Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Mol Cell Biol ; 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37528066

ABSTRACT

The super elongation complex (SEC) containing P-TEFb plays a critical role in regulating transcription elongation. AFF1 and AFF4, members of the AF4/FMR2 family, act as central scaffold proteins of SEC and are associated with various human diseases. However, their precise roles in transcriptional control remain unclear. We here reveal differences in the genomic distribution patterns of AFF1 and AFF4 around transcription start sites (TSSs). AFF1 mainly binds upstream of the TSSs, while AFF4 is enriched downstream of the TSSs. Notably, disruption of AFF4 results in slow elongation and early termination in a subset of AFF4 bound active genes, whereas AFF1 deletion leads to fast elongation and transcriptional readthrough in the same gene subset. Additionally, AFF1 knockdown increases AFF4 levels at chromatin, and vice versa. In summary, these findings demonstrate that AFF1 and AFF4 function antagonistically to regulate Pol II transcription.

2.
Nat Cell Biol ; 25(9): 1319-1331, 2023 09.
Article in English | MEDLINE | ID: mdl-37591949

ABSTRACT

LINE-1s are the major clade of retrotransposons with autonomous retrotransposition activity. Despite the potential genotoxicity, LINE-1s are highly activated in early embryos. Here we show that a subset of young LINE-1s, L1Md_Ts, are marked by the RNA polymerase II elongation factor ELL3, and function as enhancers in mouse embryonic stem cells. ELL3 depletion dislodges the DNA hydroxymethylase TET1 and the co-repressor SIN3A from L1Md_Ts, but increases the enrichment of the Bromodomain protein BRD4, leading to loss of 5hmC, gain of H3K27ac, and upregulation of the L1Md_T nearby genes. Specifically, ELL3 occupies and represses the L1Md_T-based enhancer located within Akt3, which encodes a key regulator of AKT pathway. ELL3 is required for proper ERK activation and efficient shutdown of naïve pluripotency through inhibiting Akt3 during naïve-primed transition. Our study reveals that the enhancer function of a subset of young LINE-1s controlled by ELL3 in transcription regulation and mouse early embryo development.


Subject(s)
Nuclear Proteins , Transcription Factors , Animals , Mice , 5' Untranslated Regions , Nuclear Proteins/genetics , Transcription Factors/genetics , Embryonic Stem Cells , Peptide Elongation Factors
3.
Genes Genomics ; 44(3): 379-387, 2022 03.
Article in English | MEDLINE | ID: mdl-35133623

ABSTRACT

BACKGROUND: Dynamic chromatin reorganization occurs during two waves of cell lineage specification process, blastocyst formation and gastrulation, to generate distinct cell types. Epigenetic defects have been associated with severe developmental defects and diseases. How epigenetic remodeling coordinates the two lineage specification waves is becoming uncovered, benefiting from the development and application of new technologies including low-input or single-cell epigenome analysis approached in the past few years. OBJECTIVE: In this review, we aim to highlight the most recent findings on epigenetic remodeling in cell lineage specification during blastocyst formation and gastrulation. METHODS: First, we introduce how DNA methylation dynamically changes in blastocyst formation and gastrulation and its function in transcriptional regulation lineage-specific genes. Then, we discuss widespread remodeling of histone modification at promoters and enhancers in orchestrating the trajectory of cell lineage specification. Finally, we review dynamics of chromatin accessibility and 3D structure regulating developmental gene expression and associating with specific transcription factor binding events at stage specific manner. We also highlight the key questions that remain to be answered to fully understand chromatin regulation and reorganization in lineage specification. CONCLUSION: Here, we summarize the recent advances and discoveries on epigenetic reorganization and its roles in blastocyst formation and gastrulation, and how it cooperates with the lineage specification, painting from global sequencing data from mouse in vivo tissues.


Subject(s)
Blastocyst , Epigenesis, Genetic , Animals , Blastocyst/metabolism , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Embryonic Development/genetics , Mice
4.
J Biol Chem ; 296: 100319, 2021.
Article in English | MEDLINE | ID: mdl-33493519

ABSTRACT

Upregulation of the neuropeptide neurotensin (NTS) in a subgroup of lung cancers has been linked to poor prognosis. However, the regulatory pathway centered on NTS in lung cancer remains unclear. Here we identified the NTS-specific enhancer in lung adenocarcinoma cells. The AF4/FMR2 (AFF) family protein AFF1 occupies the NTS enhancer and inhibits NTS transcription. Clustering analysis of lung adenocarcinoma gene expression data demonstrated that NTS expression is highly positively correlated with the expression of the oncogenic factor CPS1. Detailed analyses demonstrated that the IL6 pathway antagonizes NTS in regulating CPS1. Thus, our analyses revealed a novel NTS-centered regulatory axis, consisting of AFF1 as a master transcription suppressor and IL6 as an antagonist in lung adenocarcinoma cells.


Subject(s)
Adenocarcinoma of Lung/genetics , Carbamoyl-Phosphate Synthase (Ammonia)/genetics , DNA-Binding Proteins/genetics , Interleukin-6/genetics , Neurotensin/genetics , Transcriptional Elongation Factors/genetics , A549 Cells , Adenocarcinoma of Lung/pathology , Cell Proliferation/genetics , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Prognosis , Signal Transduction/genetics
5.
J Mol Cell Biol ; 12(11): 894-905, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33104214

ABSTRACT

Regulation of RNA stability plays a crucial role in gene expression control. Deadenylation is the initial rate-limiting step for the majority of RNA decay events. Here, we show that RING finger protein 219 (RNF219) interacts with the CCR4-NOT deadenylase complex. RNF219-CCR4-NOT exhibits deadenylation activity in vitro. RNA-seq analyses identify some of the 2-cell-specific genes and the neuronal genes significantly downregulated upon RNF219 knockdown, while upregulated after depletion of the CCR4-NOT subunit CNOT10 in mouse embryonic stem (ES) cells. RNF219 depletion leads to impaired neuronal lineage commitment during ES cell differentiation. Our study suggests that RNF219 is a novel interacting partner of CCR4-NOT and required for maintenance of ES cell pluripotency.


Subject(s)
Cell Differentiation , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Differentiation/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/cytology , Neurons/metabolism , Protein Binding
6.
Nat Commun ; 9(1): 1537, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29670118

ABSTRACT

H3.3 is a histone variant, which is deposited on genebodies and regulatory elements, by Hira, marking active transcription. Moreover, H3.3 is deposited on heterochromatin by Atrx/Daxx complex. The exact role of H3.3 in cell fate transition remains elusive. Here, we investigate the dynamic changes in the deposition of the histone variant H3.3 during cellular reprogramming. H3.3 maintains the identities of the parental cells during reprogramming as its removal at early time-point enhances the efficiency of the process. We find that H3.3 plays a similar role in transdifferentiation to hematopoietic progenitors and neuronal differentiation from embryonic stem cells. Contrastingly, H3.3 deposition on genes associated with the newly reprogrammed lineage is essential as its depletion at the later phase abolishes the process. Mechanistically, H3.3 deposition by Hira, and its K4 and K36 modifications are central to the role of H3.3 in cell fate conversion. Finally, H3.3 safeguards fibroblast lineage by regulating Mapk cascade and collagen synthesis.


Subject(s)
Cell Lineage , Histone Chaperones/metabolism , Histones/chemistry , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Chromatin Immunoprecipitation , Collagen/chemistry , Fibroblasts/metabolism , HEK293 Cells , Heterochromatin , Humans , MAP Kinase Signaling System , Mice , Neurons/metabolism , Nucleosomes , Protein Binding , Retroviridae/genetics , Software , Transcriptome
7.
Nat Genet ; 49(9): 1354-1363, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28740264

ABSTRACT

The transcriptional network acting downstream of LIF, WNT and MAPK-ERK to stabilize mouse embryonic stem cells (ESCs) in their naive state has been extensively characterized. However, the upstream factors regulating these three signaling pathways remain largely uncharted. PR-domain-containing proteins (PRDMs) are zinc-finger sequence-specific chromatin factors that have essential roles in embryonic development and cell fate decisions. Here we characterize the transcriptional regulator PRDM15, which acts independently of PRDM14 to regulate the naive state of mouse ESCs. Mechanistically, PRDM15 modulates WNT and MAPK-ERK signaling by directly promoting the expression of Rspo1 (R-spondin1) and Spry1 (Sprouty1). Consistent with these findings, CRISPR-Cas9-mediated disruption of PRDM15-binding sites in the Rspo1 and Spry1 promoters recapitulates PRDM15 depletion, both in terms of local chromatin organization and the transcriptional modulation of these genes. Collectively, our findings uncover an essential role for PRDM15 as a chromatin factor that modulates the transcription of upstream regulators of WNT and MAPK-ERK signaling to safeguard naive pluripotency.


Subject(s)
DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , MAP Kinase Signaling System/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Animals , Blotting, Western , Cell Line , Cell Self Renewal/genetics , Cells, Cultured , Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Humans , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Mice, Nude , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
8.
Proc Natl Acad Sci U S A ; 109(7): 2521-6, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308476

ABSTRACT

The boronic acid dipeptide bortezomib inhibits the chymotrypsin-like activity of the 26S proteasome and shows significant therapeutic efficacy in multiple myeloma. However, recent studies suggest that bortezomib may have more complex mechanisms of action in treating cancer. We report here that the endocytosis and lysosomal degradation of the receptor tyrosine kinase C-KIT are required for bortezomib- but not tyrosine kinase inhibitor imatinib-caused apoptosis of t(8;21) leukemia and gastrointestinal stromal tumor cells, suggesting that C-KIT may recruit an apoptosis initiator. We show that C-KIT binds and phosphorylates heat shock protein 90ß (Hsp90ß), which sequestrates apoptotic protease activating factor 1 (Apaf-1). Bortezomib dephosphorylates pHsp90ß and releases Apaf-1. Although the activated caspase-3 is not sufficient to cause marked apoptosis, it cleaves the t(8;21) generated acute myeloid leukemia 1-eight twenty one (AML1-ETO) and AML1-ETO9a fusion proteins, with production of cleavage fragments that perturb the functions of the parental oncoproteins and further contribute to apoptosis. Notably, bortezomib exerts potent therapeutic efficacy in mice bearing AML1-ETO9a-driven leukemia. These data show that C-KIT-pHsp90ß-Apaf-1 cascade is critical for some malignant cells to evade apoptosis, and the clinical therapeutic potentials of bortezomib in C-KIT-driven neoplasms should be further explored.


Subject(s)
Boronic Acids/pharmacology , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Leukemia/pathology , Proto-Oncogene Proteins c-kit/metabolism , Pyrazines/pharmacology , Translocation, Genetic , Apoptosis , Bortezomib , Humans , Leukemia/genetics , Phosphorylation , Protein Binding
9.
Leuk Res ; 35(2): 214-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20691473

ABSTRACT

Triptolide is a compound isolated from the traditional Chinese medicinal herb Tripterygium wilfordii that shows potent anti-tumor activities, but its effects on acute myeloid leukemia with t(8;21) remain unclear. Here we report that triptolide inhibits cell proliferation and induces apoptosis in a dose- and time-dependent manner of t(8;21)-bearing Kasumi-1, SKNO-1 and CD34+ cells harvested from bone marrow samples of patients with t(8;21) leukemia. We show that triptolide triggers cleavage of the resultant AML1-ETO fusion protein of t(8;21), and causes downregulation of C-KIT followed by inhibition of JAK-STAT signaling. Triptolide downregulates p65 and inhibits the DNA-binding activity of NF-κB. Our data indicate that triptolide might be an effective agent for t(8;21) leukemia.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Diterpenes/pharmacology , Leukemia, Myeloid, Acute/genetics , Phenanthrenes/pharmacology , Signal Transduction/drug effects , Blotting, Western , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/drug effects , Epoxy Compounds/pharmacology , Humans , Janus Kinases/drug effects , Janus Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/drug effects , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins c-kit/drug effects , RUNX1 Translocation Partner 1 Protein , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/drug effects , STAT Transcription Factors/metabolism
10.
Sheng Wu Gong Cheng Xue Bao ; 25(3): 452-6, 2009 Mar.
Article in Chinese | MEDLINE | ID: mdl-19621590

ABSTRACT

To establish a green fluorescent protein (GFP)-based cellular model for screening proteasome inhibitors from compounds including extracts from Traditional Chinese Medicinal herbs, we transfected A549 cells with lentivirus expression vector pGC-E1-ZU1-GFP, and selected clones stably expressing ZU1-GFP. The A549-ZU1-GFP cells were treated with PS-341 for 24 h, and the accumulation of GFP was analyzed by fluorescence microscope. We found that the fluorescence intensity of A549-ZU1-GFP cells treated with PS-341 was significantly increased as compared to the control. We screened for proteasome inhibitors from compounds including some from Traditional Chinese Medicinal herbs, and the data suggested a few compounds could be novel proteasome inhibitors.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Green Fluorescent Proteins/genetics , Models, Biological , Protease Inhibitors/isolation & purification , Protease Inhibitors/pharmacology , Boronic Acids/pharmacology , Bortezomib , Cell Line , Drug Evaluation, Preclinical , Drugs, Chinese Herbal/isolation & purification , Genetic Vectors/genetics , Green Fluorescent Proteins/biosynthesis , Humans , Lentivirus/genetics , Lentivirus/metabolism , Pyrazines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL