86 results on '"Arthur L. Shaffer"'
Search Results
2. Supplementary Table 7 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table 7. Targeting epigenetic resistance to BTK inhibitor treatment in ABC DLBCL, relates to Fig7
- Published
- 2023
3. Supplementary Table 1 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table 1. Epigenetic Ibrutinib Resistance in ABC DLBCL lines, , relates to Fig. 1
- Published
- 2023
4. Supplementary Figures and Legends from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Information- Figures & Legends (Supplemental Tables loaded separately)
- Published
- 2023
5. Supplementary Table 6 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table 6. RAC2 protein interactions are a marker of epigenetic ibrutinib resistance
- Published
- 2023
6. Supplementary Table 4 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table 4. Altered dependencies in ibrutinib resistant ABC DLBCL, relates to Fig. 4
- Published
- 2023
7. Supplementary Methods from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplementary Methods
- Published
- 2023
8. Supplementary Table 2 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table2. Tracking the evolution of ibrutinib resistance phenotypes, relates to Fig. 2
- Published
- 2023
9. Oligos and Primers - related to Methods from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Oligo and Primer Tables
- Published
- 2023
10. Supplementary Table 5 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table 5. RAC2 as a mediator of epigenetic ibrutinib resistance, , relates to Fig5
- Published
- 2023
11. Data from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
The use of Bruton tyrosine kinase (BTK) inhibitors to block B-cell receptor (BCR)–dependent NF-κB activation in lymphoid malignancies has been a major clinical advance, yet acquired therapeutic resistance is a recurring problem. We modeled the development of resistance to the BTK inhibitor ibrutinib in the activated B-cell (ABC) subtype of diffuse large B-cell lymphoma, which relies on chronic active BCR signaling for survival. The primary mode of resistance was epigenetic, driven in part by the transcription factor TCF4. The resultant phenotypic shift altered BCR signaling such that the GTPase RAC2 substituted for BTK in the activation of phospholipase Cγ2, thereby sustaining NF-κB activity. The interaction of RAC2 with phospholipase Cγ2 was also increased in chronic lymphocytic leukemia cells from patients with persistent or progressive disease on BTK inhibitor treatment. We identified clinically available drugs that can treat epigenetic ibrutinib resistance, suggesting combination therapeutic strategies.Significance:In diffuse large B-cell lymphoma, we show that primary resistance to BTK inhibitors is due to epigenetic rather than genetic changes that circumvent the BTK blockade. We also observed this resistance mechanism in chronic lymphocytic leukemia, suggesting that epigenetic alterations may contribute more to BTK inhibitor resistance than currently thought.See related commentary by Pasqualucci, p. 555.This article is highlighted in the In This Issue feature, p. 549
- Published
- 2023
12. Supplementary Table 3 from Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Louis M. Staudt, Adrian Wiestner, Wyndham H. Wilson, Erika M. Gaglione, Clare Sun, Inhye E. Ahn, Michael C. Kelly, Zachary Rae, Thomas Oellerich, Björn Häupl, Craig J. Thomas, Carleen Klumpp-Thomas, Crystal McKnight, Lu Chen, Kelli M. Wilson, Xiaohu Zhang, Michele Ceribelli, Sandrine Roulland, Weihong Xu, Xin Yu, Hong Zhao, Yandan Yang, Daniel E. Webster, Ryan M. Young, Jaewoo Choi, Monica Kasbekar, George W. Wright, DaWei Huang, James Q. Wang, James D. Phelan, and Arthur L. Shaffer
- Abstract
Supplemental Table 3. Epigenetic retuning of oncogenic signaling in ibrutinib resistance, relates to Fig. 3
- Published
- 2023
13. Data from TLR Signaling Is Activated in Lymph Node–Resident CLL Cells and Is Only Partially Inhibited by Ibrutinib
- Author
-
Sarah E. M. Herman, Adrian Wiestner, Louis M. Staudt, Michael J. Kruhlak, Jessica R. Iyer, Ryan M. Young, Arthur L. Shaffer, Delong Liu, Erin M. McAuley, and Eman L. Dadashian
- Abstract
Chronic lymphocytic leukemia (CLL) is a malignancy of mature B cells driven by B-cell receptor (BCR) signaling and activated primarily in the lymph node. The Bruton's tyrosine kinase (BTK) inhibitor ibrutinib effectively inhibits BCR-dependent proliferation and survival signals and has emerged as a breakthrough therapy for CLL. However, complete remissions are uncommon and are achieved only after years of continuous therapy. We hypothesized that other signaling pathways that sustain CLL cell survival are only partially inhibited by ibrutinib. In normal B cells, Toll-like receptor (TLR) signaling cooperates with BCR signaling to activate prosurvival NF-κB. Here, we show that an experimentally validated gene signature of TLR activation is overexpressed in lymph node–resident CLL cells compared with cells in the blood. Consistent with TLR activation, we detected phosphorylation of NF-κB, STAT1, and STAT3 in lymph node–resident CLL cells and in cells stimulated with CpG oligonucleotides in vitro. CpG promoted IRAK1 degradation, secretion of IL10, and extended survival of CLL cells in culture. CpG-induced TLR signaling was significantly inhibited by both an IRAK1/4 inhibitor and ibrutinib. Although inhibition of TLR signaling was incomplete with either drug, the combination achieved superior results, including more effective inhibition of TLR-mediated survival signaling. Our data suggest an important role for TLR signaling in CLL pathogenesis and in sustaining the viability of CLL cells during ibrutinib therapy. The combination of ibrutinib with a TLR pathway inhibitor could provide superior antitumor activity and should be investigated in clinical studies.Significance:CLL relies on the concomitant cooperation of B-cell receptor and Toll-like receptor signaling; inhibition of both pathways is superior to inhibition of either pathway alone.
- Published
- 2023
14. Supplementary Figures S1-S10 from TLR Signaling Is Activated in Lymph Node–Resident CLL Cells and Is Only Partially Inhibited by Ibrutinib
- Author
-
Sarah E. M. Herman, Adrian Wiestner, Louis M. Staudt, Michael J. Kruhlak, Jessica R. Iyer, Ryan M. Young, Arthur L. Shaffer, Delong Liu, Erin M. McAuley, and Eman L. Dadashian
- Abstract
Supplementary Figure S1: Dose response experiments with the IRAK1/4 inhibitor (IRAKi) in CLL cells. Supplementary Figure S2: TLR signaling is active in lymph node-resident CLL cells. Supplementary Figure S3: Flow Cytometry gating strategy. Supplementary Figure S4: Early activation of STAT3 leads to early production of serum IL-10. Supplementary Figure S5: TLR signaling induces similar levels of IRAK1 degradation and STAT signaling in vitro with CpG stimulation in primary CLL PBMCs. Supplementary Figure S6: Upstream IRAK1 degradation and downstream phosphorylation of STAT3 and IκBα are inversely related. Supplementary Figure S7: CpG induced TLR signaling promotes cell survival and activates CLL cells in vitro. Supplementary Figure S8: Inhibiting induced TLR activation downregulates expression of activation markers and prevents the survival advantage in CLL cells. Supplementary Figure S9: αIgM stimulation induces activation of BCR pathway signaling. Supplementary Figure S10: Ibrutinib inhibits both BCR and TLR signaling in activated CLL cells harvested from the lymph node.
- Published
- 2023
15. Supplementary Figure Legends from TLR Signaling Is Activated in Lymph Node–Resident CLL Cells and Is Only Partially Inhibited by Ibrutinib
- Author
-
Sarah E. M. Herman, Adrian Wiestner, Louis M. Staudt, Michael J. Kruhlak, Jessica R. Iyer, Ryan M. Young, Arthur L. Shaffer, Delong Liu, Erin M. McAuley, and Eman L. Dadashian
- Abstract
Supplementary Figure Legends S1-S10
- Published
- 2023
16. Data from Insertion of Myc into Igh Accelerates Peritoneal Plasmacytomas in Mice
- Author
-
Siegfried Janz, Louis M. Staudt, Michael Potter, Wendy duBois, Joong Su Kim, Arthur L. Shaffer, and Sung Sup Park
- Abstract
Gene-targeted mice that contain a His6-tagged mouse c-Myc cDNA, MycHis, inserted head to head into different sites of the mouse immunoglobulin heavy-chain locus, Igh, mimic the chromosomal T(12;15)(Igh-Myc) translocation that results in the activation of Myc in the great majority of mouse plasmacytomas. Mice carrying MycHis just 5′ of the intronic heavy-chain enhancer Eμ (strain iMycEμ) provide a specific model of the type of T(12;15) found in a subset (∼20%) of plasmacytomas that develop “spontaneously” in the gut-associated lymphoid tissue (GALT) of interleukin-6 transgenic BALB/c (C) mice. Here we show that the transfer of the iMycEμ transgene from a mixed genetic background of segregating C57BL/6 × 129/SvJ alleles to the background of C increased the incidence of GALT plasmacytomas by a factor of 2.5 in first-generation backcross mice (C.iMycEμ N1). Third-generation backcross mice (C.iMycEμ N3, ∼94% C alleles) were hypersusceptible to inflammation-induced peritoneal plasmacytomas (tumor incidence, 100%; mean tumor onset, 86 ± 28 days) compared with inbred C mice (tumor incidence, 5% on day 150 after tumor induction). Peritoneal plasmacytomas of C.iMycEμ N3 mice overexpressed MycHis, produced monoclonal immunoglobulin, and exhibited a unique plasma cell signature upon gene expression profiling on mouse Lymphochip cDNA microarrays. These findings indicated that the iMycEμ transgene accelerates plasmacytoma development by collaborating with tumor susceptibility alleles of strain C and circumventing the requirement for tumor precursors to acquire deregulated Myc by chromosomal translocation.
- Published
- 2023
17. Supplementary Tables 1-3, Figures 1-6 from Insertion of Myc into Igh Accelerates Peritoneal Plasmacytomas in Mice
- Author
-
Siegfried Janz, Louis M. Staudt, Michael Potter, Wendy duBois, Joong Su Kim, Arthur L. Shaffer, and Sung Sup Park
- Abstract
Supplementary Tables 1-3, Figures 1-6 from Insertion of Myc into Igh Accelerates Peritoneal Plasmacytomas in Mice
- Published
- 2023
18. A Multi-scale, Multiomic Atlas of Human Normal and Follicular Lymphoma Lymph Nodes
- Author
-
Andrea J. Radtke, Ekaterina Postovalova, Arina Varlamova, Alexander Bagaev, Maria Sorokina, Olga Kudryashova, Mark Meerson, Margarita Polyakova, Ilia Galkin, Viktor Svekolkin, Sergey Isaev, Grigory Perelman, Yaroslav Lozinsky, Ziv Yaniv, Bradley C. Lowekamp, Emily Speranza, Li Yao, Stefania Pittaluga, Arthur L. Shaffer, Danny Jonigk, James D. Phelan, Theresa Davies-Hill, Da Wei Huang, Pavel Ovcharov, Krystle Nomie, Ekaterina Nuzhdina, Nikita Kotlov, Ravshan Ataullakhanov, Nathan Fowler, Michael Kelly, Jagan Muppidi, Jeremy Davis, Jonathan M. Hernandez, Wyndham H. Wilson, Elaine S. Jaffe, Louis M. Staudt, Mark Roschewski, and Ronald N. Germain
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Abstract
SUMMARYReference atlases, molecular and spatial maps of mammalian tissues, are critical resources for discovery efforts and translational research. Their utility is dependent on operationalizing the resulting data by identifying cell types, histological patterns, and predictive biomarkers underlying health and disease. The human lymph node (LN) offers a compelling use case because of its importance in immunity, structural and cellular diversity, and neoplastic involvement. One hematological malignancy, follicular lymphoma (FL), evolves from developmentally blocked germinal center B cells residing in and trafficking through these tissues. To promote survival and immune escape, tumor B cells undergo significant genetic changes and extensively remodel the lymphoid microenvironment. Here, we present an integrated portrait of healthy and FL LNs using multiple genomic and advanced imaging technologies. By leveraging the strengths of each platform, we identified several tumor-specific features and microenvironmental patterns enriched in individuals who experience early relapse, the most high-risk of FL patients.
- Published
- 2022
19. Rewiring of B cell receptor signaling by Epstein–Barr virus LMP2A
- Author
-
Louis M. Staudt, Carmen Doebele, Hang Nguyen, Arthur L. Shaffer, Angelika Oellerich, Yanlong Ji, Thomas Oellerich, George E. Wright, Samantha Schaller, Sebastian Scheich, Richard Longnecker, Jagan R. Muppidi, Kuan-Ting Pan, Henning Urlaub, Kamonwan Fish, Hubert Serve, Federico Comoglio, and Masato Ikeda
- Subjects
0301 basic medicine ,Herpesvirus 4, Human ,B-cell receptor ,Receptors, Antigen, B-Cell ,Syk ,Apoptosis ,Biology ,medicine.disease_cause ,Viral Matrix Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,otorhinolaryngologic diseases ,medicine ,Humans ,Syk Kinase ,Phosphorylation ,Transcription factor ,Adaptor Proteins, Signal Transducing ,B-Lymphocytes ,Multidisciplinary ,NFATC Transcription Factors ,NF-kappa B ,breakpoint cluster region ,Membrane Proteins ,Tyrosine phosphorylation ,Biological Sciences ,Epstein–Barr virus ,3. Good health ,Cell biology ,stomatognathic diseases ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,TCF3 ,Signal transduction ,Signal Transduction - Abstract
Epstein-Barr virus (EBV) infects human B cells and reprograms them to allow virus replication and persistence. One key viral factor in this process is latent membrane protein 2A (LMP2A), which has been described as a B cell receptor (BCR) mimic promoting malignant transformation. However, how LMP2A signaling contributes to tumorigenesis remains elusive. By comparing LMP2A and BCR signaling in primary human B cells using phosphoproteomics and transcriptome profiling, we identified molecular mechanisms through which LMP2A affects B cell biology. Consistent with the literature, we found that LMP2A mimics a subset of BCR signaling events, including tyrosine phosphorylation of the kinase SYK, the calcium initiation complex consisting of BLNK, BTK, and PLCγ2, and its downstream transcription factor NFAT. However, the majority of LMP2A-induced signaling events markedly differed from those induced by BCR stimulation. These included differential phosphorylation of kinases, phosphatases, adaptor proteins, transcription factors such as nuclear factor κB (NF-κB) and TCF3, as well as widespread changes in the transcriptional output of LMP2A-expressing B cells. LMP2A affected apoptosis and cell-cycle checkpoints by dysregulating the expression of apoptosis regulators such as BCl-xL and the tumor suppressor retinoblastoma-associated protein 1 (RB1). LMP2A cooperated with MYC and mutant cyclin D3, two oncogenic drivers of Burkitt lymphoma, to promote proliferation and survival of primary human B cells by counteracting MYC-induced apoptosis and by inhibiting RB1 function, thereby promoting cell-cycle progression. Our results indicate that LMP2A is not a pure BCR mimic but rather rewires intracellular signaling in EBV-infected B cells that optimizes cell survival and proliferation, setting the stage for oncogenic transformation.
- Published
- 2020
20. Overcoming Acquired Epigenetic Resistance to BTK Inhibitors
- Author
-
Michael C. Kelly, Xiaohu Zhang, Kelli M. Wilson, Erika M Gaglione, Inhye E. Ahn, Zachary Rae, Lu Chen, Louis M. Staudt, Weihong Xu, Yandan Yang, James D. Phelan, Sandrine Roulland, Dan E. Webster, Arthur L. Shaffer, Björn Häupl, Hong Zhao, Xin Yu, Clare Sun, George E. Wright, Jaewoo Choi, Crystal McKnight, Da-Wei Huang, Craig J. Thomas, Ryan M. Young, Monica Kasbekar, James Q. Wang, Thomas Oellerich, Wyndham H. Wilson, Carleen Klumpp-Thomas, Adrian Wiestner, and Michele Ceribelli
- Subjects
biology ,business.industry ,Chronic lymphocytic leukemia ,breakpoint cluster region ,General Medicine ,TCF4 ,medicine.disease ,Lymphoma ,chemistry.chemical_compound ,chemistry ,immune system diseases ,Ibrutinib ,hemic and lymphatic diseases ,Cancer research ,biology.protein ,medicine ,Bruton's tyrosine kinase ,Epigenetics ,business ,Transcription factor ,Research Articles - Abstract
The use of Bruton tyrosine kinase (BTK) inhibitors to block B-cell receptor (BCR)–dependent NF-κB activation in lymphoid malignancies has been a major clinical advance, yet acquired therapeutic resistance is a recurring problem. We modeled the development of resistance to the BTK inhibitor ibrutinib in the activated B-cell (ABC) subtype of diffuse large B-cell lymphoma, which relies on chronic active BCR signaling for survival. The primary mode of resistance was epigenetic, driven in part by the transcription factor TCF4. The resultant phenotypic shift altered BCR signaling such that the GTPase RAC2 substituted for BTK in the activation of phospholipase Cγ2, thereby sustaining NF-κB activity. The interaction of RAC2 with phospholipase Cγ2 was also increased in chronic lymphocytic leukemia cells from patients with persistent or progressive disease on BTK inhibitor treatment. We identified clinically available drugs that can treat epigenetic ibrutinib resistance, suggesting combination therapeutic strategies. Significance: In diffuse large B-cell lymphoma, we show that primary resistance to BTK inhibitors is due to epigenetic rather than genetic changes that circumvent the BTK blockade. We also observed this resistance mechanism in chronic lymphocytic leukemia, suggesting that epigenetic alterations may contribute more to BTK inhibitor resistance than currently thought. See related commentary by Pasqualucci, p. 555. This article is highlighted in the In This Issue feature, p. 549
- Published
- 2021
21. In vivo CRISPR screens reveal a HIF-1α-mTOR-network regulates T follicular helper versus Th1 cells
- Author
-
Bonnie Huang, James D. Phelan, Silvia Preite, Julio Gomez-Rodriguez, Kristoffer H. Johansen, Hirofumi Shibata, Arthur L. Shaffer, Qin Xu, Brendan Jeffrey, Martha Kirby, Stacie Anderson, Yandan Yang, Selamawit Gossa, Dorian B. McGavern, Louis M. Staudt, and Pamela L. Schwartzberg
- Subjects
Multidisciplinary ,Science ,TOR Serine-Threonine Kinases ,General Physics and Astronomy ,Gene Expression ,Cell Differentiation ,General Chemistry ,T-Lymphocytes, Helper-Inducer ,Mechanistic Target of Rapamycin Complex 1 ,Th1 Cells ,Germinal Center ,Hypoxia-Inducible Factor 1, alpha Subunit ,General Biochemistry, Genetics and Molecular Biology ,Immunity, Humoral ,Gene Knockout Techniques ,Mice ,Virus Diseases ,Antibody Formation ,Animals ,Clustered Regularly Interspaced Short Palindromic Repeats ,Glycolysis - Abstract
T follicular helper (Tfh) cells provide signals to initiate and maintain the germinal center (GC) reaction and are crucial for the generation of robust, long-lived antibody responses, but how the GC microenvironment affects Tfh cells is not well understood. Here we develop an in vivo T cell-intrinsic CRISPR-knockout screen to evaluate Tfh and Th1 cells in an acute viral infection model to identify regulators of Tfh cells in their physiological setting. Using a screen of druggable-targets, alongside genetic, transcriptomic and cellular analyses, we identify a function of HIF-1α in suppressing mTORC1-mediated and Myc-related pathways, and provide evidence that VHL-mediated degradation of HIF-1α is required for Tfh development; an expanded in vivo CRISPR screen reveals multiple components of these pathways that regulate Tfh versus Th1 cells, including signaling molecules, cell-cycle regulators, nutrient transporters, metabolic enzymes and autophagy mediators. Collectively, our data serve as a resource for studying Tfh versus Th1 decisions, and implicate the VHL-HIF-1α axis in fine-tuning Tfh generation.
- Published
- 2021
22. TARGETING PROXIMAL BCR SIGNALING PATHWAY IN DIFFUSE LARGE B‐CELL LYMPHOMA
- Author
-
Thomas Oellerich, J. W Choi, Arthur L. Shaffer, S Scheich, Youwen Yang, James D. Phelan, Craig J. Thomas, X. Yu, Björ Häupl, George Wright, S Corcoran, B Wang, Michele Ceribelli, D. W Huang, and L. M. Staudt
- Subjects
Cancer Research ,Oncology ,medicine ,Cancer research ,Hematology ,General Medicine ,BCR Signaling Pathway ,Biology ,medicine.disease ,Diffuse large B-cell lymphoma - Published
- 2021
23. Taming the Heterogeneity of Aggressive Lymphomas for Precision Therapy
- Author
-
Louis M. Staudt, Roland Schmitz, Thomas Oellerich, Wyndham H. Wilson, George E. Wright, Ryan M. Young, James D. Phelan, Calvin A. Johnson, Arthur L. Shaffer, and Da-Wei Huang
- Subjects
0301 basic medicine ,Cancer Research ,Genetic heterogeneity ,B-cell receptor ,Cell Biology ,Biology ,medicine.disease ,Precision medicine ,Lymphoma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,immune system diseases ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Diffuse large B-cell lymphoma - Abstract
Genomic analyses of diffuse large B cell lymphoma (DLBCL) are revealing the genetic and phenotypic heterogeneity of these aggressive lymphomas. In part, this heterogeneity reflects the existence of distinct genetic subtypes that acquire characteristic constellations of somatic genetic alterations to converge on the DLBCL phenotype. In parallel, functional genomic screens and proteomic analyses have identified multiprotein assemblies that coordinate oncogenic survival signaling in DLBCL. In this review, we merge these recent insights into a unified conceptual framework with implications for the design of precision medicine trials in DLBCL.
- Published
- 2019
24. Regulation of B cell receptor-dependent NF-κB signaling by the tumor suppressor KLHL14
- Author
-
George E. Wright, Louis M. Staudt, Thomas Oellerich, Björn Häupl, Xin Yu, Da-Wei Huang, Zhuo Wang, Arthur L. Shaffer, James D. Phelan, Hong Zhao, Jaewoo Choi, and Ryan M. Young
- Subjects
B-cell receptor ,Receptors, Antigen, B-Cell ,Endoplasmic Reticulum ,chemistry.chemical_compound ,Piperidines ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Bruton's tyrosine kinase ,Humans ,Genes, Tumor Suppressor ,Multidisciplinary ,biology ,Chemistry ,Adenine ,breakpoint cluster region ,Intracellular Signaling Peptides and Proteins ,NF-kappa B ,Ubiquitin-Protein Ligase Complexes ,NF-κB ,CD79B ,Biological Sciences ,medicine.disease ,HEK293 Cells ,Pyrimidines ,Cell culture ,Drug Resistance, Neoplasm ,Ibrutinib ,Myeloid Differentiation Factor 88 ,Proteolysis ,Cancer research ,biology.protein ,Mutagenesis, Site-Directed ,Pyrazoles ,Lymphoma, Large B-Cell, Diffuse ,Carrier Proteins ,Diffuse large B-cell lymphoma ,CD79 Antigens ,Signal Transduction - Abstract
The KLHL14 gene acquires frequent inactivating mutations in mature B cell malignancies, especially in the MYD88 L265P , CD79B mutant (MCD) genetic subtype of diffuse large B cell lymphoma (DLBCL), which relies on B cell receptor (BCR) signaling for survival. However, the pathogenic role of KLHL14 in DLBCL and its molecular function are largely unknown. Here, we report that KLHL14 is in close proximity to the BCR in the endoplasmic reticulum of MCD cell line models and promotes the turnover of immature glycoforms of BCR subunits, reducing total cellular BCR levels. Loss of KLHL14 confers relative resistance to the Bruton tyrosine kinase (BTK) inhibitor ibrutinib and promotes assembly of the MYD88-TLR9-BCR (My-T-BCR) supercomplex, which initiates prosurvival NF-κB activation. Consequently, KLHL14 inactivation allows MCD cells to maintain NF-κB signaling in the presence of ibrutinib. These findings reinforce the central role of My-T-BCR–dependent NF-κB signaling in MCD DLBCL and suggest that the genetic status of KLHL14 should be considered in clinical trials testing inhibitors of BTK and BCR signaling mediators in DLBCL.
- Published
- 2020
25. A Prospective Study of Clonal Evolution in Follicular Lymphoma: Circulating Tumor DNA Correlates with Overall Tumor Burden and Fluctuates over Time without Therapy
- Author
-
Sarah Evans, Jagan R. Muppidi, Nathan Fowler, Amynah Pradhan, Ekaterina Postovalova, Jillian Simard, Christopher Melani, Allison Distler, Amy Hillsman, Theresa Davies-Hill, Arthur L. Shaffer, Olga Kudryashova, Mark A. Ahlman, Mark Roschewski, Wyndham H. Wilson, Nikita Kotlov, Elaine S. Jaffe, Allison P. Jacob, James D. Phelan, Louis M. Staudt, Alexander Bagaev, Stefania Pittaluga, Mark Meerson, Yandan Yang, and Rahul Lakhotia
- Subjects
business.industry ,Immunology ,Follicular lymphoma ,Tumor burden ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Somatic evolution in cancer ,Circulating tumor DNA ,medicine ,Cancer research ,business ,Prospective cohort study - Abstract
Background: Follicular lymphoma (FL) shows marked variation in clinical course including spontaneous regression and histologic transformation (HT). Watchful waiting (W&W) is routinely applied to pts with newly diagnosed FL, but monitoring strategies are not standardized. Pts with progression within 1-2y of diagnosis have worse outcomes, but the biologic basis is unclear and biologic-based classifiers are not routinely applied at diagnosis. Circulating tumor DNA (ctDNA) is a highly tumor-specific biomarker that is prognostic in aggressive B-cell lymphomas, but its ability to serially monitor FL remains undefined. We applied a next-generation sequencing assay to identify tumor clonotypes for serial monitoring of peripheral blood in pts with untreated FL as part of an ongoing prospective clinical trial [NCT03190928]. Methods: Pts with grade I-II or 3A FL are eligible if evaluable disease on CT or FDG-PET, age ≥18, ECOG ≤2, no evidence of HT, and no prior systemic therapy. Pts undergo W&W until they meet uniform protocol-defined treatment criteria and remain on study until second-line therapy. Baseline testing includes labs, peripheral blood flow cytometry, BM biopsy/aspirate, CT and FDG-PET scans, and research biopsy. Pt have clinic visits every 4m for 2y, every 6m in years 3-5, then annually. CT scans are every 8m for 2y, then annually. FDG-PET scans are at baseline, at 2y, and any time of suspected progression. Peripheral blood samples including Streck tubes (plasma) and PBMCs are drawn at each clinic visit and stored. For ctDNA analysis, tumor DNA was amplified from FFPE using locus-specific primer sets for the Ig heavy-chain and light-chain loci along with BCL1/BCL2 translocations (Adaptive Biotechnologies). Amplified products were sequenced and tumor clonotypes were identified in plasma and PBMCs. Serial tracking of ctDNA was done in plasma and blinded to clinical outcomes. Results: 77 pts enrolled between July 2017 and July 2021. Median age was 57 (range 24-83) including 14 (18%) low-risk, 29 (38%) intermediate-risk, and 34 (44%) high-risk by FLIPI. Fourteen (18%) pts had stage I-II disease. Forty-three (56%) pts had monoclonal B-cells on peripheral blood flow cytometry. Twenty-nine (38%) pts progressed requiring frontline therapy including 7 (9%) pts with HT. Twenty-five (32%) pts were monitored ≥2y with no progression including 10 (13%) pts with evidence of at least some spontaneous regression by CT. Twenty (26%) pts were on study for Conclusions: ctDNA quantified from plasma in FL mirrors TMTV. Serial monitoring of ctDNA in patients without therapy demonstrated various patterns of fluctuation, including some patients in which ctDNA became undetectable coincident with spontaneous clinical regressions. ctDNA thus provides a non-invasive platform to monitor the natural history of FL, enabling future studies of tumor immune surveillance in this disease. Figure 1 Figure 1. Disclosures Jacob: Adaptive Biotechnologies: Current Employment, Current equity holder in publicly-traded company. Bagaev: BostonGene Corp.: Current Employment, Current holder of stock options in a privately-held company, Patents & Royalties: BostonGene. Meerson: BostonGene: Current Employment, Current holder of stock options in a privately-held company, Patents & Royalties: BostonGene. Postovalova: BostonGene Corp.: Current Employment, Current holder of stock options in a privately-held company, Patents & Royalties: BostonGene. Kudryashova: BostonGene: Current Employment, Current holder of stock options in a privately-held company, Patents & Royalties: BostonGene. Kotlov: BostonGene Corp: Current Employment, Current holder of stock options in a privately-held company, Patents & Royalties. Fowler: BostonGene: Current Employment, Current holder of stock options in a privately-held company.
- Published
- 2021
26. Integration and Iteration: Using Advanced, High-Content Imaging and Single-Cell Gene Expression Analysis to Uncover Unique Aspects of Follicular Lymphoma Biology
- Author
-
Nishant Thakur, Andrea J. Radtke, Maria Tsiper, Margarita Polyakova, Felix Frenkel, Nathan Fowler, Louis M. Staudt, Olga Plotnikova, Alexander Bagaev, Arthur L. Shaffer, Ekaterina Postovalova, Stefania Pittaluga, Mark Meerson, Sergei Isaev, Pavel Ovcharov, Wyndham H. Wilson, Theresa Davies-Hill, Da-Wei Huang, Bradley C. Lowekamp, Ziv Yaniv, Michael C. Kelly, Jagan R. Muppidi, Elaine S. Jaffe, Nikita Kotlov, Ilia Galkin, Mark Roschewski, Ezzat Dadkhah, Krystle Nomie, Ronald N. Germain, Yaroslav Lozinsky, Viktor Svekolkin, Ravshan Attaulakhanov, Arina Varlamova, and Ekaterina O. Nuzhdina
- Subjects
medicine.anatomical_structure ,Immunology ,Cell ,Gene expression ,medicine ,Follicular lymphoma ,Cell Biology ,Hematology ,Computational biology ,Biology ,medicine.disease ,Biochemistry ,High content imaging - Abstract
BACKGROUND: Follicular lymphoma (FL) is an indolent malignancy of germinal center B-cell origin. FL patients experience remarkable heterogeneity in their disease trajectory, with many patients slowly progressing over several years, and a subset of patients experiencing an aggressive clinical course. Uncovering the cell-intrinsic and -extrinsic factors that govern differential progression and outcome in FL patients is thus essential. Beyond the genetic and epigenetic aberrations that contribute to FL oncogenesis, the tumor microenvironment (TME) plays an integral role in supporting the proliferation and survival of malignant cells. In a process described as "re-education", FL tumor cells may actively subvert the normal functions of non-malignant cells present in the TME, including T cells, follicular dendritic cells (FDCs), macrophages, dendritic cells, and stromal cells to support their survival and growth. METHODS: To address the importance of the TME in FL, we molecularly profiled excisional lymph node biopies from untreated FL patients using multiple platforms: bulk RNA sequencing (RNAseq), single-cell RNA sequencing (scRS), and a unique high content imaging method, Iterative Bleaching Extends MultipleXity (IBEX), which utilizes chemical bleaching to image 40+ proteins in the same tissue section by antibody staining. In combination with advanced computational tools for the quantitative analysis of cell types and distribution in tissues, we have used this approach to evaluate the TME:FL interaction within 8 FL samples, with 4 normal lymph nodes as controls. RESULTS: Both FL and TME components reconstructed from bulk RNA-seq were similar to the cellular composition revealed by scRS and IBEX analyses. Moreover, the bulk RNAseq and scRS identified the expression of genes involved in tumorigenesis and oncogenic signaling, often unique to each case. However, RNAseq-based approaches often miss important cellular and acellular components not readily extracted from dense tissues. For example, IBEX imaging can trace the pattern of blood vessels within a section, which cannot be achieved with non-imaging methods. In one case, our multi-parameter imaging studies revealed the close spatial interaction between clonal FL B cells, expressing a B-cell receptor (BCR) possessing a de novo N-linked glycosylation site introduced by somatic hypermutation, and cells expressing dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN). This contact may activate pro-survival signaling in the malignant B cells. CONCLUSIONS: Integration of bulk RNAseq, scRS, clonotype analysis, and IBEX reveals both shared and unique aspects of different FL tumors. These data highlight the importance of integrating direct tissue analysis by high-content imaging with methods examining aspects of isolated cells. This approach may provide a more complete understanding of tumor biology, which in turn will identify patients at risk for developing aggressive disease and rationally improve treatment strategies for FL. This research was supported in part by the Intramural Research Program of the NIH, NIAID and NCI Figure Disclosures Bagaev: BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Plotnikova:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Galkin:BostonGene: Current Employment, Patents & Royalties. Postovalova:BostonGene: Current Employment, Current equity holder in private company. Svekolkin:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Isaev:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Lozinsky:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Meerson:BostonGene: Current Employment. Varlamova:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Ovcharov:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Polyakova:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Nomie:BostonGene: Current Employment, Current equity holder in private company. Kotlov:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Tsiper:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Frenkel:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Attaulakhanov:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties. Fowler:BostonGene: Current Employment, Current equity holder in private company, Patents & Royalties.
- Published
- 2020
27. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma
- Author
-
Bao Tran, Wenming Xiao, Wyndham H. Wilson, Randy D. Gascoyne, Yongmei Zhao, Joseph M. Connors, Dan R. Soppet, Lisa M. Rimsza, Andreas Rosenwald, Kevin Tay Kuang Wei, Andrew D. Zelenetz, Jan Delabie, Xin Yu, Monica Kasbekar, John P. Leonard, Armando López-Guillermo, Hong Zhao, Weihong Xu, Daniel J. Hodson, James D. Phelan, Louis M. Staudt, Roland Schmitz, Bin Zhou, Stefania Pittaluga, Wei Du, Nancy L. Bartlett, Yandan Yang, Elias Campo, Calvin A. Johnson, James Q. Wang, Elaine S. Jaffe, German Ott, Wing C. Chan, Sandrine Roulland, Jyoti Shetty, Ryan M. Young, George E. Wright, Xuelu Liu, Da-Wei Huang, Arthur L. Shaffer, Universitat de Barcelona, Hodson, Daniel J [0000-0001-6225-2033], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Limfomes ,Genotype ,Cèl·lules B ,Biopsy ,Kaplan-Meier Estimate ,Epigenesis, Genetic ,Transcriptome ,03 medical and health sciences ,Genetic Heterogeneity ,0302 clinical medicine ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Genetics ,Humans ,Exome ,B cells ,Genetic heterogeneity ,business.industry ,Gene Expression Profiling ,General Medicine ,Sequence Analysis, DNA ,Amplicon ,medicine.disease ,Prognosis ,3. Good health ,Gene expression profiling ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Medical genetics ,Lymphomas ,Lymphoma, Large B-Cell, Diffuse ,business ,Diffuse large B-cell lymphoma ,Genètica - Abstract
BACKGROUND: Diffuse large B-cell lymphomas (DLBCLs) are phenotypically and genetically heterogeneous. Gene-expression profiling has identified subgroups of DLBCL (activated B-cell-like [ABC], germinal-center B-cell-like [GCB], and unclassified) according to cell of origin that are associated with a differential response to chemotherapy and targeted agents. We sought to extend these findings by identifying genetic subtypes of DLBCL based on shared genomic abnormalities and to uncover therapeutic vulnerabilities based on tumor genetics. METHODS: We studied 574 DLBCL biopsy samples using exome and transcriptome sequencing, array-based DNA copy-number analysis, and targeted amplicon resequencing of 372 genes to identify genes with recurrent aberrations. We developed and implemented an algorithm to discover genetic subtypes based on the co-occurrence of genetic alterations. RESULTS: We identified four prominent genetic subtypes in DLBCL, termed MCD (based on the co-occurrence of MYD88L265P and CD79B mutations), BN2 (based on BCL6 fusions and NOTCH2 mutations), N1 (based on NOTCH1 mutations), and EZB (based on EZH2 mutations and BCL2 translocations). Genetic aberrations in multiple genes distinguished each genetic subtype from other DLBCLs. These subtypes differed phenotypically, as judged by differences in gene-expression signatures and responses to immunochemotherapy, with favorable survival in the BN2 and EZB subtypes and inferior outcomes in the MCD and N1 subtypes. Analysis of genetic pathways suggested that MCD and BN2 DLBCLs rely on "chronic active" B-cell receptor signaling that is amenable to therapeutic inhibition. CONCLUSIONS: We uncovered genetic subtypes of DLBCL with distinct genotypic, epigenetic, and clinical characteristics, providing a potential nosology for precision-medicine strategies in DLBCL. (Funded by the Intramural Research Program of the National Institutes of Health and others.).
- Published
- 2018
28. Corrected and Republished from: BCL11A Is a Critical Component of a Transcriptional Network That Activates RAG Expression and V(D)J Recombination
- Author
-
Vishwanath R. Iyer, Baeck Seung Lee, Gregory C. Ippolito, Haley O. Tucker, Joseph D. Dekker, Barry P. Sleckman, Bum Kyu Lee, and Arthur L. Shaffer
- Subjects
0301 basic medicine ,Gene isoform ,V(D)J recombination ,hemic and immune systems ,Cell Biology ,Plasmacytoid dendritic cell ,Biology ,Recombination-activating gene ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Regulatory sequence ,RAG2 ,medicine ,Molecular Biology ,Transcription factor ,B cell - Abstract
Recombination activating gene 1 (RAG1) and RAG2 are critical enzymes for initiating variable-diversity-joining [V(D)J] segment recombination, an essential process for antigen receptor expression and lymphocyte development. The BCL11A transcription factor is required for B cell and plasmacytoid dendritic cell (pDC) development, but its molecular function(s) in early B cell fate specification and commitment is unknown. We show here that the major B cell isoform, BCL11A-XL, binds directly to the RAG1 promoter as well as directly to regulatory regions of transcription factors previously implicated in both B cell and pDC development to activate RAG1 and RAG2 gene transcription in pro- and pre-B cells. We employed BCL11A overexpression with recombination substrates to demonstrate direct consequences of BCL11A/RAG modulation on V(D)J recombination. We conclude that BCL11A is a critical component of a transcriptional network that regulates B cell fate by controlling V(D)J recombination.
- Published
- 2018
29. A multiprotein supercomplex controlling oncogenic signalling in lymphoma
- Author
-
Maryknoll Palisoc, Lu Chen, Craig J. Thomas, Michele Ceribelli, Fausto J. Rodriguez, George E. Wright, Yanlong Ji, Jan Delabie, Masao Nakagawa, Ryan M. Young, Wing C. Chan, James D. Phelan, Randy D. Gascoyne, Stephen M. Hewitt, Theresa Davies-Hill, Sandrine Roulland, Dan E. Webster, Monica Kasbekar, Fayez Estephan, Weihong Xu, German Ott, Andreas Rosenwald, Elias Campo, Thomas Oellerich, Emmanuel Bachy, Xin Yu, Lisa M. Rimsza, Hong Zhao, Louis M. Staudt, Roland Schmitz, Michael J. Kruhlak, Stefania Pittaluga, Yandan Yang, Elaine S. Jaffe, Racquel R. Valadez, Arthur L. Shaffer, Matthias Holdhoff, Da-Wei Huang, Wyndham H. Wilson, James Q. Wang, Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Service d’Hématologie [Centre Hospitalier Lyon Sud - HCL], Centre Hospitalier Lyon Sud [CHU - HCL] (CHLS), Hospices Civils de Lyon (HCL)-Hospices Civils de Lyon (HCL), Tianjin University of Science and Technology (TUST), East China Jiaotong University (ECJU), National Institutes of Health [Bethesda] (NIH), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona (UB), Metabolism Branch, National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH)-National Institutes of Health [Bethesda] (NIH)-Center for Cancer Research-National Institutes of Health, University of Toronto, and National Institutes of Health
- Subjects
0301 basic medicine ,Proteomics ,Carcinogenesis ,Biopsy ,Syk ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Piperidines ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,ComputingMilieux_MISCELLANEOUS ,Multidisciplinary ,biology ,TOR Serine-Threonine Kinases ,breakpoint cluster region ,NF-kappa B ,3. Good health ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Ibrutinib ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Female ,Lymphoma, Large B-Cell, Diffuse ,Signal Transduction ,B-cell receptor ,Receptors, Antigen, B-Cell ,Article ,03 medical and health sciences ,medicine ,Bruton's tyrosine kinase ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,B cell ,Adenine ,medicine.disease ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Pyrimidines ,chemistry ,Drug Design ,Multiprotein Complexes ,Toll-Like Receptor 9 ,Mutation ,Myeloid Differentiation Factor 88 ,biology.protein ,Cancer research ,Pyrazoles ,CRISPR-Cas Systems ,Diffuse large B-cell lymphoma - Abstract
B cell receptor (BCR) signalling has emerged as a therapeutic target in B cell lymphomas, but inhibiting this pathway in diffuse large B cell lymphoma (DLBCL) has benefited only a subset of patients1. Gene expression profiling identified two major subtypes of DLBCL, known as germinal centre B cell-like and activated B cell-like (ABC)2,3, that show poor outcomes after immunochemotherapy in ABC. Autoantigens drive BCR-dependent activation of NF-κB in ABC DLBCL through a kinase signalling cascade of SYK, BTK and PKCβ to promote the assembly of the CARD11-BCL10-MALT1 adaptor complex, which recruits and activates IκB kinase4-6. Genome sequencing revealed gain-of-function mutations that target the CD79A and CD79B BCR subunits and the Toll-like receptor signalling adaptor MYD885,7, with MYD88(L265P) being the most prevalent isoform. In a clinical trial, the BTK inhibitor ibrutinib produced responses in 37% of cases of ABC1. The most striking response rate (80%) was observed in tumours with both CD79B and MYD88(L265P) mutations, but how these mutations cooperate to promote dependence on BCR signalling remains unclear. Here we used genome-wide CRISPR-Cas9 screening and functional proteomics to determine the molecular basis of exceptional clinical responses to ibrutinib. We discovered a new mode of oncogenic BCR signalling in ibrutinib-responsive cell lines and biopsies, coordinated by a multiprotein supercomplex formed by MYD88, TLR9 and the BCR (hereafter termed the My-T-BCR supercomplex). The My-T-BCR supercomplex co-localizes with mTOR on endolysosomes, where it drives pro-survival NF-κB and mTOR signalling. Inhibitors of BCR and mTOR signalling cooperatively decreased the formation and function of the My-T-BCR supercomplex, providing mechanistic insight into their synergistic toxicity for My-T-BCR+ DLBCL cells. My-T-BCR supercomplexes characterized ibrutinib-responsive malignancies and distinguished ibrutinib responders from non-responders. Our data provide a framework for the rational design of oncogenic signalling inhibitors in molecularly defined subsets of DLBCL.
- Published
- 2018
30. Selective interleukin-1 receptor–associated kinase 4 inhibitors for the treatment of autoimmune disorders and lymphoid malignancy
- Author
-
Lixin Rui, Donna L. Romero, Divya Chaudhary, Shaughnessy Robinson, Louis M. Staudt, Rosana Kapeller, Wenyan Miao, Priscilla N. Kelly, Arthur L. Shaffer, Yibin Yang, and William F. Westlin
- Subjects
Male ,Gout ,Immunology ,Population ,Receptors, Antigen, B-Cell ,News ,Insights ,Autoimmune Diseases ,chemistry.chemical_compound ,Cell Line, Tumor ,Drug Discovery ,Agammaglobulinaemia Tyrosine Kinase ,medicine ,Animals ,Humans ,Syk Kinase ,Immunology and Allergy ,Bruton's tyrosine kinase ,Autocrine signalling ,education ,Protein Kinase Inhibitors ,Research Articles ,Mice, Inbred BALB C ,education.field_of_study ,Cell Death ,biology ,Tumor Necrosis Factor-alpha ,Brief Definitive Report ,Intracellular Signaling Peptides and Proteins ,Protein-Tyrosine Kinases ,IRAK4 ,medicine.disease ,Arthritis, Experimental ,Interleukin-1 Receptor-Associated Kinases ,Proto-Oncogene Proteins c-bcl-2 ,chemistry ,Mice, Inbred DBA ,Ibrutinib ,Myeloid Differentiation Factor 88 ,Cancer research ,biology.protein ,Lymphoma, Large B-Cell, Diffuse ,Signal transduction ,Diffuse large B-cell lymphoma ,Tyrosine kinase ,Signal Transduction - Abstract
Kelly et al. report the development of two highly selective and bioavailable small molecule IRAK4 inhibitors and show for the first time their therapeutic efficacy in autoimmune disorders and in a specific subset of diffuse large B cell lymphomas in mice., Pathological activation of the Toll-like receptor signaling adaptor protein MYD88 underlies many autoimmune and inflammatory disease states. In the activated B cell–like (ABC) subtype of diffuse large B cell lymphoma (DLBCL), the oncogenic MYD88 L265P mutation occurs in 29% of cases, making it the most prevalent activating mutation in this malignancy. IRAK4 kinase accounts for almost all of the biological functions of MYD88, highlighting IRAK4 as a therapeutic target for diseases driven by aberrant MYD88 signaling. Using innovative structure-based drug design methodologies, we report the development of highly selective and bioavailable small molecule IRAK4 inhibitors, ND-2158 and ND-2110. These small molecules suppressed LPS-induced TNF production, alleviated collagen-induced arthritis, and blocked gout formation in mouse models. IRAK4 inhibition promoted killing of ABC DLBCL lines harboring MYD88 L265P, by down-modulating survival signals, including NF-κB and autocrine IL-6/IL-10 engagement of the JAK–STAT3 pathway. In ABC DLBCL xenograft models, IRAK4 inhibition suppressed tumor growth as a single agent, and in combination with the Bruton’s tyrosine kinase (BTK) inhibitor ibrutinib or the Bcl-2 inhibitor ABT-199. Our findings support pharmacological inhibition of IRAK4 as a therapeutic strategy in autoimmune disorders, in a genetically defined population of ABC DLBCL, and possibly other malignancies dependent on aberrant MYD88 signaling.
- Published
- 2015
31. Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma
- Author
-
Thomas M. Habermann, Chih Jian Lih, Ranjana H. Advani, Davina Moussa, Andre Goy, Paul M. Barr, Rebecca Elstrom, Nathan Fowler, Fong Clow, Betty Y. Chang, Maria Fardis, Darrin M. Beaupre, Brian Munneke, John F. Gerecitano, Ryan M. Young, Wyndham H. Wilson, Louis M. Staudt, Roland Schmitz, Stefania Pittaluga, Vaishalee P. Kenkre, Yandan Yang, P. Mickey Williams, George E. Wright, Andrei R. Shustov, Julie M. Vose, Jacqueline C. Barrientos, Jesse McGreivy, Sven de Vos, Arthur L. Shaffer, and Kristie A. Blum
- Subjects
Adult ,Male ,Molecular Sequence Data ,B-cell receptor ,Receptors, Antigen, B-Cell ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,chemistry.chemical_compound ,Piperidines ,hemic and lymphatic diseases ,medicine ,Humans ,Protein Kinase Inhibitors ,Aged ,Base Sequence ,Adenine ,breakpoint cluster region ,Germinal center ,General Medicine ,Middle Aged ,CD79B ,medicine.disease ,Lymphoma ,Pyrimidines ,chemistry ,Ibrutinib ,Mutation ,Myeloid Differentiation Factor 88 ,Immunology ,Cancer research ,Pyrazoles ,Female ,Lymphoma, Large B-Cell, Diffuse ,Diffuse large B-cell lymphoma ,CD79 Antigens ,Signal Transduction - Abstract
The two major subtypes of diffuse large B cell lymphoma (DLBCL)—activated B cell–like (ABC) and germinal center B cell–like (GCB)—arise by distinct mechanisms, with ABC selectively acquiring mutations that target the B cell receptor (BCR), fostering chronic active BCR signaling(1). The ABC subtype has a ∼40% cure rate with currently available therapies, which is worse than the rate for GCB DLBCL, and highlights the need for ABC subtype-specific treatment strategies(2). We hypothesized that ABC, but not GCB, DLBCL tumors would respond to ibrutinib, an inhibitor of BCR signaling. In a phase 1/2 clinical trial that involved 80 subjects with relapsed or refractory DLBCL, ibrutinib produced complete or partial responses in 37% (14/38) of those with ABC DLBCL, but in only 5% (1/20) of subjects with GCB DLBCL (P = 0.0106). ABC tumors with BCR mutations responded to ibrutinib frequently (5/9; 55.5%), especially those with concomitant myeloid differentiation primary response 88 (MYD88) mutations (4/5; 80%), a result that is consistent with in vitro cooperation between the BCR and MYD88 pathways. However, the highest number of responses occurred in ABC tumors that lacked BCR mutations (9/29; 31%), suggesting that oncogenic BCR signaling in ABC does not require BCR mutations and might be initiated by non-genetic mechanisms. These results support the selective development of ibrutinib for the treatment of ABC DLBCL.
- Published
- 2015
32. B-Cell Receptor Signaling in Diffuse Large B-Cell lymphoma
- Author
-
Louis M. Staudt, Ryan M. Young, James D. Phelan, and Arthur L. Shaffer
- Subjects
Receptors, Antigen, B-Cell ,Biology ,Lymphocyte Activation ,Article ,chemistry.chemical_compound ,immune system diseases ,RNA interference ,hemic and lymphatic diseases ,medicine ,Humans ,B-Lymphocytes ,Effector ,NF-kappa B ,breakpoint cluster region ,Hematology ,medicine.disease ,Lymphoma ,chemistry ,Ibrutinib ,Immunology ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Signal transduction ,Diffuse large B-cell lymphoma ,Tyrosine kinase ,Signal Transduction - Abstract
The importance of understanding the genetic and biochemical basis of B cell receptor (BCR) survival signaling in diffuse large B cell lymphoma (DLBCL) is underscored by the recent clinical success of agents that target the BCR pathway. DLBCL is composed of multiple distinct molecular subtypes with divergent clinical outcomes. The activated B cell-like (ABC) subtype is the most aggressive form of DLBCL and is often resistant to standard chemotherapies. ABC DLBCL expresses numerous genes found in antigen-activated B cells, and genetic and pharmacologic studies have demonstrated that ABC DLBCL tumors are addicted to NF-κB activity. The origins of this NF-κB activity remained obscure until RNA interference screens established that the majority of ABC DLBCL cell lines rely on expression of BCR components and downstream signaling effectors for NF-κB activation. Pharmacological inhibition with ibrutinib of Bruton’s tyrosine kinase (Btk), a kinase that is required for BCR signaling to engage NF-κB, is selectively toxic for ABC DLBCL tumors; a finding that has now been translated to the clinic. These novel targets not only offer a promising new therapy options for ABC DLBCL, but also demonstrate the value of a deep molecular understanding of oncogenic signaling pathways.
- Published
- 2015
33. Cell-Intrinsic Expression of TLR9 in Autoreactive B Cells Constrains BCR/TLR7-Dependent Responses
- Author
-
Ann Marshak-Rothstein, Arthur L. Shaffer, Krishna Moody, Kerstin Nündel, Michael P. Cancro, Patricia Busto, Nathaniel M. Green, Dan Eilat, Kensuke Miyake, and Michael A. Oropallo
- Subjects
Systemic lupus erythematosus ,Cellular differentiation ,Immunology ,virus diseases ,TLR9 ,TLR7 ,Biology ,Plasma cell ,medicine.disease ,medicine.disease_cause ,Autoimmunity ,medicine.anatomical_structure ,Antigen ,immune system diseases ,medicine ,Cancer research ,Immunology and Allergy ,B cell - Abstract
Endosomal TLRs play an important role in systemic autoimmune diseases, such as systemic erythematosus lupus, in which DNA- and RNA-associated autoantigens activate autoreactive B cells through TLR9- and TLR7-dependent pathways. Nevertheless, TLR9-deficient autoimmune-prone mice develop more severe clinical disease, whereas TLR7-deficient and TLR7/9–double deficient autoimmune-prone mice develop less severe disease. To determine whether the regulatory activity of TLR9 is B cell intrinsic, we directly compared the functional properties of autoantigen-activated wild-type, TLR9-deficient, and TLR7-deficient B cells in an experimental system in which proliferation depends on BCR/TLR coengagement. In vitro, TLR9-deficient cells are less dependent on survival factors for a sustained proliferative response than are either wild-type or TLR7-deficient cells. The TLR9-deficient cells also preferentially differentiate toward the plasma cell lineage, as indicated by expression of CD138, sustained expression of IRF4, and other molecular markers of plasma cells. In vivo, autoantigen-activated TLR9-deficient cells give rise to greater numbers of autoantibody-producing cells. Our results identify distinct roles for TLR7 and TLR9 in the differentiation of autoreactive B cells that explain the capacity of TLR9 to limit, as well as TLR7 to promote, the clinical features of systemic erythematosus lupus.
- Published
- 2015
34. Targeting the HTLV-I-Regulated BATF3/IRF4 Transcriptional Network in Adult T Cell Leukemia/Lymphoma
- Author
-
Masao Nakagawa, James D. Phelan, Sigrid Dubois, Patrick L. Green, Louis M. Staudt, Michiyuki Maeda, George E. Wright, Wenming Xiao, Dan E. Webster, Holger Kohlhammer, Yandan Yang, Hee Min Yoo, Hong Zhao, Weihong Xu, Da-Wei Huang, Bonita R. Bryant, Xin Yu, Takashi Ishio, Joji Shimono, Meili Zhang, Yibin Yang, Michele Ceribelli, Emmanuel Bachy, Michael N. Petrus, Thomas A. Waldmann, Arthur L. Shaffer, and John Powell
- Subjects
0301 basic medicine ,Cancer Research ,Genes, myc ,Retroviridae Proteins ,Adult T-cell leukemia/lymphoma ,Article ,BET inhibitor ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Retrovirus ,RNA interference ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Gene Regulatory Networks ,Transcription factor ,Human T-lymphotropic virus 1 ,biology ,Proteins ,Cell Biology ,biology.organism_classification ,medicine.disease ,Chromatin ,Leukemia ,030104 developmental biology ,Basic-Leucine Zipper Transcription Factors ,Oncology ,030220 oncology & carcinogenesis ,Interferon Regulatory Factors ,Cancer research ,IRF4 - Abstract
Summary Adult T cell leukemia/lymphoma (ATLL) is a frequently incurable disease associated with the human lymphotropic virus type I (HTLV-I). RNAi screening of ATLL lines revealed that their proliferation depends on BATF3 and IRF4, which cooperatively drive ATLL-specific gene expression. HBZ, the only HTLV-I encoded transcription factor that is expressed in all ATLL cases, binds to an ATLL-specific BATF3 super-enhancer and thereby regulates the expression of BATF3 and its downstream targets, including MYC. Inhibitors of bromodomain-and-extra-terminal-domain (BET) chromatin proteins collapsed the transcriptional network directed by HBZ and BATF3, and were consequently toxic for ATLL cell lines, patient samples, and xenografts. Our study demonstrates that the HTLV-I oncogenic retrovirus exploits a regulatory module that can be attacked therapeutically with BET inhibitors.
- Published
- 2017
35. Blockade of oncogenic IκB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors
- Author
-
Lesley A. Mathews Griner, Emmanuel Normant, George E. Wright, Jonathan M. Keller, Arthur L. Shaffer, Paul Shinn, Wenming Xiao, Dongbo Liu, Craig J. Thomas, Paresma R. Patel, Peter Sandy, Michele Ceribelli, Shivangi Joshi, Marc Ferrer, Rajarshi Guha, Priscilla N. Kelly, Louis M. Staudt, Sujata Nerle, and Yibin Yang
- Subjects
BRD4 ,Cell Survival ,Cell Cycle Proteins ,Mice, SCID ,IκB kinase ,Protein Serine-Threonine Kinases ,Biology ,BET inhibitor ,Mice ,chemistry.chemical_compound ,Piperidines ,immune system diseases ,Cell Line, Tumor ,hemic and lymphatic diseases ,Animals ,Humans ,Bruton's tyrosine kinase ,Protein Kinase Inhibitors ,Multidisciplinary ,Cell Death ,Adenine ,I-Kappa-B Kinase ,Nuclear Proteins ,Drug Synergism ,Azepines ,Triazoles ,Biological Sciences ,Xenograft Model Antitumor Assays ,I-kappa B Kinase ,Protein Structure, Tertiary ,Bromodomain ,Pyrimidines ,chemistry ,Ibrutinib ,Cancer research ,biology.protein ,Pyrazoles ,Lymphoma, Large B-Cell, Diffuse ,Signal transduction ,Signal Transduction ,Transcription Factors - Abstract
In the activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL), NF-κB activity is essential for viability of the malignant cells and is sustained by constitutive activity of IκB kinase (IKK) in the cytoplasm. Here, we report an unexpected role for the bromodomain and extraterminal domain (BET) proteins BRD2 and BRD4 in maintaining oncogenic IKK activity in ABC DLBCL. IKK activity was reduced by small molecules targeting BET proteins as well as by genetic knockdown of BRD2 and BRD4 expression, thereby inhibiting downstream NF-κB-driven transcriptional programs and killing ABC DLBCL cells. Using a high-throughput platform to screen for drug-drug synergy, we observed that the BET inhibitor JQ1 combined favorably with multiple drugs targeting B-cell receptor signaling, one pathway that activates IKK in ABC DLBCL. The BTK kinase inhibitor ibrutinib, which is in clinical development for the treatment of ABC DLBCL, synergized strongly with BET inhibitors in killing ABC DLBCL cells in vitro and in a xenograft mouse model. These findings provide a mechanistic basis for the clinical development of BET protein inhibitors in ABC DLBCL, particularly in combination with other modulators of oncogenic IKK signaling.
- Published
- 2014
36. B Cells Are Not Essential for Lactobacillus-Mediated Protection against Lethal Pneumovirus Infection
- Author
-
Arthur L. Shaffer, Kimberly D. Dyer, Katia E. Garcia-Crespo, Joseph B. Domachowske, Stanislaw J. Gabryszewski, Caroline M. Percopo, and Helene F. Rosenberg
- Subjects
Immunology ,Heterologous ,Priming (immunology) ,Respiratory Mucosa ,Biology ,Article ,Virus ,Microbiology ,Proinflammatory cytokine ,Mice ,Immunity ,medicine ,Animals ,Pneumovirus Infections ,Immunology and Allergy ,Lung ,B cell ,B-Lymphocytes ,Mice, Inbred BALB C ,Pneumovirus ,Germinal center ,Antibodies, Bacterial ,Lactobacillus ,medicine.anatomical_structure ,Cytokines ,Respiratory virus - Abstract
We have shown previously that priming of respiratory mucosa with live Lactobacillus species promotes robust and prolonged survival from an otherwise lethal infection with pneumonia virus of mice, a property known as heterologous immunity. Lactobacillus priming results in a moderate reduction in virus recovery and a dramatic reduction in virus-induced proinflammatory cytokine production; the precise mechanisms underlying these findings remain to be elucidated. Because B cells have been shown to promote heterologous immunity against respiratory virus pathogens under similar conditions, in this study we explore the role of B cells in Lactobacillus-mediated protection against acute pneumovirus infection. We found that Lactobacillus-primed mice feature elevated levels of airway Igs IgG, IgA, and IgM and lung tissues with dense, B cell (B220+)–enriched peribronchial and perivascular infiltrates with germinal centers consistent with descriptions of BALT. No B cells were detected in lung tissue of Lactobacillus-primed B cell deficient μMT mice or Jh mice, and Lactobacillus-primed μMT mice had no characteristic infiltrates or airway Igs. Nonetheless, we observed diminished virus recovery and profound suppression of virus-induced proinflammatory cytokines CCL2, IFN-γ, and CXCL10 in both wild-type and Lactobacillus-primed μMT mice. Furthermore, Lactobacillus plantarum–primed, B cell–deficient μMT and Jh mice were fully protected from an otherwise lethal pneumonia virus of mice infection, as were their respective wild-types. We conclude that B cells are dispensable for Lactobacillus-mediated heterologous immunity and were not crucial for promoting survival in response to an otherwise lethal pneumovirus infection.
- Published
- 2014
37. KLHL14 Is a Tumor Suppressor in Diffuse Large B-Cell Lymphoma
- Author
-
James D. Phelan, Louis M. Staudt, Ryan M. Young, Thomas Oellerich, Da-Wei Huang, Jaewoo Choi, and Arthur L. Shaffer
- Subjects
Oncogene Proteins ,Chemistry ,Immunology ,Cancer ,Cell Biology ,Hematology ,NFKB1 ,medicine.disease ,Biochemistry ,law.invention ,Lymphoma ,Gene expression profiling ,law ,Cell culture ,medicine ,Cancer research ,Suppressor ,Diffuse large B-cell lymphoma - Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive cancer of aberrant B-lymphocytes. Although a portion of DLBCL is curable with standard immunochemotherapy, patients who fail this treatment have a poor prognosis. Recently, cancer genomics has paved the way for better understanding of the genetic basis of lymphoma pathogenesis. Characterization of point mutations and structural alterations has uncovered novel molecular targets for lymphoma therapy and provided a comprehensive view of lymphoma development. By performing multiplatform genomic analysis of DLBCL biopsy samples, we have identified KLHL14 as a recurrent target of somatic mutations in activated B-cell-like (ABC) DLBCL biopsies (10.8% of patients). KLHL14 contains a BTB (broad complex, tramtrack, and bric a brac) domain that can potentially mediate dimerization and binding to Cullin3 (CUL3)-a essential scaffold component of the Cullin-RING-based E3 ubiquitin ligase complexes. KLHL14 also contains kelch repeats that can form a B-propeller tertiary structure that can serve as a substrate-binding domain. KLHL14 is highly expressed in B-cells but is found at low levels in non-immune tissues. Deficiency of KLHL14 in mice leads to embryonic lethality while KLHL14 heterozygous mice show reduction of B-1a cells, suggesting a role for KLHL14 in B-cell homeostasis. Importantly, KLHL14 mutations are highly enriched in tumors belonging to the recently defined MCD (MYD88L265P/CD79B mutation) genetic subtype of DLBCL, the subset of ABC DLBCLs. Somatic mutations primarily localize to the N-terminus of the protein in the BTB domain and BACK (BTB and C-terminal Kelch) domain. However, the impact of these mutations as well as the molecular function of KLHL14 is largely unknown. To investigate the biological effect of KLHL14 loss of function, we used an inducible CRISPR/Cas9 system to delete KLHL14 in ABC DLBCL cell lines and monitored cell growth. Ablation of KLHL14 resulted in an increase in cell proliferation and survival, supporting a role for KLHL14 as a tumor suppressor. Next, we performed a multiplatform -omic analysis (proteomics, phosphoproteomics, ubiquitinomics, high-throughput sequencing) to explore the signaling networks and interactome of KLHL14. Whereas ectopic expression of wild-type KLHL14 altered the dynamics of tyrosine phosphorylation and ubiquitylation events in ABC DLBCL lines, KLHL14 lymphoma-associated mutant alleles had little if any effect, suggesting that they are loss-of-function variants. Gene expression profiling by RNA-sequencing revealed that KLHL14-inactivated cells have a higher NF-kB target gene expression than wild-type cells. Thus, tumor-associated inactivating mutations of KLHL14 depend on a subset of essential NF-kB-related oncoproteins for their survival and this might contribute to the proliferative advantage of DLBCL. In summary, we have uncovered a tumor suppressive function of KLHL14 and found that KLHL14 mutants promote ABC DLBCL survival by increasing NF-kB activity. These findings suggest that tumors with KLHL14 inactivating mutations may serve as a marker of resistance to anti-NF-kB treatment and provide the basis for treating MCD subtype patients with downstream NF-kB pathway inhibitors in the clinical settings. Disclosures Staudt: Nanostring: Patents & Royalties.
- Published
- 2019
38. Epigenetic gene regulation by Janus kinase 1 in diffuse large B-cell lymphoma
- Author
-
Louis M. Staudt, George E. Wright, Wenming Xiao, Yandan Yang, Michele Ceribelli, Hong Zhao, Lixin Rui, Weihong Xu, Daniel J. Hodson, Kreg M. Grindle, Yangguang Li, Da-Wei Huang, Arthur L. Shaffer, Fen Zhu, Li Lu, Amanda C. Drennan, Hodson, Daniel [0000-0001-6225-2033], and Apollo - University of Cambridge Repository
- Subjects
STAT3 Transcription Factor ,0301 basic medicine ,Apoptosis ,lymphoma ,Epigenesis, Genetic ,03 medical and health sciences ,immune system diseases ,oncogene ,Cell Line, Tumor ,hemic and lymphatic diseases ,Humans ,Medicine ,Bruton's tyrosine kinase ,histone modification ,Epigenetics ,Autocrine signalling ,STAT3 ,Regulation of gene expression ,Multidisciplinary ,biology ,Janus kinase 1 ,epigenetics ,business.industry ,Gene Expression Profiling ,Janus Kinase 1 ,Chromatin ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,JAK1 ,PNAS Plus ,biology.protein ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Janus kinase ,business - Abstract
Janus kinases (JAKs) classically signal by activating STAT transcription factors but can also regulate gene expression by epigenetically phosphorylating histone H3 on tyrosine 41 (H3Y41-P). In diffuse large B-cell lymphomas (DLBCLs), JAK signaling is a feature of the activated B-cell (ABC) subtype and is triggered by autocrine production of IL-6 and IL-10. Whether this signaling involves STAT activation, epigenetic modification of chromatin, or both mechanisms is unknown. Here we use genetic and pharmacological inhibition to show that JAK1 signaling sustains the survival of ABC DLBCL cells. Whereas STAT3 contributed to the survival of ABC DLBCL cell lines, forced STAT3 activity could not protect these cells from death following JAK1 inhibition, suggesting epigenetic JAK1 action. JAK1 regulated the expression of nearly 3,000 genes in ABC DLBCL cells, and the chromatin surrounding many of these genes was modified by H3Y41-P marks that were diminished by JAK1 inhibition. These JAK1 epigenetic target genes encode important regulators of ABC DLBCL proliferation and survival, including IRF4, MYD88, and MYC. A small molecule JAK1 inhibitor cooperated with the BTK inhibitor ibrutinib in reducing IRF4 levels and acted synergistically to kill ABC DLBCL cells, suggesting that this combination should be evaluated in clinical trials.
- Published
- 2016
39. A Druggable TCF4 and BRD4 dependent Transcriptional Network Sustains Malignancy in Blastic Plasmacytoid Dendritic Cell Neoplasm
- Author
-
Priscilla N. Kelly, Rajarshi Guha, Elaine S. Jaffe, Louis M. Staudt, Boris Reizis, Craig J. Thomas, Joel Plumas, Laurence Chaperot, Wenming Xiao, Arthur L. Shaffer, Stefania Pittaluga, George E. Wright, Zhiying Esther Hou, Marc Ferrer, Da-Wei Huang, Moses O. Evbuomwan, Xiaohu Zhang, Karthik A. Ganapathi, Stephen J. Forman, Michele Ceribelli, and Guido Marcucci
- Subjects
0301 basic medicine ,Cancer Research ,BRD4 ,Skin Neoplasms ,Cell Cycle Proteins ,HL-60 Cells ,Biology ,Malignancy ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Jurkat Cells ,Mice ,Super-enhancer ,Transcription Factor 4 ,Downregulation and upregulation ,RNA interference ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,RNA, Small Interfering ,Transcription factor ,Myeloproliferative Disorders ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Nuclear Proteins ,Cell Biology ,TCF4 ,Dendritic Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Bromodomain ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Hematologic Neoplasms ,Immunology ,Cancer research ,Transcription Factors - Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an aggressive and largely incurable hematologic malignancy originating from plasmacytoid dendritic cells (pDCs). Using RNA interference screening, we identified the E-box transcription factor TCF4 as a master regulator of the BPDCN oncogenic program. TCF4 served as a faithful diagnostic marker of BPDCN, and its downregulation caused the loss of the BPDCN-specific gene expression program and apoptosis. High-throughput drug screening revealed that bromodomain and extra-terminal domain inhibitors (BETi’s) induced BPDCN apoptosis, which was attributable to disruption of a BPDCN-specific transcriptional network controlled by TCF4-dependent super-enhancers. BETi’s retarded the growth of BPDCN xenografts, supporting their clinical evaluation in this recalcitrant malignancy.
- Published
- 2016
40. Regulation of normal B-cell differentiation and malignant B-cell survival by OCT2
- Author
-
Masao Nakagawa, James D. Phelan, Wenming Xiao, Holger Kohlhammer, Dan E. Webster, Daniel J. Hodson, Thomas A. Waldmann, Louis M. Staudt, Roland Schmitz, George E. Wright, Lixin Rui, Arthur L. Shaffer, Yandan Yang, Hodson, Daniel [0000-0001-6225-2033], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,XBP1 ,Organic Cation Transport Proteins ,Cell Survival ,Cellular differentiation ,lymphoma ,Biology ,03 medical and health sciences ,Transactivation ,Mice ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,Conditional gene knockout ,Coactivator ,medicine ,Animals ,B cell ,cancer biology ,Mice, Knockout ,B-Lymphocytes ,Multidisciplinary ,Germinal center ,Organic Cation Transporter 2 ,Cell Differentiation ,Molecular biology ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,germinal center ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Chromatin immunoprecipitation - Abstract
The requirement for the B-cell transcription factor OCT2 (octamer-binding protein 2, encoded by Pou2f2) in germinal center B cells has proved controversial. Here, we report that germinal center B cells are formed normally after depletion of OCT2 in a conditional knockout mouse, but their proliferation is reduced and in vivo differentiation to antibody-secreting plasma cells is blocked. This finding led us to examine the role of OCT2 in germinal center-derived lymphomas. shRNA knockdown showed that almost all diffuse large B-cell lymphoma (DLBCL) cell lines are addicted to the expression of OCT2 and its coactivator OCA-B. Genome-wide chromatin immunoprecipitation (ChIP) analysis and gene-expression profiling revealed the broad transcriptional program regulated by OCT2 that includes the expression of STAT3, IL-10, ELL2, XBP1, MYC, TERT, and ADA. Importantly, genetic alteration of OCT2 is not a requirement for cellular addiction in DLBCL. However, we detected amplifications of the POU2F2 locus in DLBCL tumor biopsies and a recurrent mutation of threonine 223 in the DNA-binding domain of OCT2. This neomorphic mutation subtly alters the DNA-binding preference of OCT2, leading to the transactivation of noncanonical target genes including HIF1a and FCRL3 Finally, by introducing mutations designed to disrupt the OCT2-OCA-B interface, we reveal a requirement for this protein-protein interface that ultimately might be exploited therapeutically. Our findings, combined with the predominantly B-cell-restricted expression of OCT2 and the absence of a systemic phenotype in our knockout mice, suggest that an OCT2-targeted therapeutic strategy would be efficacious in both major subtypes of DLBCL while avoiding systemic toxicity.
- Published
- 2016
41. Subtype-specific addiction of the activated B-cell subset of diffuse large B-cell lymphoma to FOXP1
- Author
-
Bum Kyu Lee, Arthur L. Shaffer, Vishwanath R. Iyer, George Georgiou, Louis M. Staudt, Wei Deng, Daechan Park, Holger Kohlhammer, Haley O. Tucker, Gregory C. Ippolito, and Joseph D. Dekker
- Subjects
0301 basic medicine ,Transcription, Genetic ,Cellular differentiation ,Biology ,Lymphocyte Activation ,03 medical and health sciences ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,Gene expression ,medicine ,Humans ,neoplasms ,B cell ,B-Lymphocytes ,Multidisciplinary ,Germinal center ,Cell Differentiation ,Forkhead Transcription Factors ,FOXP1 ,medicine.disease ,BCL6 ,Lymphoma ,Repressor Proteins ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Diffuse large B-cell lymphoma - Abstract
High expression of the forkhead box P1 (FOXP1) transcription factor distinguishes the aggressive activated B cell (ABC) diffuse large B-cell lymphoma (DLBCL) subtype from the better prognosis germinal center B-cell (GCB)-DLBCL subtype and is highly correlated with poor outcomes. A genetic or functional role for FOXP1 in lymphomagenesis, however, remains unknown. Here, we report that sustained FOXP1 expression is vital for ABC-DLBCL cell-line survival. Genome-wide analyses revealed direct and indirect FOXP1 transcriptional enforcement of ABC-DLBCL hallmarks, including the classical NF-κB and MYD88 (myeloid differentiation primary response gene 88) pathways. FOXP1 promoted gene expression underlying transition of the GCB cell to the plasmablast--the transient B-cell stage targeted in ABC-DLBCL transformation--by antagonizing pathways distinctive of GCB-DLBCL, including that of the GCB "master regulator," BCL6 (B-cell lymphoma 6). Cell-line derived FOXP1 target genes that were highly correlated with FOXP1 expression in primary DLBCL accurately segregated the corresponding clinical subtypes of a large cohort of primary DLBCL isolates and identified conserved pathways associated with ABC-DLBCL pathology.
- Published
- 2016
42. Prolonged early G1 arrest by selective CDK4/CDK6 inhibition sensitizes myeloma cells to cytotoxic killing through cell cycle–coupled loss of IRF4
- Author
-
Jun Liang, Scott Ely, Jamieson Bretz, Selina Chen-Kiang, Isan Chen, Xiangao Huang, David Jayabalan, Maurizio Di Liberto, Louis M. Staudt, Malcolm A.S. Moore, Tracey Louie, Ruben Niesvizky, Sophia Randolph, Arthur L. Shaffer, and William C. Hahn
- Subjects
Time Factors ,Immunology ,Down-Regulation ,Apoptosis ,Mice, Transgenic ,Mice, SCID ,Biochemistry ,Substrate Specificity ,Bortezomib ,Mice ,Mice, Inbred NOD ,Cyclin-dependent kinase ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Protein Kinase Inhibitors ,Multiple myeloma ,Lymphoid Neoplasia ,biology ,Cytotoxins ,Cyclin-dependent kinase 4 ,Cell Cycle ,Cyclin-Dependent Kinase 4 ,Drug Synergism ,Cyclin-Dependent Kinase 6 ,Cell Biology ,Hematology ,medicine.disease ,Boronic Acids ,G1 Phase Cell Cycle Checkpoints ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Drug Resistance, Neoplasm ,Pyrazines ,Interferon Regulatory Factors ,Cancer cell ,biology.protein ,Proteasome inhibitor ,Cancer research ,Cyclin-dependent kinase 6 ,Multiple Myeloma ,medicine.drug - Abstract
Dysregulation of cyclin-dependent kinase 4 (CDK4) and CDK6 by gain of function or loss of inhibition is common in human cancer, including multiple myeloma, but success in targeting CDK with broad-spectrum inhibitors has been modest. By selective and reversible inhibition of CDK4/CDK6, we have developed a strategy to both inhibit proliferation and enhance cytotoxic killing of cancer cells. We show that induction of prolonged early-G1 arrest (pG1) by CDK4/CDK6 inhibition halts gene expression in early-G1 and prevents expression of genes programmed for other cell-cycle phases. Removal of the early-G1 block leads to S-phase synchronization (pG1-S) but fails to completely restore scheduled gene expression. Consequently, the IRF4 protein required to protect myeloma cells from apoptosis is markedly reduced in pG1 and further in pG1-S in response to cytotoxic agents, such as the proteasome inhibitor bortezomib. The coordinated loss of IRF4 and gain of Bim sensitize myeloma tumor cells to bortezomib-induced apoptosis in pG1 in the absence of Noxa and more profoundly in pG1-S in cooperation with Noxa in vitro. Induction of pG1 and pG1-S by reversible CDK4/CDK6 inhibition further augments tumor-specific bortezomib killing in myeloma xenografts. Reversible inhibition of CDK4/CDK6 in sequential combination therapy thus represents a novel mechanism-based cancer therapy.
- Published
- 2012
43. Prostaglandin E2 Suppresses Antifungal Immunity by Inhibiting Interferon Regulatory Factor 4 Function and Interleukin-17 Expression in T Cells
- Author
-
Patricia A. Valdez, Arthur L. Shaffer, Paul J. Vithayathil, Sandip K. Datta, Brian M. Janelsins, and Peter R. Williamson
- Subjects
Cellular differentiation ,Immunology ,Biology ,Lymphocyte Activation ,Dinoprostone ,Article ,Mice ,Immunity ,medicine ,Animals ,Immunology and Allergy ,Prostaglandin E2 ,Transcription factor ,Cells, Cultured ,Interleukins ,Interleukin-17 ,Interleukin ,Cell Differentiation ,Cryptococcosis ,Cell biology ,Mice, Inbred C57BL ,Infectious Diseases ,Interferon Regulatory Factors ,Cryptococcus neoformans ,Th17 Cells ,Interleukin 17 ,Interferon regulatory factors ,IRF4 ,medicine.drug - Abstract
SummaryT helper 17 (Th17) cells play an important role in mucosal host defense through production of the signature cytokines IL-17 and IL-22. Prostaglandin E2 (PGE2) has been shown to enhance IL-17 production by mature Th17 cells. However, when present during Th17 cell differentiation, we found that PGE2 inhibited the transcription factor IRF4 and suppressed production of IL-17 but not IL-22. We show that IRF4 was required for IL-17 expression but inhibited IL-22 expression, highlighting the potential for discordant regulation of these two cytokines in Th17 cells. The pathogenic fungus Cryptococcus neoformans produces PGE2, and we found that it uses PGE2- and IRF4-dependent mechanisms to specifically inhibit induction of IL-17 during Th17 cell differentiation. Blockade of host PGE2 during infection led to increased IL-17 production from CD4+ T cells and increased survival of mice. These findings suggest that host- or pathogen-derived PGE2 can act directly on Th17 cells during differentiation to inhibit IL-17-dependent antimicrobial responses.
- Published
- 2012
- Full Text
- View/download PDF
44. A mechanistic rationale for MEK inhibitor therapy in myeloma based on blockade of MAF oncogene expression
- Author
-
Christina M. Annunziata, Laurence Lamy, Lidia Hernandez, Adriana Zingone, Elaine M. Hurt, W. Michael Kuehl, Arthur L. Shaffer, Louis M. Staudt, Lloyd T. Lam, and R. Eric Davis
- Subjects
Mitogen-Activated Protein Kinase Kinases ,MAPK/ERK pathway ,Lymphoid Neoplasia ,Transcription, Genetic ,Kinase ,MEK inhibitor ,Immunology ,Oncogene Protein v-maf ,Apoptosis ,Histone-Lysine N-Methyltransferase ,Cell Biology ,Hematology ,Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,Repressor Proteins ,Gene Expression Regulation ,Cancer research ,Humans ,Multiple Myeloma ,Protein kinase A ,Protein Kinases ,Chromatin immunoprecipitation ,Transcription factor - Abstract
Modulating aberrant transcription of oncogenes is a relatively unexplored opportunity in cancer therapeutics. In approximately 10% of multiple myelomas, the initiating oncogenic event is translocation of musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a transcriptional activator of key target genes, including cyclinD2. Our prior work showed that MAF is up-regulated in an additional 30% of multiple myeloma cases. The present study describes a common mechanism inducing MAF transcription in both instances. The second mode of MAF transcription occurred in myelomas with multiple myeloma SET domain (MMSET) translocation. MMSET knockdown decreased MAF transcription and cell viability. A small-molecule screen found an inhibitor of mitogen-activated protein kinase kinase (MEK), which activates extracellular signal-regulated kinase (ERK)-MAP kinases, reduced MAF mRNA in cells representing MMSET or MAF subgroups. ERK activates transcription of FOS, part of the AP-1 transcription factor. By chromatin immunoprecipitation, FOS bound the MAF promoter, and MEK inhibition decreased this interaction. MEK inhibition selectively induced apoptosis in MAF-expressing myelomas, and FOS inactivation was similarly toxic. Reexpression of MAF rescued cells from death induced by MMSET depletion, MEK inhibition, or FOS inactivation. The data presented herein demonstrate that the MEK-ERK pathway regulates MAF transcription, providing molecular rationale for clinical evaluation of MEK inhibitors in MAF-expressing myeloma.
- Published
- 2011
45. Oncogenically active MYD88 mutations in human lymphoma
- Author
-
Erlend B. Smeland, Elaine S. Jaffe, Richard I. Fisher, Randy D. Gascoyne, Kian-Huat Lim, Sameer Jhavar, Vu N. Ngo, George E. Wright, Louis M. Staudt, Roland Schmitz, Elias Campo, Holger Kohlhammer, Hans K. Müller-Hermelink, German Ott, Paul B. Romesser, Arthur L. Shaffer, Ryan M. Young, Lisa M. Rimsza, Yandan Yang, Denny D. Weisenburger, John Powell, James R. Cook, Jan Delabie, Wenming Xiao, Wing C. Chan, Joseph M. Connors, Weihong Xu, Raymond R. Tubbs, Andreas Rosenwald, Rita M. Braziel, and Hong Zhao
- Subjects
STAT3 Transcription Factor ,Cell Survival ,Molecular Sequence Data ,Biology ,medicine.disease_cause ,Article ,immune system diseases ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,Amino Acid Sequence ,Phosphorylation ,Kinase activity ,Janus Kinases ,Mutation ,Multidisciplinary ,Sequence Analysis, RNA ,Kinase ,Toll-Like Receptors ,NF-kappa B ,High-Throughput Nucleotide Sequencing ,Receptors, Interleukin-1 ,Lymphoma, B-Cell, Marginal Zone ,Oncogenes ,IRAK4 ,medicine.disease ,Burkitt Lymphoma ,Protein Structure, Tertiary ,Lymphoma ,Interleukin-1 Receptor-Associated Kinases ,Amino Acid Substitution ,Myeloid Differentiation Factor 88 ,Cancer research ,Cytokines ,Mutant Proteins ,RNA Interference ,Lymphoma, Large B-Cell, Diffuse ,Carcinogenesis ,Janus kinase ,Hydrophobic and Hydrophilic Interactions ,Diffuse large B-cell lymphoma ,Signal Transduction - Abstract
The activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) remains the least curable form of this malignancy despite recent advances in therapy1. Constitutive nuclear factor (NF)-κB and JAK kinase signalling promotes malignant cell survival in these lymphomas, but the genetic basis for this signalling is incompletely understood. Here we describe the dependence of ABC DLBCLs on MYD88, an adaptor protein that mediates toll and interleukin (IL)-1 receptor signalling2,3, and the discovery of highly recurrent oncogenic mutations affecting MYD88 in ABC DLBCL tumours. RNA interference screening revealed that MYD88 and the associated kinases IRAK1 and IRAK4 are essential for ABC DLBCL survival. High-throughput RNA resequencing uncovered MYD88 mutations in ABC DLBCL lines. Notably, 29% of ABC DLBCL tumours harboured the same amino acid substitution, L265P, in the MYD88 Toll/IL-1 receptor (TIR) domain at an evolutionarily invariant residue in its hydrophobic core. This mutation was rare or absent in other DLBCL subtypes and Burkitt’s lymphoma, but was observed in 9% of mucosa-associated lymphoid tissue lymphomas. At a lower frequency, additional mutations were observed in the MYD88 TIR domain, occurring in both the ABC and germinal centre B-cell-like (GCB) DLBCL subtypes. Survival of ABC DLBCL cells bearing the L265P mutation was sustained by the mutant but not the wild-type MYD88 isoform, demonstrating that L265P is a gain-of-function driver mutation. The L265P mutant promoted cell survival by spontaneously assembling a protein complex containing IRAK1 and IRAK4, leading to IRAK4 kinase activity, IRAK1 phosphorylation, NF-κB signalling, JAK kinase activation of STAT3, and secretion of IL-6, IL-10 and interferon-β. Hence, theMYD88 signalling pathway is integral to the pathogenesis of ABC DLBCL, supporting the development of inhibitors of IRAK4 kinase and other components of this pathway for the treatment of tumours bearing oncogenic MYD88 mutations.
- Published
- 2010
46. Cooperative Epigenetic Modulation by Cancer Amplicon Genes
- Author
-
Joseph M. Connors, Wing C. Chan, Peter Möller, Jan Delabie, Graham W. Slack, Andreas Rosenwald, Kerry J. Savage, David Levens, Vu N. Ngo, N. C. Tolga Emre, Erlend B. Smeland, Dennis D. Weisenburger, David G. Maloney, Lixin Rui, Georg Lenz, Elias Campo, Hong Zhao, Hye Jung Chung, Louis M. Staudt, Randy D. Gascoyne, Craig J. Thomas, George E. Wright, Christian Steidl, Michael J. Kruhlak, German Ott, Yandan Yang, John Powell, Elaine S. Jaffe, Hans K. Müller-Hermelink, R. Eric Davis, Lisa M. Rimsza, Arthur L. Shaffer, Wenming Xiao, Weihong Xu, and Laurence Lamy
- Subjects
Jumonji Domain-Containing Histone Demethylases ,Cancer Research ,Lymphoma, B-Cell ,Mediastinal Neoplasms ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Cell Line, Tumor ,hemic and lymphatic diseases ,Histone H2A ,Humans ,Epigenetics ,Phosphorylation ,030304 developmental biology ,0303 health sciences ,biology ,Cell Biology ,Epigenome ,Janus Kinase 2 ,Amplicon ,Hodgkin Disease ,Histone ,Histone phosphorylation ,Oncology ,030220 oncology & carcinogenesis ,Histone methyltransferase ,biology.protein ,Cancer research ,Chromosomes, Human, Pair 9 - Abstract
SummaryChromosome band 9p24 is frequently amplified in primary mediastinal B cell lymphoma (PMBL) and Hodgkin lymphoma (HL). To identify oncogenes in this amplicon, we screened an RNA interference library targeting amplicon genes and thereby identified JAK2 and the histone demethylase JMJD2C as essential genes in these lymphomas. Inhibition of JAK2 and JMJD2C cooperated in killing these lymphomas by decreasing tyrosine 41 phosphorylation and increasing lysine 9 trimethylation of histone H3, promoting heterochromatin formation. MYC, a major target of JAK2-mediated histone phosphorylation, was silenced after JAK2 and JMJD2C inhibition, with a corresponding increase in repressive chromatin. Hence, JAK2 and JMJD2C cooperatively remodel the PMBL and HL epigenome, offering a mechanistic rationale for the development of JAK2 and JMJD2C inhibitors in these diseases.
- Published
- 2010
47. Targeting the HTLV-I-Regulated BATF3/IRF4 Transcriptional Network in Adult T-Cell Leukemia/Lymphoma
- Author
-
Bonita R. Bryant, John Powell, Hee Min Yoo, Hong Zhao, Xin Yu, Michael N. Petrus, Michele Ceribelli, Thomas A. Waldmann, L. M. Staudt, Michiyuki Maeda, Masao Nakagawa, Yibin Yang, Meili Zhang, James D. Phelan, Weihong Xu, George E. Wright, Holger Kohlhammer, Yandan Yang, Da-Wei Huang, Wenming Xiao, and Arthur L. Shaffer
- Subjects
BRD4 ,T cell ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Gene expression profiling ,medicine.anatomical_structure ,immune system diseases ,hemic and lymphatic diseases ,BATF ,medicine ,Cancer research ,Epigenetics ,Transcription factor ,B cell ,IRF4 - Abstract
After neonatal HTLV-I infection through breast feeding, approximately 5% of HTLV-I carriers eventually develop Adult T-Cell Leukemia/Lymphoma (ATLL) with a latency of ~50 years, suggesting that acquired genetic and epigenetic changes in cellular genes act in concert with HTLV-I to initiate and maintain oncogenic transformation. We and others have recently utilized next generation sequencing technology to identify mutated genes that could be pivotal in the pathogenesis of ATLL. However, due to the complexity of genomic/epigenetic alteration in the ATLL genome, the identification of indispensable genes for proliferation and/or survival of ATLL cells remains a formidable challenge. To discover essential regulatory networks that are required for the proliferation and survival of ATLL cells, we performed a pooled shRNA screen in 8 ATLL cell lines using a library enriched for shRNAs targeting lymphoid regulatory factors and discovered that two BATF3 shRNAs and one IRF4 shRNA were highly toxic for all ATLL lines, but had little if any effect in other T cell and B cell lines. It is recently shown that a transcriptional complex of Irf4 and Batf binds to AP1-IRF composite (AICE) DNA motifs and plays key roles in the differentiation and function of certain mouse helper T cell subsets. A close paralogue of Batf, Batf3, is an indispensable transcription factor in a mouse dendritic cell subset, but also appears to play a redundant role with Batf in the differentiation of TH2 cells and can substitute for Batf in Batf knockout T cells. Our observations from shRNA screening suggested that IRF4 and BATF3 may cooperate to drive a transcriptional program that is essential for ATLL viability. We next used genome-wide chromatin precipitation (ChIP-seq) to identify the loci that are bound by BATF3 and IRF4. The set of binding peaks and the associated genes in IRF4 and BATF3 ChIP-seq intersected significantly. By integrating the ChIP-seq and gene expression profiling data of shBATF3- and shIRF4-ATLL cells, we defined a set of 68 BATF3-IRF4 direct target genes. Gene set enrichment analysis using gene expression profiling data from primary T cell lymphomas demonstrated that BATF3-IRF4 direct target genes were significantly enriched among genes that are more highly expressed in ATLL than in peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), suggesting that the BATF3 and IRF4 cooperatively regulate transcription in primary ATLL cells. HBZ is unique among HTLV-I viral proteins in being maintained in expression in all ATLL cases, suggesting that it may help maintain the malignant phenotype. Given that BATF3 and IRF4 are essential regulators in ATLL, we hypothesized possible relationship between HBZ and BATF3-IRF4 complex. We defined HBZ direct target genes by integrating the ChIP-seq and gene expression profiling data of HBZ-knockout ATLL cell lines by CRISPR/Cas9. Notably we discovered that BATF3 was among these. BATF3 mRNA and protein expression decreased following HBZ inactivation. The above considerations suggested that pharmacologic inhibition of the BATF3-IRF4 regulatory network might be a means to attack the HBZ oncogenic program therapeutically. ChIP-seq analysis of two enhancer marks, H3K27ac and BRD4, identified super-enhancers at the BATF3 locus in two ATLL cell lines. The small molecule JQ1 prevents the BET-protein BRD4 from interacting with chromatin, which is required for the function of super-enhancers. JQ1 treatment reduced BATF3 mRNA and protein levels in all ATLL lines tested, correlating with the eviction of BRD4 from the BATF3 super-enhancer. MYC mRNA and protein expression was also broadly downmodulated by JQ1. JQ1 treatment was consistently toxic for all ATLL cell lines tested at dose ranges that killed cell line models of T-ALL and DLBCL, which are known to rely on BET-proteins. In a dose-dependent manner, JQ1 also reduced the viability of primary ATLL samples and downregulated their expression BATF3 and MYC mRNA. Finally, we treated mouse xenograft models of ATLL with the BET-protein inhibitor CPI-203, a JQ1 analog with superior bioavailability in mice. In two different xenograft models, we observed significant tumor regression or growth inhibition, without evidence of systemic toxicity. Our study demonstrates that the HTLV-I virus exploits a regulatory module that can potentially be attacked therapeutically with BET protein inhibitors. Disclosures Yu: Celgene Corporation: Employment.
- Published
- 2017
48. IRF4: Immunity. Malignancy! Therapy?
- Author
-
Arthur L. Shaffer, Louis M. Staudt, Paul B. Romesser, and N. C. Tolga Emre
- Subjects
Cancer Research ,Myeloid ,Cellular differentiation ,Regulator ,Biology ,Article ,Immune system ,medicine ,Animals ,Humans ,Myeloid Cells ,Lymphocytes ,Transcription factor ,Multiple myeloma ,Immunity ,Cell Differentiation ,Dendritic Cells ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Interferon Regulatory Factors ,Immunology ,Cancer research ,Multiple Myeloma ,IRF4 ,Interferon regulatory factors - Abstract
IRF4, a member of the Interferon Regulatory Factor (IRF) family of transcription factors, is expressed in cells of the immune system, where it transduces signals from various receptors to activate or repress gene expression. IRF4 expression is a key regulator of several steps in lymphoid-, myeloid-, and dendritic-cell differentiation, including the differentiation of mature B cells into antibody-secreting plasma cells. IRF4 expression is also associated with many lymphoid malignancies, with recent evidence pointing to an essential role in multiple myeloma, a malignancy of plasma cells. Interference with IRF4 expression is lethal to multiple myeloma cells, irrespective of their genetic etiology, making IRF4 an “Achilles' heel” that may be exploited therapeutically.
- Published
- 2009
49. Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways
- Author
-
Lisa M. Rimsza, Lloyd T. Lam, Elaine S. Jaffe, Elias Campo, Sandeep S. Dave, Arthur L. Shaffer, N. C. Tolga Emre, Dennis D. Weisenburger, German Ott, Hans Konrad Müller-Hermelink, Randy D. Gascoyne, John Powell, Wing C. Chan, Holger Kohlhammer, George E. Wright, Shannon A. Carty, Jan Delabie, Wenming Xiao, Joseph M. Connors, Georg Lenz, Erlend B. Smeland, Andreas Rosenwald, R. Eric Davis, Richard I. Fisher, and Louis M. Staudt
- Subjects
Cell Survival ,Biopsy ,Copy number analysis ,Biology ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Humans ,PTEN ,neoplasms ,Chromosome Aberrations ,Oncogene Proteins ,Multidisciplinary ,Genome, Human ,Gene Expression Profiling ,Tumor Suppressor Proteins ,Germinal center ,FOXP1 ,Biological Sciences ,Amplicon ,Prognosis ,medicine.disease ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Chromosome 3 ,biology.protein ,Cancer research ,Lymphoma, Large B-Cell, Diffuse ,Diffuse large B-cell lymphoma ,Comparative genomic hybridization - Abstract
Gene-expression profiling has been used to define 3 molecular subtypes of diffuse large B-cell lymphoma (DLBCL), termed germinal center B-cell-like (GCB) DLBCL, activated B-cell-like (ABC) DLBCL, and primary mediastinal B-cell lymphoma (PMBL). To investigate whether these DLBCL subtypes arise by distinct pathogenetic mechanisms, we analyzed 203 DLBCL biopsy samples by high-resolution, genome-wide copy number analysis coupled with gene-expression profiling. Of 272 recurrent chromosomal aberrations that were associated with gene-expression alterations, 30 were used differentially by the DLBCL subtypes ( P < 0.006). An amplicon on chromosome 19 was detected in 26% of ABC DLBCLs but in only 3% of GCB DLBCLs and PMBLs. A highly up-regulated gene in this amplicon was SPIB , which encodes an ETS family transcription factor. Knockdown of SPIB by RNA interference was toxic to ABC DLBCL cell lines but not to GCB DLBCL, PMBL, or myeloma cell lines, strongly implicating SPIB as an oncogene involved in the pathogenesis of ABC DLBCL. Deletion of the INK4a / ARF tumor suppressor locus and trisomy 3 also occurred almost exclusively in ABC DLBCLs and was associated with inferior outcome within this subtype. FOXP1 emerged as a potential oncogene in ABC DLBCL that was up-regulated by trisomy 3 and by more focal high-level amplifications. In GCB DLBCL, amplification of the oncogenic mir-17–92 microRNA cluster and deletion of the tumor suppressor PTEN were recurrent, but these events did not occur in ABC DLBCL. Together, these data provide genetic evidence that the DLBCL subtypes are distinct diseases that use different oncogenic pathways.
- Published
- 2008
50. Repression of BCL-6 is required for the formation of human memory B cells in vitro
- Author
-
Yong Sung Choi, Arthur L. Shaffer, Tracy C. Kuo, Louis M. Staudt, Kathryn Calame, and Joseph Haddad
- Subjects
Transcription, Genetic ,T cell ,Cellular differentiation ,Immunology ,B-cell receptor ,Naive B cell ,Biology ,Article ,medicine ,Immunology and Allergy ,Humans ,RNA, Messenger ,Memory B cell ,B cell ,Cells, Cultured ,B-Lymphocytes ,CD40 ,Gene Expression Profiling ,Germinal center ,Cell Differentiation ,Articles ,DNA ,Molecular biology ,DNA-Binding Proteins ,Kinetics ,medicine.anatomical_structure ,Gene Expression Regulation ,biology.protein ,Proto-Oncogene Proteins c-bcl-6 ,Immunologic Memory - Abstract
Memory B cells provide rapid protection to previously encountered antigens; however, how these cells develop from germinal center B cells is not well understood. A previously described in vitro culture system using human tonsillar germinal center B cells was used to study the transcriptional changes that occur during differentiation of human memory B cells. Kinetic studies monitoring the expression levels of several known late B cell transcription factors revealed that BCL-6 is not expressed in memory B cells generated in vitro, and gene expression profiling studies confirmed that BCL-6 is not expressed in these memory B cells. Furthermore, ectopic expression of BCL-6 in human B cell cultures resulted in formation of fewer memory B cells. In addition, the expression profile of in vitro memory B cells showed a unique pattern that includes expression of genes encoding multiple costimulatory molecules and cytokine receptors, antiapoptotic proteins, T cell chemokines, and transcription factors. These studies establish new molecular criteria for defining the memory B cell stage in human B cells.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.