Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Nat Commun ; 15(1): 4609, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816425

ABSTRACT

The protection of the replication fork structure under stress conditions is essential for genome maintenance and cancer prevention. A key signaling pathway for fork protection involves TRPV2-mediated Ca2+ release from the ER, which is triggered after the generation of cytosolic DNA and the activation of cGAS/STING. This results in CaMKK2/AMPK activation and subsequent Exo1 phosphorylation, which prevent aberrant fork processing, thereby ensuring genome stability. However, it remains poorly understood how the TRPV2 channel is activated by the presence of cytosolic DNA. Here, through a genome-wide CRISPR-based screen, we identify TRPM8 channel-associated factor 1 (TCAF1) as a key factor promoting TRPV2-mediated Ca2+ release under replication stress or other conditions that activate cGAS/STING. Mechanistically, TCAF1 assists Ca2+ release by facilitating the dissociation of STING from TRPV2, thereby relieving TRPV2 repression. Consistent with this function, TCAF1 is required for fork protection, chromosomal stability, and cell survival after replication stress.


Subject(s)
Calcium , Cytosol , DNA Replication , Membrane Proteins , TRPV Cation Channels , Humans , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cytosol/metabolism , DNA/metabolism , DNA Damage , Genomic Instability , HEK293 Cells , HeLa Cells , Membrane Proteins/metabolism , Membrane Proteins/genetics , Phosphorylation , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics
2.
J Clin Invest ; 134(3)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38032734

ABSTRACT

Pancreatic ß cells are specialized for coupling glucose metabolism to insulin peptide production and secretion. Acute glucose exposure robustly and coordinately increases translation of proinsulin and proteins required for secretion of mature insulin peptide. By contrast, chronically elevated glucose levels that occur during diabetes impair ß cell insulin secretion and have been shown experimentally to suppress insulin translation. Whether translation of other genes critical for insulin secretion is similarly downregulated by chronic high glucose is unknown. Here, we used high-throughput ribosome profiling and nascent proteomics in MIN6 insulinoma cells to elucidate the genome-wide impact of sustained high glucose on ß cell mRNA translation. Before induction of ER stress or suppression of global translation, sustained high glucose suppressed glucose-stimulated insulin secretion and downregulated translation of not only insulin, but also mRNAs related to insulin secretory granule formation, exocytosis, and metabolism-coupled insulin secretion. Translation of these mRNAs was also downregulated in primary rat and human islets following ex vivo incubation with sustained high glucose and in an in vivo model of chronic mild hyperglycemia. Furthermore, translational downregulation decreased cellular abundance of these proteins. Our study uncovered a translational regulatory circuit during ß cell glucose toxicity that impairs expression of proteins with critical roles in ß cell function.


Subject(s)
Hyperglycemia , Insulin-Secreting Cells , Islets of Langerhans , Pancreatic Neoplasms , Rats , Humans , Animals , Insulin Secretion , RNA, Messenger/metabolism , Insulin/metabolism , Hyperglycemia/genetics , Hyperglycemia/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Peptides/metabolism , Pancreatic Neoplasms/metabolism , Islets of Langerhans/metabolism
3.
bioRxiv ; 2023 Sep 29.
Article in English | MEDLINE | ID: mdl-37808767

ABSTRACT

Pancreatic ß-cells are specialized for coupling glucose metabolism to insulin peptide production and secretion. Acute glucose exposure robustly and coordinately increases translation of proinsulin and proteins required for secretion of mature insulin peptide. By contrast, chronically elevated glucose levels that occur during diabetes impair ß-cell insulin secretion and have been shown experimentally to suppress insulin translation. Whether translation of other genes critical for insulin secretion are similarly downregulated by chronic high glucose is unknown. Here, we used high-throughput ribosome profiling and nascent proteomics in MIN6 insulinoma cells to elucidate the genome-wide impact of sustained high glucose on ß-cell mRNA translation. Prior to induction of ER stress or suppression of global translation, sustained high glucose suppressed glucose-stimulated insulin secretion and downregulated translation of not only insulin, but also of mRNAs related to insulin secretory granule formation, exocytosis, and metabolism-coupled insulin secretion. Translation of these mRNAs was also downregulated in primary rat and human islets following ex-vivo incubation with sustained high glucose and in an in vivo model of chronic mild hyperglycemia. Furthermore, translational downregulation decreased cellular abundance of these proteins. Our findings uncover a translational regulatory circuit during ß-cell glucose toxicity that impairs expression of proteins with critical roles in ß-cell function.

4.
Mol Cell ; 83(4): 556-573.e7, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36696898

ABSTRACT

The protection of DNA replication forks under stress is essential for genome maintenance and cancer suppression. One mechanism of fork protection involves an elevation in intracellular Ca2+ ([Ca2+]i), which in turn activates CaMKK2 and AMPK to prevent uncontrolled fork processing by Exo1. How replication stress triggers [Ca2+]i elevation is unclear. Here, we report a role of cytosolic self-DNA (cytosDNA) and the ion channel TRPV2 in [Ca2+]i induction and fork protection. Replication stress leads to the generation of ssDNA and dsDNA species that, upon translocation into cytoplasm, trigger the activation of the sensor protein cGAS and the production of cGAMP. The subsequent binding of cGAMP to STING causes its dissociation from TRPV2, leading to TRPV2 derepression and Ca2+ release from the ER, which in turn activates the downstream signaling cascade to prevent fork degradation. This Ca2+-dependent genome protection pathway is also activated in response to replication stress caused by oncogene activation.


Subject(s)
DNA , Nucleotidyltransferases , DNA/genetics , DNA/metabolism , DNA Replication , DNA, Single-Stranded , Membrane Proteins , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , TRPV Cation Channels
5.
J Biol Chem ; 298(8): 102215, 2022 08.
Article in English | MEDLINE | ID: mdl-35779634

ABSTRACT

Uncontrolled resection of replication forks under stress can cause genomic instability and influence cancer formation. Extensive fork resection has also been implicated in the chemosensitivity of "BReast CAncer gene" BRCA-deficient cancers. However, how fork resection is controlled in different genetic contexts and how it affects chromosomal stability and cell survival remains incompletely understood. Here, we report a novel function of the transcription repressor ZKSCAN3 in fork protection and chromosomal stability maintenance under replication stress. We show disruption of ZKSCAN3 function causes excessive resection of replication forks by the exonuclease Exo1 and homologous DNA recombination/repair protein Mre11 following fork reversal. Interestingly, in BRCA1-deficient cells, we found ZKSCAN3 actually promotes fork resection upon replication stress. We demonstrate these anti- and pro-resection roles of ZKSCAN3, consisting of a SCAN box, Kruppel-associated box, and zinc finger domain, are mediated by its SCAN box domain and do not require the Kruppel-associated box or zinc finger domains, suggesting that the transcriptional function of ZKSCAN3 is not involved. Furthermore, despite the severe impact on fork structure and chromosomal stability, depletion of ZKSCAN3 did not affect the short-term survival of BRCA1-proficient or BRCA1-deficient cells after treatment with cancer drugs hydroxyurea, PARPi, or cisplatin. Our findings reveal a unique relationship between ZKSCAN3 and BRCA1 in fork protection and add to our understanding of the relationships between replication fork protection, chromosomal instability, and chemosensitivity.


Subject(s)
DNA Replication , Genomic Instability , Transcription Factors/metabolism , Chromosomal Instability , Humans
6.
Cancer Res ; 81(17): 4499-4513, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34215620

ABSTRACT

Nonsense-mediated RNA decay (NMD) is recognized as an RNA surveillance pathway that targets aberrant mRNAs with premature translation termination codons (PTC) for degradation, however, its molecular mechanisms and roles in health and disease remain incompletely understood. In this study, we developed a novel reporter system to accurately measure NMD activity in individual cells. A genome-wide CRISPR-Cas9 knockout screen using this reporter system identified novel NMD-promoting factors, including multiple components of the SF3B complex and other U2 spliceosome factors. Interestingly, cells with mutations in the spliceosome genes SF3B1 and U2AF1, which are commonly found in myelodysplastic syndrome (MDS) and cancers, have overall attenuated NMD activity. Compared with wild-type (WT) cells, SF3B1- and U2AF1-mutant cells were more sensitive to NMD inhibition, a phenotype that is accompanied by elevated DNA replication obstruction, DNA damage, and chromosomal instability. Remarkably, the sensitivity of spliceosome mutant cells to NMD inhibition was rescued by overexpression of RNase H1, which removes R-loops in the genome. Together, these findings shed new light on the functional interplay between NMD and RNA splicing and suggest a novel synthetic lethal strategy for the treatment of MDS and cancers with spliceosome mutations. SIGNIFICANCE: This study has developed a novel NMD reporter system and identified a potential therapeutic approach of targeting the NMD pathway to treat cancer with spliceosome gene mutations.


Subject(s)
Mutation , Myelodysplastic Syndromes/metabolism , Nonsense Mediated mRNA Decay , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Splicing Factor U2AF/genetics , Cell Cycle , Cell Line, Tumor , Chromosomal Instability , Fluorescent Dyes , Gene Expression Regulation , Genes, Reporter , Genome-Wide Association Study , Humans , K562 Cells , RNA-Binding Proteins , RNA-Seq , Ribonuclease H/metabolism , Spliceosomes
7.
PLoS One ; 13(10): e0204978, 2018.
Article in English | MEDLINE | ID: mdl-30289931

ABSTRACT

The nonsense mediated RNA decay (NMD) pathway safeguards the integrity of the transcriptome by targeting mRNAs with premature translation termination codons (PTCs) for degradation. It also regulates gene expression by degrading a large number of non-mutant RNAs (including mRNAs and noncoding RNAs) that bear NMD-inducing features. Consequently, NMD has been shown to influence development, cellular response to stress, and clinical outcome of many genetic diseases. Small molecules that can modulate NMD activity provide critical tools for understanding the mechanism and physiological functions of NMD, and they also offer potential means for treating certain genetic diseases and cancer. Therefore, there is an intense interest in identifying small-molecule NMD inhibitors or enhancers. It was previously reported that both inhibition of NMD and treatment with the AMPK-selective inhibitor Compound C (CC) induce autophagy in human cells, raising the possibility that CC may be capable of inhibiting NMD. Here we show that CC indeed has a NMD-inhibitory activity. Inhibition of NMD by CC is, however, independent of AMPK activity. As a competitive ATP analog, CC does not affect the kinase activity of SMG1, an essential NMD factor and the only known kinase in the NMD pathway. However, CC treatment down-regulates the protein levels of several NMD factors. The induction of autophagy by CC treatment is independent of ATF4, a NMD target that has been shown to promote autophagy in response to NMD inhibition. Our results reveal a new activity of CC as a NMD inhibitor, which has implications for its use in basic research and drug development.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Nonsense Mediated mRNA Decay/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Cell Line , Humans , RNA Stability/drug effects
8.
J Biol Chem ; 292(37): 15266-15276, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28765281

ABSTRACT

Persistent DNA damage induces profound alterations in gene expression that, in turn, influence tissue homeostasis, tumorigenesis, and cancer treatment outcome. However, the underlying mechanism for gene expression reprogramming induced by persistent DNA damage remains poorly understood. Here, using a highly effective bioluminescence-based reporter system and other tools, we report that persistent DNA damage inhibits nonsense-mediated RNA decay (NMD), an RNA surveillance and gene-regulatory pathway, in noncycling cells. NMD suppression by persistent DNA damage required the activity of the p38α MAPK. Activating transcription factor 3 (ATF3), an NMD target and a key stress-inducible transcription factor, was stabilized in a p38α- and NMD-dependent manner following persistent DNA damage. Our results reveal a novel p38α-dependent pathway that regulates NMD activity in response to persistent DNA damage, which, in turn, controls ATF3 expression in affected cells.


Subject(s)
Activating Transcription Factor 3/metabolism , DNA Damage , Gene Expression Regulation , Mitogen-Activated Protein Kinase 14/metabolism , Nonsense Mediated mRNA Decay , RNA, Messenger/metabolism , Activating Transcription Factor 3/chemistry , Activating Transcription Factor 3/genetics , Biomarkers/metabolism , Bleomycin/toxicity , Cells, Cultured , Cellular Senescence , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Gamma Rays/adverse effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Genes, Reporter/drug effects , Genes, Reporter/radiation effects , HEK293 Cells , Humans , Luminescent Measurements , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics , Mutagens/toxicity , Nonsense Mediated mRNA Decay/drug effects , Nonsense Mediated mRNA Decay/radiation effects , Oxidative Stress , Protein Stability/drug effects , Protein Stability/radiation effects , RNA Interference , RNA Stability/drug effects , RNA Stability/radiation effects , RNA, Messenger/chemistry
9.
Genes Dev ; 30(8): 946-59, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27083998

ABSTRACT

Dynamic regulation of RNF168-mediated ubiquitylation of histone H2A Lys13,15 (H2AK13,15ub) at DNA double-strand breaks (DSBs) is crucial for preventing aberrant DNA repair and maintaining genome stability. However, it remains unclear which deubiquitylating enzyme (DUB) removes H2AK13,15ub. Here we show that USP51, a previously uncharacterized DUB, deubiquitylates H2AK13,15ub and regulates DNA damage response. USP51 depletion results in increased spontaneous DNA damage foci and elevated levels of H2AK15ub and impairs DNA damage response. USP51 overexpression suppresses the formation of ionizing radiation-induced 53BP1 and BRCA1 but not RNF168 foci, suggesting that USP51 functions downstream from RNF168 in DNA damage response. In vitro, USP51 binds to H2A-H2B directly and deubiquitylates H2AK13,15ub. In cells, USP51 is recruited to chromatin after DNA damage and regulates the dynamic assembly/disassembly of 53BP1 and BRCA1 foci. These results show that USP51 is the DUB for H2AK13,15ub and regulates DNA damage response.


Subject(s)
DNA Damage/physiology , DNA Repair/physiology , Histones/metabolism , Ubiquitin-Specific Proteases/metabolism , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/physiology , DNA/metabolism , DNA/radiation effects , Humans , Nuclear Proteins/metabolism , Protein Binding , Radiation, Ionizing , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
10.
DNA Repair (Amst) ; 35: 106-15, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26519824

ABSTRACT

Exonuclease 1 (Exo1) has important roles in DNA metabolic transactions that are essential for genome maintenance, telomere regulation and cancer suppression. However, the mechanisms for regulating Exo1 activity in these processes remain incompletely understood. Here, we report that Exo1 activity is regulated by a direct interaction with poly(ADP-ribose) (PAR), a prominent posttranslational modification at the sites of DNA damage. This PAR-binding activity promotes the early recruitment of Exo1 to sites of DNA damage, where it is retained through an interaction with PCNA, which interacts with the C-terminus of Exo1. The effects of both PAR and PCNA on Exo1 damage association are antagonized by the 14-3-3 adaptor proteins, which interact with the central domain of Exo1. Although PAR binding inhibits both the exonuclease activity and the 5' flap endonuclease activity of purified Exo1, the pharmacological blockade of PAR synthesis does not overtly affect DNA double-strand break end resection in a cell free Xenopus egg extract. Thus, the counteracting effects of PAR on Exo1 recruitment and enzymatic activity may enable appropriate resection of DNA ends while preventing unscheduled or improper processing of DNA breaks in cells.


Subject(s)
DNA Damage , DNA Repair , Exodeoxyribonucleases/metabolism , Flap Endonucleases/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Proliferating Cell Nuclear Antigen/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Extracts , Cell Nucleus , Glycoside Hydrolases/metabolism , HEK293 Cells , Humans , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Protein Processing, Post-Translational , Xenopus
11.
FASEB J ; 28(2): 667-75, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24158394

ABSTRACT

Endosomal sorting complexes required for transport (ESCRTs) mediate sorting of ubiquitinated membrane proteins into multivesicular bodies en route to lysosomes for degradation. A mutation in CHMP2B (CHMP2B(Intron5), an ESCRT-III component) that is associated with a hereditary form of frontotemporal dementia (FTD3) disrupts the endosomal-lysosomal pathway and causes accumulation of autophagosomes and multilamellar structures. We previously demonstrated that expression of CHMP2B(Intron5) in the Drosophila eye using GMR-Gal4 causes misregulation of the Toll receptor pathway. Here, we show that ectopic expression of CHMP2B(Intron5) using eyeless-Gal4 (ey>CHMP2B(Intron5)), a driver with different spatiotemporal expression attributes than GMR-Gal4 in the Drosophila eye, causes eye deformities when compared to expression of wild-type CHMP2B (CHMP2B(WT)) and the Drosophila homologue of CHMP2B (CG4618). In addition, ey>CHMP2B(Intron5) flies showed defects in photoreceptor cell patterning and phototactic behavior. Furthermore, ey>CHMP2B(Intron5) flies showed accumulation of Notch in enlarged endosomes and up-regulation of Notch activity. Partial loss of Notch activity in ey>CHMP2B(Intron5) flies significantly rescued eye deformities, photoreceptor patterning defect, and phototactic behavior defect, indicating that these defects are primarily due to Notch misregulation. These results demonstrate that CHMP2B(Intron5) preferentially affects different receptor signaling pathways in a cellular and developmental context-dependent manner.


Subject(s)
Drosophila Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Eye/metabolism , Eye/pathology , Frontotemporal Dementia/metabolism , Receptors, Notch/metabolism , Vesicular Transport Proteins/metabolism , Animals , Blotting, Western , Drosophila , Drosophila Proteins/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Frontotemporal Dementia/genetics , Polymerase Chain Reaction , Receptors, Notch/genetics , Vesicular Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL