C. Wedge, Laura Mudie, Jan Zuna, John Gamble, Andrea Biondi, Peter J. Campbell, Keiran Raine, Frederik W. van Delft, Papaemmanuil, E, Rapado, I, Li, Y, Potter, N, Wedge, D, Tubio, J, Alexandrov, L, Van Loo, P, Cooke, S, Marshall, J, Martincorena, I, Hinton, J, Gundem, G, Van Delft, F, Nik Zainal, S, Jones, D, Ramakrishna, M, Titley, I, Stebbings, L, Leroy, C, Menzies, A, Gamble, J, Robinson, B, Mudie, L, Raine, K, O'Meara, S, Teague, J, Butler, A, Cazzaniga, G, Biondi, A, Zuna, J, Kempski, H, Muschen, M, Ford, A, Stratton, M, Greaves, M, and Campbell, P
- Subjects
DNA Copy Number Variations ,Oncogene Proteins, Fusion ,Transcription Factor ,Sequence analysis ,Basic Helix-Loop-Helix Transcription Factor ,Molecular Sequence Data ,Biology ,Precursor B-Cell Lymphoblastic Leukemia-Lymphoma ,Genetics ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Recombination signal sequences ,Humans ,Genes, Tumor Suppressor ,Gene ,Exome ,Childhood Acute Lymphoblastic Leukemia ,DNA Copy Number Variation ,Gene Library ,Sequence Deletion ,Gene Rearrangement ,Homeodomain Proteins ,Recombination, Genetic ,Base Sequence ,V(D)J recombination ,Genetic Variation ,Homeodomain Protein ,MED/38 - PEDIATRIA GENERALE E SPECIALISTICA ,Gene rearrangement ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,V(D)J Recombination ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Leukemia ,Core Binding Factor Alpha 2 Subunit ,Human ,Transcription Factors - Abstract
The ETV6-RUNX1 fusion gene, found in 25% of childhood acute lymphoblastic leukemia (ALL) cases, is acquired in utero but requires additional somatic mutations for overt leukemia. We used exome and low-coverage whole-genome sequencing to characterize secondary events associated with leukemic transformation. RAG-mediated deletions emerge as the dominant mutational process, characterized by recombination signal sequence motifs near breakpoints, incorporation of non-templated sequence at junctions, ∼30-fold enrichment at promoters and enhancers of genes actively transcribed in B cell development and an unexpectedly high ratio of recurrent to non-recurrent structural variants. Single-cell tracking shows that this mechanism is active throughout leukemic evolution, with evidence of localized clustering and reiterated deletions. Integration of data on point mutations and rearrangements identifies ATF7IP and MGA as two new tumor-suppressor genes in ALL. Thus, a remarkably parsimonious mutational process transforms ETV6-RUNX1-positive lymphoblasts, targeting the promoters, enhancers and first exons of genes that normally regulate B cell differentiation. © 2014 Nature America, Inc
- Published
- 2013
16. Correction: Protein Phosphatase 2A as a Therapeutic Target in Small Cell Lung Cancer.
- Author
-
Mirzapoiazova T, Xiao G, Mambetsariev B, Nasser MW, Miaou E, Singhal SS, Srivastava S, Mambetsariev I, Nelson MS, Nam A, Behal A, Arvanitis LD, Atri P, Muschen M, Tissot FLH, Miser J, Kovach JS, Sattler M, Batra SK, Kulkarni P, and Salgia R
- Published
- 2022
- Full Text
- View/download PDF
17. Protein Phosphatase 2A as a Therapeutic Target in Small Cell Lung Cancer.
- Author
-
Mirzapoiazova T, Xiao G, Mambetsariev B, Nasser MW, Miaou E, Singhal SS, Srivastava S, Mambetsariev I, Nelson MS, Nam A, Behal A, Arvanitis LD, Atri P, Muschen M, Tissot FLH, Miser J, Kovach JS, Sattler M, Batra SK, Kulkarni P, and Salgia R
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis, Cell Proliferation, Humans, Lung Neoplasms enzymology, Lung Neoplasms pathology, Small Cell Lung Carcinoma enzymology, Small Cell Lung Carcinoma pathology, Tumor Cells, Cultured, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Neoplastic drug effects, Lung Neoplasms drug therapy, Piperazines pharmacology, Protein Phosphatase 2 antagonists & inhibitors, Small Cell Lung Carcinoma drug therapy
- Abstract
Protein phosphatase 2A (PP2A), a serine/threonine phosphatase involved in the regulation of apoptosis, proliferation, and DNA-damage response, is overexpressed in many cancers, including small cell lung cancer (SCLC). Here we report that LB100, a small molecule inhibitor of PP2A, when combined with platinum-based chemotherapy, synergistically elicited an antitumor response both in vitro and in vivo with no apparent toxicity. Using inductively coupled plasma mass spectrometry, we determined quantitatively that sensitization via LB100 was mediated by increased uptake of carboplatin in SCLC cells. Treatment with LB100 alone or in combination resulted in inhibition of cell viability in two-dimensional culture and three-dimensional spheroid models of SCLC, reduced glucose uptake, and attenuated mitochondrial and glycolytic ATP production. Combining LB100 with atezolizumab increased the capacity of T cells to infiltrate and kill tumor spheroids, and combining LB100 with carboplatin caused hyperphosphorylation of the DNA repair marker γH2AX and enhanced apoptosis while attenuating MET signaling and invasion through an endothelial cell monolayer. Taken together, these data highlight the translational potential of inhibiting PP2A with LB100 in combination with platinum-based chemotherapy and immunotherapy in SCLC., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
18. IKAROS and CK2 regulate expression of BCL-XL and chemosensitivity in high-risk B-cell acute lymphoblastic leukemia.
- Author
-
Song C, Ge Z, Ding Y, Tan BH, Desai D, Gowda K, Amin S, Gowda R, Robertson GP, Yue F, Huang S, Spiegelman V, Payne JL, Reeves ME, Gurel Z, Iyer S, Dhanyamraju PK, Xiang M, Kawasawa YI, Cury NM, Yunes JA, McGrath M, Schramm J, Su R, Yang Y, Zhao Z, Lyu X, Muschen M, Payne KJ, Gowda C, and Dovat S
- Subjects
- Animals, Antibiotics, Antineoplastic pharmacology, Antibiotics, Antineoplastic therapeutic use, Cell Line, Tumor, Doxorubicin pharmacology, Doxorubicin therapeutic use, Humans, Mice, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Casein Kinase II genetics, Drug Resistance, Neoplasm, Gene Expression Regulation, Leukemic drug effects, Ikaros Transcription Factor genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, bcl-X Protein genetics
- Abstract
High-risk B-cell acute lymphoblastic leukemia (B-ALL) is an aggressive disease, often characterized by resistance to chemotherapy. A frequent feature of high-risk B-ALL is loss of function of the IKAROS (encoded by the IKZF1 gene) tumor suppressor. Here, we report that IKAROS regulates expression of the BCL2L1 gene (encodes the BCL-XL protein) in human B-ALL. Gain-of-function and loss-of-function experiments demonstrate that IKAROS binds to the BCL2L1 promoter, recruits histone deacetylase HDAC1, and represses BCL2L1 expression via chromatin remodeling. In leukemia, IKAROS' function is impaired by oncogenic casein kinase II (CK2), which is overexpressed in B-ALL. Phosphorylation by CK2 reduces IKAROS binding and recruitment of HDAC1 to the BCL2L1 promoter. This results in a loss of IKAROS-mediated repression of BCL2L1 and increased expression of BCL-XL. Increased expression of BCL-XL and/or CK2, as well as reduced IKAROS expression, are associated with resistance to doxorubicin treatment. Molecular and pharmacological inhibition of CK2 with a specific inhibitor CX-4945, increases binding of IKAROS to the BCL2L1 promoter and enhances IKAROS-mediated repression of BCL2L1 in B-ALL. Treatment with CX-4945 increases sensitivity to doxorubicin in B-ALL, and reverses resistance to doxorubicin in multidrug-resistant B-ALL. Combination treatment with CX-4945 and doxorubicin show synergistic therapeutic effects in vitro and in preclinical models of high-risk B-ALL. Results reveal a novel signaling network that regulates chemoresistance in leukemia. These data lay the groundwork for clinical testing of a rationally designed, targeted therapy that combines the CK2 inhibitor, CX-4945, with doxorubicin for the treatment of hematopoietic malignancies., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
19. Extrafollicular CD4 + T-B interactions are sufficient for inducing autoimmune-like chronic graft-versus-host disease.
- Author
-
Deng R, Hurtz C, Song Q, Yue C, Xiao G, Yu H, Wu X, Muschen M, Forman S, Martin PJ, and Zeng D
- Subjects
- Animals, Chronic Disease, Germinal Center cytology, Graft vs Host Disease genetics, Humans, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Proto-Oncogene Proteins c-bcl-6 genetics, Proto-Oncogene Proteins c-bcl-6 immunology, STAT3 Transcription Factor genetics, STAT3 Transcription Factor immunology, T-Lymphocytes, Helper-Inducer immunology, Autoimmune Diseases immunology, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, Germinal Center immunology, Graft vs Host Disease immunology
- Abstract
Chronic graft-versus-host disease (cGVHD) is an autoimmune-like syndrome mediated by pathogenic CD4
+ T and B cells, but the function of extrafollicular and germinal center CD4+ T and B interactions in cGVHD pathogenesis remains largely unknown. Here we show that extrafollicular CD4+ T and B interactions are sufficient for inducing cGVHD, while germinal center formation is dispensable. The pathogenesis of cGVHD is associated with the expansion of extrafollicular CD44hi CD62lo PSGL-1lo CD4+ (PSGL-1lo CD4+ ) T cells. These cells express high levels of ICOS, and the blockade of ICOS/ICOSL interaction prevents their expansion and ameliorates cGVHD. Expansion of PSGL-1lo CD4+ T cells is also prevented by BCL6 or Stat3 deficiency in donor CD4+ T cells, with the induction of cGVHD ameliorated by BCL6 deficiency and completely suppressed by Stat3 deficiency in donor CD4+ T cells. These results support that Stat3- and BCL6-dependent extrafollicular CD4+ T and B interactions play critical functions in the pathogenesis of cGVHD.Chronic graft-versus-host disease (cGVHD) is mediated by specific CD4 and B cells, but the relative contribution of extrafollicular and germinal centre (GC) T-B interaction is unclear. Here the authors show that the extrafollicular expansion of a specific CD4 T subset is sufficient for inducing cGVHD while GC is dispensable.- Published
- 2017
- Full Text
- View/download PDF
20. Gene expression and mutation-guided synthetic lethality eradicates proliferating and quiescent leukemia cells.
- Author
-
Nieborowska-Skorska M, Sullivan K, Dasgupta Y, Podszywalow-Bartnicka P, Hoser G, Maifrede S, Martinez E, Di Marcantonio D, Bolton-Gillespie E, Cramer-Morales K, Lee J, Li M, Slupianek A, Gritsyuk D, Cerny-Reiterer S, Seferynska I, Stoklosa T, Bullinger L, Zhao H, Gorbunova V, Piwocka K, Valent P, Civin CI, Muschen M, Dick JE, Wang JC, Bhatia S, Bhatia R, Eppert K, Minden MD, Sykes SM, and Skorski T
- Subjects
- Animals, Cell Line, Tumor, Cell Transformation, Neoplastic, Cricetinae, DNA Breaks, Double-Stranded, DNA End-Joining Repair, Genes, BRCA1, Genes, BRCA2, Genes, Lethal, Genes, abl, Humans, Leukemia drug therapy, Mice, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Mouse Embryonic Stem Cells physiology, Phthalazines pharmacology, Piperazines pharmacology, Transcriptome, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Cell Proliferation, Leukemia genetics, Poly(ADP-ribose) Polymerase Inhibitors pharmacology
- Abstract
Quiescent and proliferating leukemia cells accumulate highly lethal DNA double-strand breaks that are repaired by 2 major mechanisms: BRCA-dependent homologous recombination and DNA-dependent protein kinase-mediated (DNA-PK-mediated) nonhomologous end-joining, whereas DNA repair pathways mediated by poly(ADP)ribose polymerase 1 (PARP1) serve as backups. Here we have designed a personalized medicine approach called gene expression and mutation analysis (GEMA) to identify BRCA- and DNA-PK-deficient leukemias either directly, using reverse transcription-quantitative PCR, microarrays, and flow cytometry, or indirectly, by the presence of oncogenes such as BCR-ABL1. DNA-PK-deficient quiescent leukemia cells and BRCA/DNA-PK-deficient proliferating leukemia cells were sensitive to PARP1 inhibitors that were administered alone or in combination with current antileukemic drugs. In conclusion, GEMA-guided targeting of PARP1 resulted in dual cellular synthetic lethality in quiescent and proliferating immature leukemia cells, and is thus a potential approach to eradicate leukemia stem and progenitor cells that are responsible for initiation and manifestation of the disease. Further, an analysis of The Cancer Genome Atlas database indicated that this personalized medicine approach could also be applied to treat numerous solid tumors from individual patients.
- Published
- 2017
- Full Text
- View/download PDF
21. The Public Repository of Xenografts Enables Discovery and Randomized Phase II-like Trials in Mice.
- Author
-
Townsend EC, Murakami MA, Christodoulou A, Christie AL, Köster J, DeSouza TA, Morgan EA, Kallgren SP, Liu H, Wu SC, Plana O, Montero J, Stevenson KE, Rao P, Vadhi R, Andreeff M, Armand P, Ballen KK, Barzaghi-Rinaudo P, Cahill S, Clark RA, Cooke VG, Davids MS, DeAngelo DJ, Dorfman DM, Eaton H, Ebert BL, Etchin J, Firestone B, Fisher DC, Freedman AS, Galinsky IA, Gao H, Garcia JS, Garnache-Ottou F, Graubert TA, Gutierrez A, Halilovic E, Harris MH, Herbert ZT, Horwitz SM, Inghirami G, Intlekofer AM, Ito M, Izraeli S, Jacobsen ED, Jacobson CA, Jeay S, Jeremias I, Kelliher MA, Koch R, Konopleva M, Kopp N, Kornblau SM, Kung AL, Kupper TS, LeBoeuf NR, LaCasce AS, Lees E, Li LS, Look AT, Murakami M, Muschen M, Neuberg D, Ng SY, Odejide OO, Orkin SH, Paquette RR, Place AE, Roderick JE, Ryan JA, Sallan SE, Shoji B, Silverman LB, Soiffer RJ, Steensma DP, Stegmaier K, Stone RM, Tamburini J, Thorner AR, van Hummelen P, Wadleigh M, Wiesmann M, Weng AP, Wuerthner JU, Williams DA, Wollison BM, Lane AA, Letai A, Bertagnolli MM, Ritz J, Brown M, Long H, Aster JC, Shipp MA, Griffin JD, and Weinstock DM
- Published
- 2016
- Full Text
- View/download PDF
22. Targeting casein kinase II restores Ikaros tumor suppressor activity and demonstrates therapeutic efficacy in high-risk leukemia.
- Author
-
Song C, Gowda C, Pan X, Ding Y, Tong Y, Tan BH, Wang H, Muthusami S, Ge Z, Sachdev M, Amin SG, Desai D, Gowda K, Gowda R, Robertson GP, Schjerven H, Muschen M, Payne KJ, and Dovat S
- Subjects
- Animals, Apoptosis drug effects, Casein Kinase II genetics, Casein Kinase II metabolism, Cell Proliferation drug effects, Chromatin Immunoprecipitation, Enzyme Inhibitors pharmacology, Female, Humans, Ikaros Transcription Factor genetics, Mice, Mice, Inbred NOD, Phosphatidylinositol 3-Kinases, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma drug therapy, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction drug effects, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Casein Kinase II antagonists & inhibitors, Genes, Tumor Suppressor, Ikaros Transcription Factor metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology
- Abstract
Ikaros (IKZF1) is a tumor suppressor that binds DNA and regulates expression of its target genes. The mechanism of Ikaros activity as a tumor suppressor and the regulation of Ikaros function in leukemia are unknown. Here, we demonstrate that Ikaros controls cellular proliferation by repressing expression of genes that promote cell cycle progression and the phosphatidylinositol-3 kinase (PI3K) pathway. We show that Ikaros function is impaired by the pro-oncogenic casein kinase II (CK2), and that CK2 is overexpressed in leukemia. CK2 inhibition restores Ikaros function as transcriptional repressor of cell cycle and PI3K pathway genes, resulting in an antileukemia effect. In high-risk leukemia where one IKZF1 allele has been deleted, CK2 inhibition restores the transcriptional repressor function of the remaining wild-type IKZF1 allele. CK2 inhibition demonstrated a potent therapeutic effect in a panel of patient-derived primary high-risk B-cell acute lymphoblastic leukemia xenografts as indicated by prolonged survival and a reduction of leukemia burden. We demonstrate the efficacy of a novel therapeutic approach for high-risk leukemia: restoration of Ikaros tumor suppressor activity via inhibition of CK2. These results provide a rationale for the use of CK2 inhibitors in clinical trials for high-risk leukemia, including cases with deletion of one IKZF1 allele., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
23. RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia.
- Author
-
Papaemmanuil E, Rapado I, Li Y, Potter NE, Wedge DC, Tubio J, Alexandrov LB, Van Loo P, Cooke SL, Marshall J, Martincorena I, Hinton J, Gundem G, van Delft FW, Nik-Zainal S, Jones DR, Ramakrishna M, Titley I, Stebbings L, Leroy C, Menzies A, Gamble J, Robinson B, Mudie L, Raine K, O'Meara S, Teague JW, Butler AP, Cazzaniga G, Biondi A, Zuna J, Kempski H, Muschen M, Ford AM, Stratton MR, Greaves M, and Campbell PJ
- Subjects
- Base Sequence, Basic Helix-Loop-Helix Transcription Factors genetics, DNA Copy Number Variations genetics, Gene Library, Genes, Tumor Suppressor, Humans, Molecular Sequence Data, Repressor Proteins, Sequence Analysis, DNA, Sequence Deletion genetics, Transcription Factors genetics, V(D)J Recombination genetics, Core Binding Factor Alpha 2 Subunit genetics, Gene Expression Regulation, Neoplastic genetics, Gene Rearrangement genetics, Genetic Variation, Homeodomain Proteins genetics, Oncogene Proteins, Fusion genetics, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma genetics, Recombination, Genetic genetics
- Abstract
The ETV6-RUNX1 fusion gene, found in 25% of childhood acute lymphoblastic leukemia (ALL) cases, is acquired in utero but requires additional somatic mutations for overt leukemia. We used exome and low-coverage whole-genome sequencing to characterize secondary events associated with leukemic transformation. RAG-mediated deletions emerge as the dominant mutational process, characterized by recombination signal sequence motifs near breakpoints, incorporation of non-templated sequence at junctions, ∼30-fold enrichment at promoters and enhancers of genes actively transcribed in B cell development and an unexpectedly high ratio of recurrent to non-recurrent structural variants. Single-cell tracking shows that this mechanism is active throughout leukemic evolution, with evidence of localized clustering and reiterated deletions. Integration of data on point mutations and rearrangements identifies ATF7IP and MGA as two new tumor-suppressor genes in ALL. Thus, a remarkably parsimonious mutational process transforms ETV6-RUNX1-positive lymphoblasts, targeting the promoters, enhancers and first exons of genes that normally regulate B cell differentiation.
- Published
- 2014
- Full Text
- View/download PDF
24. Crosstalk between ROR1 and the Pre-B cell receptor promotes survival of t(1;19) acute lymphoblastic leukemia.
- Author
-
Bicocca VT, Chang BH, Masouleh BK, Muschen M, Loriaux MM, Druker BJ, and Tyner JW
- Subjects
- Chromosomes, Human, Pair 1, Chromosomes, Human, Pair 19, Dasatinib, Gene Expression Regulation, Neoplastic, Gene Silencing, Humans, Phosphorylation, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt metabolism, Pyrimidines pharmacology, Signal Transduction, Thiazoles pharmacology, Translocation, Genetic, Pre-B Cell Receptors metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Receptor Tyrosine Kinase-like Orphan Receptors physiology
- Abstract
We report that t(1;19) ALL cells universally exhibit expression of and dependence on the cell surface receptor ROR1. We further identify t(1;19) ALL cell sensitivity to the kinase inhibitor dasatinib due to its inhibition of the pre-B cell receptor (pre-BCR) signaling complex. These phenotypes are a consequence of developmental arrest at an intermediate/late stage of B-lineage maturation. Additionally, inhibition of pre-BCR signaling induces further ROR1 upregulation, and we identify distinct ROR1 and pre-BCR downstream signaling pathways that are modulated in a counterbalancing manner-both leading to AKT phosphorylation. Consistent with this, AKT phosphorylation is transiently eliminated after dasatinib treatment, but is partially restored following dasatinib potentiation of ROR1 expression. Consequently, ROR1 silencing accentuates dasatinib killing of t(1;19) ALL cells., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
25. HDAC inhibitors potentiate the activity of the BCR/ABL kinase inhibitor KW-2449 in imatinib-sensitive or -resistant BCR/ABL+ leukemia cells in vitro and in vivo.
- Author
-
Nguyen T, Dai Y, Attkisson E, Kramer L, Jordan N, Nguyen N, Kolluri N, Muschen M, and Grant S
- Subjects
- Adult, Animals, Antineoplastic Combined Chemotherapy Protocols pharmacology, Benzamides, Cell Line, Tumor, Drug Synergism, Fusion Proteins, bcr-abl antagonists & inhibitors, Fusion Proteins, bcr-abl metabolism, Histone Deacetylase Inhibitors administration & dosage, Humans, Imatinib Mesylate, Indazoles administration & dosage, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Mice, Mice, Inbred BALB C, Mice, SCID, Piperazines administration & dosage, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors pharmacology, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Drug Resistance, Neoplasm drug effects, Histone Deacetylase Inhibitors pharmacology, Indazoles pharmacology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive drug therapy, Piperazines pharmacology, Piperazines therapeutic use, Pyrimidines therapeutic use
- Abstract
Purpose: The purpose of this study was to determine whether histone deacetylase (HDAC) inhibitors (HDACI) such as vorinostat or entinostat (SNDX-275) could increase the lethality of the dual Bcr/Abl-Aurora kinase inhibitor KW-2449 in various Bcr/Abl(+) human leukemia cells, including those resistant to imatinib mesylate (IM)., Experimental Design: Bcr/Abl(+) chronic myelogenous leukemia (CML) and acute lymphoblastic leukemia (ALL) cells, including those resistant to IM (T315I, E255K), were exposed to KW-2449 in the presence or absence of vorinostat or SNDX-275, after which apoptosis and effects on signaling pathways were examined. In vivo studies combining HDACIs and KW2449 were carried out by using a systemic IM-resistant ALL xenograft model., Results: Coadministration of HDACIs synergistically increased KW-2449 lethality in vitro in multiple CML and Ph(+) ALL cell types including human IM resistant cells (e.g., BV-173/E255K and Adult/T315I). Combined treatment resulted in inactivation of Bcr/Abl and downstream targets (e.g., STAT5 and CRKL), as well as increased reactive oxygen species (ROS) generation and DNA damage (γH2A.X). The latter events and cell death were significantly attenuated by free radical scavengers (TBAP). Increased lethality was also observed in primary CD34(+) cells from patients with CML, but not in normal CD34(+) cells. Finally, minimally active vorinostat or SNDX275 doses markedly increased KW2449 antitumor effects and significantly prolonged the survival of murine xenografts bearing IM-resistant ALL cells (BV173/E255K)., Conclusions: HDACIs increase KW-2449 lethality in Bcr/Abl(+) cells in association with inhibition of Bcr/Abl, generation of ROS, and induction of DNA damage. This strategy preferentially targets primary Bcr/Abl(+) hematopoietic cells and exhibits enhanced in vivo activity. Combining KW-2449 with HDACIs warrants attention in IM-resistant Bcr/Abl(+) leukemias., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
26. Launching of american journal of blood research.
- Author
-
Kwak LW, Goldenberg DM, Benz EJ Jr, Yi Q, Muschen M, and Wang D
- Published
- 2011
27. Human chromosomal translocations at CpG sites and a theoretical basis for their lineage and stage specificity.
- Author
-
Tsai AG, Lu H, Raghavan SC, Muschen M, Hsieh CL, and Lieber MR
- Subjects
- Basic Helix-Loop-Helix Transcription Factors genetics, Chromosome Breakage, Cytidine Deaminase metabolism, DNA Breaks, Double-Stranded, Genes, bcl-1, Genes, bcl-2, Homeodomain Proteins metabolism, Humans, Leukemia, Lymphoid metabolism, B-Lymphocytes metabolism, CpG Islands, Leukemia, Lymphoid genetics, Translocation, Genetic
- Abstract
We have assembled, annotated, and analyzed a database of over 1700 breakpoints from the most common chromosomal rearrangements in human leukemias and lymphomas. Using this database, we show that although the CpG dinucleotide constitutes only 1% of the human genome, it accounts for 40%-70% of breakpoints at pro-B/pre-B stage translocation regions-specifically, those near the bcl-2, bcl-1, and E2A genes. We do not observe CpG hotspots in rearrangements involving lymphoid-myeloid progenitors, mature B cells, or T cells. The stage specificity, lineage specificity, CpG targeting, and unique breakpoint distributions at these cluster regions may be explained by a lesion-specific double-strand breakage mechanism involving the RAG complex acting at AID-deaminated methyl-CpGs.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.