Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Rev Mol Cell Biol ; 20(10): 573-589, 2019 10.
Article in English | MEDLINE | ID: mdl-31270442

ABSTRACT

Chromatin is a macromolecular complex predominantly comprising DNA, histone proteins and RNA. The methylation of chromatin components is highly conserved as it helps coordinate the regulation of gene expression, DNA repair and DNA replication. Dynamic changes in chromatin methylation are essential for cell-fate determination and development. Consequently, inherited or acquired mutations in the major factors that regulate the methylation of DNA, RNA and/or histones are commonly observed in developmental disorders, ageing and cancer. This has provided the impetus for the clinical development of epigenetic therapies aimed at resetting the methylation imbalance observed in these disorders. In this Review, we discuss the cellular functions of chromatin methylation and focus on how this fundamental biological process is corrupted in cancer. We discuss methylation-based cancer therapies and provide a perspective on the emerging data from early-phase clinical trial therapies that target regulators of DNA and histone methylation. We also highlight promising therapeutic strategies, including monitoring chromatin methylation for diagnostic purposes and combination epigenetic therapy strategies that may improve immune surveillance in cancer and increase the efficacy of conventional and targeted anticancer drugs.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Histones/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , RNA Processing, Post-Transcriptional , RNA, Neoplasm/metabolism , DNA, Neoplasm/genetics , Histones/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , RNA, Neoplasm/genetics
2.
PLoS Biol ; 16(8): e2004986, 2018 08.
Article in English | MEDLINE | ID: mdl-30080881

ABSTRACT

Distinct transcriptional states are maintained through organization of chromatin, resulting from the sum of numerous repressive and active histone modifications, into tightly packaged heterochromatin versus more accessible euchromatin. Polycomb repressive complex 2 (PRC2) is the main mammalian complex responsible for histone 3 lysine 27 trimethylation (H3K27me3) and is integral to chromatin organization. Using in vitro and in vivo studies, we show that deletion of Suz12, a core component of all PRC2 complexes, results in loss of H3K27me3 and H3K27 dimethylation (H3K27me2), completely blocks normal mammary gland development, and profoundly curtails progenitor activity in 3D organoid cultures. Through the application of mammary organoids to bypass the severe phenotype associated with Suz12 loss in vivo, we have explored gene expression and chromatin structure in wild-type and Suz12-deleted basal-derived organoids. Analysis of organoids led to the identification of lineage-specific changes in gene expression and chromatin structure, inferring cell type-specific PRC2-mediated gene silencing of the chromatin state. These expression changes were accompanied by cell cycle arrest but not lineage infidelity. Together, these data indicate that canonical PRC2 function is essential for development of the mammary gland through the repression of alternate transcription programs and maintenance of chromatin states.


Subject(s)
Mammary Glands, Animal/embryology , Polycomb Repressive Complex 2/physiology , Animals , Chromatin/metabolism , Enhancer of Zeste Homolog 2 Protein/physiology , Female , Heterochromatin/metabolism , Histone Code , Histones/metabolism , Lysine/metabolism , Mammary Glands, Animal/metabolism , Methylation , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins , Primary Cell Culture , Protein Processing, Post-Translational
3.
Mol Cell ; 48(3): 343-52, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23000175

ABSTRACT

Trp63, a transcription factor related to the tumor suppressor p53, is activated by diverse stimuli and can initiate a range of cellular responses. TAp63 is the predominant Trp53 family member in primordial follicle oocyte nuclei and is essential for their apoptosis triggered by DNA damage in vivo. After γ-irradiation, induction of the proapoptotic BH3-only members Puma and Noxa was observed in primordial follicle oocytes from WT and Trp53(-/-) mice but not in those from TAp63-deficient mice. Primordial follicle oocytes from mice lacking Puma or both Puma and Noxa were protected from γ-irradiation-induced apoptosis and, remarkably, could produce healthy offspring. Hence, PUMA and NOXA are critical for DNA damage-induced, TAp63-mediated primordial follicle oocyte apoptosis. Thus, blockade of PUMA may protect fertility during cancer therapy and prevent premature menopause, improving women's health.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , DNA Damage , Fertility/genetics , Oocytes/metabolism , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/metabolism , Female , Gamma Rays , Gene Expression/radiation effects , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oocytes/cytology , Oocytes/radiation effects , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 112(27): 8409-14, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26100884

ABSTRACT

Metaplastic breast carcinoma (MBC) is a rare histological breast cancer subtype characterized by mesenchymal elements and poor clinical outcome. A large fraction of MBCs harbor defects in breast cancer 1 (BRCA1). As BRCA1 deficiency sensitizes tumors to DNA cross-linking agents and poly(ADP-ribose) polymerase (PARP) inhibitors, we sought to investigate the response of BRCA1-deficient MBCs to the PARP inhibitor olaparib. To this end, we established a genetically engineered mouse model (GEMM) for BRCA1-deficient MBC by introducing the MET proto-oncogene into a BRCA1-associated breast cancer model, using our novel female GEMM ES cell (ESC) pipeline. In contrast to carcinomas, BRCA1-deficient mouse carcinosarcomas resembling MBC show intrinsic resistance to olaparib caused by increased P-glycoprotein (Pgp) drug efflux transporter expression. Indeed, resistance could be circumvented by using another PARP inhibitor, AZD2461, which is a poor Pgp substrate. These preclinical findings suggest that patients with BRCA1-associated MBC may show poor response to olaparib and illustrate the value of GEMM-ESC models of human cancer for evaluation of novel therapeutics.


Subject(s)
BRCA1 Protein/deficiency , Mammary Neoplasms, Experimental/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , BRCA1 Protein/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinosarcoma/drug therapy , Carcinosarcoma/genetics , Carcinosarcoma/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Metaplasia , Mice, Inbred C57BL , Mice, Knockout , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Mas , Survival Analysis
5.
Genes Dev ; 24(15): 1608-13, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20679396

ABSTRACT

Although tumor development requires impaired apoptosis, we describe a novel paradigm of apoptosis-dependent tumorigenesis. Because DNA damage triggers apoptosis through p53-mediated induction of BH3-only proteins Puma and Noxa, we explored their roles in gamma-radiation-induced thymic lymphomagenesis. Surprisingly, whereas Noxa loss accelerated it, Puma loss ablated tumorigenesis. Tumor suppression by Puma deficiency reflected its protection of leukocytes from gamma-irradiation-induced death, because their glucocorticoid-mediated decimation in Puma-deficient mice activated cycling of stem/progenitor cells and restored thymic lymphomagenesis. Our demonstration that cycles of cell attrition and repopulation by stem/progenitor cells can drive tumorigenesis has parallels in human cancers, such as therapy-induced malignancies.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Apoptosis/radiation effects , Gamma Rays , Lymphoma/physiopathology , Thymus Neoplasms/physiopathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Cells, Cultured , Dexamethasone/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Leukocytes/drug effects , Leukocytes/pathology , Leukocytes/radiation effects , Lymphoma/genetics , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Survival Analysis , Thymus Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism
6.
Blood ; 122(14): 2402-11, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23966420

ABSTRACT

Caloric restriction (CR) is proposed to decrease tumorigenesis through a variety of mechanisms including effects on glycolysis. However, the understanding of how CR affects the response to cancer therapy is still rudimentary. Here, using the Eµ-Myc transgenic mouse model of B-cell lymphoma, we report that by reducing protein translation, CR can reduce expression of the prosurvival Bcl-2 family member Mcl-1 and sensitize lymphomas to ABT-737-induced death in vivo. By using Eµ-Myc lymphoma cells lacking p53, we showed that CR mimetics such as 2-deoxyglucose led to a decrease in Mcl-1 expression and sensitized lymphoma cells to ABT-737-induced death independently of p53. In keeping with this, Eµ-Myc lymphoma cells lacking the BH3-only proapoptotic members Noxa, Puma, or Bim were also sensitized by CR mimetics to ABT-737-induced death. Remarkably, neither the loss of both Puma and Noxa, the loss of both Puma and Bim, nor the loss of all three BH3-only proteins prevented sensitization to ABT-737 induced by CR mimetics. Thus, CR can influence Mcl-1 expression and sensitize cells to BH3 mimetic-induced apoptosis, independently of the main BH3-only proteins and of p53. Exploiting this may improve the efficiency of, or prevent resistance to, cancer therapy.


Subject(s)
Caloric Restriction , Drug Resistance, Neoplasm/physiology , Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Biphenyl Compounds/pharmacology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein , Nitrophenols/pharmacology , Piperazines/pharmacology , Sulfonamides/pharmacology
7.
J Biol Chem ; 288(7): 5198-209, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23243310

ABSTRACT

c-Myc transformed human Burkitt's lymphoma (BL) cells are highly sensitive to TGF-ß-induced apoptosis. Previously we demonstrated that TGF-ß-mediated cell death in BL cells is regulated via the mitochondrial intrinsic apoptosis pathway, which is dependent on the activation of BAX and/or BAK. TGF-ß directly induces transcription of the BH3-only protein BIK and represses expression of the pro-survival factor BCL-X(L) but has no effect on the direct BAX/BAK "activators" BIM or BID (tBID). Here we show that TGF-ß induces the BH3-only activator PUMA to aid induction of the intrinsic cell death pathway. TGF-ß also induced PUMA in normal germinal center CD77-positive centroblasts isolated from human tonsil tissue. PUMA was a direct TGF-ß target gene in B-cells, and we identify a putative Smad-binding region within the human PUMA promoter that recruits Smad3 and Smad4 in cells in response to TGF-ß signaling. Constitutive activity of the isolated Smad-binding region in luciferase reporter assays was dependent on Smad consensus sequences and was partially dependent on endogenous TGF-ß signaling and Smad4. Knockdown of PUMA in BL cells using lentiviral shRNA resulted in slower kinetics of the TGF-ß-mediated apoptotic response. Analysis of Eµ-Myc cell lines demonstrated that c-myc-driven murine lymphomas are also sensitive to TGF-ß-mediated apoptosis. Moreover, Puma(-/-) Eµ-Myc lines demonstrated significantly delayed kinetics of the apoptotic response when compared with wild type lymphomas. TGF-ß therefore induces a polygenic response in Myc-driven lymphomas involving transcription of PUMA, which is necessary for the rapid induction of cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cell Survival , HEK293 Cells , Humans , Kinetics , Lymphoma/metabolism , Mice , Promoter Regions, Genetic , Signal Transduction , Transcription, Genetic
8.
J Pathol ; 228(3): 300-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926799

ABSTRACT

The leucine-rich repeat-containing heterotrimeric guanine nucleotide-binding protein-coupled receptor 5 (LGR5) has been identified as a marker of cycling stem cells in several epithelial tissues, including small intestine, colon, stomach and hair follicle. To investigate whether LGR5 also marks mammary epithelial stem cells, we performed in situ lineage-tracing studies and mammary gland reconstitutions with LGR5-expressing mammary epithelial cells. Interestingly, the LGR5 progeny population in mammary epithelium switches from the luminal to the myoepithelial compartment during the first 12 days of postnatal development, likely reflecting local changes in Wnt signalling. Together, our findings point to a stage-specific contribution of LGR5-expressing cells to luminal and basal epithelial lineages during postnatal mammary gland development.


Subject(s)
Cell Lineage/physiology , Epithelial Cells/cytology , Mammary Glands, Animal/growth & development , Morphogenesis/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/physiology , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice , Mice, Knockout , Models, Animal , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/physiology , Wnt Proteins/physiology
9.
Blood ; 116(24): 5256-67, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20829369

ABSTRACT

DNA-damaging chemotherapy is the backbone of cancer treatment, although it is not clear how such treatments kill tumor cells. In nontransformed lymphoid cells, the combined loss of 2 proapoptotic p53 target genes, Puma and Noxa, induces as much resistance to DNA damage as loss of p53 itself. In Eµ-Myc lymphomas, however, lack of both Puma and Noxa resulted in no greater drug resistance than lack of Puma alone. A third B-cell lymphoma-2 homology domain (BH)3-only gene, Bim, although not a direct p53 target, was up-regulated in Eµ-Myc lymphomas incurring DNA damage, and knockdown of Bim levels markedly increased the drug resistance of Eµ-Myc/Puma(-/-)Noxa(-/-) lymphomas both in vitro and in vivo. Remarkably, c-MYC-driven lymphoma cell lines from Noxa(-/-)Puma(-/-)Bim(-/-) mice were as resistant as those lacking p53. Thus, the combinatorial action of Puma, Noxa, and Bim is critical for optimal apoptotic responses of lymphoma cells to 2 commonly used DNA-damaging chemotherapeutic agents, identifying Bim as an additional biomarker for treatment outcome in the clinic.


Subject(s)
Apoptosis Regulatory Proteins/physiology , DNA Damage , Drug Resistance, Neoplasm/genetics , Lymphoma, B-Cell/pathology , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Animals , Apoptosis , Bcl-2-Like Protein 11 , Cyclophosphamide/pharmacology , Etoposide/pharmacology , Lymphoma, B-Cell/drug therapy , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Suppressor Proteins/physiology
10.
J Cell Biol ; 176(4): 415-24, 2007 Feb 12.
Article in English | MEDLINE | ID: mdl-17283183

ABSTRACT

To identify the mechanisms of ultraviolet radiation (UVR)-induced cell death, for which the tumor suppressor p53 is essential, we have analyzed mouse embryonic fibroblasts (MEFs) and keratinocytes in mouse skin that have specific apoptotic pathways blocked genetically. Blocking the death receptor pathway provided no protection to MEFs, whereas UVR-induced apoptosis was potently inhibited by Bcl-2 overexpression, implicating the mitochondrial pathway. Indeed, Bcl-2 overexpression boosted cell survival more than p53 loss, revealing a p53-independent pathway controlled by the Bcl-2 family. Analysis of primary MEFs lacking individual members of its BH3-only subfamily identified major initiating roles for the p53 targets Noxa and Puma. In the transformed derivatives, where Puma, unexpectedly, was not induced by UVR, Noxa had the dominant role and Bim a minor role. Furthermore, loss of Noxa suppressed the formation of apoptotic keratinocytes in the skin of UV-irradiated mice. Collectively, these results demonstrate that UVR activates the Bcl-2-regulated apoptotic pathway predominantly through activation of Noxa and, depending on cellular context, Puma.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Fibroblasts/metabolism , Keratinocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Skin/metabolism , Animals , Apoptosis/radiation effects , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Fibroblasts/radiation effects , Keratinocytes/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Death Domain/antagonists & inhibitors , Receptors, Death Domain/metabolism , Signal Transduction/genetics , Signal Transduction/radiation effects , Skin/cytology , Skin/radiation effects , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ultraviolet Rays
11.
Cancer Cell ; 36(4): 385-401.e8, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31564637

ABSTRACT

Loss of MHC class I (MHC-I) antigen presentation in cancer cells can elicit immunotherapy resistance. A genome-wide CRISPR/Cas9 screen identified an evolutionarily conserved function of polycomb repressive complex 2 (PRC2) that mediates coordinated transcriptional silencing of the MHC-I antigen processing pathway (MHC-I APP), promoting evasion of T cell-mediated immunity. MHC-I APP gene promoters in MHC-I low cancers harbor bivalent activating H3K4me3 and repressive H3K27me3 histone modifications, silencing basal MHC-I expression and restricting cytokine-induced upregulation. Bivalent chromatin at MHC-I APP genes is a normal developmental process active in embryonic stem cells and maintained during neural progenitor differentiation. This physiological MHC-I silencing highlights a conserved mechanism by which cancers arising from these primitive tissues exploit PRC2 activity to enable immune evasion.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gene Expression Regulation, Neoplastic/immunology , Histocompatibility Antigens Class I/genetics , Neoplasms/immunology , Polycomb Repressive Complex 2/metabolism , Tumor Escape/genetics , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , DNA Methylation/immunology , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , Drug Resistance, Neoplasm/genetics , Epigenetic Repression/drug effects , Epigenetic Repression/immunology , Female , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histone Code/drug effects , Humans , Mice , Middle Aged , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Polycomb Repressive Complex 2/antagonists & inhibitors , T-Lymphocytes/immunology , Tumor Escape/drug effects , Xenograft Model Antitumor Assays
12.
Mol Cell Biol ; 25(19): 8732-47, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16166651

ABSTRACT

The c-Jun N-terminal protein kinase (JNK)/c-Jun and p53 pathways form distinct death-signaling modules in neurons that culminate in Bax-dependent apoptosis. To investigate whether this signaling autonomy is due to recruitment of particular BH3-only proteins, we searched for a toxic signal that would activate both pathways in the same set of neurons. We show that arsenite activates both the JNK/c-Jun and p53 pathways in cortical neurons, which together account for >95% of apoptosis, as determined by using the mixed-lineage kinase (JNK/c-Jun) pathway inhibitor CEP11004 and p53-null mice. Despite the coexistence of both pathways in at least 30% of the population, Bim mRNA and protein expression was increased only by the JNK/c-Jun signaling pathway, whereas Noxa and Puma mRNA and Puma protein expression was entirely JNK/c-Jun independent. About 50% of Puma/Noxa expression was p53 dependent, with the remaining signal being independent of both pathways and possibly facilitated by arsenite-induced reduction in P-Akt. However, functionally, Puma was predominant in mediating Bax-dependent apoptosis, as evidenced by the fact that more than 90% of apoptosis was prevented in Puma-null neurons, although Bim was still upregulated, while Bim- and Noxa-null neurons died similarly to wild-type neurons. Thus, the p53 and JNK/c-Jun pathways can activate mutually exclusive subclasses of BH3-only proteins in the same set of neurons. However, other factors besides expression may determine which BH3-only proteins mediate apoptosis.


Subject(s)
Arsenites/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Neurons/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Death , DNA Primers/chemistry , Electrophoresis, Polyacrylamide Gel , Immunoblotting , Immunohistochemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subcellular Fractions , Time Factors , Tumor Suppressor Proteins/metabolism , Up-Regulation
13.
J Neurosci ; 26(27): 7257-64, 2006 Jul 05.
Article in English | MEDLINE | ID: mdl-16822983

ABSTRACT

Neural precursor cells (NPCs) are highly sensitive to genotoxic injury, which triggers activation of the intrinsic mitochondria-dependent apoptotic pathway. This pathway is typically initiated by members of the BH3 (Bcl-2 homology 3)-only subgroup of the Bcl-2 (B-cell CLL/lymphoma 2) protein family, which are positioned upstream in the apoptotic pathway to respond to specific death stimuli. We have shown previously that NPCs deficient in the tumor suppressor protein p53 show significantly less death after exposure to genotoxic injury or to staurosporine (STS), a broad kinase inhibitor and potent apoptosis inducer. p53 has been shown to regulate the expression of both Noxa and Puma, two BH3-only proteins, although their involvement in p53-dependent cell death appears to be cell-type and stimulus specific. A systematic comparison of the relative contributions of Noxa and Puma to NPC apoptosis has not yet been performed. We hypothesized that p53-dependent transcription of Noxa and Puma leads to death in telencephalic NPCs exposed to genotoxic stress. We found that genotoxic injury induces a rapid p53-dependent increase in expression of Noxa and Puma mRNA in telencephalic NPCs. Furthermore, deficiency of either Noxa or Puma inhibited DNA damage-induced caspase-3 activation and cell death in telencephalic NPCs in vitro. However, only Puma deficiency protected telencephalic ventricular zone NPCs from death in vivo. In contrast to genotoxic injury, STS produced a p53-independent increase in Noxa and Puma expression, but neither Noxa nor Puma was required for STS-induced NPC death. Together, these experiments identify Noxa and Puma as important regulators of genotoxin-induced telencephalic NPC death.


Subject(s)
Apoptosis/physiology , Neurons/cytology , Proto-Oncogene Proteins c-bcl-2/metabolism , Stem Cells/cytology , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , DNA Damage/physiology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/physiology , Pregnancy , Proto-Oncogene Proteins c-bcl-2/genetics , Staurosporine/pharmacology , Stem Cells/physiology , Telencephalon/cytology , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Up-Regulation
14.
Mol Oncol ; 10(10): 1497-1515, 2016 12.
Article in English | MEDLINE | ID: mdl-27717710

ABSTRACT

Histone methyltransferases (HMTs) catalyze the methylation of lysine and arginine residues on histone tails and non-histone targets. These important post-translational modifications are exquisitely regulated and affect chromatin compaction and transcriptional programs leading to diverse biological outcomes. There is accumulating evidence that genetic alterations of several HMTs impinge on oncogenic or tumor-suppressor functions and influence both cancer initiation and progression. HMTs therefore represent an opportunity for therapeutic targeting in those patients with tumors in which HMTs are dysregulated, to reverse the histone marks and transcriptional programs associated with aggressive tumor behavior. In this review, we describe the known histone methyltransferases and their emerging roles in breast cancer tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast/pathology , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Protein-Arginine N-Methyltransferases/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Drug Discovery , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Code/drug effects , Histone-Lysine N-Methyltransferase/analysis , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Humans , Molecular Targeted Therapy , Protein-Arginine N-Methyltransferases/analysis , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/metabolism , Transcriptional Activation/drug effects
15.
Nat Protoc ; 10(11): 1755-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26492136

ABSTRACT

Preclinical in vivo validation of target genes for therapeutic intervention requires careful selection and characterization of the most suitable animal model in order to assess the role of these genes in a particular process or disease. To this end, genetically engineered mouse models (GEMMs) are typically used. However, the appropriate engineering of these models is often cumbersome and time consuming. Recently, we and others described a modular approach for fast-track modification of existing GEMMs by re-derivation of embryonic stem cells (ESCs) that can be modified by recombinase-mediated transgene insertion and subsequently used for the production of chimeric mice. This 'GEMM-ESC strategy' allows for rapid in vivo analysis of gene function in the chimeras and their offspring. Moreover, this strategy is compatible with CRISPR/Cas9-mediated genome editing. This protocol describes when and how to use the GEMM-ESC strategy effectively, and it provides a detailed procedure for re-deriving and manipulating GEMM-ESCs under feeder- and serum-free conditions. This strategy produces transgenic mice with the desired complex genotype faster than traditional methods: generation of validated GEMM-ESC clones for controlled transgene integration takes 9-12 months, and recombinase-mediated transgene integration and chimeric cohort production takes 2-3 months. The protocol requires skills in embryology, stem cell biology and molecular biology, and it is ideally performed within, or in close collaboration with, a transgenic facility.


Subject(s)
Embryonic Stem Cells/physiology , Gene Expression , Gene Targeting/methods , Mice, Transgenic , Proteins/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Recombination, Genetic , Transgenes
16.
EMBO Mol Med ; 6(2): 212-25, 2014 02.
Article in English | MEDLINE | ID: mdl-24401838

ABSTRACT

Human cancers modeled in Genetically Engineered Mouse Models (GEMMs) can provide important mechanistic insights into the molecular basis of tumor development and enable testing of new intervention strategies. The inherent complexity of these models, with often multiple modified tumor suppressor genes and oncogenes, has hampered their use as preclinical models for validating cancer genes and drug targets. In our newly developed approach for the fast generation of tumor cohorts we have overcome this obstacle, as exemplified for three GEMMs; two lung cancer models and one mesothelioma model. Three elements are central for this system; (i) The efficient derivation of authentic Embryonic Stem Cells (ESCs) from established GEMMs, (ii) the routine introduction of transgenes of choice in these GEMM-ESCs by Flp recombinase-mediated integration and (iii) the direct use of the chimeric animals in tumor cohorts. By applying stringent quality controls, the GEMM-ESC approach proofs to be a reliable and effective method to speed up cancer gene assessment and target validation. As proof-of-principle, we demonstrate that MycL1 is a key driver gene in Small Cell Lung Cancer.


Subject(s)
Embryonic Stem Cells/cytology , Gene Transfer Techniques , Lung Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cells, Cultured , Chimera , Clone Cells , DNA Nucleotidyltransferases/metabolism , Disease Models, Animal , Embryonic Stem Cells/metabolism , Genes, Reporter , Genomic Instability , Genotype , Germ Cells/metabolism , Humans , Luciferases/metabolism , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Oncogenes , Phenotype , Pluripotent Stem Cells/metabolism , Quality Control , Reproducibility of Results , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology
17.
Cell Rep ; 3(5): 1339-45, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23665218

ABSTRACT

Activation of apoptosis through transcriptional induction of Puma and Noxa has long been considered to constitute the critical (if not sole) process by which p53 suppresses tumor development, although G1/S boundary cell-cycle arrest via induction of the CDK inhibitor p21 has also been thought to contribute. Recent analyses of mice bearing mutations that impair p53-mediated induction of select target genes have indicated that activation of apoptosis and G1/S cell-cycle arrest may, in fact, be dispensable for p53-mediated tumor suppression. However, the expression of Puma, Noxa, and p21 was not abrogated in these mutants, only reduced; therefore, the possibility that the reduced levels of these critical effectors of p53-mediated apoptosis and G1/S-cell-cycle arrest sufficed to prevent tumorigenesis could not be excluded. To resolve this important issue, we have generated mice deficient for p21, Puma, and Noxa (p21-/-puma-/-noxa-/- mice). Cells from these mice were deficient in their ability to undergo p53-mediated apoptosis, G1/S cell-cycle arrest, and senescence. Nonetheless, these animals remained tumor free until at least 500 days, in contrast to p53-deficient mice, which had all succumbed to lymphoma or sarcoma by 250 days. Interestingly, DNA lesions induced by γ-irradiation persisted longer in p53-deficient cells compared to wild-type or p21-/-puma-/-noxa-/- cells, and the former failed to transcriptionally activate several p53 target genes implicated in DNA repair. These results demonstrate beyond a doubt that the induction of apoptosis, cell-cycle arrest, and possibly senescence is dispensable for p53-mediated suppression of spontaneous tumor development and indicate that coordination of genomic stability and possibly other processes, such as metabolic adaptation, may instead be critical.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Cells, Cultured , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/radiation effects , DNA Repair , G1 Phase Cell Cycle Checkpoints , Gamma Rays , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , S Phase Cell Cycle Checkpoints , Thymocytes/cytology , Thymocytes/metabolism , Thymocytes/radiation effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
18.
Stem Cell Investig ; 4: 45, 2017.
Article in English | MEDLINE | ID: mdl-28607919
19.
J Cell Biol ; 180(2): 341-55, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18209102

ABSTRACT

Like Bcl-2, Mcl-1 is an important survival factor for many cancers, its expression contributing to chemoresistance and disease relapse. However, unlike other prosurvival Bcl-2-like proteins, Mcl-1 stability is acutely regulated. For example, the Bcl-2 homology 3 (BH3)-only protein Noxa, which preferentially binds to Mcl-1, also targets it for proteasomal degradation. In this paper, we describe the discovery and characterization of a novel BH3-like ligand derived from Bim, Bim(S)2A, which is highly selective for Mcl-1. Unlike Noxa, Bim(S)2A is unable to trigger Mcl-1 degradation, yet, like Noxa, Bim(S)2A promotes cell killing only when Bcl-x(L) is absent or neutralized. Furthermore, killing by endogenous Bim is not associated with Mcl-1 degradation. Thus, functional inactivation of Mcl-1 does not always require its elimination. Rather, it can be efficiently antagonized by a BH3-like ligand tightly engaging its binding groove, which is confirmed here with a structural study. Our data have important implications for the discovery of compounds that might kill cells whose survival depends on Mcl-1.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cells, Cultured , Humans , Ligands , Membrane Proteins/chemistry , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/chemistry , Peptide Fragments/chemistry , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-bcl-2/chemistry , Sequence Alignment , bcl-2-Associated X Protein/metabolism
20.
Cell ; 129(7): 1337-49, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17604722

ABSTRACT

Endoplasmic reticulum (ER) stress caused by misfolded proteins or cytotoxic drugs can kill cells and although activation of this pathway has been implicated in the etiology of certain degenerative disorders its mechanism remains unresolved. Bim, a proapoptotic BH3-only member of the Bcl-2 family is required for initiation of apoptosis induced by cytokine deprivation or certain stress stimuli. Its proapoptotic activity can be regulated by several transcriptional or posttranslational mechanisms, such as ERK-mediated phosphorylation, promoting its ubiquitination and proteasomal degradation. We found that Bim is essential for ER stress-induced apoptosis in a diverse range of cell types both in culture and within the whole animal. ER stress activates Bim through two novel pathways, involving protein phosphatase 2A-mediated dephosphorylation, which prevents its ubiquitination and proteasomal degradation and CHOP-C/EBPalpha-mediated direct transcriptional induction. These results define the molecular mechanisms of ER stress-induced apoptosis and identify targets for therapeutic intervention in ER stress-related diseases.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Bcl-2-Like Protein 11 , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/ultrastructure , Enzyme Inhibitors/pharmacology , Mice , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Phosphatase 2 , Protein Structure, Tertiary/physiology , Regulatory Elements, Transcriptional/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Thapsigargin/pharmacology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL