Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Mol Cell ; 62(6): 805-807, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27315554

ABSTRACT

Evidence mounts, via two studies published in Molecular Cell (Riscal et al., 2016; Wienken et al., 2016), that chromatin-bound MDM2 impacts pluripotency and metabolism to promote survival and proliferation of cancer cells, independently of p53 degradation.


Subject(s)
Chromatin , Proto-Oncogene Proteins c-mdm2/genetics , Cell Line, Tumor , Gene Expression Regulation , Humans , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics
2.
Mol Cell ; 64(5): 967-981, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27912097

ABSTRACT

Recent evidence suggests that lncRNAs play an integral regulatory role in numerous functions, including determination of cellular identity. We determined global expression (RNA-seq) and genome-wide profiles (ChIP-seq) of histone post-translational modifications and p53 binding in human embryonic stem cells (hESCs) undergoing differentiation to define a high-confidence set of 40 lncRNAs, which are p53 transcriptional targets. We focused on lncRNAs highly expressed in pluripotent hESCs and repressed by p53 during differentiation to identify lncPRESS1 as a p53-regulated transcript that maintains hESC pluripotency in concert with core pluripotency factors. RNA-seq of hESCs depleted of lncPRESS1 revealed that lncPRESS1 controls a gene network that promotes pluripotency. Further, we found that lncPRESS1 physically interacts with SIRT6 and prevents SIRT6 chromatin localization, which maintains high levels of histone H3K56 and H3K9 acetylation at promoters of pluripotency genes. In summary, we describe a p53-regulated, pluripotency-specific lncRNA that safeguards the hESC state by disrupting SIRT6 activity.


Subject(s)
Cell Differentiation/genetics , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Histones/metabolism , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor Suppressor Protein p53/metabolism , Chromatin/metabolism , Embryonic Stem Cells/cytology , Histone Deacetylases , Histones/genetics , Humans , Pluripotent Stem Cells/cytology , Protein Processing, Post-Translational/genetics , Sirtuins/genetics , Sirtuins/metabolism , Tumor Suppressor Protein p53/genetics
3.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33653952

ABSTRACT

The protein p53 is a crucial tumor suppressor, often called "the guardian of the genome"; however, mutations transform p53 into a powerful cancer promoter. The oncogenic capacity of mutant p53 has been ascribed to enhanced propensity to fibrillize and recruit other cancer fighting proteins in the fibrils, yet the pathways of fibril nucleation and growth remain obscure. Here, we combine immunofluorescence three-dimensional confocal microscopy of human breast cancer cells with light scattering and transmission electron microscopy of solutions of the purified protein and molecular simulations to illuminate the mechanisms of phase transformations across multiple length scales, from cellular to molecular. We report that the p53 mutant R248Q (R, arginine; Q, glutamine) forms, both in cancer cells and in solutions, a condensate with unique properties, mesoscopic protein-rich clusters. The clusters dramatically diverge from other protein condensates. The cluster sizes are decoupled from the total cluster population volume and independent of the p53 concentration and the solution concentration at equilibrium with the clusters varies. We demonstrate that the clusters carry out a crucial biological function: they host and facilitate the nucleation of amyloid fibrils. We demonstrate that the p53 clusters are driven by structural destabilization of the core domain and not by interactions of its extensive unstructured region, in contradistinction to the dense liquids typical of disordered and partially disordered proteins. Two-step nucleation of mutant p53 amyloids suggests means to control fibrillization and the associated pathologies through modifying the cluster characteristics. Our findings exemplify interactions between distinct protein phases that activate complex physicochemical mechanisms operating in biological systems.


Subject(s)
Amyloid/chemistry , Mutation, Missense , Tumor Suppressor Protein p53/chemistry , Amino Acid Substitution , Amyloid/genetics , Amyloid/metabolism , Humans , MCF-7 Cells , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
4.
Genes Dev ; 29(8): 803-16, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25877919

ABSTRACT

Embryonic stem cells are maintained in a self-renewing and pluripotent state by multiple regulatory pathways. Pluripotent-specific transcriptional networks are sequentially reactivated as somatic cells reprogram to achieve pluripotency. How epigenetic regulators modulate this process and contribute to somatic cell reprogramming is not clear. Here we performed a functional RNAi screen to identify the earliest epigenetic regulators required for reprogramming. We identified components of the SAGA histone acetyltransferase complex, in particular Gcn5, as critical regulators of reprogramming initiation. Furthermore, we showed in mouse pluripotent stem cells that Gcn5 strongly associates with Myc and that, upon initiation of somatic reprogramming, Gcn5 and Myc form a positive feed-forward loop that activates a distinct alternative splicing network and the early acquisition of pluripotency-associated splicing events. These studies expose a Myc-SAGA pathway that drives expression of an essential alternative splicing regulatory network during somatic cell reprogramming.


Subject(s)
Alternative Splicing , Cellular Reprogramming/genetics , Epigenomics , Histone Acetyltransferases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Differentiation , Cell Movement/genetics , Cells, Cultured , Embryonic Stem Cells , Gene Expression Regulation, Developmental , Histone Acetyltransferases/genetics , Mice , Pluripotent Stem Cells , RNA Interference , RNA Processing, Post-Transcriptional/genetics
5.
Hepatology ; 73(2): 776-794, 2021 02.
Article in English | MEDLINE | ID: mdl-32380568

ABSTRACT

BACKGROUND AND AIMS: How Wnt signaling is orchestrated in liver regeneration and tumorigenesis remains elusive. Recently, we identified transmembrane protein 9 (TMEM9) as a Wnt signaling amplifier. APPROACH AND RESULTS: TMEM9 facilitates v-ATPase assembly for vesicular acidification and lysosomal protein degradation. TMEM9 is highly expressed in regenerating liver and hepatocellular carcinoma (HCC) cells. TMEM9 expression is enriched in the hepatocytes around the central vein and acutely induced by injury. In mice, Tmem9 knockout impairs hepatic regeneration with aberrantly increased adenomatosis polyposis coli (Apc) and reduced Wnt signaling. Mechanistically, TMEM9 down-regulates APC through lysosomal protein degradation through v-ATPase. In HCC, TMEM9 is overexpressed and necessary to maintain ß-catenin hyperactivation. TMEM9-up-regulated APC binds to and inhibits nuclear translocation of ß-catenin, independent of HCC-associated ß-catenin mutations. Pharmacological blockade of TMEM9-v-ATPase or lysosomal degradation suppresses Wnt/ß-catenin through APC stabilization and ß-catenin cytosolic retention. CONCLUSIONS: Our results reveal that TMEM9 hyperactivates Wnt signaling for liver regeneration and tumorigenesis through lysosomal degradation of APC.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Membrane Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Carbon Tetrachloride/administration & dosage , Carbon Tetrachloride/toxicity , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Cell Nucleus/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Gene Knockout Techniques , HEK293 Cells , Hep G2 Cells , Humans , Leupeptins/pharmacology , Liver Neoplasms/genetics , Liver Regeneration , Lysosomes/drug effects , Lysosomes/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Proteolysis/drug effects , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/genetics , beta Catenin/metabolism
6.
Br J Cancer ; 125(2): 176-189, 2021 07.
Article in English | MEDLINE | ID: mdl-33795809

ABSTRACT

BACKGROUND: The mechanism by which immune cells regulate metastasis is unclear. Understanding the role of immune cells in metastasis will guide the development of treatments improving patient survival. METHODS: We used syngeneic orthotopic mouse tumour models (wild-type, NOD/scid and Nude), employed knockout (CD8 and CD4) models and administered CXCL4. Tumours and lungs were analysed for cancer cells by bioluminescence, and circulating tumour cells were isolated from blood. Immunohistochemistry on the mouse tumours was performed to confirm cell type, and on a tissue microarray with 180 TNBCs for human relevance. TCGA data from over 10,000 patients were analysed as well. RESULTS: We reveal that intratumoral immune infiltration differs between metastatic and non-metastatic tumours. The non-metastatic tumours harbour high levels of CD8+ T cells and low levels of platelets, which is reverse in metastatic tumours. During tumour progression, platelets and CXCL4 induce differentiation of monocytes into myeloid-derived suppressor cells (MDSCs), which inhibit CD8+ T-cell function. TCGA pan-cancer data confirmed that CD8lowPlatelethigh patients have a significantly lower survival probability compared to CD8highPlateletlow. CONCLUSIONS: CD8+ T cells inhibit metastasis. When the balance between CD8+ T cells and platelets is disrupted, platelets produce CXCL4, which induces MDSCs thereby inhibiting the CD8+ T-cell function.


Subject(s)
Breast Neoplasms/immunology , CD4 Antigens/genetics , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/transplantation , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Platelet Factor 4/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Gene Knockout Techniques , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, Nude , Myeloid-Derived Suppressor Cells/immunology , Neoplastic Cells, Circulating/immunology , Platelet Factor 4/administration & dosage , Platelet Factor 4/pharmacology , Survival Analysis , Transplantation, Isogeneic , Xenograft Model Antitumor Assays
7.
Genome Res ; 28(2): 159-170, 2018 02.
Article in English | MEDLINE | ID: mdl-29273624

ABSTRACT

Noncoding transcription is a defining feature of active enhancers, linking transcription factor (TF) binding to the molecular mechanisms controlling gene expression. To determine the relationship between enhancer activity and biological outcomes in breast cancers, we profiled the transcriptomes (using GRO-seq and RNA-seq) and epigenomes (using ChIP-seq) of 11 different human breast cancer cell lines representing five major molecular subtypes of breast cancer, as well as two immortalized ("normal") human breast cell lines. In addition, we developed a robust and unbiased computational pipeline that simultaneously identifies putative subtype-specific enhancers and their cognate TFs by integrating the magnitude of enhancer transcription, TF mRNA expression levels, TF motif P-values, and enrichment of H3K4me1 and H3K27ac. When applied across the 13 different cell lines noted above, the Total Functional Score of Enhancer Elements (TFSEE) identified key breast cancer subtype-specific TFs that act at transcribed enhancers to dictate gene expression patterns determining growth outcomes, including Forkhead TFs, FOSL1, and PLAG1. FOSL1, a Fos family TF, (1) is highly enriched at the enhancers of triple negative breast cancer (TNBC) cells, (2) acts as a key regulator of the proliferation and viability of TNBC cells, but not Luminal A cells, and (3) is associated with a poor prognosis in TNBC breast cancer patients. Taken together, our results validate our enhancer identification pipeline and reveal that enhancers transcribed in breast cancer cells direct critical gene regulatory networks that promote pathogenesis.


Subject(s)
Carcinogenesis/genetics , Enhancer Elements, Genetic/genetics , Transcriptome/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Histones/genetics , Humans , Middle Aged , RNA, Messenger/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/pathology
8.
Nature ; 508(7495): 263-8, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24590075

ABSTRACT

Recognition of modified histones by 'reader' proteins plays a critical role in the regulation of chromatin. H3K36 trimethylation (H3K36me3) is deposited onto the nucleosomes in the transcribed regions after RNA polymerase II elongation. In yeast, this mark in turn recruits epigenetic regulators to reset the chromatin to a relatively repressive state, thus suppressing cryptic transcription. However, much less is known about the role of H3K36me3 in transcription regulation in mammals. This is further complicated by the transcription-coupled incorporation of the histone variant H3.3 in gene bodies. Here we show that the candidate tumour suppressor ZMYND11 specifically recognizes H3K36me3 on H3.3 (H3.3K36me3) and regulates RNA polymerase II elongation. Structural studies show that in addition to the trimethyl-lysine binding by an aromatic cage within the PWWP domain, the H3.3-dependent recognition is mediated by the encapsulation of the H3.3-specific 'Ser 31' residue in a composite pocket formed by the tandem bromo-PWWP domains of ZMYND11. Chromatin immunoprecipitation followed by sequencing shows a genome-wide co-localization of ZMYND11 with H3K36me3 and H3.3 in gene bodies, and its occupancy requires the pre-deposition of H3.3K36me3. Although ZMYND11 is associated with highly expressed genes, it functions as an unconventional transcription co-repressor by modulating RNA polymerase II at the elongation stage. ZMYND11 is critical for the repression of a transcriptional program that is essential for tumour cell growth; low expression levels of ZMYND11 in breast cancer patients correlate with worse prognosis. Consistently, overexpression of ZMYND11 suppresses cancer cell growth in vitro and tumour formation in mice. Together, this study identifies ZMYND11 as an H3.3-specific reader of H3K36me3 that links the histone-variant-mediated transcription elongation control to tumour suppression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/metabolism , Histones/metabolism , Lysine/metabolism , RNA Polymerase II/metabolism , Transcription Elongation, Genetic , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Carrier Proteins/chemistry , Cell Cycle Proteins , Chromatin/genetics , Chromatin/metabolism , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/metabolism , Crystallography, X-Ray , DNA-Binding Proteins , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Histones/chemistry , Humans , Methylation , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Oncogenes/genetics , Prognosis , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
9.
J Biol Chem ; 293(19): 7476-7485, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29523690

ABSTRACT

Proteins with domains that recognize and bind post-translational modifications (PTMs) of histones are collectively termed epigenetic readers. Numerous interactions between specific reader protein domains and histone PTMs and their regulatory outcomes have been reported, but little is known about how reader proteins may in turn be modulated by these interactions. Tripartite motif-containing protein 24 (TRIM24) is a histone reader aberrantly expressed in multiple cancers. Here, our investigation revealed functional cross-talk between histone acetylation and TRIM24 SUMOylation. Binding of TRIM24 to chromatin via its tandem PHD-bromodomain, which recognizes unmethylated lysine 4 and acetylated lysine 23 of histone H3 (H3K4me0/K23ac), led to TRIM24 SUMOylation at lysine residues 723 and 741. Inactivation of the bromodomain, either by mutation or with a small-molecule inhibitor, IACS-9571, abolished TRIM24 SUMOylation. Conversely, inhibition of histone deacetylation markedly increased TRIM24's interaction with chromatin and its SUMOylation. Of note, gene expression profiling of MCF7 cells expressing WT versus SUMO-deficient TRIM24 identified cell adhesion as the major pathway regulated by the cross-talk between chromatin acetylation and TRIM24 SUMOylation. In conclusion, our findings establish a new link between histone H3 acetylation and SUMOylation of the reader protein TRIM24, a functional connection that may bear on TRIM24's oncogenic function and may inform future studies of PTM cross-talk between histones and epigenetic regulators.


Subject(s)
Carrier Proteins/metabolism , Cell Adhesion , Chromatin/metabolism , Sumoylation , Acetylation , Carrier Proteins/chemistry , Epigenesis, Genetic , HEK293 Cells , Histones/metabolism , Humans , MCF-7 Cells , Oncogenes , Protein Processing, Post-Translational
10.
BMC Genomics ; 19(1): 150, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458327

ABSTRACT

BACKGROUND: Epigenetic regulators are frequently mutated or aberrantly expressed in a variety of cancers, leading to altered transcription states that result in changes in cell identity, behavior, and response to therapy. RESULTS: To define alterations in epigenetic landscapes in breast cancers, we profiled the distributions of 8 key histone modifications by ChIP-Seq, as well as primary (GRO-seq) and steady state (RNA-Seq) transcriptomes, across 13 distinct cell lines that represent 5 molecular subtypes of breast cancer and immortalized human mammary epithelial cells. DISCUSSION: Using combinatorial patterns of distinct histone modification signals, we defined subtype-specific chromatin signatures to nominate potential biomarkers. This approach identified AFAP1-AS1 as a triple negative breast cancer-specific gene associated with cell proliferation and epithelial-mesenchymal-transition. In addition, our chromatin mapping data in basal TNBC cell lines are consistent with gene expression patterns in TCGA that indicate decreased activity of the androgen receptor pathway but increased activity of the vitamin D biosynthesis pathway. CONCLUSIONS: Together, these datasets provide a comprehensive resource for histone modification profiles that define epigenetic landscapes and reveal key chromatin signatures in breast cancer cell line subtypes with potential to identify novel and actionable targets for treatment.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histones/metabolism , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Female , Gene Expression Profiling , Humans , Transcriptome
11.
Genes Dev ; 24(14): 1479-84, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20581084

ABSTRACT

Self-renewing embryonic stem (ES) cells have an exceptional need for timely biomass production, yet the transcriptional control mechanisms responsible for meeting this requirement are largely unknown. We report here that Ronin (Thap11), which is essential for the self-renewal of ES cells, binds with its transcriptional coregulator, Hcf-1, to a highly conserved enhancer element that previously lacked a recognized binding factor. The subset of genes bound by Ronin/Hcf-1 function primarily in transcription initiation, mRNA splicing, and cell metabolism; genes involved in cell signaling and cell development are conspicuously underrepresented in this target gene repertoire. Although Ronin/Hcf-1 represses the expression of some target genes, its activity at promoter sites more often leads to the up-regulation of genes essential to protein biosynthesis and energy production. We propose that Ronin/Hcf-1 controls a genetic program that contributes to the unimpeded growth of ES cells.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Host Cell Factor C1/metabolism , Animals , DNA-Binding Proteins/genetics , Energy Metabolism , Mice , Protein Biosynthesis , Protein Structure, Tertiary , Repressor Proteins , Transcription, Genetic
13.
Nature ; 538(7623): 45-46, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27626387
14.
Tumour Biol ; 37(11): 14803-14812, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27638829

ABSTRACT

Acetylated H3 lysine 23 (H3K23ac) is a specific histone post-translational modification recognized by oncoprotein TRIM24. However, it is not clear whether H3K23ac levels are correlated with TRIM24 expression and what role H3K23ac may have in cancer. In this study, we collected breast carcinoma samples from 121 patients and conducted immunohistochemistry to determine the levels of TRIM24 and H3K23ac in breast cancer. Our results demonstrated that TRIM24 expression is positively correlated with H3K23ac levels, and high levels of both TRIM24 and H3K23ac predict shorter overall survival of breast cancer patients. We also showed that both TRIM24 and H3K23ac are higher in HER2-positive patients, and their levels were positively correlated with HER2 levels in breast cancer. Moreover, TRIM24 expression is associated with estrogen receptor (ER) and progesterone receptor (PR) statuses in both our cohort and The Cancer Genome Atlas (TCGA) breast carcinoma. In summary, our results revealed an important role of TRIM24 and H3K23ac in breast cancer and provided further evidence that TRIM24 small-molecule inhibitors may benefit ER- and PR-negative or HER2-positive breast cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Carrier Proteins/metabolism , Histones/metabolism , Acetylation , Breast/pathology , Breast Neoplasms/mortality , Female , Humans , Immunohistochemistry , Middle Aged , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism
15.
Proc Natl Acad Sci U S A ; 110(43): 17284-9, 2013 Oct 22.
Article in English | MEDLINE | ID: mdl-24101509

ABSTRACT

Estrogen receptor alpha (ERα) is a ligand-activated transcription factor. Upon estrogen stimulation, ERα recruits a number of coregulators, including both coactivators and corepressors, to the estrogen response elements, modulating gene activation or repression. Most coregulator complexes contain histone-modifying enzymes to control ERα target gene expression in an epigenetic manner. In addition to histones, these epigenetic modifiers can modify nonhistone proteins including ERα, thereby constituting another layer of transcriptional regulation. Here we show that SET and MYND domain containing 2 (SMYD2), a histone H3K4 and H3K36 methyltransferase, directly methylates ERα protein at lysine 266 (K266) both in vitro and in cells. In breast cancer MCF7 cells, SMYD2 attenuates the chromatin recruitment of ERα to prevent ERα target gene activation under an estrogen-depleted condition. Importantly, the SMYD2-mediated repression of ERα target gene expression is mediated by the methylation of ERα at K266 in the nucleus, but not the methylation of histone H3K4. Upon estrogen stimulation, ERα-K266 methylation is diminished, thereby enabling p300/cAMP response element-binding protein-binding protein to acetylate ERα at K266, which is known to promote ERα transactivation activity. Our study identifies a previously undescribed inhibitory methylation event on ERα. Our data suggest that the dynamic cross-talk between SMYD2-mediated ERα protein methylation and p300/cAMP response element-binding protein-binding protein-dependent ERα acetylation plays an important role in fine-tuning the functions of ERα at chromatin and the estrogen-induced gene expression profiles.


Subject(s)
Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic/genetics , Histone-Lysine N-Methyltransferase/metabolism , Lysine/metabolism , Acetylation , Amino Acid Sequence , Blotting, Western , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Lysine/genetics , MCF-7 Cells , Methylation/drug effects , Mutation , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , p300-CBP Transcription Factors/metabolism
16.
PLoS Genet ; 8(6): e1002770, 2012.
Article in English | MEDLINE | ID: mdl-22737085

ABSTRACT

Gene duplication is a powerful driver of evolution. Newly duplicated genes acquire new roles that are relevant to fitness, or they will be lost over time. A potential path to functional relevance is mutation of the coding sequence leading to the acquisition of novel biochemical properties, as analyzed here for the highly homologous paralogs Foxa1 and Foxa2 transcriptional regulators. We determine by genome-wide location analysis (ChIP-Seq) that, although Foxa1 and Foxa2 share a large fraction of binding sites in the liver, each protein also occupies distinct regulatory elements in vivo. Foxa1-only sites are enriched for p53 binding sites and are frequently found near genes important to cell cycle regulation, while Foxa2-restricted sites show only a limited match to the forkhead consensus and are found in genes involved in steroid and lipid metabolism. Thus, Foxa1 and Foxa2, while redundant during development, have evolved divergent roles in the adult liver, ensuring the maintenance of both genes during evolution.


Subject(s)
DNA-Binding Proteins , Hepatocyte Nuclear Factor 3-alpha , Hepatocyte Nuclear Factor 3-beta , Liver , Transcription, Genetic , Amino Acid Sequence , Animals , Binding Sites , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Development/genetics , Gene Duplication , Gene Expression Regulation , Genes, p53/genetics , Genome , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Liver/growth & development , Liver/metabolism , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Nucleotide Motifs , Sequence Homology
17.
Adv Sci (Weinh) ; : e2309220, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023197

ABSTRACT

Gut microbiome composition is tied to diseases ranging from arthritis to cancer to depression. However, mechanisms of action are poorly understood, limiting development of relevant therapeutics. Organ-on-chip platforms, which model minimal functional units of tissues and can tightly control communication between them, are ideal platforms to study these relationships. Many gut microbiome models are published to date but devices are typically fabricated using oxygen permeable polydimethylsiloxane, requiring interventions to support anaerobic bacteria. To address this challenge, a platform is developed where the chips are fabricated entirely from gas-impermeable polycarbonate without tapes or gaskets. These chips replicate polarized villus-like structures of the native tissue. Further, they enable co-cultures of commensal anaerobic bacteria Blautia coccoides on the surface of gut epithelia for two days within a standard incubator. Another complication of commonly used materials in organ-on-chip devices is high ad-/absorption, limiting applications in high-resolution microscopy and biomolecule interaction studies. For future communication studies between gut microbiota and distal tumors, an additional polycarbonate chip design is developed to support hydrogel-embedded tissue culture. These chips enable high-resolution microscopy with all relevant processing done on-chip. Designed for facile linking, this platform will make a variety of mechanistic studies possible.

18.
iScience ; 26(7): 107126, 2023 Jul 21.
Article in English | MEDLINE | ID: mdl-37426340

ABSTRACT

TRIM24 is an oncogenic chromatin reader that is frequently overexpressed in human tumors and associated with poor prognosis. However, TRIM24 is rarely mutated, duplicated, or rearranged in cancer. This raises questions about how TRIM24 is regulated and what changes in its regulation are responsible for its overexpression. Here, we perform a genome-wide CRISPR-Cas9 screen by fluorescence-activated cell sorting (FACS) that nominated 220 negative regulators and elucidated a regulatory network that includes the KAP1 corepressor, CNOT deadenylase, and GID/CTLH E3 ligase. Knocking out required components of these three complexes caused TRIM24 overexpression, confirming their negative regulation of TRIM24. Our findings identify regulators of TRIM24 that nominate previously unexplored contexts for this oncoprotein in biology and disease. These findings were enabled by SLIDER, a new scoring system designed and vetted in our study as a broadly applicable tool for analysis of CRISPR screens performed by FACS.

19.
Cancer Res ; 82(16): 2824-2825, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35971677

ABSTRACT

Activation of p53 regulates a transcriptional program that can cause cell cycle arrest, senescence, apoptosis, and ferroptosis, which are potent tumor suppressive mechanisms. Unexpectedly, Makino and colleagues show in this issue of Cancer Research that the constitutive activation of p53 in murine hepatocytes leads to tumor development. Detailed analyses indicate that p53 activation leads to loss of hepatocytes, increased expression of chemokines and humoral factors, and expansion of the hepatic progenitor cell population. These progenitor cells are highly proliferative, show chromosomal instability, and eventually transform. In chronic liver disease in humans, activation of p53 is associated with increased liver cancer development. This study highlights the complexity and non-cell autonomous nature of the physiologic p53 response. See related article by Makino et al., p. 2860.


Subject(s)
Liver Neoplasms , Tumor Suppressor Protein p53 , Animals , Apoptosis , Carcinogenesis , Hepatocytes/metabolism , Humans , Liver Neoplasms/pathology , Mice , Tumor Suppressor Protein p53/metabolism
20.
J Cell Sci ; 122(Pt 22): 4049-61, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19843587

ABSTRACT

Catenins of the p120 subclass display an array of intracellular localizations and functions. Although the genetic knockout of mouse delta-catenin results in mild cognitive dysfunction, we found severe effects of its depletion in Xenopus. delta-catenin in Xenopus is transcribed as a full-length mRNA, or as three (or more) alternatively spliced isoforms designated A, B and C. Further structural and functional complexity is suggested by three predicted and alternative translation initiation sites. Transcript analysis suggests that each splice isoform is expressed during embryogenesis, with the B and C transcript levels varying according to developmental stage. Unlike the primarily neural expression of delta-catenin reported in mammals, delta-catenin is detectable in most adult Xenopus tissues, although it is enriched in neural structures. delta-catenin associates with classical cadherins, with crude embryo fractionations further revealing non-plasma-membrane pools that might be involved in cytoplasmic and/or nuclear functions. Depletion of delta-catenin caused gastrulation defects, phenotypes that were further enhanced by co-depletion of the related p120-catenin. Depletion was significantly rescued by titrated p120-catenin expression, suggesting that these catenins have shared roles. Biochemical assays indicated that delta-catenin depletion results in reduced cadherin levels and cell adhesion, as well as perturbation of RhoA and Rac1. Titrated doses of C-cadherin, dominant-negative RhoA or constitutively active Rac1 significantly rescued delta-catenin depletion. Collectively, our experiments indicate that delta-catenin has an essential role in amphibian development, and has functional links to cadherins and Rho-family GTPases.


Subject(s)
Cadherins/metabolism , Catenins/genetics , Catenins/metabolism , Gastrula/metabolism , Xenopus laevis/embryology , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Alternative Splicing , Animals , Base Sequence , Cell Adhesion , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gastrula/ultrastructure , Gastrulation/physiology , Gene Knockout Techniques , Molecular Sequence Data , Neurulation/physiology , Xenopus laevis/metabolism , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL