Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jae-Woong Lee is active.

Publication


Featured researches published by Jae-Woong Lee.


Nature | 2015

Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia.

Zhengshan Chen; Seyedmehdi Shojaee; Maike Buchner; Huimin Geng; Jae-Woong Lee; Lars Klemm; Björn Titz; Thomas G. Graeber; Eugene Park; Ying Xim Tan; Anne B. Satterthwaite; Elisabeth Paietta; Stephen P. Hunger; Cheryl L. Willman; Ari Melnick; Mignon L. Loh; Jae U. Jung; John E. Coligan; Silvia Bolland; Tak W. Mak; Andre Limnander; Hassan Jumaa; Michael Reth; Arthur Weiss; Clifford A. Lowell; Markus Müschen

B cells are selected for an intermediate level of B-cell antigen receptor (BCR) signalling strength: attenuation below minimum (for example, non-functional BCR) or hyperactivation above maximum (for example, self-reactive BCR) thresholds of signalling strength causes negative selection. In ∼25% of cases, acute lymphoblastic leukaemia (ALL) cells carry the oncogenic BCR-ABL1 tyrosine kinase (Philadelphia chromosome positive), which mimics constitutively active pre-BCR signalling. Current therapeutic approaches are largely focused on the development of more potent tyrosine kinase inhibitors to suppress oncogenic signalling below a minimum threshold for survival. We tested the hypothesis that targeted hyperactivation—above a maximum threshold—will engage a deletional checkpoint for removal of self-reactive B cells and selectively kill ALL cells. Here we find, by testing various components of proximal pre-BCR signalling in mouse BCR–ABL1 cells, that an incremental increase of Syk tyrosine kinase activity was required and sufficient to induce cell death. Hyperactive Syk was functionally equivalent to acute activation of a self-reactive BCR on ALL cells. Despite oncogenic transformation, this basic mechanism of negative selection was still functional in ALL cells. Unlike normal pre-B cells, patient-derived ALL cells express the inhibitory receptors PECAM1, CD300A and LAIR1 at high levels. Genetic studies revealed that Pecam1, Cd300a and Lair1 are critical to calibrate oncogenic signalling strength through recruitment of the inhibitory phosphatases Ptpn6 (ref. 7) and Inpp5d (ref. 8). Using a novel small-molecule inhibitor of INPP5D (also known as SHIP1), we demonstrated that pharmacological hyperactivation of SYK and engagement of negative B-cell selection represents a promising new strategy to overcome drug resistance in human ALL.


Cancer Cell | 2015

Self-Enforcing Feedback Activation between BCL6 and Pre-B Cell Receptor Signaling Defines a Distinct Subtype of Acute Lymphoblastic Leukemia

Huimin Geng; Christian Hurtz; Kyle Lenz; Zhengshan Chen; Dirk Baumjohann; Sarah K. Thompson; Natalya A. Goloviznina; Wei Yi Chen; Jianya Huan; Dorian LaTocha; Erica Ballabio; Gang Xiao; Jae-Woong Lee; Anne Deucher; Zhongxia Qi; Eugene Park; Chuanxin Huang; Rahul Nahar; Soo Mi Kweon; Seyedmehdi Shojaee; Lai N. Chan; Jingwei Yu; Steven M. Kornblau; Janetta Jacoba Bijl; B. Hilda Ye; K. Mark Ansel; Elisabeth Paietta; Ari Melnick; Stephen P. Hunger; Peter Kurre

Studying 830 pre-B ALL cases from four clinical trials, we found that human ALL can be divided into two fundamentally distinct subtypes based on pre-BCR function. While absent in the majority of ALL cases, tonic pre-BCR signaling was found in 112 cases (13.5%). In these cases, tonic pre-BCR signaling induced activation of BCL6, which in turn increased pre-BCR signaling output at the transcriptional level. Interestingly, inhibition of pre-BCR-related tyrosine kinases reduced constitutive BCL6 expression and selectively killed patient-derived pre-BCR(+) ALL cells. These findings identify a genetically and phenotypically distinct subset of human ALL that critically depends on tonic pre-BCR signaling. In vivo treatment studies suggested that pre-BCR tyrosine kinase inhibitors are useful for the treatment of patients with pre-BCR(+) ALL.


Nature | 2017

Metabolic gatekeeper function of B-lymphoid transcription factors

Lai N. Chan; Zhengshan Chen; Daniel Braas; Jae-Woong Lee; Gang Xiao; Huimin Geng; Kadriye Nehir Cosgun; Christian Hurtz; Seyedmehdi Shojaee; Valeria Cazzaniga; Hilde Schjerven; Thomas Ernst; Andreas Hochhaus; Steven M. Kornblau; Marina Konopleva; Miles A. Pufall; Giovanni Cazzaniga; Grace J. Liu; Thomas A. Milne; H. Phillip Koeffler; Theodora S. Ross; Isidro Sánchez-García; Arndt Borkhardt; Keith R. Yamamoto; Ross A. Dickins; Thomas G. Graeber; Markus Müschen

B-lymphoid transcription factors, such as PAX5 and IKZF1, are critical for early B-cell development, yet lesions of the genes encoding these transcription factors occur in over 80% of cases of pre-B-cell acute lymphoblastic leukaemia (ALL). The importance of these lesions in ALL has, until now, remained unclear. Here, by combining studies using chromatin immunoprecipitation with sequencing and RNA sequencing, we identify a novel B-lymphoid program for transcriptional repression of glucose and energy supply. Our metabolic analyses revealed that PAX5 and IKZF1 enforce a state of chronic energy deprivation, resulting in constitutive activation of the energy-stress sensor AMPK. Dominant-negative mutants of PAX5 and IKZF1, however, relieved this glucose and energy restriction. In a transgenic pre-B ALL mouse model, the heterozygous deletion of Pax5 increased glucose uptake and ATP levels by more than 25-fold. Reconstitution of PAX5 and IKZF1 in samples from patients with pre-B ALL restored a non-permissive state and induced energy crisis and cell death. A CRISPR/Cas9-based screen of PAX5 and IKZF1 transcriptional targets identified the products of NR3C1 (encoding the glucocorticoid receptor), TXNIP (encoding a glucose-feedback sensor) and CNR2 (encoding a cannabinoid receptor) as central effectors of B-lymphoid restriction of glucose and energy supply. Notably, transport-independent lipophilic methyl-conjugates of pyruvate and tricarboxylic acid cycle metabolites bypassed the gatekeeper function of PAX5 and IKZF1 and readily enabled leukaemic transformation. Conversely, pharmacological TXNIP and CNR2 agonists and a small-molecule AMPK inhibitor strongly synergized with glucocorticoids, identifying TXNIP, CNR2 and AMPK as potential therapeutic targets. Furthermore, our results provide a mechanistic explanation for the empirical finding that glucocorticoids are effective in the treatment of B-lymphoid but not myeloid malignancies. Thus, B-lymphoid transcription factors function as metabolic gatekeepers by limiting the amount of cellular ATP to levels that are insufficient for malignant transformation.


Nature | 2018

Author Correction: Metabolic gatekeeper function of B-lymphoid transcription factors

Lai N. Chan; Zhengshan Chen; Daniel Braas; Jae-Woong Lee; Gang Xiao; Huimin Geng; Kadriye Nehir Cosgun; Christian Hurtz; Seyedmehdi Shojaee; Valeria Cazzaniga; Hilde Schjerven; Thomas Ernst; Andreas Hochhaus; Steven M. Kornblau; Marina Konopleva; Miles A. Pufall; Giovanni Cazzaniga; Grace J. Liu; Thomas A. Milne; H. Phillip Koeffler; Theodora S. Ross; Isidro Sánchez-García; Arndt Borkhardt; Keith R. Yamamoto; Ross A. Dickins; Thomas G. Graeber; Markus Müschen

Author(s): 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; 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.


Cancer Research | 2017

Abstract 93: Transcriptional control of glucocorticoid responses in leukemia

Lai N. Chan; Zhengshan Chen; Gang Xiao; Jae-Woong Lee; Kadriye Nehir Cosgun; Huimin Geng; Valeria Cazzaniga; Hilde Schjerven; Ross A. Dickins; Markus Müschen

Glucocorticoids (GCs) are central to all major therapy regimens for pre-B cell-derived acute lymphoblastic leukemia (ALL), but have no activity in myeloid leukemia. Such divergent responses represent an empirically established clinical standard; however, neither the mechanism by which GCs induce cell death nor the biological basis for the distinct responses in B-cell and myeloid leukemias is clear. Studying patient-derived samples revealed that NR3C1 (glucocorticoid receptor) levels were 6- to 20-fold higher in pre-B ALL compared to chronic myeloid leukemia (CML). High levels of Nr3c1 were reduced upon B- to myeloid-lineage conversion, suggesting that regulation of NR3C1 expression and GC responsiveness depend on a B-cell transcriptional program. B-cell transcription factors (e.g. PAX5, IKZF1) are critical for B-cell development, yet they are genetically lesioned in more than 80% of pre-B ALL cases. Despite such high frequency, the significance of these inactivating lesions remains elusive. Combining ChIP-seq and RNA-seq analyses, we identified a novel B-cell transcriptional program for activation of NR3C1 and its transcriptional target TXNIP (a negative regulator of glucose uptake). Reconstitution of PAX5 or IKZF1 expression in haploinsufficient patient-derived pre-B ALL cells increased NR3C1 and TXNIP levels. Conversely, expression of dominant negative mutant of PAX5 or IKZF1 abolished NR3C1 expression. Loss of Nr3c1 or Txnip in murine BCR-ABL1-driven pre-B ALL cells resulted in survival advantage in competitive growth assays. Importantly, loss of Nr3c1 or Txnip significantly elevated glucose uptake, lactate production and cellular ATP levels. These findings suggest that GCs induce cell death by exacerbating glucose and energy depletion. Notably, reconstitution of PAX5 or IKZF1 rendered haploinsufficient patient-derived pre-B ALL cells more sensitive to dexamethasone (dex) treatment. In contrast, dominant-negative PAX5 or IKZF1 largely de-sensitized pre-B ALL cells expressing wildtype PAX5 or IKZF1. These findings suggest that B-cell transcription factors set the threshold for GC responsiveness in pre-B ALL. Since relapsed ALL cells often acquire GC resistance, drug-combinations may be useful to prevent GC-resistance. As expected, loss of Nr3c1 abrogated responses to GCs. Interestingly, loss of Txnip also largely rescued GC-induced cell death in pre-B ALL cells. On this basis, we tested drug interactions between GCs and TXNIP agonists, 3-O-methylglucose (3-OMG) and D-allose. Treating patient-derived GC-refractory pre-B ALL cells with 3-OMG or D-allose strongly synergized with GC-treatment. Collectively, our findings provide a mechanistic explanation for the empiric finding that GCs are effective in the treatment of B-cell but not myeloid malignancies, and identify TXNIP as a novel therapeutic target in pre-B ALL. Note: This abstract was not presented at the meeting. Citation Format: Lai N. Chan, Zhengshan Chen, Gang Xiao, Jae Woong Lee, Kadriye Nehir Cosgun, Huimin Geng, Valeria Cazzaniga, Hilde Schjerven, Ross A. Dickins, Markus Muschen. Transcriptional control of glucocorticoid responses in leukemia [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 93. doi:10.1158/1538-7445.AM2017-93


Nature | 2016

Corrigendum: Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia

Zhengshan Chen; Seyedmehdi Shojaee; Maike Buchner; Huimin Geng; Jae-Woong Lee; Lars Klemm; Björn Titz; Thomas G. Graeber; Eugene Park; Ying Xim Tan; Anne B. Satterthwaite; Elisabeth Paietta; Stephen P. Hunger; Cheryl L. Willman; Ari Melnick; Mignon L. Loh; Jae U. Jung; John E. Coligan; Silvia Bolland; Tak W. Mak; Andre Limnander; Hassan Jumaa; Michael Reth; Arthur Weiss; Clifford A. Lowell; Markus Müschen

Corrigendum: Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia


Nature | 2016

Erratum: Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia (Nature (2015) 521 (357-361) DOI:10.1038/nature14231)

Zhengshan Chen; Seyedmehdi Shojaee; Maike Buchner; Huimin Geng; Jae-Woong Lee; Lars Klemm; Björn Titz; Thomas G. Graeber; Eugene Park; Ying Xim Tan; Anne B. Satterthwaite; Elisabeth Paietta; Stephen P. Hunger; Cheryl L. Willman; Ari Melnick; Mignon L. Loh; Jae U. Jung; John E. Coligan; Silvia Bolland; Tak W. Mak; Andre Limnander; Hassan Jumaa; Michael Reth; Arthur Weiss; Clifford A. Lowell; Markus Müschen

Corrigendum: Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia


Nature | 2016

Erratum: Corrigendum: Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia

Zhengshan Chen; Seyedmehdi Shojaee; Maike Buchner; Huimin Geng; Jae-Woong Lee; Lars Klemm; Björn Titz; Thomas G. Graeber; Eugene Park; Ying Xim Tan; Anne B. Satterthwaite; Elisabeth Paietta; Stephen P. Hunger; Cheryl L. Willman; Ari Melnick; Mignon L. Loh; Jae U. Jung; John E. Coligan; Silvia Bolland; Tak W. Mak; Andre Limnander; Hassan Jumaa; Michael Reth; Arthur Weiss; Clifford A. Lowell; Markus Müschen

Corrigendum: Signalling thresholds and negative B-cell selection in acute lymphoblastic leukaemia


Cancer Research | 2015

Abstract 2075: Signaling thresholds and negative B cell selection in acute lymphoblastic leukemia

Zhengshan Chen; Seyedmehdi Shojaee; Maike Buchner; Huimin Geng; Jae-Woong Lee; Lars Klemm; Eugene Park; Ying Xim Tan; Anne B. Satterthwaite; Elisabeth Paietta; Stephen P. Hunger; Mignon L. Loh; Jae U. Jung; John E. Coligan; Silvia Bolland; Tak W. Mak; Andre Limnander; Hassan Jumaa; Michael Reth; Arthur Weiss; Clifford A. Lowell; Markus Müschen

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Introduction: Unlike other cell types, B cells are selected for an intermediate level of signaling strength. Critical survival and proliferation signals emanate from the (pre-) B cell receptor (BCR): Both attenuation below minimum (e.g. non-functional pre-BCR) and hyperactivation above maximum (e.g. autoreactive pre-BCR) thresholds of signaling strength trigger negative selection and cell death. The oncogenic BCR-ABL1 tyrosine kinase mimics active pre-BCR signaling in Ph+ acute lymphoblastic leukemia (ALL) which defines the ALL subgroup with the worst clinical outcome. Current therapy approaches are largely focused on the development of more potent tyrosine kinase inhibitors (TKI) to suppress oncogenic signaling. However resistance to TKI is developed invariably. Here, we test the hypothesis that targeting hyperactivation above a maximum threshold will selectively kill Ph+ ALL cells, similar to removal of self-reactive B cells. Results: The Ph+ ALL cells don not express ITAM (immunoreceptor tyrosine-based activation motif) receptor Igα or Igβ on the cell surface, indicating defects for a functional pre-BCR. Reconstitution of ITAM receptor was sufficient to induce cell death through increasing pre-BCR signaling strength indicated by phosphorylation of SYK, SRC, BTK and PLCγ2. TKI-treatment, while designed to kill leukemia cells, seemingly paradoxically rescued Ph+ ALL cells in this experimental setting. Surprisingly, patient-derived Ph+ ALL cells express the ITIM (immunoreceptor tyrosine-based inhibitory motif) receptors PECAM1, CD300A and LAIR1 at high levels compared to normal pre-B cells. Importantly, high expression levels of ITIM-receptors are predictive of poor outcome in two clinical trials, including both pediatric and adult ALL patients. Genetic studies revealed that Pecam1, Cd300a and Lair1 were critical to calibrate pre-BCR signaling strength through recruitment of the inhibitory phosphatases Ptpn6 (Shp1) and Inpp5d (Ship1). Genetic deletion of Lair1, Ptpn6 or Inpp5d in BCR-ABL1 ALL caused cell death in vitro and in vivo through hyperactivation of pre-BCR signaling. Testing various components of proximal pre-BCR signaling, we found that an incremental increase of SYK tyrosine kinase activity was required and sufficient to induce cell death. Hyperactive SYK was functionally equivalent to acute activation of a self-reactive BCR on ALL cells. Using chimeric PECAM1, CD300A and LAIR1 receptor decoys and a novel small molecule inhibitor of INPP5D, we demonstrated that pharmacological hyperactivation of pre-BCR signaling and engagement of negative B cell selection represents a promising new strategy to overcome drug-resistance in human Ph+ ALL. Conclusion: These results indicated that inhibitory receptors and downstream phosphatases are critical regulators of pre-BCR signaling strength in Ph+ ALL, and identified targeting hyperactivation of pre-BCR signaling as a potential novel class of therapeutic strategy. Note: This abstract was not presented at the meeting. Citation Format: Zhengshan Chen, Seyedmehdi Shojaee, Maike Buchner, Huimin Geng, Jae Woong Lee, Lars Klemm, Eugene Park, Ying Xim Tan, Anne Satterthwaite, Elisabeth Paietta, Stephen P. Hunger, Mignon L. Loh, Jae U. Jung, John E. Coligan, Silvia Bolland, Tak W. Mak, Andre Limnander, Hassan Jumaa, Michael Reth, Arthur Weiss, Clifford A. Lowell, Markus Muschen. Signaling thresholds and negative B cell selection in acute lymphoblastic leukemia. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2075. doi:10.1158/1538-7445.AM2015-2075


Blood | 2013

Personalized synthetic lethality induced by targeting RAD52 in leukemias identified by gene mutation and expression profile

Kimberly Cramer-Morales; Margaret Nieborowska-Skorska; Kara A. Scheibner; Michelle Padget; David Irvine; Tomasz Sliwinski; Kimberly Haas; Jae-Woong Lee; Huimin Geng; Darshan Roy; Artur Slupianek; Feyruz V. Rassool; Mariusz A. Wasik; Wayne E. Childers; Mhairi Copland; Markus Müschen; Curt I. Civin; Tomasz Skorski

Collaboration


Dive into the Jae-Woong Lee's collaboration.

Top Co-Authors

Avatar

Huimin Geng

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhengshan Chen

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gang Xiao

University of California

View shared research outputs
Top Co-Authors

Avatar

Eugene Park

University of California

View shared research outputs
Top Co-Authors

Avatar

Lars Klemm

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lai N. Chan

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge