48 results on '"Barbash O"'
Search Results
2. 456MO METEOR-1: A phase I study of the safety and efficacy of the protein arginine methyltransferase 5 (PRMT5) inhibitor GSK3326595 in advanced solid tumors
- Author
-
Postel-Vinay, S., primary, Italiano, A., additional, Martin Romano, P., additional, Cassier, P.A., additional, Siu, L.L., additional, Lossos, I.S., additional, Hilton, J.F., additional, Mckean, M.A., additional, Strauss, J., additional, Falchook, G.S., additional, de Jonge, M.J.A., additional, Opdam, F.L., additional, Rasco, D., additional, Vermaat, J.S., additional, Crossman, T., additional, Zajac, M., additional, Hainline, A., additional, Kremer, B., additional, Barbash, O., additional, and Gounder, M.M., additional
- Published
- 2022
- Full Text
- View/download PDF
3. Phosphorylation-dependent regulation of SCFFbx4 dimerization and activity involves a novel component, 14-3-3ɛ
- Author
-
Barbash, O, Lee, E K, and Diehl, J A
- Published
- 2011
- Full Text
- View/download PDF
4. Lysine 269 is essential for cyclin D1 ubiquitylation by the SCFFbx4/αB-crystallin ligase and subsequent proteasome-dependent degradation
- Author
-
Barbash, O, Egan, E, Pontano, L L, Kosak, J, and Diehl, J A
- Published
- 2009
- Full Text
- View/download PDF
5. 310P Longitudinal evaluation of circulating tumour DNA in early breast cancer using a plasma-only methylation-based assay
- Author
-
Elliott, M., Antras, J. Fuentes, Dou, A., Veitch, Z.W., Bedard, P.L., Amir, E., Nadler, M., Van de Laar, E., Yu, C., Annan, M., Silvestro, A., Zhang, Q., Cheikh, R., Kim, J., Barbash, O., Siu, L.L., Berman, H., and Cescon, D.
- Published
- 2023
- Full Text
- View/download PDF
6. Identification of mutations that disrupt phosphorylation-dependent nuclear export of cyclin D1
- Author
-
Benzeno, S, Lu, F, Guo, M, Barbash, O, Zhang, F, Herman, J G, Klein, P S, Rustgi, A, and Diehl, J A
- Published
- 2006
- Full Text
- View/download PDF
7. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia
- Author
-
Bell, CC, Fenne, KA, Chan, Y-C, Rambow, F, Yeung, MM, Vassiliadis, D, Lara, L, Yeh, P, Martelotto, LG, Rogiers, A, Kremer, BE, Barbash, O, Mohammad, HP, Johanson, TM, Burr, ML, Dhar, A, Karpinich, N, Tian, L, Tyler, DS, MacPherson, L, Shi, J, Pinnawala, N, Fong, CY, Papenfuss, AT, Grimmond, SM, Dawson, S-J, Allan, RS, Kruger, RG, Vakoc, CR, Goode, DL, Naik, SH, Gilan, O, Lam, EYN, Marine, J-C, Prinjha, RK, Dawson, MA, Bell, CC, Fenne, KA, Chan, Y-C, Rambow, F, Yeung, MM, Vassiliadis, D, Lara, L, Yeh, P, Martelotto, LG, Rogiers, A, Kremer, BE, Barbash, O, Mohammad, HP, Johanson, TM, Burr, ML, Dhar, A, Karpinich, N, Tian, L, Tyler, DS, MacPherson, L, Shi, J, Pinnawala, N, Fong, CY, Papenfuss, AT, Grimmond, SM, Dawson, S-J, Allan, RS, Kruger, RG, Vakoc, CR, Goode, DL, Naik, SH, Gilan, O, Lam, EYN, Marine, J-C, Prinjha, RK, and Dawson, MA
- Abstract
Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.
- Published
- 2019
8. Smyd3 links lysine methylation of map3k2 to ras-driven cancer
- Author
-
Mazur, P.K., Reynoird, N., Khatri, P., Jansen, P. W., Wilkinson, A.W., Liu, S., Barbash, O., Aller, G.S. Van, Huddleston, M., Dhanak, D., Tummino, P.J., Kruger, R.G., Garcia, B.A., Butte, A.J., Vermeulen, M., Sage, J., Gozani, O., Mazur, P.K., Reynoird, N., Khatri, P., Jansen, P. W., Wilkinson, A.W., Liu, S., Barbash, O., Aller, G.S. Van, Huddleston, M., Dhanak, D., Tummino, P.J., Kruger, R.G., Garcia, B.A., Butte, A.J., Vermeulen, M., Sage, J., and Gozani, O.
- Abstract
Contains fulltext : 135402.pdf (Publisher’s version ) (Closed access)
- Published
- 2014
9. Phosphorylation-dependent regulation of SCFFbx4 dimerization and activity involves a novel component, 14-3-3ɛ.
- Author
-
Barbash, O, Lee, E K, and Diehl, J A
- Subjects
- *
PHOSPHORYLATION , *UBIQUITIN , *LIGASES , *ESOPHAGEAL cancer , *GENETIC mutation , *CYCLIN-dependent kinases , *GLYCOGEN synthase kinase-3 - Abstract
Fbx4 is an F-box constituent of Skp-Cullin-F-box (SCF) ubiquitin ligases that directs ubiquitylation of cyclin D1. Ubiquitylation of cyclin D1 requires phosphorylation of both cyclin D1 and Fbx4 by GSK3β. GSK3β-mediated phosphorylation of Fbx4 Ser12 during the G1/S transition regulates Fbx4 dimerization, which in turn governs Fbx4-driven E3 ligase activity. In esophageal carcinomas that overexpress cyclin D1, Fbx4 is subject to inactivating mutations that specifically disrupt dimerization, highlighting the biological significance of this regulatory mechanism. In an effort to elucidate the mechanisms that regulate dimerization, we sought to identify proteins that differentially bind to wild-type Fbx4 versus a cancer-derived dimerization-deficient mutant. We provide evidence that phosphorylation of Ser12 generates a docking site for 14-3-3ɛ. 14-3-3ɛ binds to endogenous Fbx4 and this association is impaired by mutations that target either Ser8 or Ser12 in Fbx4, suggesting that this N-terminal motif in Fbx4 directs its interaction with 14-3-3ɛ. Knockdown of 14-3-3ɛ inhibited Fbx4 dimerization, reduced SCFFbx4 E3 ligase activity and stabilized cyclin D1. Collectively, the current results suggest a model wherein 14-3-3ɛ binds to Ser12-phosphorylated Fbx4 to mediate dimerization and function. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
10. Lysine 269 is essential for cyclin D1 ubiquitylation by the SCFFbx4/αB-crystallin ligase and subsequent proteasome-dependent degradation.
- Author
-
Barbash, O., Egan, E., Pontano, L. L., Kosak, J., and Diehl, J. A.
- Subjects
AMINO acids ,UBIQUITIN ,GENETIC toxicology ,CELL transformation ,DNA damage ,BIOCHEMICAL genetics ,GENETIC mutation - Abstract
Protein ubiquitylation is a complex enzymatic process that results in the covalent attachment of ubiquitin, through Gly-76 of ubiquitin, to an ɛNH2 group of an internal lysine residue in a given substrate. Although E3 ligases frequently use lysines adjacent to the degron within the substrate, many substrates can be targeted to the proteasome through the polyubiquitylation of any lysine. We have assessed the role of lysine residues proximal to the cyclin D1 phosphodegron for ubiquitylation by the SCF
Fbx4/αB-crystallin ubiquitin ligase and subsequent proteasome-dependent degradation of cyclin D1. The work described herein reveals a requisite role for Lys-269 (K269) for the rapid polyubiquitin-mediated degradation of cyclin D1. Mutation of Lys-269, which is proximal to the phosphodegron sequence surrounding Thr-286 in cyclin D1, not only stabilizes cyclin D1 but also triggers cyclin D1 accumulation within the nucleus, thereby promoting cell transformation. In addition, D1-K269R is resistant to genotoxic stress-induced degradation, similar to non-phosphorylatable D1-T286A, supporting the critical role for the post-translational regulation of cyclin D1 in response to DNA-damaging agents. Strikingly, although mutation of lysine 269 to arginine inhibits cyclin D1 degradation, it does not inhibit cyclin D1 ubiquitylation in vivo, showing that ubiquitylation of a specific lysine can influence substrate targeting to the 26S proteasome. [ABSTRACT FROM AUTHOR]- Published
- 2009
- Full Text
- View/download PDF
11. [DNA methylation profiling of the vascular tissues in the setting of atherosclerosis]
- Author
-
Nazarenko, M. S., Anton Markov, Lebedev, I. N., Sleptsov, A. A., Frolov, A. V., Barbash, O. L., and Puzyrev, V. P.
12. SCF Fbx4/alphaB-crystallin cyclin D1 ubiquitin ligase: a license to destroy
- Author
-
Lin Douglas I, Barbash Olena, and Diehl J Alan
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Cyclin D1 is an allosteric regulator for cyclin-dependent kinases 4 and 6 (CDK4/6). The cyclin D/CDK4 kinase promotes G1/S transition through the posttranslational modification and the subsequent inactivation of the retinoblastoma (Rb) protein and related family members (p107 and p130). Accumulation of cyclin D1 is tightly regulated through various mechanisms including transcription, protein localization and ubiquitin-dependent proteolysis. While regulators of cyclin D1 gene expression have been under considerable scrutiny, the identity of the protein complex that targets cyclin D1 protein for degradation, the putative E3 ubiquitin ligase, has remained obscure. In a recent report, Lin et al 1 describe the identification and characterization of a novel SCF, wherein FBX4 and αB-crystallin serve as specificity factors that direct ubiquitination of phosphorylated cyclin D1. As cyclin D1 overexpression in human cancer has been postulated to occur through the loss of degradation machinery, the identification of the SCFFbx4/αB-crystallin ligase will allow new experimental approaches that address mechanisms of cyclin D1 overexpression in human cancer.
- Published
- 2007
- Full Text
- View/download PDF
13. Acute resistance to BET inhibitors remodels compensatory transcriptional programs via p300 coactivation.
- Author
-
Shah V, Giotopoulos G, Osaki H, Meyerhöfer M, Meduri E, Gallego-Crespo A, Behrendt MA, Saura-Pañella M, Tarkar A, Schubert B, Yun H, Horton SJ, Agrawal-Singh S Dr, Hähnel PS, Basheer F, Lugo D, Eleftheriadou I, Barbash O, Dhar A, Kühn M, Guezguez B, Theobald M, Kindler T, Gallipoli P, Yeh PS, Dawson MA, Prinjha RK, Huntly BJ, and Sasca D
- Abstract
Initial clinical trials with drugs targeting epigenetic modulators - such as bromodomain and extraterminal protein (BET) inhibitors - demonstrate modest results in acute myeloid leukemia (AML). A major reason for this involves an increased transcriptional plasticity within AML, which allows cells to escape the therapeutic pressure. In this study, we investigated immediate epigenetic and transcriptional responses following BET inhibition and could demonstrate that BET inhibitor-mediated release of BRD4 from chromatin is accompanied by an acute compensatory feedback that attenuates down-regulation, or even increases expression, of specific transcriptional modules. This adaptation is marked at key AML maintenance genes and is mediated by p300, suggesting a rational therapeutic opportunity to improve outcomes by combining BET- and p300- inhibition. p300 activity is required during all steps of resistance adaptation, however, the specific transcriptional programs that p300 regulates to induce resistance to BET inhibition differ in part between AML subtypes. As a consequence, in some AMLs the requirement for p300 is highest during earlier stages of resistance to BET inhibition, where p300 regulates transitional transcriptional patterns that allow leukemia-homeostatic adjustments. In other AMLs, p300 shapes a linear resistance to BET inhibition and remains crucial throughout all stages of the evolution of resistance. Altogether, our study elucidates the mechanisms that underlie an "acute" state of resistance to BET inhibition, achieved through p300 activity, and how these mechanisms remodel to mediate "chronic" resistance. Importantly, our data also suggest that sequential treatment with BET- and p300-inhibition may prevent resistance development, thereby improving outcomes., (Copyright © 2024 American Society of Hematology.)
- Published
- 2024
- Full Text
- View/download PDF
14. Potential synergistic effect between niraparib and statins in ovarian cancer clinical trials.
- Author
-
Zhang H, Rutkowska A, González-Martín A, Mirza MR, Monk BJ, Vergote I, Pothuri B, Graybill WAS, Goessel C, Barbash O, Bergamini G, and Feng B
- Abstract
This study investigates the potential clinical synergy between the poly(adenosine 5'-diphosphate [ADP]-ribose) polymerase (PARP) inhibitor niraparib (Zejula®) and concomitant statins, exploring their combined effects on progression-free survival (PFS) in ovarian cancer patients. We retrospectively analysed niraparib registrational clinical trials in ovarian cancer to investigate potential interactions between niraparib and statins. In the PRIMA trial, patients receiving niraparib demonstrated improved PFS compared to those on placebo (hazard ratio=0.62; P<0.001; mPFS 13.8 months vs 8.2 months). The post hoc analysis revealed that patients receiving maintenance niraparib who reported concomitant use of statins exhibited significantly improved PFS compared to those on placebo with concomitant statins (hazard ratio=0.34; P<0.001; mPFS 18.2 months vs 6.0 months). Notably, the improved efficacy in the two-arm comparison of concomitant statin patients were much better than in the two-arm comparison of those patients without statin, as reflected in the niraparib:statin interaction (P=0.005). These findings suggest novel opportunities in oncology for the use of statins in combination therapies with PARP inhibitors and emphasize the need for further investigation.
- Published
- 2024
- Full Text
- View/download PDF
15. Phase I/II study of the clinical activity and safety of GSK3326595 in patients with myeloid neoplasms.
- Author
-
Watts J, Minden MD, Bachiashvili K, Brunner AM, Abedin S, Crossman T, Zajac M, Moroz V, Egger JL, Tarkar A, Kremer BE, Barbash O, and Borthakur G
- Abstract
Background: GSK3326595 is a potent, selective, reversible protein arginine methyltransferase 5 (PRMT5) inhibitor under investigation for treatment of myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia (CMML), and acute myeloid leukemia (AML). In preclinical models of AML, PRMT5 inhibition decreased proliferation and increased cell death, supporting additional clinical research in myeloid neoplasms., Objectives: To determine the clinical activity, safety, tolerability, dosing, additional measures of clinical activity, pharmacokinetics, and pharmacodynamics of GSK3326595., Design: In part 1 of this open-label, multicenter, multipart, phase I/II study, adults with relapsed/refractory myeloid neoplasms (e.g., MDS, CMML, and AML) received monotherapy with 400 or 300 mg oral GSK3326595 once daily. Study termination occurred prior to part 2 enrollment., Methods: Clinical activity was determined by the clinical benefit rate (CBR; proportion of patients achieving complete remission (CR), complete marrow remission (mCR), partial remission, stable disease (SD) >8 weeks, or hematologic improvement). Adverse events (AEs) were assessed by incidence and severity. Exploratory examination of spliceosome mutations was performed to determine the relationship between genomic profiles and clinical response to GSK3326595., Results: Thirty patients with a median age of 73.5 years (range, 47-90) were enrolled; 13 (43%) and 17 (57%) received 400 and 300 mg of GSK3326595, respectively. Five (17%) patients met CBR criteria: 4 (13%) with SD >8 weeks and 1 (3%) achieving mCR. Of five patients with clinical benefit: three had SRSF2 mutation, one U2AF1, and one was splicing factor wild-type. Frequent GSK3326595-related AEs were decreased platelet count (27%), dysgeusia (23%), fatigue (20%), and nausea (20%). GSK3326595 had rapid absorption, with a T
max of approximately 2 h and a terminal half-life of 4-6 h., Conclusion: GSK3326595 monotherapy had limited clinical activity in heavily pretreated patients despite robust target engagement. The safety profile was broadly consistent with other published PRMT5 inhibitor studies., Trial Registration: ClinicalTrials.gov: NCT03614728., (© The Author(s), 2024.)- Published
- 2024
- Full Text
- View/download PDF
16. A Phase I/II Study of GSK525762 Combined with Fulvestrant in Patients with Hormone Receptor-positive/HER2-negative Advanced or Metastatic Breast Cancer.
- Author
-
Cescon DW, Hilton J, Morales Murilo S, Layman RM, Pluard T, Yeo B, Park IH, Provencher L, Kim SB, Im YH, Wyce A, Krishnatry AS, Hicks K, Zhang Q, Barbash O, Khaled A, Horner T, Dhar A, Oliveira M, and Sparano JA
- Subjects
- Humans, Female, Fulvestrant, Nuclear Proteins, Receptor, ErbB-2 metabolism, Transcription Factors, Disease Progression, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bromodomain Containing Proteins, Cell Cycle Proteins, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Benzodiazepines
- Abstract
Purpose: Endocrine-based therapy is the initial primary treatment option for hormone receptor-positive and human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer (mBC). However, patients eventually experience disease progression due to resistance to endocrine therapy. Molibresib (GSK525762) is a small-molecule inhibitor of bromodomain and extraterminal (BET) family proteins (BRD2, BRD3, BRD4, and BRDT). Preclinical data suggested that the combination of molibresib with endocrine therapy might overcome endocrine resistance. This study aimed to investigate the safety, tolerability, pharmacokinetics, pharmacodynamics, and efficacy [objective response rate (ORR)] of molibresib combined with fulvestrant in women with HR+/HER2- mBC., Patients and Methods: In this phase I/II dose-escalation and dose-expansion study, patients received oral molibresib 60 or 80 mg once daily in combination with intramuscular fulvestrant. Patients enrolled had relapsed/refractory, advanced/metastatic HR+/HER2- breast cancer with disease progression on prior treatment with an aromatase inhibitor, with or without a cyclin-dependent kinase 4/6 inhibitor., Results: The study included 123 patients. The most common treatment-related adverse events (AE) were nausea (52%), dysgeusia (49%), and fatigue (45%). At a 60-mg dosage of molibresib, >90% of patients experienced treatment-related AE. Grade 3 or 4 treatment-related AE were observed in 47% and 48% of patients treated with molibresib 60 mg and molibresib 80 mg, respectively. The ORR was 13% [95% confidence interval (CI), 8-20], not meeting the 25% threshold for proceeding to phase II. Among 82 patients with detected circulating tumor DNA and clinical outcome at study enrollment, a strong association was observed between the detection of copy-number amplification and poor progression-free survival (HR, 2.89; 95% CI, 1.73-4.83; P < 0.0001)., Conclusions: Molibresib in combination with fulvestrant did not demonstrate clinically meaningful activity in this study., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
17. A Phase I/II Open-Label Study of Molibresib for the Treatment of Relapsed/Refractory Hematologic Malignancies.
- Author
-
Dawson MA, Borthakur G, Huntly BJP, Karadimitris A, Alegre A, Chaidos A, Vogl DT, Pollyea DA, Davies FE, Morgan GJ, Glass JL, Kamdar M, Mateos MV, Tovar N, Yeh P, Delgado RG, Basheer F, Marando L, Gallipoli P, Wyce A, Krishnatry AS, Barbash O, Bakirtzi E, Ferron-Brady G, Karpinich NO, McCabe MT, Foley SW, Horner T, Dhar A, Kremer BE, and Dickinson M
- Subjects
- Humans, Lymphoma, Non-Hodgkin drug therapy, Hematologic Neoplasms drug therapy, Leukemia, Myeloid, Acute drug therapy, Thrombocytopenia
- Abstract
Purpose: Molibresib is a selective, small molecule inhibitor of the bromodomain and extra-terminal (BET) protein family. This was an open-label, two-part, Phase I/II study investigating molibresib monotherapy for the treatment of hematological malignancies (NCT01943851)., Patients and Methods: Part 1 (dose escalation) determined the recommended Phase 2 dose (RP2D) of molibresib in patients with acute myeloid leukemia (AML), Non-Hodgkin lymphoma (NHL), or multiple myeloma. Part 2 (dose expansion) investigated the safety and efficacy of molibresib at the RP2D in patients with relapsed/refractory myelodysplastic syndrome (MDS; as well as AML evolved from antecedent MDS) or cutaneous T-cell lymphoma (CTCL). The primary endpoint in Part 1 was safety and the primary endpoint in Part 2 was objective response rate (ORR)., Results: There were 111 patients enrolled (87 in Part 1, 24 in Part 2). Molibresib RP2Ds of 75 mg daily (for MDS) and 60 mg daily (for CTCL) were selected. Most common Grade 3+ adverse events included thrombocytopenia (37%), anemia (15%), and febrile neutropenia (15%). Six patients achieved complete responses [3 in Part 1 (2 AML, 1 NHL), 3 in Part 2 (MDS)], and 7 patients achieved partial responses [6 in Part 1 (4 AML, 2 NHL), 1 in Part 2 (MDS)]. The ORRs for Part 1, Part 2, and the total study population were 10% [95% confidence interval (CI), 4.8-18.7], 25% (95% CI, 7.3-52.4), and 13% (95% CI, 6.9-20.6), respectively., Conclusions: While antitumor activity was observed with molibresib, use was limited by gastrointestinal and thrombocytopenia toxicities. Investigations of molibresib as part of combination regimens may be warranted., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
18. TP53 mutations and RNA-binding protein MUSASHI-2 drive resistance to PRMT5-targeted therapy in B-cell lymphoma.
- Author
-
Erazo T, Evans CM, Zakheim D, Chu EL, Refermat AY, Asgari Z, Yang X, Da Silva Ferreira M, Mehta S, Russo MV, Knezevic A, Zhang XP, Chen Z, Fennell M, Garippa R, Seshan V, de Stanchina E, Barbash O, Batlevi CL, Leslie CS, Melnick AM, Younes A, and Kharas MG
- Subjects
- Cell Line, Tumor, Humans, Mutation, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases metabolism, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Tumor Suppressor Protein p53 genetics, Lymphoma genetics, Lymphoma, B-Cell
- Abstract
To identify drivers of sensitivity and resistance to Protein Arginine Methyltransferase 5 (PRMT5) inhibition, we perform a genome-wide CRISPR/Cas9 screen. We identify TP53 and RNA-binding protein MUSASHI2 (MSI2) as the top-ranked sensitizer and driver of resistance to specific PRMT5i, GSK-591, respectively. TP53 deletion and TP53
R248W mutation are biomarkers of resistance to GSK-591. PRMT5 expression correlates with MSI2 expression in lymphoma patients. MSI2 depletion and pharmacological inhibition using Ro 08-2750 (Ro) both synergize with GSK-591 to reduce cell growth. Ro reduces MSI2 binding to its global targets and dual treatment of Ro and PRMT5 inhibitors result in synergistic gene expression changes including cell cycle, P53 and MYC signatures. Dual MSI2 and PRMT5 inhibition further blocks c-MYC and BCL-2 translation. BCL-2 depletion or inhibition with venetoclax synergizes with a PRMT5 inhibitor by inducing reduced cell growth and apoptosis. Thus, we propose a therapeutic strategy in lymphoma that combines PRMT5 with MSI2 or BCL-2 inhibition., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
19. Inhibiting Type I Arginine Methyltransferase Activity Promotes T Cell-Mediated Antitumor Immune Responses.
- Author
-
Fedoriw A, Shi L, O'Brien S, Smitheman KN, Wang Y, Hou J, Sherk C, Rajapurkar S, Laraio J, Williams LJ, Xu C, Han G, Feng Q, Bedford MT, Wang L, Barbash O, Kruger RG, Hwu P, Mohammad HP, and Peng W
- Subjects
- Animals, Arginine, Humans, Immunity, Mice, Intracellular Signaling Peptides and Proteins, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases metabolism
- Abstract
Protein arginine methyltransferases (PRMT) are a widely expressed class of enzymes responsible for catalyzing arginine methylation on numerous protein substrates. Among them, type I PRMTs are responsible for generating asymmetric dimethylarginine. By controlling multiple basic cellular processes, such as DNA damage responses, transcriptional regulation, and mRNA splicing, type I PRMTs contribute to cancer initiation and progression. A type I PRMT inhibitor, GSK3368715, has been developed and has entered clinical trials for solid and hematologic malignancies. Although type I PRMTs have been reported to play roles in modulating immune cell function, the immunologic role of tumor-intrinsic pathways controlled by type I PRMTs remains uncharacterized. Here, our The Cancer Genome Atlas dataset analysis revealed that expression of type I PRMTs associated with poor clinical response and decreased immune infiltration in patients with melanoma. In cancer cell lines, inhibition of type I PRMTs induced an IFN gene signature, amplified responses to IFN and innate immune signaling, and decreased expression of the immunosuppressive cytokine VEGF. In immunocompetent mouse tumor models, including a model of T-cell exclusion that represents a common mechanism of anti-programmed cell death protein 1 (PD-1) resistance in humans, type I PRMT inhibition increased T-cell infiltration, produced durable responses dependent on CD8+ T cells, and enhanced efficacy of anti-PD-1 therapy. These data indicate that type I PRMT inhibition exhibits immunomodulatory properties and synergizes with immune checkpoint blockade (ICB) to induce durable antitumor responses in a T cell-dependent manner, suggesting that type I PRMT inhibition can potentiate an antitumor immunity in refractory settings., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
20. Homologous Recombination Deficiency: Concepts, Definitions, and Assays.
- Author
-
Stewart MD, Merino Vega D, Arend RC, Baden JF, Barbash O, Beaubier N, Collins G, French T, Ghahramani N, Hinson P, Jelinic P, Marton MJ, McGregor K, Parsons J, Ramamurthy L, Sausen M, Sokol ES, Stenzinger A, Stires H, Timms KM, Turco D, Wang I, Williams JA, Wong-Ho E, and Allen J
- Subjects
- BRCA1 Protein genetics, DNA Repair, Female, Homologous Recombination genetics, Humans, Poly(ADP-ribose) Polymerases genetics, Recombinational DNA Repair genetics, Ovarian Neoplasms drug therapy, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use
- Abstract
Background: Homologous recombination deficiency (HRD) is a phenotype that is characterized by the inability of a cell to effectively repair DNA double-strand breaks using the homologous recombination repair (HRR) pathway. Loss-of-function genes involved in this pathway can sensitize tumors to poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibitors and platinum-based chemotherapy, which target the destruction of cancer cells by working in concert with HRD through synthetic lethality. However, to identify patients with these tumors, it is vital to understand how to best measure homologous repair (HR) status and to characterize the level of alignment in these measurements across different diagnostic platforms. A key current challenge is that there is no standardized method to define, measure, and report HR status using diagnostics in the clinical setting., Methods: Friends of Cancer Research convened a consortium of project partners from key healthcare sectors to address concerns about the lack of consistency in the way HRD is defined and methods for measuring HR status., Results: This publication provides findings from the group's discussions that identified opportunities to align the definition of HRD and the parameters that contribute to the determination of HR status. The consortium proposed recommendations and best practices to benefit the broader cancer community., Conclusion: Overall, this publication provides additional perspectives for scientist, physician, laboratory, and patient communities to contextualize the definition of HRD and various platforms that are used to measure HRD in tumors., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
21. A Chemical Acetylation-Based Mass Spectrometry Platform for Histone Methylation Profiling.
- Author
-
Zappacosta F, Wagner CD, Della Pietra A 3rd, Gerhart SV, Keenan K, Korenchuck S, Quinn CJ, Barbash O, McCabe MT, and Annan RS
- Subjects
- Acetylation, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Humans, Mass Spectrometry, Methylation, Histones metabolism
- Abstract
Histones are highly posttranslationally modified proteins that regulate gene expression by modulating chromatin structure and function. Acetylation and methylation are the most abundant histone modifications, with methylation occurring on lysine (mono-, di-, and trimethylation) and arginine (mono- and dimethylation) predominately on histones H3 and H4. In addition, arginine dimethylation can occur either symmetrically (SDMA) or asymmetrically (ADMA) conferring different biological functions. Despite the importance of histone methylation on gene regulation, characterization and quantitation of this modification have proven to be quite challenging. Great advances have been made in the analysis of histone modification using both bottom-up and top-down mass spectrometry (MS). However, MS-based analysis of histone posttranslational modifications (PTMs) is still problematic, due both to the basic nature of the histone N-terminal tails and to the combinatorial complexity of the histone PTMs. In this report, we describe a simplified MS-based platform for histone methylation analysis. The strategy uses chemical acetylation with d
0 -acetic anhydride to collapse all the differently acetylated histone forms into one form, greatly reducing the complexity of the peptide mixture and improving sensitivity for the detection of methylation via summation of all the differently acetylated forms. We have used this strategy for the robust identification and relative quantitation of H4R3 methylation, for which stoichiometry and symmetry status were determined, providing an antibody-independent evidence that H4R3 is a substrate for both Type I and Type II PRMTs. Additionally, this approach permitted the robust detection of H4K5 monomethylation, a very low stoichiometry methylation event (0.02% methylation). In an independent example, we developed an in vitro assay to profile H3K27 methylation and applied it to an EZH2 mutant xenograft model following small-molecule inhibition of the EZH2 methyltransferase. These specific examples highlight the utility of this simplified MS-based approach to quantify histone methylation profiles., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
22. Limited antitumor activity of combined BET and MEK inhibition in neuroblastoma.
- Author
-
Healy JR, Hart LS, Shazad AL, Gagliardi ME, Tsang M, Elias J, Ruden J, Farrel A, Rokita JL, Li Y, Wyce A, Barbash O, Batra V, Samanta M, Maris JM, and Schnepp RW
- Subjects
- Animals, Apoptosis, Cell Proliferation, Female, Humans, Mice, Mice, SCID, Neuroblastoma metabolism, Neuroblastoma pathology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Benzodiazepines pharmacology, MAP Kinase Kinase 1 antagonists & inhibitors, Neuroblastoma drug therapy, Proteins antagonists & inhibitors, Pyridones pharmacology, Pyrimidinones pharmacology
- Abstract
Background: The treatment of high-risk neuroblastoma continues to present a formidable challenge to pediatric oncology. Previous studies have shown that Bromodomain and extraterminal (BET) inhibitors can inhibit MYCN expression and suppress MYCN-amplified neuroblastoma in vivo. Furthermore, alterations within RAS-MAPK (mitogen-activated protein kinase) signaling play significant roles in neuroblastoma initiation, maintenance, and relapse, and mitogen-activated extracellular signal-regulated kinase (MEK) inhibitors demonstrate efficacy in subsets of neuroblastoma preclinical models. Finally, hyperactivation of RAS-MAPK signaling has been shown to promote resistance to BET inhibitors. Therefore, we examined the antitumor efficacy of combined BET/MEK inhibition utilizing I-BET726 or I-BET762 and trametinib in high-risk neuroblastoma., Procedure: Utilizing a panel of genomically annotated neuroblastoma cell line models, we investigated the in vitro effects of combined BET/MEK inhibition on cell proliferation and apoptosis. Furthermore, we evaluated the effects of combined inhibition in neuroblastoma xenograft models., Results: Combined BET and MEK inhibition demonstrated synergistic effects on the growth and survival of a large panel of neuroblastoma cell lines through augmentation of apoptosis. A combination therapy slowed tumor growth in a non-MYCN-amplified, NRAS-mutated neuroblastoma xenograft model, but had no efficacy in an MYCN-amplified model harboring a loss-of-function mutation in NF1., Conclusions: Combinatorial BET and MEK inhibition was synergistic in the vast majority of neuroblastoma cell lines in the in vitro setting but showed limited antitumor activity in vivo. Collectively, these data do not support clinical development of this combination in high-risk neuroblastoma., (© 2020 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
23. Phase 1 Study of Molibresib (GSK525762), a Bromodomain and Extra-Terminal Domain Protein Inhibitor, in NUT Carcinoma and Other Solid Tumors.
- Author
-
Piha-Paul SA, Hann CL, French CA, Cousin S, Braña I, Cassier PA, Moreno V, de Bono JS, Harward SD, Ferron-Brady G, Barbash O, Wyce A, Wu Y, Horner T, Annan M, Parr NJ, Prinjha RK, Carpenter CL, Hilton J, Hong DS, Haas NB, Markowski MC, Dhar A, O'Dwyer PJ, and Shapiro GI
- Abstract
Background: Bromodomain and extra-terminal domain proteins are promising epigenetic anticancer drug targets. This first-in-human study evaluated the safety, recommended phase II dose, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the bromodomain and extra-terminal domain inhibitor molibresib (GSK525762) in patients with nuclear protein in testis (NUT) carcinoma (NC) and other solid tumors., Methods: This was a phase I and II, open-label, dose-escalation study. Molibresib was administered orally once daily. Single-patient dose escalation (from 2 mg/d) was conducted until the first instance of grade 2 or higher drug-related toxicity, followed by a 3 + 3 design. Pharmacokinetic parameters were obtained during weeks 1 and 3. Circulating monocyte chemoattractant protein-1 levels were measured as a pharmacodynamic biomarker., Results: Sixty-five patients received molibresib. During dose escalation, 11% experienced dose-limiting toxicities, including six instances of grade 4 thrombocytopenia, all with molibresib 60-100 mg. The most frequent treatment-related adverse events of any grade were thrombocytopenia (51%) and gastrointestinal events, including nausea, vomiting, diarrhea, decreased appetite, and dysgeusia (22%-42%), anemia (22%), and fatigue (20%). Molibresib demonstrated an acceptable safety profile up to 100 mg; 80 mg once daily was selected as the recommended phase II dose. Following single and repeat dosing, molibresib showed rapid absorption and elimination (maximum plasma concentration: 2 hours; t
1/2 : 3-7 hours). Dose-dependent reductions in circulating monocyte chemoattractant protein-1 levels were observed. Among 19 patients with NC, four achieved either confirmed or unconfirmed partial response, eight had stable disease as best response, and four were progression-free for more than 6 months., Conclusions: Once-daily molibresib was tolerated at doses demonstrating target engagement. Preliminary data indicate proof-of-concept in NC., (© The Author(s) 2019. Published by Oxford University Press.)- Published
- 2019
- Full Text
- View/download PDF
24. Therapeutic Targeting of RNA Splicing Catalysis through Inhibition of Protein Arginine Methylation.
- Author
-
Fong JY, Pignata L, Goy PA, Kawabata KC, Lee SC, Koh CM, Musiani D, Massignani E, Kotini AG, Penson A, Wun CM, Shen Y, Schwarz M, Low DH, Rialdi A, Ki M, Wollmann H, Mzoughi S, Gay F, Thompson C, Hart T, Barbash O, Luciani GM, Szewczyk MM, Wouters BJ, Delwel R, Papapetrou EP, Barsyte-Lovejoy D, Arrowsmith CH, Minden MD, Jin J, Melnick A, Bonaldi T, Abdel-Wahab O, and Guccione E
- Subjects
- Animals, Antineoplastic Agents pharmacokinetics, Catalysis, Enzyme Inhibitors pharmacokinetics, Ethylenediamines pharmacokinetics, Gene Expression Regulation, Neoplastic, Gene Regulatory Networks, Humans, K562 Cells, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mice, Inbred C57BL, Mice, Transgenic, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases metabolism, Pyrroles pharmacokinetics, RNA, Neoplasm genetics, Repressor Proteins antagonists & inhibitors, Repressor Proteins metabolism, THP-1 Cells, Tumor Cells, Cultured, U937 Cells, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Ethylenediamines pharmacology, Leukemia, Myeloid, Acute drug therapy, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Pyrroles pharmacology, RNA Splicing drug effects, RNA, Neoplasm metabolism
- Abstract
Cancer-associated mutations in genes encoding RNA splicing factors (SFs) commonly occur in leukemias, as well as in a variety of solid tumors, and confer dependence on wild-type splicing. These observations have led to clinical efforts to directly inhibit the spliceosome in patients with refractory leukemias. Here, we identify that inhibiting symmetric or asymmetric dimethylation of arginine, mediated by PRMT5 and type I protein arginine methyltransferases (PRMTs), respectively, reduces splicing fidelity and results in preferential killing of SF-mutant leukemias over wild-type counterparts. These data identify genetic subsets of cancer most likely to respond to PRMT inhibition, synergistic effects of combined PRMT5 and type I PRMT inhibition, and a mechanistic basis for the therapeutic efficacy of PRMT inhibition in cancer., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2019
- Full Text
- View/download PDF
25. Anti-tumor Activity of the Type I PRMT Inhibitor, GSK3368715, Synergizes with PRMT5 Inhibition through MTAP Loss.
- Author
-
Fedoriw A, Rajapurkar SR, O'Brien S, Gerhart SV, Mitchell LH, Adams ND, Rioux N, Lingaraj T, Ribich SA, Pappalardi MB, Shah N, Laraio J, Liu Y, Butticello M, Carpenter CL, Creasy C, Korenchuk S, McCabe MT, McHugh CF, Nagarajan R, Wagner C, Zappacosta F, Annan R, Concha NO, Thomas RA, Hart TK, Smith JJ, Copeland RA, Moyer MP, Campbell J, Stickland K, Mills J, Jacques-O'Hagan S, Allain C, Johnston D, Raimondi A, Porter Scott M, Waters N, Swinger K, Boriack-Sjodin A, Riera T, Shapiro G, Chesworth R, Prinjha RK, Kruger RG, Barbash O, and Mohammad HP
- Subjects
- Alternative Splicing, Antineoplastic Agents chemistry, Biomarkers, Cell Line, Tumor, Drug Synergism, Enzyme Inhibitors chemistry, Humans, Methylation, Models, Molecular, Molecular Conformation, Molecular Structure, Protein Binding, Protein-Arginine N-Methyltransferases chemistry, Substrate Specificity, Antineoplastic Agents pharmacology, Enzyme Inhibitors pharmacology, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Purine-Nucleoside Phosphorylase deficiency
- Abstract
Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia.
- Author
-
Bell CC, Fennell KA, Chan YC, Rambow F, Yeung MM, Vassiliadis D, Lara L, Yeh P, Martelotto LG, Rogiers A, Kremer BE, Barbash O, Mohammad HP, Johanson TM, Burr ML, Dhar A, Karpinich N, Tian L, Tyler DS, MacPherson L, Shi J, Pinnawala N, Yew Fong C, Papenfuss AT, Grimmond SM, Dawson SJ, Allan RS, Kruger RG, Vakoc CR, Goode DL, Naik SH, Gilan O, Lam EYN, Marine JC, Prinjha RK, and Dawson MA
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Bone Marrow pathology, CRISPR-Cas Systems genetics, Cell Line, Tumor, Epigenesis, Genetic drug effects, Female, HEK293 Cells, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Mice, Mice, Inbred C57BL, Sequence Analysis, RNA, Single-Cell Analysis, Trans-Activators genetics, Trans-Activators metabolism, Transcription, Genetic drug effects, Treatment Outcome, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Gene Expression Regulation, Leukemic drug effects, Leukemia, Myeloid, Acute drug therapy, Trans-Activators antagonists & inhibitors
- Abstract
Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.
- Published
- 2019
- Full Text
- View/download PDF
27. Targeting epigenetic modifications in cancer therapy: erasing the roadmap to cancer.
- Author
-
Mohammad HP, Barbash O, and Creasy CL
- Subjects
- DNA (Cytosine-5-)-Methyltransferases antagonists & inhibitors, Drug Discovery, Histone Acetyltransferases antagonists & inhibitors, Histone Deacetylase Inhibitors therapeutic use, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Humans, Molecular Targeted Therapy, Neoplasms genetics, Antineoplastic Agents therapeutic use, Enzyme Inhibitors therapeutic use, Epigenesis, Genetic, Neoplasms drug therapy, Protein-Arginine N-Methyltransferases therapeutic use
- Abstract
Epigenetic dysregulation is a common feature of most cancers, often occurring directly through alteration of epigenetic machinery. Over the last several years, a new generation of drugs directed at epigenetic modulators have entered clinical development, and results from these trials are now being disclosed. Unlike first-generation epigenetic therapies, these new agents are selective, and many are targeted to proteins which are mutated or translocated in cancer. This review will provide a summary of the epigenetic modulatory agents currently in clinical development and discuss the opportunities and challenges in their development. As these drugs advance in the clinic, drug discovery has continued with a focus on both novel and existing epigenetic targets. We will provide an overview of these efforts and the strategies being employed.
- Published
- 2019
- Full Text
- View/download PDF
28. "Z4" Complex Member Fusions in NUT Carcinoma: Implications for a Novel Oncogenic Mechanism.
- Author
-
Shiota H, Elya JE, Alekseyenko AA, Chou PM, Gorman SA, Barbash O, Becht K, Danga K, Kuroda MI, Nardi V, and French CA
- Subjects
- Adolescent, Cell Line, Tumor, Cell Nucleus genetics, Cell Nucleus metabolism, Cell Survival, DNA-Binding Proteins metabolism, Female, High-Throughput Nucleotide Sequencing, Humans, In Situ Hybridization, Fluorescence, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Oncogene Proteins, Fusion metabolism, Sequence Analysis, DNA, Carcinoma, Squamous Cell genetics, DNA-Binding Proteins genetics, Neoplasm Proteins genetics, Nuclear Proteins genetics, Oncogene Proteins, Fusion genetics, Tumor Suppressor Proteins metabolism
- Abstract
Nuclear protein in testis (NUT) carcinoma (NC) is a rare, distinctly aggressive subtype of squamous carcinoma defined by the presence of NUT -fusion oncogenes resulting from chromosomal translocation. In most cases, the NUT gene ( NUTM1 ) is fused to bromodomain containing 4 ( BRD4 ) forming the BRD4-NUT oncogene. Here, a novel fusion partner to NUT was discovered using next-generation sequencing and FISH from a young patient with an undifferentiated malignant round cell tumor. Interestingly, the NUT fusion identified involved ZNF592, a zinc finger containing protein, which was previously identified as a component of the BRD4-NUT complex. In BRD4-NUT-expressing NC cells, wild-type ZNF592 and other associated "Z4" complex proteins, including ZNF532 and ZMYND8, colocalize with BRD4-NUT in characteristic nuclear foci. Furthermore, ectopic expression of BRD4-NUT in a non-NC cell line induces sequestration of Z4 factors to BRD4-NUT foci. Finally, the data demonstrate the specific dependency of NC cells on Z4 modules, ZNF532 and ZNF592. IMPLICATIONS: This study establishes the oncogenic role of Z4 factors in NC, offering potential new targeted therapeutic strategies in this incurable cancer. Visual Overview: http://mcr.aacrjournals.org/content/molcanres/16/12/1826/F1.large.jpg., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
29. Activation of the p53-MDM4 regulatory axis defines the anti-tumour response to PRMT5 inhibition through its role in regulating cellular splicing.
- Author
-
Gerhart SV, Kellner WA, Thompson C, Pappalardi MB, Zhang XP, Montes de Oca R, Penebre E, Duncan K, Boriack-Sjodin A, Le B, Majer C, McCabe MT, Carpenter C, Johnson N, Kruger RG, and Barbash O
- Subjects
- Alternative Splicing genetics, Antineoplastic Agents, Arginine analogs & derivatives, Arginine metabolism, Cell Cycle drug effects, Cell Cycle genetics, Cell Cycle Proteins, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Enzyme Inhibitors pharmacology, Humans, Nuclear Proteins genetics, Protein Isoforms genetics, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Proto-Oncogene Proteins genetics, Tumor Suppressor Protein p53 genetics, snRNP Core Proteins metabolism, Nuclear Proteins metabolism, Protein-Arginine N-Methyltransferases metabolism, Proto-Oncogene Proteins metabolism, RNA Splicing genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Evasion of the potent tumour suppressor activity of p53 is one of the hurdles that must be overcome for cancer cells to escape normal regulation of cellular proliferation and survival. In addition to frequent loss of function mutations, p53 wild-type activity can also be suppressed post-translationally through several mechanisms, including the activity of PRMT5. Here we describe broad anti-proliferative activity of potent, selective, reversible inhibitors of protein arginine methyltransferase 5 (PRMT5) including GSK3326595 in human cancer cell lines representing both hematologic and solid malignancies. Interestingly, PRMT5 inhibition activates the p53 pathway via the induction of alternative splicing of MDM4. The MDM4 isoform switch and subsequent p53 activation are critical determinants of the response to PRMT5 inhibition suggesting that the integrity of the p53-MDM4 regulatory axis defines a subset of patients that could benefit from treatment with GSK3326595.
- Published
- 2018
- Full Text
- View/download PDF
30. MEK inhibitors overcome resistance to BET inhibition across a number of solid and hematologic cancers.
- Author
-
Wyce A, Matteo JJ, Foley SW, Felitsky DJ, Rajapurkar SR, Zhang XP, Musso MC, Korenchuk S, Karpinich NO, Keenan KM, Stern M, Mathew LK, McHugh CF, McCabe MT, Tummino PJ, Kruger RG, Carpenter C, and Barbash O
- Abstract
BET inhibitors exhibit broad activity in cancer models, making predictive biomarkers challenging to define. Here we investigate the biomarkers of activity of the clinical BET inhibitor GSK525762 (I-BET; I-BET762) across cancer cell lines and demonstrate that KRAS mutations are novel resistance biomarkers. This finding led us to combine BET with RAS pathway inhibition using MEK inhibitors to overcome resistance, which resulted in synergistic effects on growth and survival in RAS pathway mutant models as well as a subset of cell lines lacking RAS pathway mutations. GSK525762 treatment up-regulated p-ERK1/2 levels in both RAS pathway wild-type and mutant cell lines, suggesting that MEK/ERK pathway activation may also be a mechanism of adaptive BET inhibitor resistance. Importantly, gene expression studies demonstrated that the BET/MEK combination uniquely sustains down-regulation of genes associated with mitosis, leading to prolonged growth arrest that is not observed with either single agent therapy. These studies highlight a potential to enhance the clinical benefit of BET and MEK inhibitors and provide a strong rationale for clinical evaluation of BET/MEK combination therapies in cancer.
- Published
- 2018
- Full Text
- View/download PDF
31. PRMT5 Is a Critical Regulator of Breast Cancer Stem Cell Function via Histone Methylation and FOXP1 Expression.
- Author
-
Chiang K, Zielinska AE, Shaaban AM, Sanchez-Bailon MP, Jarrold J, Clarke TL, Zhang J, Francis A, Jones LJ, Smith S, Barbash O, Guccione E, Farnie G, Smalley MJ, and Davies CC
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Proliferation, Enzyme Inhibitors pharmacology, Epigenesis, Genetic, Female, Forkhead Transcription Factors metabolism, Humans, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells physiology, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Protein-Arginine N-Methyltransferases metabolism, Repressor Proteins metabolism, Breast Neoplasms genetics, Forkhead Transcription Factors genetics, Histone Code, Neoplastic Stem Cells metabolism, Protein-Arginine N-Methyltransferases genetics, Repressor Proteins genetics
- Abstract
Breast cancer progression, treatment resistance, and relapse are thought to originate from a small population of tumor cells, breast cancer stem cells (BCSCs). Identification of factors critical for BCSC function is therefore vital for the development of therapies. Here, we identify the arginine methyltransferase PRMT5 as a key in vitro and in vivo regulator of BCSC proliferation and self-renewal and establish FOXP1, a winged helix/forkhead transcription factor, as a critical effector of PRMT5-induced BCSC function. Mechanistically, PRMT5 recruitment to the FOXP1 promoter facilitates H3R2me2s, SET1 recruitment, H3K4me3, and gene expression. Our findings are clinically significant, as PRMT5 depletion within established tumor xenografts or treatment of patient-derived BCSCs with a pre-clinical PRMT5 inhibitor substantially reduces BCSC numbers. Together, our findings highlight the importance of PRMT5 in BCSC maintenance and suggest that small-molecule inhibitors of PRMT5 or downstream targets could be an effective strategy eliminating this cancer-causing population., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Targeting Histone Methylation in Cancer.
- Author
-
McCabe MT, Mohammad HP, Barbash O, and Kruger RG
- Subjects
- Antimetabolites, Antineoplastic pharmacology, Antimetabolites, Antineoplastic therapeutic use, Enzyme Inhibitors pharmacology, Enzyme Inhibitors therapeutic use, Histone Demethylases genetics, Histone Demethylases metabolism, Histone-Lysine N-Methyltransferase genetics, Histones metabolism, Humans, Methylation drug effects, Neoplasms genetics, Protein Processing, Post-Translational drug effects, Epigenesis, Genetic, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Histones genetics, Molecular Targeted Therapy methods, Neoplasms drug therapy
- Abstract
Most, if not all, human cancers exhibit altered epigenetic signatures that promote aberrant gene expression that contributes to cellular transformation. Historically, attempts to pharmacologically intervene in this process have focused on DNA methylation and histone acetylation. More recently, genome-wide studies have identified histone and chromatin regulators as one of the most frequently dysregulated functional classes in a wide range of cancer types. These findings have provided numerous potential therapeutic targets including many that affect histone methylation. These include histone lysine methyltransferases such as enhancer of zeste homolog 2 and DOT1L, protein arginine methyltransferases such as protein arginine methyltransferase 5, and histone lysine demethylases such as lysine-specific demethylase 1. This review presents the rationale for targeting histone methylation in oncology and provides an update on a few key targets that are being investigated in the clinic.
- Published
- 2017
- Full Text
- View/download PDF
33. A selective inhibitor of PRMT5 with in vivo and in vitro potency in MCL models.
- Author
-
Chan-Penebre E, Kuplast KG, Majer CR, Boriack-Sjodin PA, Wigle TJ, Johnston LD, Rioux N, Munchhof MJ, Jin L, Jacques SL, West KA, Lingaraj T, Stickland K, Ribich SA, Raimondi A, Scott MP, Waters NJ, Pollock RM, Smith JJ, Barbash O, Pappalardi M, Ho TF, Nurse K, Oza KP, Gallagher KT, Kruger R, Moyer MP, Copeland RA, Chesworth R, and Duncan KW
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Cell Death drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Crystallography, X-Ray, Dose-Response Relationship, Drug, Humans, Inhibitory Concentration 50, Isoquinolines chemistry, Isoquinolines therapeutic use, Lymphoma, Mantle-Cell drug therapy, Lymphoma, Mantle-Cell enzymology, Male, Methylation, Mice, Inbred Strains, Models, Molecular, Molecular Structure, Protein Binding, Pyrimidines chemistry, Pyrimidines therapeutic use, Xenograft Model Antitumor Assays, snRNP Core Proteins metabolism, Antineoplastic Agents pharmacology, Isoquinolines pharmacology, Lymphoma, Mantle-Cell pathology, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
Protein arginine methyltransferase-5 (PRMT5) is reported to have a role in diverse cellular processes, including tumorigenesis, and its overexpression is observed in cell lines and primary patient samples derived from lymphomas, particularly mantle cell lymphoma (MCL). Here we describe the identification and characterization of a potent and selective inhibitor of PRMT5 with antiproliferative effects in both in vitro and in vivo models of MCL. EPZ015666 (GSK3235025) is an orally available inhibitor of PRMT5 enzymatic activity in biochemical assays with a half-maximal inhibitory concentration (IC50) of 22 nM and broad selectivity against a panel of other histone methyltransferases. Treatment of MCL cell lines with EPZ015666 led to inhibition of SmD3 methylation and cell death, with IC50 values in the nanomolar range. Oral dosing with EPZ015666 demonstrated dose-dependent antitumor activity in multiple MCL xenograft models. EPZ015666 represents a validated chemical probe for further study of PRMT5 biology and arginine methylation in cancer and other diseases.
- Published
- 2015
- Full Text
- View/download PDF
34. SMYD3 links lysine methylation of MAP3K2 to Ras-driven cancer.
- Author
-
Mazur PK, Reynoird N, Khatri P, Jansen PW, Wilkinson AW, Liu S, Barbash O, Van Aller GS, Huddleston M, Dhanak D, Tummino PJ, Kruger RG, Garcia BA, Butte AJ, Vermeulen M, Sage J, and Gozani O
- Subjects
- Adenocarcinoma enzymology, Adenocarcinoma genetics, Adenocarcinoma metabolism, Adenocarcinoma pathology, Adenocarcinoma of Lung, Animals, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Disease Models, Animal, Humans, Lung Neoplasms enzymology, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, MAP Kinase Kinase Kinase 2 chemistry, MAP Kinase Kinase Kinases chemistry, Methylation, Mice, Mitogen-Activated Protein Kinases metabolism, Oncogene Protein p21(ras) genetics, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Protein Phosphatase 2 antagonists & inhibitors, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins A-raf metabolism, Signal Transduction, Cell Transformation, Neoplastic metabolism, Histone-Lysine N-Methyltransferase metabolism, Lysine metabolism, MAP Kinase Kinase Kinase 2 metabolism, MAP Kinase Kinase Kinases metabolism, Oncogene Protein p21(ras) metabolism
- Abstract
Deregulation of lysine methylation signalling has emerged as a common aetiological factor in cancer pathogenesis, with inhibitors of several histone lysine methyltransferases (KMTs) being developed as chemotherapeutics. The largely cytoplasmic KMT SMYD3 (SET and MYND domain containing protein 3) is overexpressed in numerous human tumours. However, the molecular mechanism by which SMYD3 regulates cancer pathways and its relationship to tumorigenesis in vivo are largely unknown. Here we show that methylation of MAP3K2 by SMYD3 increases MAP kinase signalling and promotes the formation of Ras-driven carcinomas. Using mouse models for pancreatic ductal adenocarcinoma and lung adenocarcinoma, we found that abrogating SMYD3 catalytic activity inhibits tumour development in response to oncogenic Ras. We used protein array technology to identify the MAP3K2 kinase as a target of SMYD3. In cancer cell lines, SMYD3-mediated methylation of MAP3K2 at lysine 260 potentiates activation of the Ras/Raf/MEK/ERK signalling module and SMYD3 depletion synergizes with a MEK inhibitor to block Ras-driven tumorigenesis. Finally, the PP2A phosphatase complex, a key negative regulator of the MAP kinase pathway, binds to MAP3K2 and this interaction is blocked by methylation. Together, our results elucidate a new role for lysine methylation in integrating cytoplasmic kinase-signalling cascades and establish a pivotal role for SMYD3 in the regulation of oncogenic Ras signalling.
- Published
- 2014
- Full Text
- View/download PDF
35. Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762.
- Author
-
Chaidos A, Caputo V, Gouvedenou K, Liu B, Marigo I, Chaudhry MS, Rotolo A, Tough DF, Smithers NN, Bassil AK, Chapman TD, Harker NR, Barbash O, Tummino P, Al-Mahdi N, Haynes AC, Cutler L, Le B, Rahemtulla A, Roberts I, Kleijnen M, Witherington JJ, Parr NJ, Prinjha RK, and Karadimitris A
- Subjects
- Animals, Antineoplastic Agents pharmacology, Apoptosis drug effects, Benzodiazepines pharmacology, Cell Cycle Checkpoints drug effects, Down-Regulation drug effects, Heterocyclic Compounds, 4 or More Rings pharmacology, Humans, Mice, Multiple Myeloma genetics, Multiple Myeloma pathology, Proto-Oncogene Proteins c-myc genetics, RNA-Binding Proteins genetics, Transcription Factors, Transcriptional Activation drug effects, Tumor Cells, Cultured, Antineoplastic Agents therapeutic use, Benzodiazepines therapeutic use, Heterocyclic Compounds, 4 or More Rings therapeutic use, Multiple Myeloma drug therapy
- Abstract
The bromodomain and extraterminal (BET) protein BRD2-4 inhibitors hold therapeutic promise in preclinical models of hematologic malignancies. However, translation of these data to molecules suitable for clinical development has yet to be accomplished. Herein we expand the mechanistic understanding of BET inhibitors in multiple myeloma by using the chemical probe molecule I-BET151. I-BET151 induces apoptosis and exerts strong antiproliferative effect in vitro and in vivo. This is associated with contrasting effects on oncogenic MYC and HEXIM1, an inhibitor of the transcriptional activator P-TEFb. I-BET151 causes transcriptional repression of MYC and MYC-dependent programs by abrogating recruitment to the chromatin of the P-TEFb component CDK9 in a BRD2-4-dependent manner. In contrast, transcriptional upregulation of HEXIM1 is BRD2-4 independent. Finally, preclinical studies show that I-BET762 has a favorable pharmacologic profile as an oral agent and that it inhibits myeloma cell proliferation, resulting in survival advantage in a systemic myeloma xenograft model. These data provide a strong rationale for extending the clinical testing of the novel antimyeloma agent I-BET762 and reveal insights into biologic pathways required for myeloma cell proliferation.
- Published
- 2014
- Full Text
- View/download PDF
36. Inhibition of BET bromodomain proteins as a therapeutic approach in prostate cancer.
- Author
-
Wyce A, Degenhardt Y, Bai Y, Le B, Korenchuk S, Crouthame MC, McHugh CF, Vessella R, Creasy CL, Tummino PJ, and Barbash O
- Subjects
- Animals, Apoptosis drug effects, Cell Growth Processes physiology, Cell Line, Tumor, Down-Regulation, Gene Expression Profiling, Humans, Male, Mice, Mice, SCID, Prostatic Neoplasms, Castration-Resistant enzymology, Prostatic Neoplasms, Castration-Resistant pathology, Xenograft Model Antitumor Assays, Benzodiazepines pharmacology, Nuclear Proteins antagonists & inhibitors, Prostatic Neoplasms, Castration-Resistant drug therapy, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
BET (bromodomain and extra-terminal) proteins regulate gene expression through their ability to bind to acetylated chromatin and subsequently activate RNA PolII-driven transcriptional elongation. Small molecule BET inhibitors prevent binding of BET proteins to acetylated histones and inhibit transcriptional activation of BET target genes. BET inhibitors attenuate cell growth and survival in several hematologic cancer models, partially through the down-regulation of the critical oncogene, MYC. We hypothesized that BET inhibitors will regulate MYC expression in solid tumors that frequently over-express MYC. Here we describe the effects of the highly specific BET inhibitor, I-BET762, on MYC expression in prostate cancer models. I-BET762 potently reduced MYC expression in prostate cancer cell lines and a patient-derived tumor model with subsequent inhibition of cell growth and reduction of tumor burden in vivo. Our data suggests that I-BET762 effects are partially driven by MYC down-regulation and underlines the critical importance of additional mechanisms of I-BET762 induced phenotypes.
- Published
- 2013
- Full Text
- View/download PDF
37. The FBXO4 tumor suppressor functions as a barrier to BRAFV600E-dependent metastatic melanoma.
- Author
-
Lee EK, Lian Z, D'Andrea K, Letrero R, Sheng W, Liu S, Diehl JN, Pytel D, Barbash O, Schuchter L, Amaravaradi R, Xu X, Herlyn M, Nathanson KL, and Diehl JA
- Subjects
- Amino Acid Substitution, Animals, Cell Line, Tumor, Cyclin D1 analysis, Cyclin D1 metabolism, F-Box Proteins genetics, Gene Deletion, Humans, Melanoma genetics, Melanoma metabolism, Mice, Point Mutation, Proto-Oncogene Proteins B-raf metabolism, Tumor Suppressor Proteins genetics, Ubiquitination, F-Box Proteins metabolism, Melanoma pathology, Proto-Oncogene Proteins B-raf genetics, Tumor Suppressor Proteins metabolism
- Abstract
Cyclin D1-cyclin-dependent kinase 4/6 (CDK4/6) dysregulation is a major contributor to melanomagenesis. Clinical evidence has revealed that p16(INK4A), an allosteric inhibitor of CDK4/6, is inactivated in over half of human melanomas, and numerous animal models have demonstrated that p16(INK4A) deletion promotes melanoma. FBXO4, a specificity factor for the E3 ligase that directs timely cyclin D1 proteolysis, has not been studied in melanoma. We demonstrate that Fbxo4 deficiency induces Braf-driven melanoma and that this phenotype depends on cyclin D1 accumulation in mice, underscoring the importance of this ubiquitin ligase in tumor suppression. Furthermore, we have identified a substrate-binding mutation, FBXO4 I377M, that selectively disrupts cyclin D1 degradation while preserving proteolysis of the other known FBXO4 substrate, TRF1. The I377M mutation and Fbxo4 deficiency result in nuclear accumulation of cyclin D1, a key transforming neoplastic event. Collectively, these data provide evidence that FBXO4 dysfunction, as a mechanism for cyclin D1 overexpression, is a contributor to human malignancy.
- Published
- 2013
- Full Text
- View/download PDF
38. BET inhibition silences expression of MYCN and BCL2 and induces cytotoxicity in neuroblastoma tumor models.
- Author
-
Wyce A, Ganji G, Smitheman KN, Chung CW, Korenchuk S, Bai Y, Barbash O, Le B, Craggs PD, McCabe MT, Kennedy-Wilson KM, Sanchez LV, Gosmini RL, Parr N, McHugh CF, Dhanak D, Prinjha RK, Auger KR, and Tummino PJ
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Antineoplastic Agents toxicity, Apoptosis drug effects, Apoptosis genetics, Benzodiazepines chemistry, Benzodiazepines toxicity, Cell Cycle Proteins, Cell Proliferation drug effects, Cluster Analysis, Disease Models, Animal, Female, Gene Expression Profiling, Gene Regulatory Networks, Humans, Kinetics, Mice, Models, Molecular, Molecular Conformation, N-Myc Proto-Oncogene Protein, Neuroblastoma drug therapy, Neuroblastoma pathology, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Protein Binding, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, RNA-Binding Proteins chemistry, RNA-Binding Proteins metabolism, Signal Transduction drug effects, Transcription Factors chemistry, Transcription Factors metabolism, Tumor Burden drug effects, Tumor Burden genetics, Xenograft Model Antitumor Assays, Benzodiazepines pharmacology, Gene Expression Regulation, Neoplastic drug effects, Gene Silencing, Neuroblastoma genetics, Neuroblastoma metabolism, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins genetics, Oncogene Proteins genetics, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 genetics, RNA-Binding Proteins antagonists & inhibitors, Transcription Factors antagonists & inhibitors
- Abstract
BET family proteins are epigenetic regulators known to control expression of genes involved in cell growth and oncogenesis. Selective inhibitors of BET proteins exhibit potent anti-proliferative activity in a number of hematologic cancer models, in part through suppression of the MYC oncogene and downstream Myc-driven pathways. However, little is currently known about the activity of BET inhibitors in solid tumor models, and whether down-regulation of MYC family genes contributes to sensitivity. Here we provide evidence for potent BET inhibitor activity in neuroblastoma, a pediatric solid tumor associated with a high frequency of MYCN amplifications. We treated a panel of neuroblastoma cell lines with a novel small molecule inhibitor of BET proteins, GSK1324726A (I-BET726), and observed potent growth inhibition and cytotoxicity in most cell lines irrespective of MYCN copy number or expression level. Gene expression analyses in neuroblastoma cell lines suggest a role of BET inhibition in apoptosis, signaling, and N-Myc-driven pathways, including the direct suppression of BCL2 and MYCN. Reversal of MYCN or BCL2 suppression reduces the potency of I-BET726-induced cytotoxicity in a cell line-specific manner; however, neither factor fully accounts for I-BET726 sensitivity. Oral administration of I-BET726 to mouse xenograft models of human neuroblastoma results in tumor growth inhibition and down-regulation MYCN and BCL2 expression, suggesting a potential role for these genes in tumor growth. Taken together, our data highlight the potential of BET inhibitors as novel therapeutics for neuroblastoma, and suggest that sensitivity is driven by pleiotropic effects on cell growth and apoptotic pathways in a context-specific manner.
- Published
- 2013
- Full Text
- View/download PDF
39. [DNA methylation profiling of the vascular tissues in the setting of atherosclerosis].
- Author
-
Nazarenko MS, Markov AV, Lebedev IN, Sleptsov AA, Frolov AV, Barbash OL, and Puzyrev VP
- Subjects
- Atherosclerosis genetics, Atherosclerosis pathology, Female, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Male, Mammary Arteries pathology, Middle Aged, Proteins genetics, Proteins metabolism, Saphenous Vein pathology, Atherosclerosis metabolism, CpG Islands, DNA Methylation, Mammary Arteries metabolism, Saphenous Vein metabolism
- Abstract
To date the question of epigenetic mechanisms of gene regulation in the context of cardiovascular diseases is of a considerable interest. Here, for the first time DNA methylation profiles of vascular tissues of atherosclerotic patients have been analyzed with using the microarray Infinium HumanMethylation27 BeadChip ("Illumina", USA). As the result, within 286 genes 314 CpG-sites that varied significantly in the DNA methylation level between the tissue samples of carotid (in the area of atherosclerotic plaques and nearby macroscopically intact tissues of the vascular wall) and mammary arteries as well saphenous veins have been identified. The most pronounced differences in the methylation level were registered for CpG-sites of homeobox genes HOXA2 and HOXD4 as well as imprinted gene MEST. In particular, these genes were found to be hypomethylated in the carotid atherosclerotic plaques compared to their methylation patterns in intact tissues of internal mammary arteries and saphenous veins.
- Published
- 2013
- Full Text
- View/download PDF
40. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation.
- Author
-
Van Aller GS, Reynoird N, Barbash O, Huddleston M, Liu S, Zmoos AF, McDevitt P, Sinnamon R, Le B, Mas G, Annan R, Sage J, Garcia BA, Tummino PJ, Gozani O, and Kruger RG
- Subjects
- Animals, Blotting, Western, Chromatin genetics, Chromatin metabolism, Enzyme Activation, Fibroblasts metabolism, Fibroblasts pathology, Genetic Complementation Test, HeLa Cells, Histone-Lysine N-Methyltransferase genetics, Histones genetics, Humans, Methylation, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutagenesis, Site-Directed, Peptide Library, Phenotype, Plasmids genetics, Plasmids metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Substrate Specificity, Gene Expression Regulation, Neoplastic, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Lysine metabolism
- Abstract
Smyd3 is a lysine methyltransferase implicated in chromatin and cancer regulation. Here we show that Smyd3 catalyzes histone H4 methylation at lysine 5 (H4K5me). This novel histone methylation mark is detected in diverse cell types and its formation is attenuated by depletion of Smyd3 protein. Further, Smyd3-driven cancer cell phenotypes require its enzymatic activity. Thus, Smyd3, via H4K5 methylation, provides a potential new link between chromatin dynamics and neoplastic disease.
- Published
- 2012
- Full Text
- View/download PDF
41. The Fbx4 tumor suppressor regulates cyclin D1 accumulation and prevents neoplastic transformation.
- Author
-
Vaites LP, Lee EK, Lian Z, Barbash O, Roy D, Wasik M, Klein-Szanto AJ, Rustgi AK, and Diehl JA
- Subjects
- Animals, Cell Cycle, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cell Nucleus metabolism, Cell Proliferation, Cells, Cultured, Cyclin D1 biosynthesis, Cyclin D1 genetics, DNA Damage, Fibroblasts metabolism, Gene Knockout Techniques, Mice, Mice, Transgenic, Neoplasms genetics, Proto-Oncogene Proteins p21(ras) metabolism, Cell Transformation, Neoplastic, Cyclin D1 metabolism, F-Box Proteins genetics, F-Box Proteins metabolism, SKP Cullin F-Box Protein Ligases metabolism
- Abstract
Skp1-Cul1-F-box (SCF) E3 ubiquitin ligase complexes modulate the accumulation of key cell cycle regulatory proteins. Following the G(1)/S transition, SCF(Fbx4) targets cyclin D1 for proteasomal degradation, a critical event necessary for DNA replication fidelity. Deregulated cyclin D1 drives tumorigenesis, and inactivating mutations in Fbx4 have been identified in human cancer, suggesting that Fbx4 may function as a tumor suppressor. Fbx4(+/-) and Fbx4(-/-) mice succumb to multiple tumor phenotypes, including lymphomas, histiocytic sarcomas and, less frequently, mammary and hepatocellular carcinomas. Tumors and premalignant tissue from Fbx4(+/-) and Fbx4(-/-) mice exhibit elevated cyclin D1, an observation consistent with cyclin D1 as a target of Fbx4. Molecular dissection of the Fbx4 regulatory network in murine embryonic fibroblasts (MEFs) revealed that loss of Fbx4 results in cyclin D1 stabilization and nuclear accumulation throughout cell division. Increased proliferation in early passage primary MEFs is antagonized by DNA damage checkpoint activation, consistent with nuclear cyclin D1-driven genomic instability. Furthermore, Fbx4(-/-) MEFs exhibited increased susceptibility to Ras-dependent transformation in vitro, analogous to tumorigenesis observed in mice. Collectively, these data reveal a requisite role for the SCF(Fbx4) E3 ubiquitin ligase in regulating cyclin D1 accumulation, consistent with tumor suppressive function in vivo.
- Published
- 2011
- Full Text
- View/download PDF
42. Genotoxic stress-induced cyclin D1 phosphorylation and proteolysis are required for genomic stability.
- Author
-
Pontano LL, Aggarwal P, Barbash O, Brown EJ, Bassing CH, and Diehl JA
- Subjects
- 3T3 Cells, Animals, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins, Cell Line, Cyclin D1 genetics, DNA-Binding Proteins, Glycogen Synthase Kinase 3, Humans, Mice, Phosphorylation, Proteasome Endopeptidase Complex metabolism, Protein Processing, Post-Translational, Protein Serine-Threonine Kinases, S Phase, Tumor Suppressor Proteins, Ubiquitination, Cyclin D1 metabolism, DNA Damage, Genomic Instability
- Abstract
While mitogenic induction of cyclin D1 contributes to cell cycle progression, ubiquitin-mediated proteolysis buffers this accumulation and prevents aberrant proliferation. Because the failure to degrade cyclin D1 during S-phase triggers DNA rereplication, we have investigated cellular regulation of cyclin D1 following genotoxic stress. These data reveal that expression of cyclin D1 alleles refractory to phosphorylation- and ubiquitin-mediated degradation increase the frequency of chromatid breaks following DNA damage. Double-strand break-dependent cyclin D1 degradation requires ATM and GSK3beta, which in turn mediate cyclin D1 phosphorylation. Phosphorylated cyclin D1 is targeted for proteasomal degradation after ubiquitylation by SCF(Fbx4-alphaBcrystallin). Loss of Fbx4-dependent degradation triggers radio-resistant DNA synthesis, thereby sensitizing cells to S-phase-specific chemotherapeutic intervention. These data suggest that failure to degrade cyclin D1 compromises the intra-S-phase checkpoint and suggest that cyclin D1 degradation is a vital cellular response necessary to prevent genomic instability following genotoxic insult.
- Published
- 2008
- Full Text
- View/download PDF
43. SCF(Fbx4/alphaB-crystallin) E3 ligase: when one is not enough.
- Author
-
Barbash O and Diehl JA
- Subjects
- Animals, Cell Cycle, Cyclin D1 genetics, Cyclin D1 metabolism, F-Box Proteins genetics, Humans, Models, Biological, Neoplasms metabolism, Phosphorylation, alpha-Crystallin B Chain genetics, F-Box Proteins metabolism, SKP Cullin F-Box Protein Ligases metabolism, Ubiquitin-Protein Ligases metabolism, alpha-Crystallin B Chain metabolism
- Abstract
Cell cycle progression is determined by the balance of positive regulators, cyclin-dependent-protein kinases (cdks) relative to negative regulators, cyclin-dependent kinase inhibitors (ckis). D-type cyclins, (D1, D2, D3) are expressed in a tissue-specific manner and are the first cyclins to be expressed during the cell cycle. Of the three D-type cyclins, cyclin D1 is most frequently overexpressed in human cancer. The mechanisms of cyclin D1 overexpression can be attributed to gene amplification, transcriptional activation and altered protein degradation; of these, inhibition of ubiquitin-dependent proteolysis of cyclin D1 is thought to be a primary mechanism of cyclin D1 overexpression in human tumors. Because the identity of the regulators of cyclin D1 proteolysis were largely undefined until recently, it had not been possible to determine whether this regulatory network was directly targeted in primary cancer. Cyclin D1 proteolysis requires phosphorylation by GSK3beta at Thr-286; additional work recently established that p286-D1 is a substrate for the SCF(Fbx4/alphaB-crystallin) E3 ligase. This discovery has facilitated an analysis of SCF(Fbx4/alphaB-crystallin) ligase in human cancers. This recent work revealed that Fbx4 is subject to mutational inactivation in human cancer, resulting in the accumulation of cyclin D1. Molecular analysis of this ligase has revealed striking regulatory features that contribute to regulated cyclin D1 accumulation and support the idea that Fbx4 is a bona fide tumor suppressor.
- Published
- 2008
- Full Text
- View/download PDF
44. Mutations in Fbx4 inhibit dimerization of the SCF(Fbx4) ligase and contribute to cyclin D1 overexpression in human cancer.
- Author
-
Barbash O, Zamfirova P, Lin DI, Chen X, Yang K, Nakagawa H, Lu F, Rustgi AK, and Diehl JA
- Subjects
- Animals, Cell Cycle, Cell Nucleus metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cyclin D, Cyclins genetics, Dimerization, Esophageal Neoplasms enzymology, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology, F-Box Proteins genetics, Female, Glycogen Synthase Kinase 3 metabolism, Glycogen Synthase Kinase 3 beta, Humans, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Mice, Mice, Inbred NOD, Mice, SCID, NIH 3T3 Cells, Phosphorylation, Protein Structure, Tertiary, Proto-Oncogene Proteins c-akt metabolism, Serine, Transfection, Up-Regulation, ras Proteins metabolism, Cyclins metabolism, Esophageal Neoplasms metabolism, F-Box Proteins metabolism, Gene Expression Regulation, Neoplastic, Mutation, SKP Cullin F-Box Protein Ligases metabolism
- Abstract
SCF(Fbx4) was recently identified as the E3 ligase for cyclin D1. We now describe cell-cycle-dependent phosphorylation and dimerization of Fbx4 that is regulated by GSK3beta and is defective in human cancer. We present data demonstrating that a pathway involving Ras-Akt-GSK3beta controls the temporal phosphorylation and dimerization of the SCF(Fbx4) E3 ligase. Inhibition of Fbx4 activity results in accumulation of nuclear cyclin D1 and oncogenic transformation. The importance of this regulatory pathway for normal cell growth is emphasized by the prevalence of mutations in Fbx4 in human cancer that impair dimerization. Collectively, these data reveal that inactivation of the cyclin D1 E3 ligase likely contributes to cyclin D1 overexpression in a significant fraction of human cancer.
- Published
- 2008
- Full Text
- View/download PDF
45. SCF Fbx4/alphaB-crystallin cyclin D1 ubiquitin ligase: a license to destroy.
- Author
-
Barbash O, Lin DI, and Diehl JA
- Abstract
Cyclin D1 is an allosteric regulator for cyclin-dependent kinases 4 and 6 (CDK4/6). The cyclin D/CDK4 kinase promotes G1/S transition through the posttranslational modification and the subsequent inactivation of the retinoblastoma (Rb) protein and related family members (p107 and p130). Accumulation of cyclin D1 is tightly regulated through various mechanisms including transcription, protein localization and ubiquitin-dependent proteolysis. While regulators of cyclin D1 gene expression have been under considerable scrutiny, the identity of the protein complex that targets cyclin D1 protein for degradation, the putative E3 ubiquitin ligase, has remained obscure. In a recent report, Lin et al describe the identification and characterization of a novel SCF, wherein FBX4 and alphaB-crystallin serve as specificity factors that direct ubiquitination of phosphorylated cyclin D1. As cyclin D1 overexpression in human cancer has been postulated to occur through the loss of degradation machinery, the identification of the SCFFbx4/alphaB-crystallin ligase will allow new experimental approaches that address mechanisms of cyclin D1 overexpression in human cancer.
- Published
- 2007
- Full Text
- View/download PDF
46. Phosphorylation-dependent ubiquitination of cyclin D1 by the SCF(FBX4-alphaB crystallin) complex.
- Author
-
Lin DI, Barbash O, Kumar KG, Weber JD, Harper JW, Klein-Szanto AJ, Rustgi A, Fuchs SY, and Diehl JA
- Subjects
- Animals, Catalysis, Cytoplasm metabolism, F-Box Proteins chemistry, F-Box Proteins genetics, G1 Phase physiology, Gene Expression Regulation, Neoplastic, Humans, Mice, NIH 3T3 Cells, Neoplasms genetics, Phosphorylation, Phosphothreonine metabolism, Protein Binding, Protein Transport, Proto-Oncogene Mas, RNA, Messenger genetics, RNA, Messenger metabolism, Thermodynamics, alpha-Crystallin B Chain genetics, Cyclin D1 metabolism, F-Box Proteins metabolism, SKP Cullin F-Box Protein Ligases metabolism, Ubiquitin metabolism, alpha-Crystallin B Chain metabolism
- Abstract
Growth factor-dependent accumulation of the cyclin D1 proto-oncogene is balanced by its rapid phosphorylation-dependent proteolysis. Degradation is triggered by threonine 286 phosphorylation, which promotes its ubiquitination by an unknown E3 ligase. We demonstrate that Thr286-phosphorylated cyclin D1 is recognized by a Skp1-Cul1-F box (SCF) ubiquitin ligase where FBX4 and alphaB crystallin govern substrate specificity. Overexpression of FBX4 and alphaB crystallin triggered cyclin D1 ubiquitination and increased cyclin D1 turnover. Impairment of SCF(FBX4-alphaB crystallin) function attenuated cyclin D1 ubiquitination, promoting cyclin D1 overexpression and accelerated cell-cycle progression. Purified SCF(FBX4-alphaB crystallin) catalyzed polyubiquitination of cyclin D1 in vitro. Consistent with a putative role for a cyclin D1 E3 ligase in tumorigenesis, FBX4 and alphaB crystallin expression was reduced in tumor-derived cell lines and a subset of primary human cancers that overexpress cyclin D1. We conclude that SCF(FBX4-alphaB crystallin) is an E3 ubiquitin ligase that promotes ubiquitin-dependent degradation of Thr286-phosphorylated cyclin D1.
- Published
- 2006
- Full Text
- View/download PDF
47. The oxidative stressor arsenite activates vascular endothelial growth factor mRNA transcription by an ATF4-dependent mechanism.
- Author
-
Roybal CN, Hunsaker LA, Barbash O, Vander Jagt DL, and Abcouwer SF
- Subjects
- Acetylcysteine pharmacology, Activating Transcription Factor 4, Animals, Antioxidants pharmacology, Binding Sites, Cell Line, Cell Line, Transformed, DNA metabolism, Embryo, Mammalian, Endoplasmic Reticulum Chaperone BiP, Eukaryotic Initiation Factor-2 metabolism, Fibroblasts, Gene Expression, Heat-Shock Proteins genetics, Heme Oxygenase (Decyclizing), Humans, Mice, Mice, Knockout, Molecular Chaperones genetics, Oxidative Stress, Oxygenases genetics, Phosphorylation, Pigment Epithelium of Eye, Promoter Regions, Genetic genetics, RNA, Messenger analysis, Rats, Trans-Activators deficiency, Trans-Activators genetics, Trans-Activators physiology, Transcription Factors genetics, Transfection, Arsenites pharmacology, Gene Expression Regulation drug effects, Transcription Factors physiology, Transcription, Genetic drug effects, Vascular Endothelial Growth Factor A genetics
- Abstract
Aberrant retinal expression of vascular endothelial growth factor (VEGF) leading to neovascularization is a central feature of age-related macular degeneration and diabetic retinopathy, two leading causes of vision loss. Oxidative stress is suggested to occur in retinal tissue during age-related macular degeneration and diabetic retinopathy and is suspected in the mechanism of VEGF expression in these diseases. Arsenite, a thiol-reactive oxidative stressor, induces VEGF expression by a HIF-1alpha-independent mechanism. Previously, we demonstrated that homocysteine, an endoplasmic reticulum stressor, increases VEGF transcription by a mechanism dependent upon activating transcription factor ATF4. Because ATF4 is expressed in response to oxidative stress, we hypothesized that ATF4 was also responsible for increased VEGF transcription in response to arsenite. We now show that arsenite increased steady state levels of VEGF mRNA and activated transcription from a VEGF promoter construct. Arsenite induced eIF2alpha phosphorylation, resulting in increased ATF4 protein levels. Inactivation or loss of ATF4 greatly diminished the VEGF response to arsenite treatment. Overexpression of ATF4 was sufficient to activate the VEGF promoter, and arsenite cooperated with exogenous ATF4 to further activate the promoter. A complex containing ATF4 binds a DNA element at +1767 bp relative to the VEGF transcription start site, and DNA binding activity is increased by arsenite treatment. In addition, the ability of a thiol antioxidant, N-acetylcysteine, to inhibit the effect of arsenite on VEGF expression coincided with its ability to inhibit phosphorylation of eIF2alpha and ATF4 protein expression. Thus, arsenite-induced up-regulation of VEGF gene transcription occurs by an ATF4-dependent mechanism.
- Published
- 2005
- Full Text
- View/download PDF
48. Expression of angiogenic factors vascular endothelial growth factor and interleukin-8/CXCL8 is highly responsive to ambient glutamine availability: role of nuclear factor-kappaB and activating protein-1.
- Author
-
Bobrovnikova-Marjon EV, Marjon PL, Barbash O, Vander Jagt DL, and Abcouwer SF
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms metabolism, Cell Line, Tumor, Chemokine CXCL1, Chemokines, CXC genetics, Chemokines, CXC physiology, Curcumin pharmacology, DNA, Neoplasm genetics, DNA, Neoplasm metabolism, Gene Expression Regulation, Neoplastic, Glutamine metabolism, Humans, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins physiology, Interleukin-8 genetics, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Oligonucleotide Array Sequence Analysis, RNA, Messenger biosynthesis, RNA, Messenger genetics, RNA, Messenger metabolism, Transcription Factor AP-1 antagonists & inhibitors, Transcription Factor AP-1 metabolism, Transcription, Genetic, Up-Regulation, Vascular Endothelial Growth Factor A genetics, Chemokines, CXC biosynthesis, Glutamine deficiency, Intercellular Signaling Peptides and Proteins biosynthesis, Interleukin-8 biosynthesis, NF-kappa B physiology, Transcription Factor AP-1 physiology, Vascular Endothelial Growth Factor A biosynthesis
- Abstract
Vascular endothelial growth factor (VEGF) and interleukin-8/CXCL8 (IL-8) are prominent pro-angiogenic and pro-metastatic proteins that represent negative prognostic factors in many types of cancer. Hypoxia is thought to be the primary environmental cause of VEGF and IL-8 expression in solid tumors. We hypothesized that a lack of nutrients other than oxygen could stimulate the expression of these factors and previously demonstrated that expression of VEGF and IL-8 is responsive to amino acid deprivation. In the present study, we examined the effect of glutamine availability on the expression of these factors as well as the role of transcription factors NFkappaB and activating protein-1 (AP-1) in the response of TSE human breast carcinoma cells to glutamine deprivation. VEGF and IL-8 secretion and mRNA levels were dramatically induced by glutamine deprivation. mRNA stabilization contributed to this response. Glutamine deprivation increased NFkappaB (p65/p50) and AP-1 (Fra-1/c-Jun+JunD) DNA-binding activities. Blocking NFkappaB and AP-1 activation with curcumin as well as expression of dominant inhibitors, inhibitor of nuclear factor-kappaB (IkappaB) super repressor (IkappaBM), and a mutant form of c-Fos (A-Fos) demonstrated that the activation of NFkappaB and AP-1 transcription factors was necessary for the induction of IL-8 expression but dispensable for the induction of VEGF expression. A macro-array containing 111 NFkappaB target genes identified a total of 17 that were up-regulated 2-fold or more in response to glutamine deprivation. These included growth regulated oncogene alpha (GROalpha/GRO1/CXCL1), another neutrophil chemoattractant implicated in tumor angiogenesis and metastasis.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.