Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Rep ; 41(10): 111749, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36476850

ABSTRACT

Co-transcriptional R loops arise from stalling of RNA polymerase, leading to the formation of stable DNA:RNA hybrids. Unresolved R loops promote genome instability but are counteracted by helicases and nucleases. Here, we show that branchpoint translocases are a third class of R-loop-displacing enzyme in vitro. In cells, deficiency in the Fanconi-anemia-associated branchpoint translocase FANCM causes R-loop accumulation, particularly after treatment with DNA:RNA-hybrid-stabilizing agents. This correlates with FANCM localization at R-loop-prone regions of the genome. Moreover, other branchpoint translocases associated with human disease, such as SMARCAL1 and ZRANB3, and those from lower organisms can also remove R loops in vitro. Branchpoint translocases are more potent than helicases in resolving R loops, indicating their evolutionary important role in R-loop suppression. In human cells, FANCM, SMARCAL1, and ZRANB3 depletion causes additive effects on R-loop accumulation and DNA damage. Our work reveals a mechanistic basis for R-loop displacement that is linked to genome stability.


Subject(s)
R-Loop Structures , RNA , Humans , DNA Helicases/genetics
2.
Cell Death Dis ; 9(9): 844, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154459

ABSTRACT

The loss of p53 function is a central event in the genesis of osteosarcoma (OS). How mutation of p53 enables OS development from osteoblastic lineage cells is poorly understood. We and others have reported a key role for elevated and persistent activation of the cAMP/PKA/Creb1 pathway in maintenance of OS. In view of the osteoblast lineage being the cell of origin of OS, we sought to determine how these pathways interact within the context of the normal osteoblast. Normal osteoblasts (p53 WT) rapidly underwent apoptosis in response to acute elevation of cAMP levels or activity, whereas p53-deficient osteoblasts tolerated this aberrant cAMP/Creb level and activity. Using the p53 activating small-molecule Nutlin-3a and cAMP/Creb1 activator forskolin, we addressed the question of how p53 responds to the activation of cAMP. We observed that p53 acts dominantly to protect cells from excessive cAMP accumulation. We identify a Creb1-Cbp complex that functions together with and interacts with p53. Finally, translating these results we find that a selective small-molecule inhibitor of the Creb1-Cbp interaction demonstrates selective toxicity to OS cells where this pathway is constitutively active. This highlights the cAMP/Creb axis as a potentially actionable therapeutic vulnerability in p53-deficient tumors such as OS. These results define a mechanism through which p53 protects normal osteoblasts from excessive or abnormal cAMP accumulation, which becomes fundamentally compromised in OS.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Osteoblasts/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Membrane Proteins/metabolism , Mice , Osteosarcoma/metabolism , Signal Transduction/physiology
3.
J Bone Miner Res ; 33(12): 2202-2213, 2018 12.
Article in English | MEDLINE | ID: mdl-30040873

ABSTRACT

Retinoic acid receptor (RAR) signaling regulates bone structure and hematopoiesis through intrinsic and extrinsic mechanisms. This study aimed to establish how early in the osteoblast lineage loss of RARγ (Rarg) disrupts the bone marrow microenvironment. Bone structure was analyzed by micro-computed tomography (µCT) in Rarg-/- mice and mice with Rarg conditional deletion in Osterix-Cre-targeted osteoblast progenitors or Prrx1-Cre-targeted mesenchymal stem cells. Rarg-/- tibias exhibited less trabecular and cortical bone and impaired longitudinal and radial growth. The trabecular bone and longitudinal, but not radial, growth defects were recapitulated in Prrx1:RargΔ/Δ mice but not Osx1:RargΔ/Δ mice. Although both male and female Prrx1:RargΔ/Δ mice had low trabecular bone mass, males exhibited increased numbers of trabecular osteoclasts and Prrx1:RargΔ/Δ females had impaired mineral deposition. Both male and female Prrx1:RargΔ/Δ growth plates were narrower than controls and their epiphyses contained hypertrophic chondrocyte islands. Flow cytometry revealed that male Prrx1:RargΔ/Δ bone marrow exhibited elevated pro-B and pre-B lymphocyte numbers, accompanied by increased Cxcl12 expression in bone marrow cells. Prrx1:RargΔ/Δ bone marrow also had elevated megakaryocyte-derived Vegfa expression accompanied by smaller sinusoidal vessels. Thus, RARγ expression by Prrx1-Cre-targeted cells directly regulates endochondral bone formation and indirectly regulates tibial vascularization. Furthermore, RARγ expression by Prrx1-Cre-targeted cells extrinsically regulates osteoclastogenesis and B lymphopoiesis in male mice. © 2018 American Society for Bone and Mineral Research.


Subject(s)
B-Lymphocytes/metabolism , Bone and Bones/metabolism , Lymphopoiesis , Mesenchymal Stem Cells/metabolism , Neovascularization, Physiologic , Receptors, Retinoic Acid/metabolism , Animals , Bone Development , Bone Marrow/blood supply , Bone Marrow/pathology , Cancellous Bone/metabolism , Cortical Bone/metabolism , Female , Male , Mice, Transgenic , Organ Size , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , Tibia/pathology , Retinoic Acid Receptor gamma
4.
J Cell Biochem ; 119(6): 4241-4250, 2018 06.
Article in English | MEDLINE | ID: mdl-29236321

ABSTRACT

Osteosarcoma (OS) is the most common cancer of bone in children and young adults. Despite extensive research efforts, there has been no significant improvement in patient outcome for many years. An improved understanding of the biology of this cancer and how genes frequently mutated contribute to OS may help improve outcomes for patients. While our knowledge of the mutational burden of OS is approaching saturation, our understanding of how these mutations contribute to OS initiation and maintenance is less clear. Murine models of OS have now been demonstrated to be highly valid recapitulations of human OS. These models were originally based on the frequent disruption of p53 and Rb in familial OS syndromes, which are also common mutations in sporadic OS. They have been applied to significantly improve our understanding about the functions of recurrently mutated genes in disease. The murine models can be used as a platform for preclinical testing and identifying new therapeutic targets, in addition to testing the role of additional mutations in vivo. Most recently these models have begun to be used for discovery based approaches and screens, which hold significant promise in furthering our understanding of the genetic and therapeutic sensitivities of OS. In this review, we discuss the mouse models of OS that have been reported in the last 3-5 years and newly identified pathways from these studies. Finally, we discuss the preclinical utilization of the mouse models of OS for identifying and validating actionable targets to improve patient outcome.


Subject(s)
Bone Neoplasms , Neoplasms, Experimental , Osteosarcoma , Retinoblastoma Protein , Tumor Suppressor Protein p53 , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Mice , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Elife ; 52016 04 12.
Article in English | MEDLINE | ID: mdl-27070462

ABSTRACT

Mutations in the P53 pathway are a hallmark of human cancer. The identification of pathways upon which p53-deficient cells depend could reveal therapeutic targets that may spare normal cells with intact p53. In contrast to P53 point mutations in other cancer, complete loss of P53 is a frequent event in osteosarcoma (OS), the most common cancer of bone. The consequences of p53 loss for osteoblastic cells and OS development are poorly understood. Here we use murine OS models to demonstrate that elevated Pthlh (Pthrp), cAMP levels and signalling via CREB1 are characteristic of both p53-deficient osteoblasts and OS. Normal osteoblasts survive depletion of both PTHrP and CREB1. In contrast, p53-deficient osteoblasts and OS depend upon continuous activation of this pathway and undergo proliferation arrest and apoptosis in the absence of PTHrP or CREB1. Our results identify the PTHrP-cAMP-CREB1 axis as an attractive pathway for therapeutic inhibition in OS.


Subject(s)
Bone Neoplasms/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP/metabolism , Gene Expression Regulation, Neoplastic , Osteosarcoma/genetics , Parathyroid Hormone-Related Protein/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Osteoblasts/metabolism , Osteoblasts/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Parathyroid Hormone-Related Protein/metabolism , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Protein p53/deficiency
6.
Cell Rep ; 14(6): 1488-1499, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26832406

ABSTRACT

How MYC promotes the development of cancer remains to be fully understood. Here, we report that the Zn(2+)-finger transcription factor ASCIZ (ATMIN, ZNF822) synergizes with MYC to activate the expression of dynein light chain (DYNLL1, LC8) in the murine Eµ-Myc model of lymphoma. Deletion of Asciz or Dynll1 prevented the abnormal expansion of pre-B cells in pre-cancerous Eµ-Myc mice and potentiated the pro-apoptotic activity of MYC in pre-leukemic immature B cells. Constitutive loss of Asciz or Dynll1 delayed lymphoma development in Eµ-Myc mice, and induced deletion of Asciz in established lymphomas extended the survival of tumor-bearing mice. We propose that ASCIZ-dependent upregulation of DYNLL1 levels is essential for the development and expansion of MYC-driven lymphomas by enabling the survival of pre-neoplastic and malignant cells.


Subject(s)
Dyneins/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Precursor Cells, B-Lymphoid/pathology , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/genetics , Animals , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Cytoplasmic Dyneins , Disease Models, Animal , Dyneins/deficiency , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Mice , Precursor Cells, B-Lymphoid/immunology , Proto-Oncogene Proteins c-myc/immunology , Signal Transduction , Survival Analysis , Transcription Factors/deficiency
7.
Sci Rep ; 5: 10120, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25944566

ABSTRACT

Osteosarcoma (OS) survival rates have plateaued in part due to a lack of new therapeutic options. Here we demonstrate that bromodomain inhibitors (BETi), JQ1, I-BET151, I-BET762, exert potent anti-tumour activity against primary and established OS cell lines, mediated by inhibition of BRD4. Strikingly, unlike previous observations in long-term established human OS cell lines, the antiproliferative activity of JQ1 in primary OS cells was driven by the induction of apoptosis, not cell cycle arrest. In further contrast, JQ1 activity in OS was mediated independently of MYC downregulation. We identified that JQ1 suppresses the transcription factor FOSL1 by displacement of BRD4 from its locus. Loss of FOSL1 phenocopied the antiproliferative effects of JQ1, identifying FOSL1 suppression as a potential novel therapeutic approach for OS. As a monotherapy JQ1 demonstrated significant anti-tumour activity in vivo in an OS graft model. Further, combinatorial treatment approaches showed that JQ1 increased the sensitivity of OS cells to doxorubicin and induced potent synergistic activity when rationally combined with CDK inhibitors. The greater level of activity achieved with the combination of BETi with CDK inhibitors demonstrates the efficacy of this combination therapy. Taken together, our studies show that BET inhibitors are a promising new therapeutic for OS.


Subject(s)
Apoptosis/drug effects , Osteosarcoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Azepines/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Down-Regulation/drug effects , Drug Synergism , Gene Knockdown Techniques , Humans , Mice , Osteosarcoma/drug therapy , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factors/metabolism , Triazoles/pharmacology
8.
PLoS Genet ; 11(4): e1005160, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25859855

ABSTRACT

RECQL4 mutations are associated with Rothmund Thomson Syndrome (RTS), RAPADILINO Syndrome and Baller-Gerold Syndrome. These patients display a range of benign skeletal abnormalities such as low bone mass. In addition, RTS patients have a highly increased incidence of osteosarcoma (OS). The role of RECQL4 in normal adult bone development and homeostasis is largely uncharacterized and how mutation of RECQL4 contributes to OS susceptibility is not known. We hypothesised that Recql4 was required for normal skeletal development and both benign and malignant osteoblast function, which we have tested in the mouse. Recql4 deletion in vivo at the osteoblastic progenitor stage of differentiation resulted in mice with shorter bones and reduced bone volume, assessed at 9 weeks of age. This was associated with an osteoblast intrinsic decrease in mineral apposition rate and bone formation rate in the Recql4-deficient cohorts. Deletion of Recql4 in mature osteoblasts/osteocytes in vivo, however, did not cause a detectable phenotype. Acute deletion of Recql4 in primary osteoblasts or shRNA knockdown in an osteoblastic cell line caused failed proliferation, accompanied by cell cycle arrest, induction of apoptosis and impaired differentiation. When cohorts of animals were aged long term, the loss of Recql4 alone was not sufficient to initiate OS. We then crossed the Recql4fl/fl allele to a fully penetrant OS model (Osx-Cre p53fl/fl). Unexpectedly, the Osx-Cre p53fl/flRecql4fl/fl (dKO) animals had a significantly increased OS-free survival compared to Osx-Cre p53fl/fl or Osx-Cre p53fl/flRecql4fl/+ (het) animals. The extended survival was explained when the Recql4 status in the tumors that arose was assessed, and in no case was there complete deletion of Recql4 in the dKO OS. These data provide a mechanism for the benign skeletal phenotypes of RECQL4 mutation syndromes. We propose that tumor suppression and osteosarcoma susceptibility are most likely a function of mutant, not null, alleles of RECQL4.


Subject(s)
Bone Neoplasms/genetics , Osteoblasts/metabolism , Osteogenesis , Osteosarcoma/genetics , RecQ Helicases/metabolism , Animals , Bone Neoplasms/metabolism , Cell Proliferation , Gene Deletion , Mice , Mice, Inbred C57BL , Osteosarcoma/metabolism , RecQ Helicases/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Mol Cell Oncol ; 1(4): e965624, 2014.
Article in English | MEDLINE | ID: mdl-27308361

ABSTRACT

In osteosarcoma, knockdown of the parathyroid hormone-related protein (PTHrP) receptor reduces activation through cyclic AMP-dependent protein kinase A (PKA) and substantially decreases tumor differentiation, invasion, and proliferation in vivo. These findings complement other evidence supporting a central role of the PKA pathway in osteosarcoma biology and pathogenesis.

10.
Nucleic Acids Res ; 40(4): e30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22156059

ABSTRACT

Chromatin immunoprecipitation coupled with massive parallel sequencing (ChIP-seq) is increasingly used to map protein-chromatin interactions at global scale. The comparison of ChIP-seq profiles for RNA polymerase II (PolII) established in different biological contexts, such as specific developmental stages or specific time-points during cell differentiation, provides not only information about the presence/accumulation of PolII at transcription start sites (TSSs) but also about functional features of transcription, including PolII stalling, pausing and transcript elongation. However, annotation and normalization tools for comparative studies of multiple samples are currently missing. Here, we describe the R-package POLYPHEMUS, which integrates TSS annotation with PolII enrichment over TSSs and coding regions, and normalizes signal intensity profiles. Thereby POLYPHEMUS facilitates to extract information about global PolII action to reveal changes in the functional state of genes. We validated POLYPHEMUS using a kinetic study on retinoic acid-induced differentiation and a publicly available data set from a comparative PolII ChIP-seq profiling in Caenorhabditis elegans. We demonstrate that POLYPHEMUS corrects the data sets by normalizing for technical variation between samples and reveal the potential of the algorithm in comparing multiple data sets to infer features of transcription regulation from dynamic PolII binding profiles.


Subject(s)
Chromatin Immunoprecipitation , High-Throughput Nucleotide Sequencing , RNA Polymerase II/metabolism , Software , Algorithms , Animals , Binding Sites , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Cell Differentiation , Chromatin/genetics , Data Interpretation, Statistical , Sequence Analysis, DNA , Transcription Initiation Site
11.
Mol Syst Biol ; 7: 538, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21988834

ABSTRACT

Retinoic acid (RA) triggers physiological processes by activating heterodimeric transcription factors (TFs) comprising retinoic acid receptor (RARα, ß, γ) and retinoid X receptor (RXRα, ß, γ). How a single signal induces highly complex temporally controlled networks that ultimately orchestrate physiological processes is unclear. Using an RA-inducible differentiation model, we defined the temporal changes in the genome-wide binding patterns of RARγ and RXRα and correlated them with transcription regulation. Unexpectedly, both receptors displayed a highly dynamic binding, with different RXRα heterodimers targeting identical loci. Comparison of RARγ and RXRα co-binding at RA-regulated genes identified putative RXRα-RARγ target genes that were validated with subtype-selective agonists. Gene-regulatory decisions during differentiation were inferred from TF-target gene information and temporal gene expression. This analysis revealed six distinct co-expression paths of which RXRα-RARγ is associated with transcription activation, while Sox2 and Egr1 were predicted to regulate repression. Finally, RXRα-RARγ regulatory networks were reconstructed through integration of functional co-citations. Our analysis provides a dynamic view of RA signalling during cell differentiation, reveals RAR heterodimer dynamics and promiscuity, and predicts decisions that diversify the RA signal into distinct gene-regulatory programs.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Genomics/methods , Receptors, Retinoic Acid/metabolism , Retinoid X Receptor alpha/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Embryonic Stem Cells/cytology , Gene Expression Profiling , Mice , Molecular Sequence Data , Protein Binding/genetics , Receptor Cross-Talk/drug effects , Receptors, Retinoic Acid/genetics , Retinoid X Receptor alpha/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Retinoic Acid Receptor gamma
12.
Nat Methods ; 8(7): 565-7, 2011 Jun 05.
Article in English | MEDLINE | ID: mdl-21642965

ABSTRACT

Genome-wide profiling of transcription factors based on massive parallel sequencing of immunoprecipitated chromatin (ChIP-seq) requires nanogram amounts of DNA. Here we describe a high-fidelity, single-tube linear DNA amplification method (LinDA) for ChIP-seq and reChIP-seq with picogram DNA amounts obtained from a few thousand cells. This amplification technology will facilitate global analyses of transcription-factor binding and chromatin with very small cell populations, such as stem or cancer-initiating cells.


Subject(s)
Chromatin Immunoprecipitation , Chromatin/metabolism , DNA/analysis , Nucleic Acid Amplification Techniques/methods , Sequence Analysis, DNA/methods , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line, Tumor , Chromatin/genetics , DNA/genetics , Humans , Mice
13.
Genes Dev ; 25(11): 1132-46, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21632823

ABSTRACT

Multiple signaling pathways ultimately modulate the epigenetic information embedded in the chromatin of gene promoters by recruiting epigenetic enzymes. We found that, in estrogen-regulated gene programming, the acetyltransferase CREB-binding protein (CBP) is specifically and exclusively methylated by the coactivator-associated arginine methyltransferase (CARM1) in vivo. CARM1-dependent CBP methylation and p160 coactivators were required for estrogen-induced recruitment to chromatin targets. Notably, methylation increased the histone acetyltransferase (HAT) activity of CBP and stimulated its autoacetylation. Comparative genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) studies revealed a variety of patterns by which p160, CBP, and methyl-CBP (meCBP) are recruited (or not) by estrogen to chromatin targets. Moreover, significant target gene-specific variation in the recruitment of (1) the p160 RAC3 protein, (2) the fraction of a given meCBP species within the total CBP, and (3) the relative recruitment of different meCBP species suggests the existence of a target gene-specific "fingerprint" for coregulator recruitment. Crossing ChIP-seq and transcriptomics profiles revealed the existence of meCBP "hubs" within the network of estrogen-regulated genes. Together, our data provide evidence for an unprecedented mechanism by which CARM1-dependent CBP methylation results in gene-selective association of estrogen-recruited meCBP species with different HAT activities and specifies distinct target gene hubs, thus diversifying estrogen receptor programming.


Subject(s)
CREB-Binding Protein/metabolism , Chromatin/metabolism , Estrogens/metabolism , Gene Expression Regulation , Acetylation , Binding Sites , Cell Line, Tumor , Coenzymes/metabolism , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genome/genetics , Histone Acetyltransferases/metabolism , Humans , Methylation , Protein Binding/drug effects , Protein-Arginine N-Methyltransferases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...