Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4819, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844464

ABSTRACT

Neuronal differentiation requires building a complex intracellular architecture, and therefore the coordinated regulation of defined sets of genes. RNA-binding proteins (RBPs) play a key role in this regulation. However, while their action on individual mRNAs has been explored in depth, the mechanisms used to coordinate gene expression programs shaping neuronal morphology are poorly understood. To address this, we studied how the paradigmatic RBP IMP1 (IGF2BP1), an essential developmental factor, selects and regulates its RNA targets during the human neuronal differentiation. We perform a combination of system-wide and molecular analyses, revealing that IMP1 developmentally transitions to and directly regulates the expression of mRNAs encoding essential regulators of the microtubule network, a key component of neuronal morphology. Furthermore, we show that m6A methylation drives the selection of specific IMP1 mRNA targets and their protein expression during the developmental transition from neural precursors to neurons, providing a molecular principle for the onset of target selectivity.


Subject(s)
Cell Differentiation , Microtubules , Neurons , RNA, Messenger , RNA-Binding Proteins , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Microtubules/metabolism , Neurons/metabolism , Neurons/cytology , Cell Differentiation/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Methylation , Neurogenesis/genetics , Adenosine/metabolism , Adenosine/analogs & derivatives , Gene Expression Regulation, Developmental
2.
Wellcome Open Res ; 8: 403, 2023.
Article in English | MEDLINE | ID: mdl-38074197

ABSTRACT

Background: CD4 + Th1 cells producing IFN-γ are required to eradicate intracellular pathogens, however if uncontrolled these cells can cause immunopathology. The cytokine IL-10 is produced by multiple immune cells including Th1 cells during infection and regulates the immune response to minimise collateral host damage. In this study we aimed to elucidate the transcriptional network of genes controlling the expression of Il10 and proinflammatory cytokines, including Ifng in Th1 cells differentiated from mouse naive CD4 + T cells. Methods: We applied computational analysis of gene regulation derived from temporal profiling of gene expression clusters obtained from bulk RNA sequencing (RNA-seq) of flow cytometry sorted naïve CD4 + T cells from mouse spleens differentiated in vitro into Th1 effector cells with IL-12 and IL-27 to produce Ifng and Il10, compared to IL-27 alone which express Il10 only , or IL-12 alone which express Ifng and no Il10, or medium control driven-CD4 + T cells which do not express effector cytokines . Data were integrated with analysis of active genomic regions from these T cells using an assay for transposase-accessible chromatin with sequencing (ATAC)-seq, integrated with literature derived-Chromatin-immunoprecipitation (ChIP)-seq data and the RNA-seq data, to elucidate the transcriptional network of genes controlling expression of Il10 and pro-inflammatory effector genes in Th1 cells. The co-dominant role for the transcription factors, Prdm1 (encoding Blimp-1) and Maf (encoding c-Maf) , in cytokine gene regulation in Th1 cells, was confirmed using T cells obtained from mice with T-cell specific deletion of these transcription factors. Results: We show that the transcription factors Blimp-1 and c-Maf each have unique and common effects on cytokine gene regulation and not only co-operate to induce Il10 gene expression in IL-12 plus IL-27 differentiated mouse Th1 cells, but additionally directly negatively regulate key proinflammatory cytokines including Ifng, thus providing mechanisms for reinforcement of regulated Th1 cell responses. Conclusions: These data show that Blimp-1 and c-Maf positively and negatively regulate a network of both unique and common anti-inflammatory and pro-inflammatory genes to reinforce a Th1 response in mice that will eradicate pathogens with minimum immunopathology.

3.
Genome Res ; 33(9): 1497-1512, 2023 09.
Article in English | MEDLINE | ID: mdl-37582635

ABSTRACT

Neurons are morphologically complex cells that rely on the compartmentalization of protein expression to develop and maintain their cytoarchitecture. The targeting of RNA transcripts to axons is one of the mechanisms that allows rapid local translation of proteins in response to extracellular signals. 3' Untranslated regions (UTRs) of mRNA are noncoding sequences that play a critical role in determining transcript localization and translation by interacting with specific RNA-binding proteins (RBPs). However, how 3' UTRs contribute to mRNA metabolism and the nature of RBP complexes responsible for these functions remains elusive. We performed 3' end sequencing of RNA isolated from cell bodies and axons of sympathetic neurons exposed to either nerve growth factor (NGF) or neurotrophin 3 (NTF3, also known as NT-3). NGF and NTF3 are growth factors essential for sympathetic neuron development through distinct signaling mechanisms. Whereas NTF3 acts mostly locally, NGF signal is retrogradely transported from axons to cell bodies. We discovered that both NGF and NTF3 affect transcription and alternative polyadenylation in the nucleus and induce the localization of specific 3' UTR isoforms to axons, including short 3' UTR isoforms found exclusively in axons. The integration of our data with CLIP sequencing data supports a model whereby long 3' UTR isoforms associate with RBP complexes in the nucleus and, upon reaching the axons, are remodeled locally into shorter isoforms. Our findings shed new light into the complex relationship between nuclear polyadenylation, mRNA localization, and local 3' UTR remodeling in developing neurons.


Subject(s)
Axons , Nerve Growth Factor , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , 3' Untranslated Regions , Axons/metabolism , Protein Isoforms/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Nat Commun ; 14(1): 2176, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37080969

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) causes motor neuron degeneration, with 97% of cases exhibiting TDP-43 proteinopathy. Elucidating pathomechanisms has been hampered by disease heterogeneity and difficulties accessing motor neurons. Human induced pluripotent stem cell-derived motor neurons (iPSMNs) offer a solution; however, studies have typically been limited to underpowered cohorts. Here, we present a comprehensive compendium of 429 iPSMNs from 15 datasets, and 271 post-mortem spinal cord samples. Using reproducible bioinformatic workflows, we identify robust upregulation of p53 signalling in ALS in both iPSMNs and post-mortem spinal cord. p53 activation is greatest with C9orf72 repeat expansions but is weakest with SOD1 and FUS mutations. TDP-43 depletion potentiates p53 activation in both post-mortem neuronal nuclei and cell culture, thereby functionally linking p53 activation with TDP-43 depletion. ALS iPSMNs and post-mortem tissue display enrichment of splicing alterations, somatic mutations, and gene fusions, possibly contributing to the DNA damage response.


Subject(s)
Amyotrophic Lateral Sclerosis , DNA-Binding Proteins , Genomic Instability , Transcriptome , Alternative Splicing/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cadaver , Cohort Studies , Datasets as Topic , DNA Damage , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Fusion , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Mutation , Spinal Cord/metabolism , Transcriptome/genetics , Humans
6.
Genome Res ; 32(10): 1808-1825, 2022 10.
Article in English | MEDLINE | ID: mdl-36180233

ABSTRACT

Intron retention (IR) is now recognized as a dominant splicing event during motor neuron (MN) development; however, the role and regulation of intron-retaining transcripts (IRTs) localized to the cytoplasm remain particularly understudied. Here we show that IR is a physiological process that is spatiotemporally regulated during MN lineage restriction and that IRTs in the cytoplasm are detected in as many as 13% (n = 2297) of the genes expressed during this process. We identify a major class of cytoplasmic IRTs that are not associated with reduced expression of their own genes but instead show a high capacity for RNA-binding protein and miRNA occupancy. Finally, we show that ALS-causing VCP mutations lead to a selective increase in cytoplasmic abundance of this particular class of IRTs, which in turn temporally coincides with an increase in the nuclear expression level of predicted miRNA target genes. Altogether, our study identifies a previously unrecognized class of cytoplasmic intronic sequences with potential regulatory function beyond gene expression.


Subject(s)
MicroRNAs , Motor Neurons , Humans , Introns , Cytoplasm/genetics , Cytoplasm/metabolism , Neurogenesis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism
7.
Neuropathol Appl Neurobiol ; 48(2): e12770, 2022 02.
Article in English | MEDLINE | ID: mdl-34595747

ABSTRACT

AIMS: Although morphological attributes of cells and their substructures are recognised readouts of physiological or pathophysiological states, these have been relatively understudied in amyotrophic lateral sclerosis (ALS) research. METHODS: In this study, we integrate multichannel fluorescence high-content microscopy data with deep learning imaging methods to reveal-directly from unsegmented images-novel neurite-associated morphological perturbations associated with (ALS-causing) VCP-mutant human motor neurons (MNs). RESULTS: Surprisingly, we reveal that previously unrecognised disease-relevant information is withheld in broadly used and often considered 'generic' biological markers of nuclei (DAPI) and neurons ( ß III-tubulin). Additionally, we identify changes within the information content of ALS-related RNA binding protein (RBP) immunofluorescence imaging that is captured in VCP-mutant MN cultures. Furthermore, by analysing MN cultures exposed to different extrinsic stressors, we show that heat stress recapitulates key aspects of ALS. CONCLUSIONS: Our study therefore reveals disease-relevant information contained in a range of both generic and more specific fluorescent markers and establishes the use of image-based deep learning methods for rapid, automated and unbiased identification of biological hypotheses.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Deep Learning , Motor Neurons/metabolism , Neurites/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Phenotype
8.
Brain ; 144(7): 1985-1993, 2021 08 17.
Article in English | MEDLINE | ID: mdl-33693641

ABSTRACT

We recently described aberrantly increased cytoplasmic SFPQ intron-retaining transcripts (IRTs) and concurrent SFPQ protein mislocalization as new hallmarks of amyotrophic lateral sclerosis (ALS). However, the generalizability and potential roles of cytoplasmic IRTs in health and disease remain unclear. Here, using time-resolved deep sequencing of nuclear and cytoplasmic fractions of human induced pluripotent stem cells undergoing motor neurogenesis, we reveal that ALS-causing VCP gene mutations lead to compartment-specific aberrant accumulation of IRTs. Specifically, we identify >100 IRTs with increased cytoplasmic abundance in ALS samples. Furthermore, these aberrant cytoplasmic IRTs possess sequence-specific attributes and differential predicted binding affinity to RNA binding proteins. Remarkably, TDP-43, SFPQ and FUS-RNA binding proteins known for nuclear-to-cytoplasmic mislocalization in ALS-abundantly and specifically bind to this aberrant cytoplasmic pool of IRTs. Our data are therefore consistent with a novel role for cytoplasmic IRTs in regulating compartment-specific protein abundance. This study provides new molecular insight into potential pathomechanisms underlying ALS and highlights aberrant cytoplasmic IRTs as potential therapeutic targets.


Subject(s)
Amyotrophic Lateral Sclerosis , Cytoplasm/metabolism , Introns , RNA-Binding Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Humans , Mutation , Valosin Containing Protein/genetics
9.
Brain Pathol ; 31(4): e12937, 2021 07.
Article in English | MEDLINE | ID: mdl-33576079

ABSTRACT

Histopathological analysis of tissue sections is invaluable in neurodegeneration research. However, cell-to-cell variation in both the presence and severity of a given phenotype is a key limitation of this approach, reducing the signal to noise ratio and leaving unresolved the potential of single-cell scoring for a given disease attribute. Here, we tested different machine learning methods to analyse high-content microscopy measurements of hundreds of motor neurons (MNs) from amyotrophic lateral sclerosis (ALS) post-mortem tissue sections. Furthermore, we automated the identification of phenotypically distinct MN subpopulations in VCP- and SOD1-mutant transgenic mice, revealing common morphological cellular phenotypes. Additionally we established scoring metrics to rank cells and tissue samples for both disease probability and severity. By adapting this paradigm to human post-mortem tissue, we validated our core finding that morphological descriptors robustly discriminate ALS from control healthy tissue at single cell resolution. Determining disease presence, severity and unbiased phenotypes at single cell resolution might prove transformational in our understanding of ALS and neurodegeneration more broadly.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , Spinal Cord/pathology , Animals , Mice , Mice, Transgenic , Mitochondria/pathology , Motor Neurons/metabolism , Phenotype , Superoxide Dismutase/metabolism
10.
Cell Rep ; 34(8): 108778, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33626357

ABSTRACT

The 3' untranslated regions (3' UTRs) of messenger RNAs (mRNAs) are non-coding sequences involved in many aspects of mRNA metabolism, including intracellular localization and translation. Incorrect processing and delivery of mRNA cause severe developmental defects and have been implicated in many neurological disorders. Here, we use deep sequencing to show that in sympathetic neuron axons, the 3' UTRs of many transcripts undergo cleavage, generating isoforms that express the coding sequence with a short 3' UTR and stable 3' UTR-derived fragments of unknown function. Cleavage of the long 3' UTR of Inositol Monophosphatase 1 (IMPA1) mediated by a protein complex containing the endonuclease argonaute 2 (Ago2) generates a translatable isoform that is necessary for maintaining the integrity of sympathetic neuron axons. Thus, our study provides a mechanism of mRNA metabolism that simultaneously regulates local protein synthesis and generates an additional class of 3' UTR-derived RNAs.


Subject(s)
3' Untranslated Regions , Axons/enzymology , Cell Body/enzymology , Phosphoric Monoester Hydrolases/metabolism , RNA, Messenger/metabolism , Superior Cervical Ganglion/enzymology , Transcription, Genetic , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , ELAV-Like Protein 4/genetics , ELAV-Like Protein 4/metabolism , Female , Gene Expression Regulation, Enzymologic , Male , PC12 Cells , Phosphoric Monoester Hydrolases/genetics , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , Polyadenylation , Protein Biosynthesis , Protein Isoforms , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/cytology , Trans-Activators/genetics , Trans-Activators/metabolism
11.
Sci Rep ; 10(1): 21635, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33303789

ABSTRACT

Human mammary epithelial cells can proliferate and reorganize into polarized multi-cellular constructs in-vitro, thereby functioning as an important model system in recapitulating key steps of in-vivo morphogenesis. Current approaches to constructing such three-dimensional mimics of the in-vivo microenvironment have involved the use of complex and ill-defined naturally derived matrices, whose properties are difficult to manipulate independently, and which have therefore limited our ability to understand the extrinsic regulation of morphogenesis. Here, we employ an automated, high-throughput approach to array modular building blocks of synthetic components, and develop a systematic approach to analyze colonies resulting from these varied microenvironmental combinations. This methodology allows us to systematically map the relationship between microenvironmental properties and ensuing morphogenetic phenotypes. Our analysis reveals that apico-basal polarity of mammary epithelial cells occurs within a narrow range of matrix stiffness, and that phenotypic homogeneity is favored in matrices which are insensitive to MMP-mediated degradation. Furthermore, combinations of extracellular proteins in the matrix finely tune the morphology of the mammary colonies, suggesting that subtle disregulations of the microenvironment may play a significant role in pathological disease states. This approach, which leverages the combinatorial possibilities of modular synthetic artificial extracellular matrices with an automated technology platform, demonstrates how morphogenesis can be assessed systematically in 3D, and provides new insights into mammary epithelial multicellularity.


Subject(s)
Mammary Glands, Human/cytology , Morphogenesis , Cell Polarity , Cell Proliferation , Extracellular Matrix/metabolism , Humans , Mammary Glands, Human/metabolism
13.
Brain ; 142(9): 2572-2580, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31368485

ABSTRACT

Mutations causing amyotrophic lateral sclerosis (ALS) clearly implicate ubiquitously expressed and predominantly nuclear RNA binding proteins, which form pathological cytoplasmic inclusions in this context. However, the possibility that wild-type RNA binding proteins mislocalize without necessarily becoming constituents of cytoplasmic inclusions themselves remains relatively unexplored. We hypothesized that nuclear-to-cytoplasmic mislocalization of the RNA binding protein fused in sarcoma (FUS), in an unaggregated state, may occur more widely in ALS than previously recognized. To address this hypothesis, we analysed motor neurons from a human ALS induced-pluripotent stem cell model caused by the VCP mutation. Additionally, we examined mouse transgenic models and post-mortem tissue from human sporadic ALS cases. We report nuclear-to-cytoplasmic mislocalization of FUS in both VCP-mutation related ALS and, crucially, in sporadic ALS spinal cord tissue from multiple cases. Furthermore, we provide evidence that FUS protein binds to an aberrantly retained intron within the SFPQ transcript, which is exported from the nucleus into the cytoplasm. Collectively, these data support a model for ALS pathogenesis whereby aberrant intron retention in SFPQ transcripts contributes to FUS mislocalization through their direct interaction and nuclear export. In summary, we report widespread mislocalization of the FUS protein in ALS and propose a putative underlying mechanism for this process.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Induced Pluripotent Stem Cells/metabolism , RNA-Binding Protein FUS/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Biomarkers/metabolism , Cell Nucleus/chemistry , Cell Nucleus/genetics , Cytoplasm/chemistry , Cytoplasm/genetics , Female , Humans , Induced Pluripotent Stem Cells/chemistry , Induced Pluripotent Stem Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Binding Protein FUS/analysis , RNA-Binding Protein FUS/genetics
14.
Nat Immunol ; 20(3): 374, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30733606

ABSTRACT

In the version of this article initially published, the Supplementary Data file was an incorrect version. The correct version is now provided. The error has been corrected in the HTML and PDF version of the article.

15.
Nat Commun ; 9(1): 2010, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29789581

ABSTRACT

Mutations causing amyotrophic lateral sclerosis (ALS) strongly implicate ubiquitously expressed regulators of RNA processing. To understand the molecular impact of ALS-causing mutations on neuronal development and disease, we analysed transcriptomes during in vitro differentiation of motor neurons (MNs) from human control and patient-specific VCP mutant induced-pluripotent stem cells (iPSCs). We identify increased intron retention (IR) as a dominant feature of the splicing programme during early neural differentiation. Importantly, IR occurs prematurely in VCP mutant cultures compared with control counterparts. These aberrant IR events are also seen in independent RNAseq data sets from SOD1- and FUS-mutant MNs. The most significant IR is seen in the SFPQ transcript. The SFPQ protein binds extensively to its retained intron, exhibits lower nuclear abundance in VCP mutant cultures and is lost from nuclei of MNs in mouse models and human sporadic ALS. Collectively, we demonstrate SFPQ IR and nuclear loss as molecular hallmarks of familial and sporadic ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Motor Neurons/metabolism , PTB-Associated Splicing Factor/genetics , RNA Splicing , Spinal Cord/metabolism , Valosin Containing Protein/genetics , Aged , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Differentiation , Disease Models, Animal , Exons , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Introns , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Motor Neurons/pathology , PTB-Associated Splicing Factor/metabolism , Primary Cell Culture , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Sequence Analysis, RNA , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Valosin Containing Protein/metabolism
16.
Nat Immunol ; 19(5): 497-507, 2018 05.
Article in English | MEDLINE | ID: mdl-29662170

ABSTRACT

The transcription factor c-Maf induces the anti-inflammatory cytokine IL-10 in CD4+ T cells in vitro. However, the global effects of c-Maf on diverse immune responses in vivo are unknown. Here we found that c-Maf regulated IL-10 production in CD4+ T cells in disease models involving the TH1 subset of helper T cells (malaria), TH2 cells (allergy) and TH17 cells (autoimmunity) in vivo. Although mice with c-Maf deficiency targeted to T cells showed greater pathology in TH1 and TH2 responses, TH17 cell-mediated pathology was reduced in this context, with an accompanying decrease in TH17 cells and increase in Foxp3+ regulatory T cells. Bivariate genomic footprinting elucidated the c-Maf transcription-factor network, including enhanced activity of NFAT; this led to the identification and validation of c-Maf as a negative regulator of IL-2. The decreased expression of the gene encoding the transcription factor RORγt (Rorc) that resulted from c-Maf deficiency was dependent on IL-2, which explained the in vivo observations. Thus, c-Maf is a positive and negative regulator of the expression of cytokine-encoding genes, with context-specific effects that allow each immune response to occur in a controlled yet effective manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Interleukin-2/biosynthesis , Proto-Oncogene Proteins c-maf/immunology , Animals , Interleukin-2/immunology , Mice
17.
Cell Rep ; 19(9): 1739-1749, 2017 05 30.
Article in English | MEDLINE | ID: mdl-28564594

ABSTRACT

Motor neurons (MNs) and astrocytes (ACs) are implicated in the pathogenesis of amyotrophic lateral sclerosis (ALS), but their interaction and the sequence of molecular events leading to MN death remain unresolved. Here, we optimized directed differentiation of induced pluripotent stem cells (iPSCs) into highly enriched (> 85%) functional populations of spinal cord MNs and ACs. We identify significantly increased cytoplasmic TDP-43 and ER stress as primary pathogenic events in patient-specific valosin-containing protein (VCP)-mutant MNs, with secondary mitochondrial dysfunction and oxidative stress. Cumulatively, these cellular stresses result in synaptic pathology and cell death in VCP-mutant MNs. We additionally identify a cell-autonomous VCP-mutant AC survival phenotype, which is not attributable to the same molecular pathology occurring in VCP-mutant MNs. Finally, through iterative co-culture experiments, we uncover non-cell-autonomous effects of VCP-mutant ACs on both control and mutant MNs. This work elucidates molecular events and cellular interplay that could guide future therapeutic strategies in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Astrocytes/pathology , Models, Biological , Motor Neurons/pathology , Valosin Containing Protein/metabolism , Cell Survival , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum Stress , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/ultrastructure , Mutation/genetics , Nerve Degeneration/pathology , Neurogenesis , Oxidative Stress , Phenotype , Synapses/pathology
18.
Toxicol Sci ; 139(2): 501-11, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24690595

ABSTRACT

The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) are closely related nuclear receptors involved in drug metabolism and play important roles in the mechanism of phenobarbital (PB)-induced rodent nongenotoxic hepatocarcinogenesis. Here, we have used a humanized CAR/PXR mouse model to examine potential species differences in receptor-dependent mechanisms underlying liver tissue molecular responses to PB. Early and late transcriptomic responses to sustained PB exposure were investigated in liver tissue from double knock-out CAR and PXR (CAR(KO)-PXR(KO)), double humanized CAR and PXR (CAR(h)-PXR(h)), and wild-type C57BL/6 mice. Wild-type and CAR(h)-PXR(h) mouse livers exhibited temporally and quantitatively similar transcriptional responses during 91 days of PB exposure including the sustained induction of the xenobiotic response gene Cyp2b10, the Wnt signaling inhibitor Wisp1, and noncoding RNA biomarkers from the Dlk1-Dio3 locus. Transient induction of DNA replication (Hells, Mcm6, and Esco2) and mitotic genes (Ccnb2, Cdc20, and Cdk1) and the proliferation-related nuclear antigen Mki67 were observed with peak expression occurring between 1 and 7 days PB exposure. All these transcriptional responses were absent in CAR(KO)-PXR(KO) mouse livers and largely reversible in wild-type and CAR(h)-PXR(h) mouse livers following 91 days of PB exposure and a subsequent 4-week recovery period. Furthermore, PB-mediated upregulation of the noncoding RNA Meg3, which has recently been associated with cellular pluripotency, exhibited a similar dose response and perivenous hepatocyte-specific localization in both wild-type and CAR(h)-PXR(h) mice. Thus, mouse livers coexpressing human CAR and PXR support both the xenobiotic metabolizing and the proliferative transcriptional responses following exposure to PB.


Subject(s)
Cell Cycle/drug effects , Liver/drug effects , Phenobarbital/toxicity , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcriptome/drug effects , Animals , Cell Cycle/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Constitutive Androstane Receptor , Gene Expression Profiling , Humans , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenobarbital/pharmacokinetics , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Species Specificity , Xenobiotics/pharmacokinetics , Xenobiotics/toxicity
19.
Int J Cancer ; 135(7): 1574-85, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24535843

ABSTRACT

The process of hepatocarcinogenesis in the diethylnitrosamine (DEN) initiation/phenobarbital (PB) promotion mouse model involves the selective clonal outgrowth of cells harboring oncogene mutations in Ctnnb1, while spontaneous or DEN-only-induced tumors are often Ha-ras- or B-raf-mutated. The molecular mechanisms and pathways underlying these different tumor sub-types are not well characterized. Their identification may help identify markers for xenobiotic promoted versus spontaneously occurring liver tumors. Here, we have characterized mouse liver tumors harboring either Ctnnb1 or Ha-ras mutations via integrated molecular profiling at the transcriptional, translational and post-translational levels. In addition, metabolites of the intermediary metabolism were quantified by high resolution (1)H magic angle nuclear magnetic resonance. We have identified tumor genotype-specific differences in mRNA and miRNA expression, protein levels, post-translational modifications, and metabolite levels that facilitate the molecular and biochemical stratification of tumor phenotypes. Bioinformatic integration of these data at the pathway level led to novel insights into tumor genotype-specific aberrant cell signaling and in particular to a better understanding of alterations in pathways of the cell intermediary metabolism, which are driven by the constitutive activation of the ß-Catenin and Ha-ras oncoproteins in tumors of the two genotypes.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Genes, ras/genetics , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Metabolomics , Mutation/genetics , beta Catenin/genetics , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Metabolic Networks and Pathways , Mice , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Protein Processing, Post-Translational , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
20.
Nucleic Acids Res ; 42(7): 4180-95, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464994

ABSTRACT

Gene regulatory interactions underlying the early stages of non-genotoxic carcinogenesis are poorly understood. Here, we have identified key candidate regulators of phenobarbital (PB)-mediated mouse liver tumorigenesis, a well-characterized model of non-genotoxic carcinogenesis, by applying a new computational modeling approach to a comprehensive collection of in vivo gene expression studies. We have combined our previously developed motif activity response analysis (MARA), which models gene expression patterns in terms of computationally predicted transcription factor binding sites with singular value decomposition (SVD) of the inferred motif activities, to disentangle the roles that different transcriptional regulators play in specific biological pathways of tumor promotion. Furthermore, transgenic mouse models enabled us to identify which of these regulatory activities was downstream of constitutive androstane receptor and ß-catenin signaling, both crucial components of PB-mediated liver tumorigenesis. We propose novel roles for E2F and ZFP161 in PB-mediated hepatocyte proliferation and suggest that PB-mediated suppression of ESR1 activity contributes to the development of a tumor-prone environment. Our study shows that combining MARA with SVD allows for automated identification of independent transcription regulatory programs within a complex in vivo tissue environment and provides novel mechanistic insights into PB-mediated hepatocarcinogenesis.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Phenobarbital/toxicity , Transcription, Genetic/drug effects , Animals , Binding Sites , Cell Proliferation/drug effects , Computational Biology/methods , Computer Simulation , Constitutive Androstane Receptor , Gene Regulatory Networks , Liver/drug effects , Liver/metabolism , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Male , Mice , Nucleotide Motifs , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcription Factors/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...