Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS J ; 288(23): 6828-6843, 2021 12.
Article in English | MEDLINE | ID: mdl-34258867

ABSTRACT

Adverse fetal environment, in particular a shortage or excess of nutrients, is associated with increased risks of metabolic diseases later in life. However, the molecular mechanisms underlying this developmental origin of adult diseases remain unclear. Here, we directly tested the role of mitochondrial stress in mediating fetal programming in mice by enzymatically depleting mtDNA in zygotes. mtDNA-targeted plasmid microinjection is used to reduce embryonic mtDNA copy number directly, followed by embryo transfer. Mice with reduced zygote mtDNA copy number were born morphologically normal and showed no accelerated body weight gain. However, at 5 months of age these mice showed markedly increased hepatic lipidosis and became glucose-intolerant. Hepatic mRNA and protein expressions of peroxisome proliferator-activated receptor α (Pparα), a key transcriptional regulator of lipid metabolism, were significantly decreased as a result of increased DNA methylation in its proximal regulatory region. These results indicate that perturbation of mitochondrial function around the periconceptional period causes hypermethylation and thus suppressed expression of PPARα in fetal liver, leading to impaired hepatic lipid metabolism. Our findings provide the first direct evidence that mitochondrial stress mediates epigenetic changes associated with fetal programming of adult diseases in a mammalian system.


Subject(s)
DNA Copy Number Variations , DNA, Mitochondrial/genetics , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Lipid Metabolism/genetics , Lipolysis/genetics , Liver/metabolism , Age Factors , Animals , DNA Methylation , Embryo, Mammalian/embryology , Epigenomics/methods , Female , Gene Expression Regulation, Developmental , Liver/embryology , Male , Membrane Potential, Mitochondrial/genetics , Mice, Inbred ICR , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/physiology , Oxygen Consumption/genetics , PPAR alpha/genetics , PPAR alpha/metabolism , RNA-Seq/methods
2.
J Clin Invest ; 128(4): 1442-1457, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29360641

ABSTRACT

Aberrant activation of MAPK signaling leads to the activation of oncogenic transcriptomes. How MAPK signaling is coupled with the transcriptional response in cancer is not fully understood. In 2 MAPK-activated tumor types, gastrointestinal stromal tumor and melanoma, we found that ETV1 and other Pea3-ETS transcription factors are critical nuclear effectors of MAPK signaling that are regulated through protein stability. Expression of stabilized Pea3-ETS factors can partially rescue the MAPK transcriptome and cell viability after MAPK inhibition. To identify the players involved in this process, we performed a pooled genome-wide RNAi screen using a fluorescence-based ETV1 protein stability sensor and identified COP1, DET1, DDB1, UBE3C, PSMD4, and COP9 signalosome members. COP1 or DET1 loss led to decoupling between MAPK signaling and the downstream transcriptional response, where MAPK inhibition failed to destabilize Pea3 factors and fully inhibit the MAPK transcriptome, thus resulting in decreased sensitivity to MAPK pathway inhibitors. We identified multiple COP1 and DET1 mutations in human tumors that were defective in the degradation of Pea3-ETS factors. Two melanoma patients had de novo DET1 mutations arising after vemurafenib treatment. These observations indicate that MAPK signaling-dependent regulation of Pea3-ETS protein stability is a key signaling node in oncogenesis and therapeutic resistance to MAPK pathway inhibition.


Subject(s)
Carrier Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Melanoma/metabolism , Mutation , Proto-Oncogene Proteins c-ets/metabolism , Transcriptome/drug effects , Ubiquitin-Protein Ligases/metabolism , Vemurafenib/pharmacology , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Mice , Mice, SCID , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
3.
Cancer Discov ; 8(2): 234-251, 2018 02.
Article in English | MEDLINE | ID: mdl-29162563

ABSTRACT

The cellular context that integrates upstream signaling and downstream nuclear response dictates the oncogenic behavior and shapes treatment responses in distinct cancer types. Here, we uncover that in gastrointestinal stromal tumor (GIST), the forkhead family member FOXF1 directly controls the transcription of two master regulators, KIT and ETV1, both required for GIST precursor-interstitial cells of Cajal lineage specification and GIST tumorigenesis. Further, FOXF1 colocalizes with ETV1 at enhancers and functions as a pioneer factor that regulates the ETV1-dependent GIST lineage-specific transcriptome through modulation of the local chromatin context, including chromatin accessibility, enhancer maintenance, and ETV1 binding. Functionally, FOXF1 is required for human GIST cell growth in vitro and murine GIST tumor growth and maintenance in vivo The simultaneous control of the upstream signaling and nuclear response sets up a unique regulatory paradigm and highlights the critical role of FOXF1 in enforcing the GIST cellular context for highly lineage-restricted clinical behavior and treatment response.Significance: We uncover that FOXF1 defines the core-regulatory circuitry in GIST through both direct transcriptional regulation and pioneer factor function. The unique and simultaneous control of signaling and transcriptional circuitry by FOXF1 sets up an enforced transcriptional addiction to FOXF1 in GIST, which can be exploited diagnostically and therapeutically. Cancer Discov; 8(2); 234-51. ©2017 AACR.See related commentary by Lee and Duensing, p. 146This article is highlighted in the In This Issue feature, p. 127.


Subject(s)
Forkhead Transcription Factors/genetics , Gastrointestinal Stromal Tumors/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Animals , Biomarkers, Tumor , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gastrointestinal Stromal Tumors/metabolism , Gene Expression Profiling , Heterografts , Humans , Protein Binding , Signal Transduction , Transcription Factors/genetics , Transcriptome
4.
Cancer Discov ; 5(3): 304-15, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25572173

ABSTRACT

UNLABELLED: Gastrointestinal stromal tumor (GIST), originating from the interstitial cells of Cajal (ICC), is characterized by frequent activating mutations of the KIT receptor tyrosine kinase. Despite the clinical success of imatinib, which targets KIT, most patients with advanced GIST develop resistance and eventually die of the disease. The ETS family transcription factor ETV1 is a master regulator of the ICC lineage. Using mouse models of Kit activation and Etv1 ablation, we demonstrate that ETV1 is required for GIST initiation and proliferation in vivo, validating it as a therapeutic target. We further uncover a positive feedback circuit where MAP kinase activation downstream of KIT stabilizes the ETV1 protein, and ETV1 positively regulates KIT expression. Combined targeting of ETV1 stability by imatinib and MEK162 resulted in increased growth suppression in vitro and complete tumor regression in vivo. The combination strategy to target ETV1 may provide an effective therapeutic strategy in GIST clinical management. SIGNIFICANCE: ETV1 is a lineage-specific oncogenic transcription factor required for the growth and survival of GIST. We describe a novel strategy of targeting ETV1 protein stability by the combination of MEK and KIT inhibitors that synergistically suppress tumor growth. This strategy has the potential to change first-line therapy in GIST clinical management.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/metabolism , Gastrointestinal Stromal Tumors/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Drug Synergism , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
5.
Cell ; 159(1): 176-187, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25201530

ABSTRACT

The lack of in vitro prostate cancer models that recapitulate the diversity of human prostate cancer has hampered progress in understanding disease pathogenesis and therapy response. Using a 3D organoid system, we report success in long-term culture of prostate cancer from biopsy specimens and circulating tumor cells. The first seven fully characterized organoid lines recapitulate the molecular diversity of prostate cancer subtypes, including TMPRSS2-ERG fusion, SPOP mutation, SPINK1 overexpression, and CHD1 loss. Whole-exome sequencing shows a low mutational burden, consistent with genomics studies, but with mutations in FOXA1 and PIK3R1, as well as in DNA repair and chromatin modifier pathways that have been reported in advanced disease. Loss of p53 and RB tumor suppressor pathway function are the most common feature shared across the organoid lines. The methodology described here should enable the generation of a large repertoire of patient-derived prostate cancer lines amenable to genetic and pharmacologic studies.


Subject(s)
Culture Techniques , Organoids , Prostatic Neoplasms/pathology , Heterografts , Humans , Male , Neoplasm Metastasis/pathology , Organoids/pathology , Pharmacology/methods , Tumor Suppressor Proteins/metabolism
6.
Nat Med ; 19(8): 1023-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23817021

ABSTRACT

Studies of ETS-mediated prostate oncogenesis have been hampered by a lack of suitable experimental systems. Here we describe a new conditional mouse model that shows robust, homogenous ERG expression throughout the prostate. When combined with homozygous Pten loss, the mice developed accelerated, highly penetrant invasive prostate cancer. In mouse prostate tissue, ERG markedly increased androgen receptor (AR) binding. Robust ERG-mediated transcriptional changes, observed only in the setting of Pten loss, included the restoration of AR transcriptional output and upregulation of genes involved in cell death, migration, inflammation and angiogenesis. Similarly, ETS variant 1 (ETV1) positively regulated the AR cistrome and transcriptional output in ETV1-translocated, PTEN-deficient human prostate cancer cells. In two large clinical cohorts, expression of ERG and ETV1 correlated with higher AR transcriptional output in PTEN-deficient prostate cancer specimens. We propose that ETS factors cause prostate-specific transformation by altering the AR cistrome, priming the prostate epithelium to respond to aberrant upstream signals such as PTEN loss.


Subject(s)
Cell Transformation, Neoplastic/pathology , Genes/genetics , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-ets/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Disease Models, Animal , Histones/metabolism , Humans , Lysine/metabolism , Male , Mice , Oncogene Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Phenotype , Principal Component Analysis , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Transcription Factors/metabolism , Transcriptional Regulator ERG , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...