GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 618-618
    Abstract: β-catenin acts as a co-activator for the T-cell factor (TCF) 4/lymphoid enhancer factor (LEF) 1 bipartite transcription factor at the promoters of the WNT-β-catenin target genes, including cyclin D1, c-Myc and survivin. The canonical WNT-β-catenin pathway is documented to be essential for self-renewal, growth and survival of the AML stem and blast progenitor cells (BPCs), which has also been correlated with a poor prognosis in AML. In AML stem/BPCs expressing mutant FLT3-ITD, increased PI3K/AKT activity causes phosphorylation and inactivation of GSK3β, thereby preventing degradation, promoting stabilization and nuclear localization of β-catenin. Additionally, FLT3 can also directly mediate the tyrosine phosphorylation of β-catenin, thereby stabilizing and promoting the nuclear localization and binding of β-catenin to TCF4. TBL1 (transducin beta-like) is an adaptor protein, which binds to nuclear β-catenin and promotes its co-factor activity with TCF4/LEF1 in mediating transcription of the target genes, including c-Myc, cyclin D1 and survivin. Therefore, we hypothesized that targeted disruption of TBL1-β-catenin binding or depletion of TBL1 would abrogate the pro-growth and oncogenic signaling of β-catenin in AML BPCs, especially those expressing FLT3-ITD. Here, we demonstrate that treatment with 20 to 100 nM of BC2059 (β-Cat Pharmaceuticals), a small molecule, anthraquinone oxime-analog, disrupts the binding of β-catenin to TBL1 (by anti-TBL1 pull down and immunofluorescence analyses) and promotes proteasomal degradation of β-catenin, thereby attenuating the nuclear levels of β-catenin in the cultured (OCI-AML3, MOLM13 and MV4-11), as well as in primary (p) AML BPCs. Concomitantly, BC2059 treatment inhibited the mRNA and protein expression of c-Myc, cyclin D1 and survivin, while de-repressing p21 and Axin2. BC2059 also dose dependently inhibited growth and induced apoptosis of cultured and CD34+ pAML BPCs expressing FLT3-ITD (40 to 60%), but not of normal CD34+ bone marrow progenitor cells (p 〈 0.01). Transient knockdown of TBL1 or beta catenin (60 to 70%) by lentivirus-transduced shRNA caused loss of viability in MOLM13 cells, which was significantly enhanced by treatment with BC2059 (p 〈 0.01). BC2059 also induced apoptosis of MOLM13-TKIR cells that were isolated in vitro to exhibit resistance to FLT3 antagonists (approximately 50-fold). Notably, BC2059 treatment (10 mg/kg, t.i.w., by IV injection) also exerted potent in vivo anti-AML activity and significantly improved the survival of immune depleted mice engrafted with cultured and patient-derived pAML BPCs (p 〈 0.001). Since compared to the control OCI-AML3 cells, BC2059 demonstrated significantly greater lethality against the OCI-AML3 cells ectopically overexpressing FLT3-ITD (approximately 8-fold), we hypothesized that co-treatment with a FLT3 antagonist would further reduce the nuclear levels of β-catenin and enhance the lethal activity of FLT3-antagonist against AML BPCs expressing FLT3-ITD. Indeed, co-treatment with BC2059 (50 nM) and the FLT3-antagonist quizartinib or ponatinib (100 to 200 nM), versus each agent alone, caused more reduction in the nuclear levels and binding of β-catenin to TBL1 (by confocal immunofluorescence analysis). This was associated with greater decline in the expression of c-Myc, cyclin D1 and survivin, but increase in the levels of p21 and BIM. Compared to each agent alone, co-treatment with BC2059 and quizartinib or ponatinib also synergistically induced apoptosis of the FLT3-ITD expressing cultured (MOLM13 and MV4-11) and pAML BPCs (combination indices of 〈 1.0, by isobologram analyses) but not of normal CD34+ progenitor cells. Treatment with BC2059 (25 to 100 nM) also significantly increased the apoptosis observed by the shRNA mediated incomplete knockdown of TBL1 or β-catenin (approximately 70%) in MOLM13 cells (p 〈 0.01). Collectively, our findings support that targeted inhibition of the levels and binding of β-catenin to TBL by BC2059 and FLT3-antagonist is a promising approach to exert lethal activity against AML BPCs expressing FLT3-ITD. Further pre-clinical development of this combination therapy against FLT3-ITD expressing AML is progressing. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2623-2623
    Abstract: We have previously reported that treatment with BET-protein bromodomain antagonist (BA) reduces the enhancer/promoter occupancy of BRD4 and induces the expression of HEXIM1, which sequesters and inhibits pTEFb (a complex of CDK9 and cyclin T). pTEFb phosphorylates serine 2 in the C-terminal heptad repeats in the stalled RNA pol II (RNAP2), causing its pause-release and the mRNA transcript elongation. While relatively sparing normal bone marrow progenitor cells, BA treatment attenuates the expression of cMYC, BCL2 and CDK4/6 and induces apoptosis of cultured (OCI-AML3, MOLM13 and MV4-11) and primary AML blast progenitor cells (BPCs), including those expressing FLT3-ITD. Because of its ability to inhibit CDK9 and pTEFb, we determined whether co-treatment with flavopiridol (FP), a known inhibitor of CDK9, would enhance the activity of BA against the cultured and primary AML BPCs. Our findings demonstrate that co-treatment with BA and FP synergistically induced apoptosis of the cultured and primary AML BPCs (CI of & lt; 1.0). Combination of BA and FP, versus each drug alone, was associated with greater attenuation of the levels of p-RNAP2, cMYC, BCL-2 and CDK4/6 but greater induction of HEXIM1 and p21 levels. Notably, co-treatment with FP and BA markedly reduced the levels of the anti-apoptotic protein MCL1, which is known to be transcriptionally regulated by pTEFb-mediated phosphorylation of p-RNAP2. However, treatment with BA and FP did not affect the levels of cyclin T or CDK9. Co-treatment with BA and FP also synergistically induced apoptosis of MOLM13-TKIR cells (expressing FLT3-ITD) that exhibit & gt; 100-fold in vitro resistance to FLT3 tyrosine kinase inhibitors. We next determined whether HEXIM1 levels regulate the activity of BA and FP in the AML BPCs. While shRNA-mediated stable knockdown of HEXIM1 significantly reduced the lethal activity of BA in OCI-AML3/HKD and MOLM13/HKD cells, tetracycline-inducible ectopic overexpression of HEXIM1 significantly enhanced BA-induced apoptosis of MOLM13 and OCI-AML3 cells. Notably, the significant improvement in the median survival of NOD/SCID mice treated with JQ1 (50 mg/kg/5 days per week/for 3 weeks) was not observed in the similarly treated mice engrafted with MOLM13/HKD cells (p & lt; 0.01). The lethality induced by co-treatment with JQ1 and FP was significantly reduced in MOLM13/HKD versus MOLM13 cells; conversely, it was significantly increased in MOLM13 cells with ectopic overexpression of HEXIM1 (p & lt; 0.01). These findings indicate that co-treatment with BA and FP exerts synergistic lethality, and BA-mediated induction of HEXIM1 is mechanistically involved in mediating the lethal effects of BA and FP against human AML BPCs. Citation Format: Santhana G. T. Devaraj, Bhavin Shah, Warren Fiskus, Baohua Sun, Saikat Saha, Sai Ravi Pingali, Swaminathan P. Iyer, Sunil Sharma, James E. Bradner, Kapil N. Bhalla. Molecular mechanism of synergy between BET-protein bromodomain antagonist (BA) and pTEFb kinase inhibitor against human AML blast progenitor cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2623. doi:10.1158/1538-7445.AM2015-2623
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Oncotarget, Impact Journals, LLC, Vol. 5, No. 14 ( 2014-07-30), p. 5637-5650
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2014
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3534-3534
    Abstract: The BET (bromodomain and extra terminal) protein family, including BRD4 are chromatin reader proteins that bind to acetylated lysines on histone proteins and recruit transcriptional regulatory complexes to gene promoters, thereby coupling histone acetylation to RNA polymerase II (RNAP2)-mediated transcript elongation. Preferential binding of BRD4 to clustered enhancers or ‘super enhancers’ enables BRD4, through the mediator complex, to regulate the transcription of specific oncogenes, including c-MYC, BCL2 and CDK4/6 in AML blast progenitor cells (BPCs). Through its C-terminal domain (CTD), BRD4 recruits pTEFb (positive transcription elongation factor b), the heterodimer composed of cyclin T and CDK9, which phosphorylates the heptad repeats on the CTD of RNAP2 at serine 2. The kinase activity of CDK9 in pTEFb is also required for the phosphorylation of DSIF and NELF, which abrogates their inhibitory hold on the pause-release of RNAP2 and productive transcript elongation of specific oncogenes and their target genes. pTEFb exists in two forms, the catalytically active form, which is associated with BRD4 and RNAP2, and a catalytically inactive form associated with 7SK snRNP (small nuclear ribonucleoprotein) and the inhibitory protein HEXIM1 (hexamethylene bis-acetamide or HMBA-inducible protein 1). The transition of pTEFb between the inactive and active complex is dynamic and regulated by cell growth and stress signaling. HEXIM1 binds to and sequesters pTEFb, thereby inhibiting its kinase activity for RNAP II. We recently reported (Mol Cancer Ther, 2014) that the BRD4 antagonist JQ1 and I-BET151 alone, and synergistically with the pan-histone deacetylase inhibitor panobinostat, inhibits the growth and induces apoptosis in cultured (OCI-AML3 and MOLM13) and patient-derived primary AML (pAML) BPCs. JQ1 treatment also improved the survival of the immune-depleted mice engrafted with MOLM13 or pAML BPCs. Notably, the lethal activity of JQ1 was associated with marked induction of HEXIM1. Therefore, we hypothesized that HEXIM1 induction, by sequestering pTEFb and inhibiting RNAP2, contributes to the growth inhibitory and lethal effects of JQ1. We additionally hypothesized that co-treatment with those agents that further augment HEXIM1 induction would synergize with JQ1 against AML BPCs. To validate these hypotheses, we achieved a stable lentivirus mediated knockdown (KD) of the mRNA and protein expressions of HEXIM1 by approximately 90% in OCI-AML3 and MOLM13 cells, as well as transient KD in pAML BPCs. Compared to the parental control cells, OCI-AML3/HKD and MOLM13/HKD cells showing HEXIM1 knockdown (HKD) exhibited reduced binding of HEXIM1 with cyclin T (as revealed by cyclin T pull-down and immunofluorescence microscopy), but showed augmented suspension culture growth and increased protein levels (by Western blot) of cyclin T, pRNAP2 and cMYC levels. However, in contrast to the parental control cells, JQ1 (1000 nM) treatment-mediated HEXIM1 induction and binding to cyclin T was markedly inhibited in the HKD AML BPCs, which was associated with significant inhibition of JQ1-induced pRNAP2, as well as differentiation and apoptosis of HKD BPCs (p 〈 0.01). Notably, whereas treatment with JQ1 alone improved the median survival of NOD/SCID mice engrafted with MOLM13 BPCs, this survival improvement was significantly impaired in the mice engrafted with MOLM13/HKD AML BPCs (p 〈 0.02). This indicated that HEXIM1-induction mechanistically contributes to JQ1-induced growth inhibition, differentiation and apoptosis of AML BPCs. Further, the synergistic apoptosis observed in MOLM13, OCI-AML3 and primary AML cells induced by co-treatment with JQ1 (250 to 1000 nM) and PS (5 to 10 nM) or HMBA (1 to 2 mM) was also associated with greater induction of HEXIM1. However, this synergy was impaired in the cultured and pAML BPCs with HEXIM1 knockdown. These findings confirm that HEXIM1 induction is a biomarker and plays a mechanistic role in mediating growth inhibition, differentiation and apoptosis induced by BRD4 antagonist in human AML BPCs. These findings support, and we have embarked upon, studies involving future pre-clinical development of those agents that in combination significantly augment BRD4 antagonist-induced HEXIM1 in human AML BPCs. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 918-918
    Abstract: Poor-risk Mantle Cell Lymphoma (MCL) demonstrate several genetic alterations involving cyclin D1, RB1, ATM, p53, INK4a/ARF, BMI1, HDM2, CDK4, CDKN2A, NOTCH1 & 2, c-MYC, BCL2 and BIM, as well as in the Toll like receptor (TLR), B-cell receptor (BCR) and NFkB signaling genes. Activated BCR signaling is also associated with downstream pro-growth and pro-survival activity of NFκB in the poor-risk MCL. Pre-clinical and clinical studies have shown that ibrutinib, a selective, orally bioavailable, irreversible inhibitor of BTK is active against B cell neoplasms, especially MCL. Treatment with ibrutinib induces clinical responses, but long-term durable remissions and cure remains uncertain. BET (bromodomain and extra terminal) protein family members, including BRD4, bind to acetylated lysine(s) on the histone proteins, help assemble transcriptional co-regulators at the transcriptionally active target oncogene ‘super’ enhancers and promoters, and regulate the expression of important MCL-relevant oncogenes, e.g., c-MYC, BCL-2, CDK4/6 and cyclin D1. Additionally, BRD4 has been shown to bind acetylated RelA and is essential for NFκB activity. Here, we determined that treatment with the prototype BET protein antagonist JQ1 (250 to 2000 nM) or I-BET151, which disrupts the binding of BRD4 to acetylated histones, dose-dependently inhibited growth and exerted lethal activity against cultured (MO2058 and Mino, Z138 and JeKo-1) and patient-derived, primary MCL (pMCL) cells. Treatment with JQ1 reduced the occupancy of BRD4 and RNA pol II (RNAP2) on the promoters of c-MYC and BCL2, as well as attenuated the mRNA (by qPCR) and protein expression (by Western blot) of c-MYC, BCL2, MCL-1 and, CDK4/6, without affecting the levels of SOX11, but induced the levels of HEXIM1, p21, p27 and cleaved PARP in MCL cells. Importantly, JQ1 treatment reduced p-BTK, BTK and p-PLCγ2 levels in MCL cells. JQ1 treatment also attenuated the nuclear RelA (by Western and confocal immunofluorescence analyses) and inhibited the expression of several NFkB target genes, including XIAP, Iκbα, cFLIP and cIAP2 expressions. Treatment with ibrutinib (2 to 10 µM) attenuated p-BTK, p-PLCγ2, p-AKT and c-MYC levels but induced p21 levels, as well as induced apoptosis of cultured (MO2058, Mino and Z138 and JeKo-1 cells) and pMCL cells. Importantly, co-treatment with JQ1 and ibrutinib markedly inhibited p-BTK, BTK, p-PLCγ2, p-AKT and nuclear RelA levels and synergistically induced apoptosis of the cultured (JeKo-1, MO2058 and Mino) and pMCL cells (CI values 〈 1.0 by isobologram analysis). Further, following tail-vein infusion and engraftment of Mino cells (3 million cells/mouse) in the bone marrow and spleen of NOD/SCID mice, as compared to each agent alone, co-treatment with JQ1 (50 mg/kg/day, IP) and ibrutinib (25 mg/kg/day IP) significantly improved the survival of the NOD/SCID mice (p 〈 0.01). We also isolated ibrutinib-resistant Mino/IR cells ( 〉 20 fold resistant), following a continuous exposure of the parental Mino cells in culture to escalating levels of ibrutinib, which exhibited high levels of p-BTK, p-PLCγ2, c-MYC and CDK6. Co-treatment with JQ1 and the pan-histone deacetylase inhibitor panobinostat (PS) synergistically induced apoptosis of not only Mino but also Mino/IR cells. We also isolated carfilzomib-resistant JeKo-1/CZR cells ( 〉 10 fold resistant). As compared to JeKo-1, JeKo-1/CZR cells demonstrated higher protein levels of proteasome subunits and increased base-line and lesser EGCG (Epigallocatechin-3-gallate)-suppressible proteasome activity. Mutations in the PSMB5 were not detected. JeKo-1/CZR cells also displayed higher levels of c-MYC, cyclin D1, MCL-1 and CDK4. Notably, co-treatment with JQ1 and PS or ibrutinib also synergistically induced apoptosis of not only JeKo-1 but also JeKo-1/CZR cells (CI values 〈 1.0 by isobologram analysis). Taken together, these pre-clinical findings demonstrate synergistic preclinical activity of BET protein (BA) antagonist-based combinations with ibrutinib, panobinostat or carfilzomib against human MCL cells. Collectively, they also define potential BA-based combinations to overcome resistance to ibrutinib and carfilzomib in human MCL. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 126, No. 13 ( 2015-09-24), p. 1565-1574
    Abstract: BA reduces MYC, CDK4/6, nuclear RelA, and BTK expression and is synergistically lethal with ibrutinib in MCL cells. Cotreatment with BA and inhibitor of BCL2, CDK4/6, or histone deacetylases is synergistically lethal against ibrutinib-resistant MCL cells.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2612-2612
    Abstract: Mantle Cell Lymphoma (MCL) cells demonstrate genetic alterations involving cyclin D1, CDK4, CDKN2A, MYC, BCL2, as well as in the Toll-like receptor, B-cell receptor (BCR) and NFkB signaling genes. Increased BCR signaling and transcriptional activity of NFkB is documented in MCL cells. Ibrutinib (IB) is an inhibitor of Bruton's tyrosine kinase (BTK) clinically effective against MCL, but long-term durable remissions and cure following IB treatment remains uncertain. We have previously reported that treatment with the BET protein bromodomain antagonist (BA) JQ1 disrupts the binding of the BET-protein BRD4 to the enhancers and promoters and inhibits the mRNA and protein levels of the oncogenes MYC, BCL-2 and CDK4/6 in MCL cells. BRD4 has also been shown to be essential for NFkB activity. Consistent with this, here, we determined that treatment with JQ1 (250 to 2000 nM) reduced the nuclear localization of RelA and attenuated the expression of the NFkB target genes, including TNFα, TNFAIP3 (A20), c-FLIP, cIAP2, XIAP and Bcl-xL in the cultured MCL Mino and MO2058 cells. Notably, JQ1 treatment also reduced the mRNA and protein levels of BTK, along with inhibition of the levels of p-BTK, p-PLCγ2 and p-AKT levels in the MCL cells. Concomitantly, JQ1 dose-dependently inhibited cell growth and exerted lethal activity against cultured (MO2058 and Mino, Z138 and JeKo-1) and patient-derived, primary MCL (pMCL) cells, while relatively sparing normal CD19+ B cells and CD34+ bone marrow progenitor cells. Importantly, co-treatment with JQ1 and IB also markedly inhibited the nuclear RelA levels and attenuated the levels of p-BTK and BTK and reduced NFkB target gene expressions. Co-treatment with JQ1 and IB also synergistically induced apoptosis of the cultured and pMCL cells (CI values & lt; 1.0 by isobologram analysis). Following tail-vein infusion and engraftment of the Mino cells in the bone marrow and spleen of NOD/SCID mice, co-treatment with JQ1 (50 mg/kg/day, IP) and IB (25 mg/kg/day IP) versus treatment with each agent alone significantly improved the median survival of the mice (p & lt; 0.01). We isolated in vitro IB-resistant Mino/IR cells ( & gt; 20-fold resistant), following continuous exposure to IB. As compared to the parental control Mino, Mino/IR cells expressed markedly higher levels of AKT, CDK6, BCL2, Bcl-xL, and XIAP. JQ1 treatment (1.0 μM) was able to inhibit the growth and induce apoptosis of Mino/IR cells. Notably, co-treatment with JQ1 and panobinostat (pan-histone deacetylase inhibitor) or palbociclib (CDK4/6 inhibitor) or ABT199 (BCL2 antagonist), or carfilzomib (CZ) (proteasome inhibitor) synergistically induced apoptosis of not only Mino but also Mino/IR cells. Taken together, these findings identify BA-based rational combinations that need to be further evaluated for their in vivo efficacy against IB-sensitive and IB-resistant MCL. Citation Format: Baohua Sun, Bhavin Shah, Warren Fiskus, Jun Qi, Santhana G. T. Devaraj, Swaminathan P. Iyer, Sunil Sharma, James E. Bradner, Youli Zu, Kapil N. Bhalla. BET-protein bromodomain antagonist-based combinations against ibrutinb-sensitive or resistant human Mantle Cell Lymphoma cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2612. doi:10.1158/1538-7445.AM2015-2612
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 3686-3686
    Abstract: The BET protein BRD4 binds to acetylated lysines on the histone proteins, recruits pTEFb kinase, a complex of cyclin T and CDK9, to gene promoters to phosphorylate serine 2 of the C-terminus of RNA pol (RNAP) II. This promotes the pause release of RNAP II, with the elongation and expression of the mRNA transcripts of several oncogenes as well as their target genes, which are essential for the growth and survival of AML cells. We have recently determined that the BET protein antagonist JQ1, which disrupts the binding of BRD4 to acetylated lysines, potently induces apoptosis in cultured (OCI-AML3 and MOLM13) and patient-derived primary AML blast progenitor cells. JQ1 treatment also improved the survival of the immune-depleted mice engrafted with MOLM13 or primary AML cells. The lethal activity of JQ1 is associated with marked induction of hexamethylene bisacetamide (HMBA)-inducible protein 1 (HEXIM1) and p21, while MYC, BCL2 and CDK4/6 expressions are concomitantly downregulated in the cultured and primary AML cells. HEXIM1 binds and sequesters pTEFb, thereby inhibiting its kinase activity for RNAP II. To determine the role of HEXIM1 induction on the lethal effects of JQ1, we achieved a stable lentivirus mediated knockdown (KD) of the mRNA and protein expressions of HEXIM1 by approximately 90% in OCI-AML3 and MOLM13 cells. There was no significant difference in the protein expressions of BRD4, MYC and BCL2, or of cyclin T and CDK9 levels in the nuclear fraction in the AML cells with HEXIM1 knockdown (HKD) versus the AML cells expressing non-targeted (NT) shRNA. However, in the HKD cells, higher levels of the complex of cyclin T with CDK9 as pTEFb were detected in the immunoprecipitates of cyclin T. As compared to the NT AML, JQ1 (250 to 2000 nM)-induced HEXIM1 and apoptosis was reduced in HKD AML cells. This was associated with abrogation of JQ1-mediated HEXIM1 and p21 induction in the HKD AML cells. Similarly, HMBA (5 mM)-induced HEXIM1 and apoptosis was also inhibited in the HKD versus NT AML cells. Co-treatment with JQ1 and pan-HDAC inhibitor panobinostat (PS) synergistically induced apoptosis of cultured (MOLM13 and OCI-AML3) and primary AML blast progenitor cells, as well as conferred superior in vivo survival on the mice engrafted with cultured and primary AML blast progenitor cells. Importantly, co-treatment with JQ1 and PS also synergistically induced apoptosis of the cultured HKD AML cells. These findings indicate that HEXIM1 induction is a biomarker and plays a mechanistic role in the lethal activity of BRD4 antagonist against human AML cells. Citation Format: Santhana G. T. Devaraj, Warren Fiskus, Sunil Sharma, Jun Qi, Bhavin Shah, Leasha J. Schaub, Melissa Rodriguez, Ka Liu, Swaminathan P. Iyer, James E. Bradner, Kapil N. Bhalla. HEXIM1 induction exerts a mechanistic role and is a biomarker of lethal activity of BRD4 antagonist against human AML cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3686. doi:10.1158/1538-7445.AM2014-3686
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Virology, Elsevier BV, Vol. 366, No. 2 ( 2007-09), p. 277-292
    Type of Medium: Online Resource
    ISSN: 0042-6822
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2007
    detail.hit.zdb_id: 1471925-3
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal of Biological Chemistry, Elsevier BV, Vol. 282, No. 44 ( 2007-11), p. 32208-32221
    Type of Medium: Online Resource
    ISSN: 0021-9258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2007
    detail.hit.zdb_id: 2141744-1
    detail.hit.zdb_id: 1474604-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...