Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Clin Invest ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38875287

ABSTRACT

It is unknown which post-transcriptional regulatory mechanisms are required for oncogenic competence. Here, we show that the LIN28 family of RNA-binding proteins (RBPs), which facilitate post-transcriptional RNA metabolism within ribonucleoprotein networks, are essential for the initiation of diverse oncotypes of hepatocellular carcinoma (HCC). In HCC models driven by NRASG12V/Tp53, CTNNB1/YAP/Tp53, or AKT/Tp53, mice without Lin28a and Lin28b were markedly impaired in cancer initiation. We biochemically defined an oncofetal regulon of 15 factors connected to Lin28 through direct mRNA and protein interactions. Interestingly, all were RBPs and only 1 of 15 is a Let-7 target. Polysome profiling and reporter assays showed that LIN28B directly increased the translation of 8 of these 15 RBPs. As expected, overexpression of LIN28B and IGFBP1-3 were able to genetically rescue cancer initiation. Using this platform to probe components downstream of LIN28, we found that 8 target RBPs were able to restore NRASG12V/Tp53 cancer formation in Lin28a/b deficient mice. Furthermore, these LIN28B targets promote cancer initiation through an increase in protein synthesis. LIN28B, central to an RNP regulon that increases translation of RBPs, is important for tumor initiation in the liver.

2.
Mob DNA ; 14(1): 18, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37990347

ABSTRACT

In November 2022 the first Dark Genome Symposium was held in Boston, USA. The meeting was hosted by Rome Therapeutics and Enara Bio, two biotechnology companies working on translating our growing understanding of this vast genetic landscape into therapies for human disease. The spirit and ambition of the meeting was one of shared knowledge, looking to strengthen the network of researchers engaged in the field. The meeting opened with a welcome from Rosana Kapeller and Kevin Pojasek followed by a first session of field defining talks from key academics in the space. A series of panels, bringing together academia and industry views, were then convened covering a wide range of pertinent topics. Finally, Richard Young and David Ting gave their views on the future direction and promise for patient impact inherent in the growing understanding of the Dark Genome.

3.
Mol Cancer Ther ; 16(11): 2586-2597, 2017 11.
Article in English | MEDLINE | ID: mdl-28835384

ABSTRACT

The EZH2 small-molecule inhibitor tazemetostat (EPZ-6438) is currently being evaluated in phase II clinical trials for the treatment of non-Hodgkin lymphoma (NHL). We have previously shown that EZH2 inhibitors display an antiproliferative effect in multiple preclinical models of NHL, and that models bearing gain-of-function mutations in EZH2 were consistently more sensitive to EZH2 inhibition than lymphomas with wild-type (WT) EZH2 Here, we demonstrate that cell lines bearing EZH2 mutations show a cytotoxic response, while cell lines with WT-EZH2 show a cytostatic response and only tumor growth inhibition without regression in a xenograft model. Previous work has demonstrated that cotreatment with tazemetostat and glucocorticoid receptor agonists lead to a synergistic antiproliferative effect in both mutant and wild-type backgrounds, which may provide clues to the mechanism of action of EZH2 inhibition in WT-EZH2 models. Multiple agents that inhibit the B-cell receptor pathway (e.g., ibrutinib) were found to have synergistic benefit when combined with tazemetostat in both mutant and WT-EZH2 backgrounds of diffuse large B-cell lymphomas (DLBCL). The relationship between B-cell activation and EZH2 inhibition is consistent with the proposed role of EZH2 in B-cell maturation. To further support this, we observe that cell lines treated with tazemetostat show an increase in the B-cell maturation regulator, PRDM1/BLIMP1, and gene signatures corresponding to more advanced stages of maturation. These findings suggest that EZH2 inhibition in both mutant and wild-type backgrounds leads to increased B-cell maturation and a greater dependence on B-cell activation signaling. Mol Cancer Ther; 16(11); 2586-97. ©2017 AACR.


Subject(s)
Benzamides/administration & dosage , Enhancer of Zeste Homolog 2 Protein/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Pyrazoles/administration & dosage , Pyridones/administration & dosage , Pyrimidines/administration & dosage , Adenine/analogs & derivatives , Animals , B-Lymphocytes/drug effects , Biphenyl Compounds , Cell Proliferation/drug effects , DNA Methylation/drug effects , Drug Synergism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Morpholines , Mutation , Piperidines , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
Mol Cancer Ther ; 16(5): 850-860, 2017 05.
Article in English | MEDLINE | ID: mdl-28292935

ABSTRACT

The SWI/SNF complex is a major regulator of gene expression and is increasingly thought to play an important role in human cancer, as evidenced by the high frequency of subunit mutations across virtually all cancer types. We previously reported that in preclinical models, malignant rhabdoid tumors, which are deficient in the SWI/SNF core component INI1 (SMARCB1), are selectively killed by inhibitors of the H3K27 histone methyltransferase EZH2. Given the demonstrated antagonistic activities of the SWI/SNF complex and the EZH2-containing PRC2 complex, we investigated whether additional cancers with SWI/SNF mutations are sensitive to selective EZH2 inhibition. It has been recently reported that ovarian cancers with dual loss of the redundant SWI/SNF components SMARCA4 and SMARCA2 are characteristic of a rare rhabdoid-like subtype known as small-cell carcinoma of the ovary hypercalcemic type (SCCOHT). Here, we provide evidence that a subset of commonly used ovarian carcinoma cell lines were misdiagnosed and instead were derived from a SCCOHT tumor. We also demonstrate that tazemetostat, a potent and selective EZH2 inhibitor currently in phase II clinical trials, induces potent antiproliferative and antitumor effects in SCCOHT cell lines and xenografts deficient in both SMARCA2 and SMARCA4. These results exemplify an additional class of rhabdoid-like tumors that are dependent on EZH2 activity for survival. Mol Cancer Ther; 16(5); 850-60. ©2017 AACR.


Subject(s)
Carcinoma, Small Cell/drug therapy , DNA Helicases/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Rhabdoid Tumor/drug therapy , Transcription Factors/genetics , Animals , Carcinoma, Small Cell/diagnosis , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/genetics , Diagnosis, Differential , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone-Lysine N-Methyltransferase/genetics , Humans , Hypercalcemia/diagnosis , Hypercalcemia/drug therapy , Hypercalcemia/genetics , Hypercalcemia/pathology , Mice , Mutation , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Rhabdoid Tumor/diagnosis , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Xenograft Model Antitumor Assays
6.
PLoS One ; 11(7): e0158888, 2016.
Article in English | MEDLINE | ID: mdl-27391784

ABSTRACT

The catalytic activities of covalent and ATP-dependent chromatin remodeling are central to regulating the conformational state of chromatin and the resultant transcriptional output. The enzymes that catalyze these activities are often contained within multiprotein complexes in nature. Two such multiprotein complexes, the polycomb repressive complex 2 (PRC2) methyltransferase and the SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeler have been reported to act in opposition to each other during development and homeostasis. An imbalance in their activities induced by mutations/deletions in complex members (e.g. SMARCB1) has been suggested to be a pathogenic mechanism in certain human cancers. Here we show that preclinical models of synovial sarcoma-a cancer characterized by functional SMARCB1 loss via its displacement from the SWI/SNF complex through the pathognomonic SS18-SSX fusion protein-display sensitivity to pharmacologic inhibition of EZH2, the catalytic subunit of PRC2. Treatment with tazemetostat, a clinical-stage, selective and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity reverses a subset of synovial sarcoma gene expression and results in concentration-dependent cell growth inhibition and cell death specifically in SS18-SSX fusion-positive cells in vitro. Treatment of mice bearing either a cell line or two patient-derived xenograft models of synovial sarcoma leads to dose-dependent tumor growth inhibition with correlative inhibition of trimethylation levels of the EZH2-specific substrate, lysine 27 on histone H3. These data demonstrate a dependency of SS18-SSX-positive, SMARCB1-deficient synovial sarcomas on EZH2 enzymatic activity and suggests the potential utility of EZH2-targeted drugs in these genetically defined cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Sarcoma, Synovial/drug therapy , Animals , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Sarcoma, Synovial/genetics , Sarcoma, Synovial/metabolism , Xenograft Model Antitumor Assays
8.
J Med Chem ; 59(4): 1556-64, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26769278

ABSTRACT

Posttranslational methylation of histones plays a critical role in gene regulation. Misregulation of histone methylation can lead to oncogenic transformation. Enhancer of Zeste homologue 2 (EZH2) methylates histone 3 at lysine 27 (H3K27) and abnormal methylation of this site is found in many cancers. Tazemetostat, an EHZ2 inhibitor in clinical development, has shown activity in both preclinical models of cancer as well as in patients with lymphoma or INI1-deficient solid tumors. Herein we report the structure-activity relationships from identification of an initial hit in a high-throughput screen through selection of tazemetostat for clinical development. The importance of several methyl groups to the potency of the inhibitors is highlighted as well as the importance of balancing pharmacokinetic properties with potency.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histones/metabolism , Methylation/drug effects , Polycomb Repressive Complex 2/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Drug Discovery , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Polycomb Repressive Complex 2/metabolism , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/pharmacokinetics , Structure-Activity Relationship
9.
Nat Med ; 21(11): 1344-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26437366

ABSTRACT

The tumor suppressors BAP1 and ASXL1 interact to form a polycomb deubiquitinase complex that removes monoubiquitin from histone H2A lysine 119 (H2AK119Ub). However, BAP1 and ASXL1 are mutated in distinct cancer types, consistent with independent roles in regulating epigenetic state and malignant transformation. Here we demonstrate that Bap1 loss in mice results in increased trimethylated histone H3 lysine 27 (H3K27me3), elevated enhancer of zeste 2 polycomb repressive complex 2 subunit (Ezh2) expression, and enhanced repression of polycomb repressive complex 2 (PRC2) targets. These findings contrast with the reduction in H3K27me3 levels seen with Asxl1 loss. Conditional deletion of Bap1 and Ezh2 in vivo abrogates the myeloid progenitor expansion induced by Bap1 loss alone. Loss of BAP1 results in a marked decrease in H4K20 monomethylation (H4K20me1). Consistent with a role for H4K20me1 in the transcriptional regulation of EZH2, expression of SETD8-the H4K20me1 methyltransferase-reduces EZH2 expression and abrogates the proliferation of BAP1-mutant cells. Furthermore, mesothelioma cells that lack BAP1 are sensitive to EZH2 pharmacologic inhibition, suggesting a novel therapeutic approach for BAP1-mutant malignancies.


Subject(s)
Gene Expression Regulation, Neoplastic , Histones/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Animals , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Knockout Techniques , HEK293 Cells , Histone Code , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunoprecipitation , Leukemia/genetics , Mesothelioma/genetics , Methylation , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
10.
ACS Med Chem Lett ; 6(5): 491-5, 2015 May 14.
Article in English | MEDLINE | ID: mdl-26005520

ABSTRACT

Inhibitors of the protein methyltransferase Enhancer of Zeste Homolog 2 (EZH2) may have significant therapeutic potential for the treatment of B cell lymphomas and other cancer indications. The ability of the scientific community to explore fully the spectrum of EZH2-associated pathobiology has been hampered by the lack of in vivo-active tool compounds for this enzyme. Here we report the discovery and characterization of EPZ011989, a potent, selective, orally bioavailable inhibitor of EZH2 with useful pharmacokinetic properties. EPZ011989 demonstrates significant tumor growth inhibition in a mouse xenograft model of human B cell lymphoma. Hence, this compound represents a powerful tool for the expanded exploration of EZH2 activity in biology.

11.
PLoS One ; 9(12): e111840, 2014.
Article in English | MEDLINE | ID: mdl-25493630

ABSTRACT

Patients with non-Hodgkin lymphoma (NHL) are treated today with a cocktail of drugs referred to as CHOP (Cyclophosphamide, Hydroxyldaunorubicin, Oncovin, and Prednisone). Subsets of patients with NHL of germinal center origin bear oncogenic mutations in the EZH2 histone methyltransferase. Clinical testing of the EZH2 inhibitor EPZ-6438 has recently begun in patients. We report here that combining EPZ-6438 with CHOP in preclinical cell culture and mouse models results in dramatic synergy for cell killing in EZH2 mutant germinal center NHL cells. Surprisingly, we observe that much of this synergy is due to Prednisolone - a glucocorticoid receptor agonist (GRag) component of CHOP. Dramatic synergy was observed when EPZ-6438 is combined with Prednisolone alone, and a similar effect was observed with Dexamethasone, another GRag. Remarkably, the anti-proliferative effect of the EPZ-6438+GRag combination extends beyond EZH2 mutant-bearing cells to more generally impact germinal center NHL. These preclinical data reveal an unanticipated biological intersection between GR-mediated gene regulation and EZH2-mediated chromatin remodeling. The data also suggest the possibility of a significant and practical benefit of combining EZH2 inhibitors and GRag that warrants further investigation in a clinical setting.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Lymphoma, Non-Hodgkin/drug therapy , Pyridones/pharmacology , Animals , Biphenyl Compounds , Cell Line, Tumor , Cyclophosphamide/pharmacology , Dexamethasone/pharmacology , Doxorubicin/pharmacology , Drug Evaluation, Preclinical , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucocorticoids/pharmacology , Humans , Lymphoma, Non-Hodgkin/metabolism , Mice, SCID , Morpholines , Neoplasm Transplantation , Prednisolone/pharmacology , Prednisone/pharmacology , Random Allocation , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Vincristine/pharmacology
12.
ACS Chem Biol ; 9(11): 2459-64, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25154026

ABSTRACT

EZH2 and EZH1 are protein methyltransferases (PMTs) responsible for histone H3, lysine 27 (H3K27) methylation. Trimethylation of H3K27 (H3K27me3) is a hallmark of many cancers, including non-Hodgkin lymphoma (NHL). Heterozygous EZH2 point mutations at Tyr641, Ala677, and Ala687 have been observed in NHL. The Tyr641 mutations enhance activity on H3K27me2 but have weak or no activity on unmethylated H3K27, whereas the Ala677 and Ala687 mutations use substrates of all methylation states effectively. It has been proposed that enzymatic coupling of the wild-type and mutant enzymes leads to the oncogenic H3K27me3 mark in mutant-bearing NHL. We show that coupling with the wild-type enzyme is needed to achieve H3K27me3 for several mutants, but that others are capable of achieving H3K27me3 on their own. All forms of PRC2 (wild-type and mutants) display kinetic signatures that are consistent with a distributive mechanism of catalysis.


Subject(s)
Mutation , Neoplasms/genetics , Polycomb Repressive Complex 2/chemistry , Enhancer of Zeste Homolog 2 Protein , Humans , Polycomb Repressive Complex 2/genetics
13.
Mol Cancer Ther ; 13(4): 842-54, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24563539

ABSTRACT

Mutations within the catalytic domain of the histone methyltransferase EZH2 have been identified in subsets of patients with non-Hodgkin lymphoma (NHL). These genetic alterations are hypothesized to confer an oncogenic dependency on EZH2 enzymatic activity in these cancers. We have previously reported the discovery of EPZ005678 and EPZ-6438, potent and selective S-adenosyl-methionine-competitive small molecule inhibitors of EZH2. Although both compounds are similar with respect to their mechanism of action and selectivity, EPZ-6438 possesses superior potency and drug-like properties, including good oral bioavailability in animals. Here, we characterize the activity of EPZ-6438 in preclinical models of NHL. EPZ-6438 selectively inhibits intracellular lysine 27 of histone H3 (H3K27) methylation in a concentration- and time-dependent manner in both EZH2 wild-type and mutant lymphoma cells. Inhibition of H3K27 trimethylation (H3K27Me3) leads to selective cell killing of human lymphoma cell lines bearing EZH2 catalytic domain point mutations. Treatment of EZH2-mutant NHL xenograft-bearing mice with EPZ-6438 causes dose-dependent tumor growth inhibition, including complete and sustained tumor regressions with correlative diminution of H3K27Me3 levels in tumors and selected normal tissues. Mice dosed orally with EPZ-6438 for 28 days remained tumor free for up to 63 days after stopping compound treatment in two EZH2-mutant xenograft models. These data confirm the dependency of EZH2-mutant NHL on EZH2 activity and portend the utility of EPZ-6438 as a potential treatment for these genetically defined cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lymphoma, Non-Hodgkin/drug therapy , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Pyridones/pharmacology , Animals , Apoptosis/drug effects , Biphenyl Compounds , Catalytic Domain/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , Lymphoma, Non-Hodgkin/pathology , Male , Mice , Mice, SCID , Molecular Sequence Data , Morpholines , Point Mutation , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays
14.
Proc Natl Acad Sci U S A ; 110(19): 7922-7, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23620515

ABSTRACT

Inactivation of the switch/sucrose nonfermentable complex component SMARCB1 is extremely prevalent in pediatric malignant rhabdoid tumors (MRTs) or atypical teratoid rhabdoid tumors. This alteration is hypothesized to confer oncogenic dependency on EZH2 in these cancers. We report the discovery of a potent, selective, and orally bioavailable small-molecule inhibitor of EZH2 enzymatic activity, (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide). The compound induces apoptosis and differentiation specifically in SMARCB1-deleted MRT cells. Treatment of xenograft-bearing mice with (N-((4,6-dimethyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-5-(ethyl(tetrahydro-2H-pyran-4-yl)amino)-4-methyl-4'-(morpholinomethyl)-[1,1'-biphenyl]-3-carboxamide) leads to dose-dependent regression of MRTs with correlative diminution of intratumoral trimethylation levels of lysine 27 on histone H3, and prevention of tumor regrowth after dosing cessation. These data demonstrate the dependency of SMARCB1 mutant MRTs on EZH2 enzymatic activity and portend the utility of EZH2-targeted drugs for the treatment of these genetically defined cancers.


Subject(s)
Apoptosis , Neoplasms/therapy , Polycomb Repressive Complex 2/antagonists & inhibitors , Rhabdoid Tumor/enzymology , Rhabdoid Tumor/genetics , Animals , Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Design , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Gene Expression Profiling , HEK293 Cells , Histones/metabolism , Humans , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/pathology , Pyridines/pharmacology
15.
Vertex ; 24(112): 426-33, 2013.
Article in Spanish | MEDLINE | ID: mdl-24511559

ABSTRACT

In this paper, we will describe the core themes behind the practice of Intentional Peer Support (IPS), offering a unique perspective on what has commonly been described as "mental illness" and the power dynamics inherent in traditional helping relationships. Through intentional conversations that explore "how we've come to know what we know" and challenge the naming of our experience by others, we begin to find new ways of understanding and responding to our own and other people's experiences. In addition, we emphasize the importance of mutuality in relationships. Both people share responsibility for the relationship, and no one is assumed to be the sole holder of "truth". Mutuality becomes harder but even more critical to practice when we begin to fear for "safety" of the other. It is our belief that as we practice IPS across all relationships in our lives, we can begin to tackle some of the complex ways in which language, roles, power and culture have contributed to our own sense of internalized oppression in any form.


Subject(s)
Mental Disorders/therapy , Peer Group , Social Support , Humans , Intention
16.
Vertex rev. argent. psiquiatr ; 24(112): 426-33, 2013 Nov-Dec.
Article in Spanish | LILACS, BINACIS | ID: biblio-1176945

ABSTRACT

In this paper, we will describe the core themes behind the practice of Intentional Peer Support (IPS), offering a unique perspective on what has commonly been described as "mental illness" and the power dynamics inherent in traditional helping relationships. Through intentional conversations that explore "how we’ve come to know what we know" and challenge the naming of our experience by others, we begin to find new ways of understanding and responding to our own and other people’s experiences. In addition, we emphasize the importance of mutuality in relationships. Both people share responsibility for the relationship, and no one is assumed to be the sole holder of "truth". Mutuality becomes harder but even more critical to practice when we begin to fear for "safety" of the other. It is our belief that as we practice IPS across all relationships in our lives, we can begin to tackle some of the complex ways in which language, roles, power and culture have contributed to our own sense of internalized oppression in any form.


Subject(s)
Social Support , Peer Group , Mental Disorders/therapy , Humans , Intention
17.
Vertex ; 24(112): 426-33, 2013 Nov-Dec.
Article in Spanish | BINACIS | ID: bin-132743

ABSTRACT

In this paper, we will describe the core themes behind the practice of Intentional Peer Support (IPS), offering a unique perspective on what has commonly been described as "mental illness" and the power dynamics inherent in traditional helping relationships. Through intentional conversations that explore "how weve come to know what we know" and challenge the naming of our experience by others, we begin to find new ways of understanding and responding to our own and other peoples experiences. In addition, we emphasize the importance of mutuality in relationships. Both people share responsibility for the relationship, and no one is assumed to be the sole holder of "truth". Mutuality becomes harder but even more critical to practice when we begin to fear for "safety" of the other. It is our belief that as we practice IPS across all relationships in our lives, we can begin to tackle some of the complex ways in which language, roles, power and culture have contributed to our own sense of internalized oppression in any form.


Subject(s)
Mental Disorders/therapy , Peer Group , Social Support , Humans , Intention
18.
Nat Chem Biol ; 8(11): 890-6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023262

ABSTRACT

EZH2 catalyzes trimethylation of histone H3 lysine 27 (H3K27). Point mutations of EZH2 at Tyr641 and Ala677 occur in subpopulations of non-Hodgkin's lymphoma, where they drive H3K27 hypertrimethylation. Here we report the discovery of EPZ005687, a potent inhibitor of EZH2 (K(i) of 24 nM). EPZ005687 has greater than 500-fold selectivity against 15 other protein methyltransferases and has 50-fold selectivity against the closely related enzyme EZH1. The compound reduces H3K27 methylation in various lymphoma cells; this translates into apoptotic cell killing in heterozygous Tyr641 or Ala677 mutant cells, with minimal effects on the proliferation of wild-type cells. These data suggest that genetic alteration of EZH2 (for example, mutations at Tyr641 or Ala677) results in a critical dependency on enzymatic activity for proliferation (that is, the equivalent of oncogene addiction), thus portending the clinical use of EZH2 inhibitors for cancers in which EZH2 is genetically altered.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histones/metabolism , Indazoles/pharmacology , Lymphoma/drug therapy , Lymphoma/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , Pyridones/pharmacology , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enhancer of Zeste Homolog 2 Protein , Enzyme Inhibitors/chemistry , Histones/chemistry , Humans , Indazoles/chemistry , Lymphoma/enzymology , Lymphoma/genetics , Lysine/metabolism , Methylation/drug effects , Molecular Structure , Point Mutation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Pyridones/chemistry , Structure-Activity Relationship
19.
FEBS Lett ; 586(19): 3448-51, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22850114

ABSTRACT

Heterozygous point mutations at Y641 and A677 in the EZH2 SET domain are prevalent in about 10-24% of Non-Hodgkin lymphomas (NHL). Previous studies indicate that these are gain-of-function mutations leading to the hypertrimethylation of H3K27. These EZH2 mutations may drive the proliferation of lymphoma and make EZH2 a molecular target for patients harboring these mutations. Here, another EZH2 SET domain point mutation, A687V, occurring in about 1-2% of lymphoma patients, is also shown to be a gain-of-function mutation that greatly enhances its ability to perform dimethylation relative to wild-type EZH2 and is equally proficient at catalyzing trimethylation. We propose that A687V EZH2 also leads to hypertrimethylation of H3K27 and may thus be a driver mutation in NHL.


Subject(s)
Lymphoma, Non-Hodgkin/enzymology , Lymphoma, Non-Hodgkin/genetics , Mutant Proteins/genetics , Neoplasm Proteins/genetics , Point Mutation , Polycomb Repressive Complex 2/genetics , Enhancer of Zeste Homolog 2 Protein , Heterozygote , Histones/chemistry , Histones/metabolism , Humans , Kinetics , Methylation , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
FEBS Lett ; 585(19): 3011-4, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21856302

ABSTRACT

Mutations at tyrosine 641 (Y641F, Y641N, Y641S and Y641H) in the SET domain of EZH2 have been identified in patients with certain subtypes of non-Hodgkin lymphoma (NHL). These mutations were shown to change the substrate specificity of EZH2 for various methylation states of lysine 27 on histone H3 (H3K27). An additional mutation at EZH2 Y641 to cysteine (Y641C) was also found in one patient with NHL and in SKM-1 cells derived from a patient with myelodisplastic syndrome (MDS). The Y641C mutation has been reported to dramatically reduce enzymatic activity. Here, we demonstrate that while the Y641C mutation ablates enzymatic activity against unmethylated and monomethylated H3K27, it is superior to wild-type in catalyzing the formation of trimethylated H3K27 from the dimethylated precursor.


Subject(s)
DNA-Binding Proteins/metabolism , Histones/metabolism , Lysine/metabolism , Mutation , Substrate Specificity/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Cysteine/chemistry , Cysteine/metabolism , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Histones/genetics , Humans , Lysine/chemistry , Methylation , Molecular Sequence Data , Polycomb Repressive Complex 2 , Sequence Analysis, DNA , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...