Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Oncogene ; 37(14): 1911-1925, 2018 04.
Article in English | MEDLINE | ID: mdl-29367767

ABSTRACT

The N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) mouse model is an attractive model system of muscle-invasive bladder cancer (MIBC) as it recapitulates the histology of human tumors in a background with intact immune system. However, it was unknown whether this carcinogen-induced model also mimicked human MIBC at the molecular and mutational level. In our study, we analyzed gene expression and mutational landscape of the BBN model by next-generation sequencing followed by a bioinformatic comparison to human MIBC using data from The Cancer Genome Atlas and other repositories. BBN tumors showed overexpression of markers of basal cancer subtype, and had a high mutation burden with frequent Trp53 (80%), Kmt2d (70%), and Kmt2c (90%) mutations by exome sequencing, similar to human MIBC. Many variants corresponded to human cancer hotspot mutations, supporting their role as driver mutations. We extracted two novel mutational signatures from the BBN mouse genomes. The integrated analysis of mutation frequencies and signatures highlighted the contribution of aberrations to chromatin regulators and genetic instability in the BBN tumors. Together, our study revealed several similarities between human MIBC and the BBN mouse model, providing a strong rationale for its use in molecular and drug discovery studies.


Subject(s)
Carcinogens , Carcinoma, Transitional Cell/chemically induced , Carcinoma, Transitional Cell/pathology , Muscle Neoplasms/secondary , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/pathology , Animals , Carcinoma, Transitional Cell/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Microarray Analysis , Muscle Neoplasms/chemically induced , Muscle Neoplasms/genetics , Mutation , Neoplasm Invasiveness , Transcriptome , Urinary Bladder Neoplasms/genetics
2.
Cell Rep ; 21(3): 628-640, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29045832

ABSTRACT

Loss or inactivation of the histone H3K27 demethylase UTX occurs in several malignancies, including multiple myeloma (MM). Using an isogenic cell system, we found that loss of UTX leads to deactivation of gene expression ultimately promoting the proliferation, clonogenicity, adhesion, and tumorigenicity of MM cells. Moreover, UTX mutant cells showed increased in vitro and in vivo sensitivity to inhibition of EZH2, a histone methyltransferase that generates H3K27me3. Such sensitivity was related to a decrease in the levels of IRF4 and c-MYC and an activation of repressors of IRF4 characteristic of germinal center B cells such as BCL6 and IRF1. Rebalance of H3K27me3 levels at specific genes through EZH2 inhibitors may be a therapeutic strategy in MM cases harboring UTX mutations.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Demethylases/deficiency , Multiple Myeloma/pathology , Nuclear Proteins/deficiency , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Clone Cells , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases/metabolism , Histones/metabolism , Indazoles/pharmacology , Interferon Regulatory Factors/metabolism , Lysine/metabolism , Methylation , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/genetics , Mutation/genetics , Nuclear Proteins/metabolism , Phenotype , Pyridones/pharmacology , Transcription, Genetic/drug effects
3.
PLoS One ; 8(9): e75012, 2013.
Article in English | MEDLINE | ID: mdl-24040378

ABSTRACT

There is compelling in vivo evidence from reports on human genetic mutations and transgenic mice that some microRNAs (miRNAs) play an important functional role in regulating skeletal development and growth. A number of published in vitro studies also point toward a role for miRNAs in controlling chondrocyte gene expression and differentiation. However, information on miRNAs that may regulate a specific phase of chondrocyte differentiation (i.e. production of progenitor, differentiated or hypertrophic chondrocytes) is lacking. To attempt to bridge this knowledge gap, we have investigated miRNA expression patterns in human embryonic cartilage tissue. Specifically, a developmental time point was selected, prior to endochondral ossification in the embryonic limb, to permit analysis of three distinct populations of chondrocytes. The location of chondroprogenitor cells, differentiated chondrocytes and hypertrophic chondrocytes in gestational day 54-56 human embryonic limb tissue sections was confirmed both histologically and by specific collagen expression patterns. Laser capture microdissection was utilized to separate the three chondrocyte populations and a miRNA profiling study was carried out using TaqMan® OpenArray® Human MicroRNA Panels (Applied Biosystems®). Here we report on abundantly expressed miRNAs in human embryonic cartilage tissue and, more importantly, we have identified miRNAs that are significantly differentially expressed between precursor, differentiated and hypertrophic chondrocytes by 2-fold or more. Some of the miRNAs identified in this study have been described in other aspects of cartilage or bone biology, while others have not yet been reported in chondrocytes. Finally, a bioinformatics approach was applied to begin to decipher developmental cellular pathways that may be regulated by groups of differentially expressed miRNAs during distinct stages of chondrogenesis. Data obtained from this work will serve as an important resource of information for the field of cartilage biology and will enhance our understanding of miRNA-driven mechanisms regulating cartilage and endochondral bone development, regeneration and repair.


Subject(s)
Cartilage/embryology , Cartilage/metabolism , Chondrocytes/cytology , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Bone and Bones/metabolism , Cell Differentiation , Chondrocytes/metabolism , Computational Biology , Femur/embryology , Gene Expression Profiling , Humans , Laser Capture Microdissection , Microscopy, Fluorescence , Regeneration , Stem Cells/cytology , Tibia/embryology
4.
BMC Genomics ; 14: 84, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23387820

ABSTRACT

BACKGROUND: The regenerative response of Schwann cells after peripheral nerve injury is a critical process directly related to the pathophysiology of a number of neurodegenerative diseases. This SC injury response is dependent on an intricate gene regulatory program coordinated by a number of transcription factors and microRNAs, but the interactions among them remain largely unknown. Uncovering the transcriptional and post-transcriptional regulatory networks governing the Schwann cell injury response is a key step towards a better understanding of Schwann cell biology and may help develop novel therapies for related diseases. Performing such comprehensive network analysis requires systematic bioinformatics methods to integrate multiple genomic datasets. RESULTS: In this study we present a computational pipeline to infer transcription factor and microRNA regulatory networks. Our approach combined mRNA and microRNA expression profiling data, ChIP-Seq data of transcription factors, and computational transcription factor and microRNA target prediction. Using mRNA and microRNA expression data collected in a Schwann cell injury model, we constructed a regulatory network and studied regulatory pathways involved in Schwann cell response to injury. Furthermore, we analyzed network motifs and obtained insights on cooperative regulation of transcription factors and microRNAs in Schwann cell injury recovery. CONCLUSIONS: This work demonstrates a systematic method for gene regulatory network inference that may be used to gain new information on gene regulation by transcription factors and microRNAs.


Subject(s)
Computational Biology/methods , Gene Regulatory Networks , MicroRNAs/genetics , MicroRNAs/metabolism , Peripheral Nerve Injuries/pathology , Schwann Cells/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Early Growth Response Protein 2/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genomics , Humans , Mice , Myelin Sheath/physiology , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Blood ; 121(9): 1633-43, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23297133

ABSTRACT

Acute myeloid leukemia (AML) is characterized by dysregulated gene expression and abnormal patterns of DNA methylation; the relationship between these events is unclear. Many AML patients are now being treated with hypomethylating agents, such as decitabine (DAC), although the mechanisms by which it induces remissions remain unknown. The goal of this study was to use a novel stromal coculture assay that can expand primary AML cells to identify the immediate changes induced by DAC with a dose (100nM) that decreases total 5-methylcytosine content and reactivates imprinted genes (without causing myeloid differentiation, which would confound downstream genomic analyses). Using array-based technologies, we found that DAC treatment caused global hypomethylation in all samples (with a preference for regions with higher levels of baseline methylation), yet there was limited correlation between changes in methylation and gene expression. Moreover, the patterns of methylation and gene expression across the samples were primarily determined by the intrinsic properties of the primary cells, rather than DAC treatment. Although DAC induces hypomethylation, we could not identify canonical target genes that are altered by DAC in primary AML cells, suggesting that the mechanism of action of DAC is more complex than previously recognized.


Subject(s)
Azacitidine/analogs & derivatives , Gene Expression Regulation, Leukemic/drug effects , Leukemia, Myeloid, Acute/genetics , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/administration & dosage , Azacitidine/pharmacology , Cells, Cultured , CpG Islands/drug effects , CpG Islands/genetics , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Dose-Response Relationship, Drug , Gene Expression Profiling , Genome, Human/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Microarray Analysis , Primary Cell Culture , Time Factors
6.
PLoS One ; 7(10): e46529, 2012.
Article in English | MEDLINE | ID: mdl-23056333

ABSTRACT

Because PML-RARA-induced acute promyelocytic leukemia (APL) is a morphologically differentiated leukemia, many groups have speculated about whether its leukemic cell of origin is a committed myeloid precursor (e.g. a promyelocyte) versus an hematopoietic stem/progenitor cell (HSPC). We originally targeted PML-RARA expression with CTSG regulatory elements, based on the early observation that this gene was maximally expressed in cells with promyelocyte morphology. Here, we show that both Ctsg, and PML-RARA targeted to the Ctsg locus (in Ctsg-PML-RARA mice), are expressed in the purified KLS cells of these mice (KLS = Kit(+)Lin(-)Sca(+), which are highly enriched for HSPCs), and this expression results in biological effects in multi-lineage competitive repopulation assays. Further, we demonstrate the transcriptional consequences of PML-RARA expression in Ctsg-PML-RARA mice in early myeloid development in other myeloid progenitor compartments [common myeloid progenitors (CMPs) and granulocyte/monocyte progenitors (GMPs)], which have a distinct gene expression signature compared to wild-type (WT) mice. Although PML-RARA is indeed expressed at high levels in the promyelocytes of Ctsg-PML-RARA mice and alters the transcriptional signature of these cells, it does not induce their self-renewal. In sum, these results demonstrate that in the Ctsg-PML-RARA mouse model of APL, PML-RARA is expressed in and affects the function of multipotent progenitor cells. Finally, since PML/Pml is normally expressed in the HSPCs of both humans and mice, and since some human APL samples contain TCR rearrangements and express T lineage genes, we suggest that the very early hematopoietic expression of PML-RARA in this mouse model may closely mimic the physiologic expression pattern of PML-RARA in human APL patients.


Subject(s)
Cathepsin G/genetics , Hematopoietic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Animals , Base Sequence , Cell Separation , DNA Primers , Flow Cytometry , Gene Expression Profiling , Mice , Polymerase Chain Reaction
7.
Cell ; 150(2): 264-78, 2012 Jul 20.
Article in English | MEDLINE | ID: mdl-22817890

ABSTRACT

Most mutations in cancer genomes are thought to be acquired after the initiating event, which may cause genomic instability and drive clonal evolution. However, for acute myeloid leukemia (AML), normal karyotypes are common, and genomic instability is unusual. To better understand clonal evolution in AML, we sequenced the genomes of M3-AML samples with a known initiating event (PML-RARA) versus the genomes of normal karyotype M1-AML samples and the exomes of hematopoietic stem/progenitor cells (HSPCs) from healthy people. Collectively, the data suggest that most of the mutations found in AML genomes are actually random events that occurred in HSPCs before they acquired the initiating mutation; the mutational history of that cell is "captured" as the clone expands. In many cases, only one or two additional, cooperating mutations are needed to generate the malignant founding clone. Cells from the founding clone can acquire additional cooperating mutations, yielding subclones that can contribute to disease progression and/or relapse.


Subject(s)
Clonal Evolution , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , Aged , DNA Mutational Analysis , Disease Progression , Female , Genome-Wide Association Study , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/physiopathology , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Recurrence , Skin/metabolism , Young Adult
8.
JAMA ; 305(15): 1568-76, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21505135

ABSTRACT

CONTEXT: The identification of patients with inherited cancer susceptibility syndromes facilitates early diagnosis, prevention, and treatment. However, in many cases of suspected cancer susceptibility, the family history is unclear and genetic testing of common cancer susceptibility genes is unrevealing. OBJECTIVE: To apply whole-genome sequencing to a patient without any significant family history of cancer but with suspected increased cancer susceptibility because of multiple primary tumors to identify rare or novel germline variants in cancer susceptibility genes. DESIGN, SETTING, AND PARTICIPANT: Skin (normal) and bone marrow (leukemia) DNA were obtained from a patient with early-onset breast and ovarian cancer (negative for BRCA1 and BRCA2 mutations) and therapy-related acute myeloid leukemia (t-AML) and analyzed with the following: whole-genome sequencing using paired-end reads, single-nucleotide polymorphism (SNP) genotyping, RNA expression profiling, and spectral karyotyping. MAIN OUTCOME MEASURES: Structural variants, copy number alterations, single-nucleotide variants, and small insertions and deletions (indels) were detected and validated using the described platforms. RESULTS; Whole-genome sequencing revealed a novel, heterozygous 3-kilobase deletion removing exons 7-9 of TP53 in the patient's normal skin DNA, which was homozygous in the leukemia DNA as a result of uniparental disomy. In addition, a total of 28 validated somatic single-nucleotide variations or indels in coding genes, 8 somatic structural variants, and 12 somatic copy number alterations were detected in the patient's leukemia genome. CONCLUSION: Whole-genome sequencing can identify novel, cryptic variants in cancer susceptibility genes in addition to providing unbiased information on the spectrum of mutations in a cancer genome.


Subject(s)
Genes, p53/genetics , Genetic Predisposition to Disease , Leukemia, Myeloid, Acute/genetics , Sequence Analysis, DNA , Sequence Deletion , Adult , Age of Onset , Breast Neoplasms/therapy , Cystadenocarcinoma, Serous/therapy , DNA, Neoplasm/genetics , Female , Genome, Human/genetics , Humans , Leukemia, Myeloid, Acute/etiology , Ovarian Neoplasms/therapy , Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics
9.
J Clin Invest ; 121(4): 1445-55, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21436584

ABSTRACT

Acute promyelocytic leukemia (APL) is a subtype of acute myeloid leukemia (AML). It is characterized by the t(15;17)(q22;q11.2) chromosomal translocation that creates the promyelocytic leukemia-retinoic acid receptor α (PML-RARA) fusion oncogene. Although this fusion oncogene is known to initiate APL in mice, other cooperating mutations, as yet ill defined, are important for disease pathogenesis. To identify these, we used a mouse model of APL, whereby PML-RARA expressed in myeloid cells leads to a myeloproliferative disease that ultimately evolves into APL. Sequencing of a mouse APL genome revealed 3 somatic, nonsynonymous mutations relevant to APL pathogenesis, of which 1 (Jak1 V657F) was found to be recurrent in other affected mice. This mutation was identical to the JAK1 V658F mutation previously found in human APL and acute lymphoblastic leukemia samples. Further analysis showed that JAK1 V658F cooperated in vivo with PML-RARA, causing a rapidly fatal leukemia in mice. We also discovered a somatic 150-kb deletion involving the lysine (K)-specific demethylase 6A (Kdm6a, also known as Utx) gene, in the mouse APL genome. Similar deletions were observed in 3 out of 14 additional mouse APL samples and 1 out of 150 human AML samples. In conclusion, whole genome sequencing of mouse cancer genomes can provide an unbiased and comprehensive approach for discovering functionally relevant mutations that are also present in human leukemias.


Subject(s)
Leukemia, Promyelocytic, Acute/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , DNA, Neoplasm/genetics , Disease Progression , Humans , Janus Kinase 1/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, Experimental/genetics , Mice , Mice, 129 Strain , Molecular Sequence Data , Mutation , Oncogene Proteins, Fusion/genetics , Polymorphism, Single Nucleotide , Sequence Deletion , Sequence Homology, Amino Acid
10.
Blood ; 117(8): 2460-8, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21190992

ABSTRACT

RARA (retinoic acid receptor alpha) haploinsufficiency is an invariable consequence of t(15;17)(q22;q21) translocations in acute promyelocytic leukemia (APL). Retinoids and RARA activity have been implicated in hematopoietic self-renewal and neutrophil maturation. We and others therefore predicted that RARA haploinsufficiency would contribute to APL pathogenesis. To test this hypothesis, we crossed Rara(+/-) mice with mice expressing PML (promyelocytic leukemia)-RARA from the cathepsin G locus (mCG-PR). We found that Rara haploinsufficiency cooperated with PML-RARA, but only modestly influenced the preleukemic and leukemic phenotype. Bone marrow from mCG-PR(+/-) × Rara(+/-) mice had decreased numbers of mature myeloid cells, increased ex vivo myeloid cell proliferation, and increased competitive advantage after transplantation. Rara haploinsufficiency did not alter mCG-PR-dependent leukemic latency or penetrance, but did influence the distribution of leukemic cells; leukemia in mCG-PR(+/-) × Rara(+/-) mice presented more commonly with low to normal white blood cell counts and with myeloid infiltration of lymph nodes. APL cells from these mice were responsive to all-trans retinoic acid and had virtually no differences in expression profiling compared with tumors arising in mCG-PR(+/-) × Rara(+/+) mice. These data show that Rara haploinsufficiency (like Pml haploinsufficiency and RARA-PML) can cooperate with PML-RARA to influence the pathogenesis of APL in mice, but that PML-RARA is the t(15;17) disease-initiating mutation.


Subject(s)
Haploinsufficiency/physiology , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Receptors, Retinoic Acid/genetics , Animals , Bone Marrow/pathology , Gene Expression Profiling , Mice , Mice, Mutant Strains , Myeloid Cells/pathology , Oncogene Proteins, Fusion/genetics , Phenotype , Retinoic Acid Receptor alpha
11.
N Engl J Med ; 363(25): 2424-33, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21067377

ABSTRACT

BACKGROUND: The genetic alterations responsible for an adverse outcome in most patients with acute myeloid leukemia (AML) are unknown. METHODS: Using massively parallel DNA sequencing, we identified a somatic mutation in DNMT3A, encoding a DNA methyltransferase, in the genome of cells from a patient with AML with a normal karyotype. We sequenced the exons of DNMT3A in 280 additional patients with de novo AML to define recurring mutations. RESULTS: A total of 62 of 281 patients (22.1%) had mutations in DNMT3A that were predicted to affect translation. We identified 18 different missense mutations, the most common of which was predicted to affect amino acid R882 (in 37 patients). We also identified six frameshift, six nonsense, and three splice-site mutations and a 1.5-Mbp deletion encompassing DNMT3A. These mutations were highly enriched in the group of patients with an intermediate-risk cytogenetic profile (56 of 166 patients, or 33.7%) but were absent in all 79 patients with a favorable-risk cytogenetic profile (P<0.001 for both comparisons). The median overall survival among patients with DNMT3A mutations was significantly shorter than that among patients without such mutations (12.3 months vs. 41.1 months, P<0.001). DNMT3A mutations were associated with adverse outcomes among patients with an intermediate-risk cytogenetic profile or FLT3 mutations, regardless of age, and were independently associated with a poor outcome in Cox proportional-hazards analysis. CONCLUSIONS: DNMT3A mutations are highly recurrent in patients with de novo AML with an intermediate-risk cytogenetic profile and are independently associated with a poor outcome. (Funded by the National Institutes of Health and others.).


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Adult , DNA Methylation , DNA Methyltransferase 3A , DNA Mutational Analysis/methods , Female , Frameshift Mutation , Gene Expression , Humans , Karyotyping , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Nucleic Acid Amplification Techniques , Prognosis , Proportional Hazards Models , Survival Analysis
12.
PLoS One ; 5(8): e12464, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20805992

ABSTRACT

Different fusion oncogenes in acute myeloid leukemia (AML) have distinct clinical and laboratory features suggesting different modes of malignant transformation. Here we compare the in vitro effects of representatives of 4 major groups of AML fusion oncogenes on primary human CD34+ cells. As expected from their clinical similarities, MLL-AF9 and NUP98-HOXA9 had very similar effects in vitro. They both caused erythroid hyperplasia and a clear block in erythroid and myeloid maturation. On the other hand, AML1-ETO and PML-RARA had only modest effects on myeloid and erythroid differentiation. All oncogenes except PML-RARA caused a dramatic increase in long-term proliferation and self-renewal. Gene expression profiling revealed two distinct temporal patterns of gene deregulation. Gene deregulation by MLL-AF9 and NUP98-HOXA9 peaked 3 days after transduction. In contrast, the vast majority of gene deregulation by AML1-ETO and PML-RARA occurred within 6 hours, followed by a dramatic drop in the numbers of deregulated genes. Interestingly, the p53 inhibitor MDM2 was upregulated by AML1-ETO at 6 hours. Nutlin-3, an inhibitor of the interaction between MDM2 and p53, specifically inhibited the proliferation and self-renewal of primary human CD34+ cells transduced with AML1-ETO, suggesting that MDM2 upregulation plays a role in cell transformation by AML1-ETO. These data show that differences among AML fusion oncogenes can be recapitulated in vitro using primary human CD34+ cells and that early gene expression profiling in these cells can reveal potential drug targets in AML.


Subject(s)
Antigens, CD34/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Oncogene Fusion , Oncogenes/genetics , Cell Differentiation/genetics , Cell Line , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...