Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 12(6): 1560-1579, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35311997

ABSTRACT

Pharmacologic inhibition of epigenetic enzymes can have therapeutic benefit against hematologic malignancies. In addition to affecting tumor cell growth and proliferation, these epigenetic agents may induce antitumor immunity. Here, we discovered a novel immunoregulatory mechanism through inhibition of histone deacetylases (HDAC). In models of acute myeloid leukemia (AML), leukemia cell differentiation and therapeutic benefit mediated by the HDAC inhibitor (HDACi) panobinostat required activation of the type I interferon (IFN) pathway. Plasmacytoid dendritic cells (pDC) produced type I IFN after panobinostat treatment, through transcriptional activation of IFN genes concomitant with increased H3K27 acetylation at these loci. Depletion of pDCs abrogated panobinostat-mediated induction of type I IFN signaling in leukemia cells and impaired therapeutic efficacy, whereas combined treatment with panobinostat and IFNα improved outcomes in preclinical models. These discoveries offer a new therapeutic approach for AML and demonstrate that epigenetic rewiring of pDCs enhances antitumor immunity, opening the possibility of exploiting this approach for immunotherapies. SIGNIFICANCE: We demonstrate that HDACis induce terminal differentiation of AML through epigenetic remodeling of pDCs, resulting in production of type I IFN that is important for the therapeutic effects of HDACis. The study demonstrates the important functional interplay between the immune system and leukemias in response to HDAC inhibition. This article is highlighted in the In This Issue feature, p. 1397.


Subject(s)
Leukemia, Myeloid, Acute , Cell Differentiation , Dendritic Cells , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Panobinostat/pharmacology
2.
Cancer Immunol Res ; 10(1): 87-107, 2022 01.
Article in English | MEDLINE | ID: mdl-34782346

ABSTRACT

Targeting chromatin binding proteins and modifying enzymes can concomitantly affect tumor cell proliferation and survival, as well as enhance antitumor immunity and augment cancer immunotherapies. By screening a small-molecule library of epigenetics-based therapeutics, BET (bromo- and extra-terminal domain) inhibitors (BETi) were identified as agents that sensitize tumor cells to the antitumor activity of CD8+ T cells. BETi modulated tumor cells to be sensitized to the cytotoxic effects of the proinflammatory cytokine TNF. By preventing the recruitment of BRD4 to p65-bound cis-regulatory elements, BETi suppressed the induction of inflammatory gene expression, including the key NF-κB target genes BIRC2 (cIAP1) and BIRC3 (cIAP2). Disruption of prosurvival NF-κB signaling by BETi led to unrestrained TNF-mediated activation of the extrinsic apoptotic cascade and tumor cell death. Administration of BETi in combination with T-cell bispecific antibodies (TCB) or immune-checkpoint blockade increased bystander killing of tumor cells and enhanced tumor growth inhibition in vivo in a TNF-dependent manner. This novel epigenetic mechanism of immunomodulation may guide future use of BETi as adjuvants for immune-oncology agents.


Subject(s)
Antineoplastic Agents/administration & dosage , Colorectal Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
Mol Cell ; 81(10): 2183-2200.e13, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34019788

ABSTRACT

To separate causal effects of histone acetylation on chromatin accessibility and transcriptional output, we used integrated epigenomic and transcriptomic analyses following acute inhibition of major cellular lysine acetyltransferases P300 and CBP in hematological malignancies. We found that catalytic P300/CBP inhibition dynamically perturbs steady-state acetylation kinetics and suppresses oncogenic transcriptional networks in the absence of changes to chromatin accessibility. CRISPR-Cas9 screening identified NCOR1 and HDAC3 transcriptional co-repressors as the principal antagonists of P300/CBP by counteracting acetylation turnover kinetics. Finally, deacetylation of H3K27 provides nucleation sites for reciprocal methylation switching, a feature that can be exploited therapeutically by concomitant KDM6A and P300/CBP inhibition. Overall, this study indicates that the steady-state histone acetylation-methylation equilibrium functions as a molecular rheostat governing cellular transcription that is amenable to therapeutic exploitation as an anti-cancer regimen.


Subject(s)
Biocatalysis , Histones/metabolism , Oncogenes , Transcription, Genetic , p300-CBP Transcription Factors/metabolism , Acetylation , Cell Line , Chromatin/metabolism , Co-Repressor Proteins/metabolism , Conserved Sequence , Evolution, Molecular , Gene Regulatory Networks , Genome , Histone Deacetylases/metabolism , Humans , Kinetics , Methylation , Models, Biological , RNA Polymerase II/metabolism
4.
Sci Adv ; 6(18)2020 05.
Article in English | MEDLINE | ID: mdl-32917631

ABSTRACT

The RNA polymerase II (POLII)-driven transcription cycle is tightly regulated at distinct checkpoints by cyclin-dependent kinases (CDKs) and their cognate cyclins. The molecular events underpinning transcriptional elongation, processivity, and the CDK-cyclin pair(s) involved remain poorly understood. Using CRISPR-Cas9 homology-directed repair, we generated analog-sensitive kinase variants of CDK12 and CDK13 to probe their individual and shared biological and molecular roles. Single inhibition of CDK12 or CDK13 induced transcriptional responses associated with cellular growth signaling pathways and/or DNA damage, with minimal effects on cell viability. In contrast, dual kinase inhibition potently induced cell death, which was associated with extensive genome-wide transcriptional changes including widespread use of alternative 3' polyadenylation sites. At the molecular level, dual kinase inhibition resulted in the loss of POLII CTD phosphorylation and greatly reduced POLII elongation rates and processivity. These data define substantial redundancy between CDK12 and CDK13 and identify both as fundamental regulators of global POLII processivity and transcription elongation.

5.
Methods Mol Biol ; 1725: 201-227, 2018.
Article in English | MEDLINE | ID: mdl-29322420

ABSTRACT

This chapter details a compendium of protocols that collectively enable the reader to perform a pooled shRNA and/or CRISPR screen-with methods to identify and validate positive controls and subsequent hits; establish a viral titer in the cell line of choice; create and screen libraries, sequence strategies, and bioinformatics resources to analyze outcomes. Collectively, this provides an overarching resource from the start to finish of a screening project, making this technology possible in all laboratories.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Library , High-Throughput Screening Assays , Lentivirus/genetics , RNA, Small Interfering/genetics , Computational Biology , HEK293 Cells , Humans
6.
Cell Rep ; 18(9): 2162-2174, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28249162

ABSTRACT

BET inhibitors (BETi) target bromodomain-containing proteins and are currently being evaluated as anti-cancer agents. We find that maximal therapeutic effects of BETi in a Myc-driven B cell lymphoma model required an intact host immune system. Genome-wide analysis of the BETi-induced transcriptional response identified the immune checkpoint ligand Cd274 (Pd-l1) as a Myc-independent, BETi target-gene. BETi directly repressed constitutively expressed and interferon-gamma (IFN-γ) induced CD274 expression across different human and mouse tumor cell lines and primary patient samples. Mechanistically, BETi decreased Brd4 occupancy at the Cd274 locus without any change in Myc occupancy, resulting in transcriptional pausing and rapid loss of Cd274 mRNA production. Finally, targeted inhibition of the PD-1/PD-L1 axis by combining anti-PD-1 antibodies and the BETi JQ1 caused synergistic responses in mice bearing Myc-driven lymphomas. Our data uncover an interaction between BETi and the PD-1/PD-L1 immune-checkpoint and provide mechanistic insight into the transcriptional regulation of CD274.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/immunology , Immune System/immunology , Nuclear Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , Transcription Factors/immunology , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/immunology , Humans , Interferon-gamma/immunology , Ligands , Lymphoma, B-Cell/immunology , Mice , Proto-Oncogene Proteins c-myc/immunology , RNA, Messenger/immunology , Transcription, Genetic/immunology
7.
Mol Cancer Ther ; 15(9): 2030-41, 2016 09.
Article in English | MEDLINE | ID: mdl-27406984

ABSTRACT

Targeting BET bromodomain proteins using small molecules is an emerging anticancer strategy with clinical evaluation of at least six inhibitors now underway. Although MYC downregulation was initially proposed as a key mechanistic property of BET inhibitors, recent evidence suggests that additional antitumor activities are important. Using the Eµ-Myc model of B-cell lymphoma, we demonstrate that BET inhibition with JQ1 is a potent inducer of p53-independent apoptosis that occurs in the absence of effects on Myc gene expression. JQ1 skews the expression of proapoptotic (Bim) and antiapoptotic (BCL-2/BCL-xL) BCL-2 family members to directly engage the mitochondrial apoptotic pathway. Consistent with this, Bim knockout or Bcl-2 overexpression inhibited apoptosis induction by JQ1. We identified lymphomas that were either intrinsically resistant to JQ1-mediated death or acquired resistance following in vivo exposure. Strikingly, in both instances BCL-2 was strongly upregulated and was concomitant with activation of RAS pathways. Eµ-Myc lymphomas engineered to express activated Nras upregulated BCL-2 and acquired a JQ1 resistance phenotype. These studies provide important information on mechanisms of apoptosis induction and resistance to BET-inhibition, while providing further rationale for the translation of BET inhibitors in aggressive B-cell lymphomas. Mol Cancer Ther; 15(9); 2030-41. ©2016 AACR.


Subject(s)
Apoptosis/genetics , Azepines/pharmacology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Triazoles/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/genetics , Genes, myc , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Mice , Multigene Family , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
8.
Blood ; 126(21): 2392-403, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26447190

ABSTRACT

Histone deacetylase (HDAC) inhibitors (HDACis) have demonstrated activity in hematological and solid malignancies. Vorinostat, romidepsin, belinostat, and panobinostat are Food and Drug Administration-approved for hematological malignancies and inhibit class II and/or class I HDACs, including HDAC1, 2, 3, and 6. We combined genetic and pharmacological approaches to investigate whether suppression of individual or multiple Hdacs phenocopied broad-acting HDACis in 3 genetically distinct leukemias and lymphomas. Individual Hdacs were depleted in murine acute myeloid leukemias (MLL-AF9;Nras(G12D); PML-RARα acute promyelocytic leukemia [APL] cells) and Eµ-Myc lymphoma in vitro and in vivo. Strikingly, Hdac3-depleted cells were selected against in competitive assays for all 3 tumor types. Decreased proliferation following Hdac3 knockdown was not prevented by BCL-2 overexpression, caspase inhibition, or knockout of Cdkn1a in Eµ-Myc lymphoma, and depletion of Hdac3 in vivo significantly reduced tumor burden. Interestingly, APL cells depleted of Hdac3 demonstrated a more differentiated phenotype. Consistent with these genetic studies, the HDAC3 inhibitor RGFP966 reduced proliferation of Eµ-Myc lymphoma and induced differentiation in APL. Genetic codepletion of Hdac1 with Hdac2 was pro-apoptotic in Eµ-Myc lymphoma in vitro and in vivo and was phenocopied by the HDAC1/2-specific agent RGFP233. This study demonstrates the importance of HDAC3 for the proliferation of leukemia and lymphoma cells, suggesting that HDAC3-selective inhibitors could prove useful for the treatment of hematological malignancies. Moreover, our results demonstrate that codepletion of Hdac1 with Hdac2 mediates a robust pro-apoptotic response. Our integrated genetic and pharmacological approach provides important insights into the individual or combinations of HDACs that could be prioritized for targeting in a range of hematological malignancies.


Subject(s)
Histone Deacetylases/metabolism , Leukemia, Promyelocytic, Acute/enzymology , Leukemia, Promyelocytic, Acute/genetics , Lymphoma/enzymology , Lymphoma/genetics , Animals , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/pathology , Lymphoma/drug therapy , Lymphoma/pathology , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
Mol Cancer Ther ; 12(12): 2709-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24092806

ABSTRACT

Histone deacetylase inhibitors (HDACi) are anticancer agents that induce hyperacetylation of histones, resulting in chromatin remodeling and transcriptional changes. In addition, nonhistone proteins, such as the chaperone protein Hsp90, are functionally regulated through hyperacetylation mediated by HDACis. Histone acetylation is thought to be primarily regulated by HDACs 1, 2, and 3, whereas the acetylation of Hsp90 has been proposed to be specifically regulated through HDAC6. We compared the molecular and biologic effects induced by an HDACi with broad HDAC specificity (vorinostat) with agents that predominantly inhibited selected class I HDACs (MRLB-223 and romidepsin). MRLB-223, a potent inhibitor of HDACs 1 and 2, killed tumor cells using the same apoptotic pathways as the HDAC 1, 2, 3, 6, and 8 inhibitor vorinostat. However, vorinostat induced histone hyperacetylation and killed tumor cells more rapidly than MRLB-223 and had greater therapeutic efficacy in vivo. FDCP-1 cells dependent on the Hsp90 client protein Bcr-Abl for survival, were killed by all HDACis tested, concomitant with caspase-dependent degradation of Bcr-Abl. These studies provide evidence that inhibition of HDAC6 and degradation of Bcr-Abl following hyperacetylation of Hsp90 is likely not a major mechanism of action of HDACis as had been previously posited.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Acetylation/drug effects , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Enzyme Activation/drug effects , Fusion Proteins, bcr-abl/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Lymphoma/drug therapy , Lymphoma/metabolism , Lymphoma/mortality , Lymphoma/pathology , Mice , Vorinostat , Xenograft Model Antitumor Assays
10.
Cancer Res ; 73(24): 7265-76, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24158093

ABSTRACT

Cell-intrinsic effects such as induction of apoptosis and/or inhibition of cell proliferation have been proposed as the major antitumor responses to histone deacetylase inhibitors (HDACi). These compounds can also mediate immune-modulatory effects that may contribute to their anticancer effects. However, HDACi can also induce anti-inflammatory, and potentially immunosuppressive, outcomes. We therefore sought to clarify the role of the immune system in mediating the efficacy of HDACi in a physiologic setting, using preclinical, syngeneic murine models of hematologic malignancies and solid tumors. We showed an intact immune system was required for the robust anticancer effects of the HDACi vorinostat and panobinostat against a colon adenocarcinoma and two aggressive models of leukemia/lymphoma. Importantly, although HDACi-treated immunocompromised mice bearing established lymphoma succumbed to disease significantly earlier than tumor bearing, HDACi-treated wild-type (WT) mice, treatment with the conventional chemotherapeutic etoposide equivalently enhanced the survival of both strains. IFN-γ and tumor cell signaling through IFN-γR were particularly important for the anticancer effects of HDACi, and vorinostat and IFN-γ acted in concert to enhance the immunogenicity of tumor cells. Furthermore, we show that a combination of vorinostat with α-galactosylceramide (α-GalCer), an IFN-γ-inducing agent, was significantly more potent against established lymphoma than vorinostat treatment alone. Intriguingly, B cells, but not natural killer cells or CD8(+) T cells, were implicated as effectors of the vorinostat antitumor immune response. Together, our data suggest HDACi are immunostimulatory during cancer treatment and that combinatorial therapeutic regimes with immunotherapies should be considered in the clinic.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Immune System/physiology , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/immunology , Receptors, Interferon/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Animals , Apoptosis/drug effects , Apoptosis/immunology , Cell Growth Processes/drug effects , Cell Growth Processes/immunology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Immune System/drug effects , Immunotherapy , Leukemia, Lymphoid/drug therapy , Leukemia, Lymphoid/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Interferon gamma Receptor
11.
Cancer Res ; 71(10): 3603-15, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21398407

ABSTRACT

The concept of personalized anticancer therapy is based on the use of targeted therapeutics through in-depth knowledge of the molecular mechanisms of action of these agents when used alone and in combination. We have identified the apoptotic proteins and pathways necessary for synergistic tumor cell apoptosis and in vivo antitumor responses seen when the HDAC inhibitor vorinostat is combined with the BH3-mimetic ABT-737 in lymphomas overexpressing Bcl-2. Vorinostat "primes" tumors overexpressing Bcl-2 for rapid ABT-737-mediated apoptosis by inducing expression of the BH3-only gene bmf. Moreover, these synergistic effects of vorinostat/ABT-737 were blunted in cells with an inactive p53 pathway or in cells lacking expression of the p53 target gene, noxa. These studies show the important and complex functional interaction between specific proapoptotic BH3-only proteins and the BH3-mimetic compound ABT-737 and provide the most comprehensive functional link between tumor genotype and the apoptotic and therapeutic effects of HDACi combined with ABT-737.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Animals , Apoptosis , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Female , Genes, p53 , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nitrophenols/pharmacology , Peptide Fragments/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins/metabolism , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/genetics , Vorinostat
12.
J Immunol ; 185(1): 367-75, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530266

ABSTRACT

Asymmetric cell division is a potential means by which cell fate choices during an immune response are orchestrated. Defining the molecular mechanisms that underlie asymmetric division of T cells is paramount for determining the role of this process in the generation of effector and memory T cell subsets. In other cell types, asymmetric cell division is regulated by conserved polarity protein complexes that control the localization of cell fate determinants and spindle orientation during division. We have developed a tractable, in vitro model of naive CD8(+) T cells undergoing initial division while attached to dendritic cells during Ag presentation to investigate whether similar mechanisms might regulate asymmetric division of T cells. Using this system, we show that direct interactions with APCs provide the cue for polarization of T cells. Interestingly, the immunological synapse disseminates before division even though the T cells retain contact with the APC. The cue from the APC is translated into polarization of cell fate determinants via the polarity network of the Par3 and Scribble complexes, and orientation of the mitotic spindle during division is orchestrated by the partner of inscuteable/G protein complex. These findings suggest that T cells have selectively adapted a number of evolutionarily conserved mechanisms to generate diversity through asymmetric cell division.


Subject(s)
Antigen Presentation/immunology , Cell Division/immunology , Conserved Sequence/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Adhesion/immunology , Cell Polarity/immunology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocyte Subsets/metabolism
13.
Blood ; 114(2): 380-93, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19383971

ABSTRACT

LAQ824 and LBH589 (panobinostat) are histone deacetylase inhibitors (HDACi) developed as cancer therapeutics and we have used the Emu-myc lymphoma model to identify the molecular events required for their antitumor effects. Induction of tumor cell death was necessary for these agents to mediate therapeutic responses in vivo and both HDACi engaged the intrinsic apoptotic cascade that did not require p53. Death receptor pathway blockade had no effect on the therapeutic activities of LAQ824 and LBH589; however, overexpression of Bcl-2 or Bcl-X(L) protected lymphoma cells from HDACi-induced killing and suppressed their therapeutic activities. Deletion of Apaf-1 or Caspase-9 delayed HDACi-induced lymphoma killing in vitro and in vivo, associated with suppression of many biochemical indicators of apoptosis, but did not provide long-term resistance to these agents and failed to inhibit their therapeutic activities. Emu-myc lymphomas lacking a functional apoptosome displayed morphologic and biochemical features of autophagy after treatment with LAQ824 and LBH589, indicating that, in the absence of a complete intrinsic apoptosis pathway involving apoptosome formation, these HDACi can still mediate a therapeutic response. Our data indicate that damage to the mitochondria is the key event necessary for LAQ824 and LBH589 to mediate tumor cell death and a robust therapeutic response.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Lymphoma/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Histone Deacetylases/metabolism , Humans , Indoles , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neoplasm Transplantation , Panobinostat , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Survival Rate , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Blood ; 113(9): 1982-91, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19060243

ABSTRACT

The apoptotic and therapeutic activities of the histone deacetylase inhibitor (HDACi) vorinostat are blocked by overexpression of Bcl-2 or Bcl-X(L). Herein, we used the small molecule inhibitor ABT-737 to restore sensitivity of Emu-myc lymphomas overexpressing Bcl-2 or Bcl-X(L) to vorinostat and valproic acid (VPA). Combining low-dose ABT-737 with vorinostat or VPA resulted in synergistic apoptosis of these cells. ABT-737 was ineffective against Emu-myc/Mcl-1 and Emu-myc/A1 cells either as a single agent or in combination with HDACi. However, in contrast to the reported binding specificity data, Emu-myc/Bcl-w lymphomas were insensitive to ABT-737 used alone or in combination with HDACi, indicating that the regulatory activity of ABT-737 is restricted to Bcl-2 and Bcl-X(L). Emu-myc lymphomas that expressed Bcl-2 throughout the tumorigenesis process were especially sensitive to ABT-737, while those forced to overexpress Mcl-1 were not. This supports the notion that tumor cells "addicted" to ABT-737 target proteins (ie, Bcl-2 or Bcl-X(L)) are likely to be the most sensitive target cell population. Our studies provide important preclinical data on the binding specificity of ABT-737 and its usefulness against primary hematologic malignancies when used as a single agent and in combination with HDACi.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/administration & dosage , Histone Deacetylase Inhibitors , Lymphoma/drug therapy , Nitrophenols/pharmacology , Sulfonamides/pharmacology , Animals , Biphenyl Compounds/administration & dosage , Cell Survival/drug effects , Drug Delivery Systems , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Synergism , Genes, bcl-2 , Genes, myc , Hydroxamic Acids/administration & dosage , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitrophenols/administration & dosage , Piperazines/administration & dosage , Piperazines/pharmacology , Substrate Specificity , Sulfonamides/administration & dosage , Vorinostat
15.
Mol Cancer Ther ; 7(5): 1066-79, 2008 May.
Article in English | MEDLINE | ID: mdl-18483296

ABSTRACT

Histone deacetylase inhibitors (HDACi) are compounds that target the epigenome and cause tumor cell-selective apoptosis. A large number of these agents that have different chemical structures and can target multiple HDACs are being testing in clinical trials and vorinostat is now an approved drug for the treatment of cutaneous T-cell lymphoma. Although these agents are showing promise for the treatment of hematologic malignancies, it is possible that different drugs may have different mechanistic, biological, and therapeutic activities. When comparing an HDACi belonging to the hydroxamic acid class of compounds (vorinostat) with a cyclic tetrapeptide (romidepsin), we showed that these agents regulate the expression of a common set of cellular genes, but certain genes specifically responded to each agent. Using the Emu-myc mouse model of B-cell lymphoma, we showed previously that overexpression of the prosurvival proteins Bcl-2 and Bcl-XL inhibited the apoptotic and therapeutic activities of the vorinostat. Herein, we compared and contrasted the apoptotic-inducing activities of the hydroxamic acid oxamflatin with romidepsin. Like vorinostat, oxamflatin was unable to kill lymphomas overexpressing Bcl-2 and Bcl-XL, indicating that these proteins can generally protect cells against this class of HDACi. In contrast, romidepsin was able to induce apoptosis in lymphomas overexpressing Bcl-2 with delayed kinetics of cell death and could mediate therapeutic responses against these lymphomas. However, romidepsin was inactive when Bcl-XL was overexpressed. These data provide strong support that HDACi of different chemical classes may have subtle yet potentially important differences in their molecular and biological activities.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis , Depsipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Animals , Antibiotics, Antineoplastic/therapeutic use , Depsipeptides/therapeutic use , Dose-Response Relationship, Drug , Enzyme Inhibitors/therapeutic use , Hydroxamic Acids/pharmacology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/enzymology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-X Protein/metabolism
16.
Immunity ; 20(2): 153-65, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14975238

ABSTRACT

To determine the importance of suppressor of cytokine signaling-3 (SOCS3) in the regulation of hematopoietic growth factor signaling generally, and of G-CSF-induced cellular responses specifically, we created mice in which the Socs3 gene was deleted in all hematopoietic cells. Although normal until young adulthood, these mice then developed neutrophilia and a spectrum of inflammatory pathologies. When stimulated with G-CSF in vitro, SOCS3-deficient cells of the neutrophilic granulocyte lineage exhibited prolonged STAT3 activation and enhanced cellular responses to G-CSF, including an increase in cloning frequency, survival, and proliferative capacity. Consistent with the in vitro findings, mutant mice injected with G-CSF displayed enhanced neutrophilia, progenitor cell mobilization, and splenomegaly, but unexpectedly also developed inflammatory neutrophil infiltration into multiple tissues and consequent hind-leg paresis. We conclude that SOCS3 is a key negative regulator of G-CSF signaling in myeloid cells and that this is of particular significance during G-CSF-driven emergency granulopoiesis.


Subject(s)
Granulocyte Colony-Stimulating Factor/immunology , Granulocytes/immunology , Inflammation/pathology , Repressor Proteins/immunology , Transcription Factors/immunology , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/drug effects , Granulocytes/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Inflammation/immunology , Mice , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/drug effects , Trans-Activators/immunology , Trans-Activators/metabolism , Transcription Factors/deficiency
17.
Immunol Cell Biol ; 80(3): 231-40, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12067410

ABSTRACT

The haematopoietic-specific RhoGTPase, Rac2, has been indirectly implicated in T-lymphocyte development and function, and as a pivotal regulator of T Helper 1 (T(H)1) responses. In other haematopoietic cells it regulates cytoskeletal rearrangement downstream of extracellular signals. Here we demonstrate that Rac2 deficiency results in an abnormal distribution of T lymphocytes in vivo and defects in T-lymphocyte migration and filamentous actin generation in response to chemoattractants in vitro. To investigate the requirement for Rac2 in IFN-gamma production and TH1 responses in vivo, Rac2-deficient mice were challenged with Leishmania major and immunized with ovalbumin-expressing cytomegalovirus. Despite a minor skewing towards a T(H)2 phenotype, Rac2-deficient mice displayed no increased susceptibility to L. major infection. Cytotoxic T-lymphocyte responses to cytomegalovirus and ovalbumin were also normal. Although Rac2 is required for normal T-lymphocyte migration, its role in the generation of T(H)1 responses to infection in vivo is largely redundant.


Subject(s)
Chemotaxis, Leukocyte , Peptidylprolyl Isomerase , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , rac GTP-Binding Proteins/physiology , Actins/metabolism , Animals , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Chemokine CCL19 , Chemokines, CC , Cytomegalovirus/immunology , Fibronectins/metabolism , Immunoglobulin G/classification , Immunoglobulin G/immunology , Immunophilins/metabolism , Integrin beta1/metabolism , Leishmania major/cytology , Leishmania major/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Th1 Cells/chemistry , rac GTP-Binding Proteins/deficiency , RAC2 GTP-Binding Protein
18.
J Immunol ; 168(7): 3376-86, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11907095

ABSTRACT

We have defined roles for the hemopoietic-specific Rho guanosine triphosphatase, Rac2, in B lymphocyte development and function through examination of rac2(-/-) mice. Rac2-deficient mice displayed peripheral blood B lymphocytosis and marked reductions in peritoneal cavity B-1a lymphocytes, marginal zone B lymphocytes, and IgM-secreting plasma cells as well as reduced concentrations of serum IgM and IgA. The rac2(-/-) B lymphocytes exhibited reduced calcium flux following coligation of B cell AgR and CD19 and reduced chemotaxis in chemokine gradients. T cell-independent responses to DNP-dextran were of reduced magnitude, but normal kinetics, in rac2(-/-) mice, while T-dependent responses to nitrophenyl-keyhole limpet hemocyanin were subtly abnormal. Rac2 is therefore an essential element in regulating B lymphocyte functions and maintaining B lymphocyte populations in vivo.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Chemotaxis, Leukocyte/immunology , Receptors, Antigen, B-Cell/metabolism , rac GTP-Binding Proteins/physiology , Actins/antagonists & inhibitors , Actins/metabolism , Animals , Antibody-Producing Cells/pathology , Antigens, CD19/immunology , Antigens, CD19/metabolism , Antigens, T-Independent/pharmacology , B-Lymphocyte Subsets/pathology , Calcium/antagonists & inhibitors , Calcium/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Migration Inhibition , Chemokines/pharmacology , Down-Regulation/genetics , Down-Regulation/immunology , Haptens , Hemocyanins/pharmacology , Immunoglobulin A/blood , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/blood , Ligands , Lymphocyte Activation/genetics , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, B-Cell/physiology , Up-Regulation/genetics , Up-Regulation/immunology , rac GTP-Binding Proteins/deficiency , rac GTP-Binding Proteins/genetics , RAC2 GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...