Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
J Neurosci ; 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227158

ABSTRACT

Cochlear hair cells (HCs) sense sound waves and allow us to hear. Loss of HCs will cause irreversible sensorineural hearing loss. It is well known that DNA damage repair plays a critical role in protecting cells in many organs. However, how HCs respond to DNA damage and how defective DNA damage repair contributes to hearing loss remain elusive.In this study, we showed that cisplatin induced DNA damage in outer hair cells (OHCs) and promoted OHC loss, leading to hearing loss in mice of either sex. Cisplatin induced the expression of Brca1, a DNA damage repair factor, in OHCs. Deficiency of Brca1 induced OHC and hearing loss, and further promoted cisplatin-induced DNA damage in OHCs, accelerating OHC loss. This study provides the first in vivo evidence demonstrating that cisplatin mainly induces DNA damage in OHCs and that BRCA1 promotes repair of DNA damage in OHCs and prevents hearing loss. Our findings not only demonstrate that DNA-damage inducible agent generates DNA damage in postmitotic HCs, but also suggest that DNA repair factors, like BRCA1, protect postmitotic HCs from DNA-damage induced cell death and hearing loss.Significance statement Sensorineural hearing loss is the most severe hearing loss caused by irreversible loss of cochlear hair cells. Hair cells are vulnerable to aging and ototoxic drug. Though DNA damage repair plays a critical role in protecting cells in many organs, it is poorly understood how DNA damage is repaired in hair cells. This study provides the first in vivo evidence demonstrating that cisplatin mainly induces DNA damage in outer hair cells and that BRCA1 promotes repair of DNA damage in outer hair cells and prevents outer hair cell loss as well as hearing loss.

2.
Plants (Basel) ; 13(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39065532

ABSTRACT

Drought is a critical factor constraining plant growth in arid regions. However, the performance and adaptive mechanism of Atriplex canescens (A. canescens) under drought stress remain unclear. Hence, a three-year experiment with three drought gradients was performed in a common garden, and the leaf functional traits, biomass and biomass partitioning patterns of A. canescens were investigated. The results showed that drought stress had significant effects on A. canescens leaf functional traits. A. canescens maintained the content of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD), but the peroxidase (POD) and catalase (CAT) activity decreased, and the content of proline (Pro) and soluble sugar (SS) increased only under heavy drought stress. Under drought stress, the leaves became smaller but denser, the specific leaf area (SLA) decreased, but the dry matter content (LDMC) maintained stability. Total biomass decreased 60% to 1758 g under heavy drought stress and the seed and leaf biomass was only 10% and 20% of non-stress group, but there had no significant difference on root biomass. More biomass was allocated to root under drought stress. The root biomass allocation ratio was doubled from 9.62% to 19.81% under heavy drought, and the root/shoot ratio (R/S) increased from 0.11 to 0.25. The MDA was significantly and negatively correlated with biomass, while the SPAD was significantly and positively correlated with total and aboveground organs biomass. The POD, CAT, Pro and SS had significant correlations with root and seed allocation ratio. The leaf morphological traits related to leaf shape and weight had significant correlations with total and aboveground biomass and biomass allocation. Our study demonstrated that under drought stress, A. canescens made tradeoffs between growth potential and drought tolerance and evolved with a conservative strategy. These findings provide more information for an in-depth understanding of the adaption strategies of A. canescens to drought stress and provide potential guidance for planting and sustainable management of A. canescens in arid and semi-arid regions.

3.
Proc Natl Acad Sci U S A ; 121(24): e2404383121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38843184

ABSTRACT

Transcription is extremely important for cellular processes but can be hindered by RNA polymerase II (RNAPII) pausing and stalling. Cockayne syndrome protein B (CSB) promotes the progression of paused RNAPII or initiates transcription-coupled nucleotide excision repair (TC-NER) to remove stalled RNAPII. However, the specific mechanism by which CSB initiates TC-NER upon damage remains unclear. In this study, we identified the indispensable role of the ARK2N-CK2 complex in the CSB-mediated initiation of TC-NER. The ARK2N-CK2 complex is recruited to damage sites through CSB and then phosphorylates CSB. Phosphorylation of CSB enhances its binding to stalled RNAPII, prolonging the association of CSB with chromatin and promoting CSA-mediated ubiquitination of stalled RNAPII. Consistent with this finding, Ark2n-/- mice exhibit a phenotype resembling Cockayne syndrome. These findings shed light on the pivotal role of the ARK2N-CK2 complex in governing the fate of RNAPII through CSB, bridging a critical gap necessary for initiating TC-NER.


Subject(s)
Cockayne Syndrome , DNA Helicases , DNA Repair Enzymes , DNA Repair , Poly-ADP-Ribose Binding Proteins , RNA Polymerase II , DNA Repair Enzymes/metabolism , DNA Repair Enzymes/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , Humans , Animals , Mice , DNA Helicases/metabolism , DNA Helicases/genetics , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , Transcription, Genetic , Phosphorylation , Casein Kinase II/metabolism , Casein Kinase II/genetics , Mice, Knockout , DNA Damage , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , Chromatin/metabolism , Ubiquitination , Excision Repair
4.
Article in English | MEDLINE | ID: mdl-38862431

ABSTRACT

Ribonuclease P (RNase P) was first described in the 1970's as an endoribonuclease acting in the maturation of precursor transfer RNAs (tRNAs). More recent studies, however, have uncovered non-canonical roles for RNase P and its components. Here, we review the recent progress of its involvement in chromatin assembly, DNA damage response, and maintenance of genome stability with implications in tumorigenesis. The possibility of RNase P as a therapeutic target in cancer is also discussed.


Subject(s)
Neoplasms , RNA Precursors , RNA, Transfer , Ribonuclease P , Ribonuclease P/metabolism , Ribonuclease P/genetics , Humans , RNA, Transfer/metabolism , RNA, Transfer/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/enzymology , RNA Precursors/metabolism , RNA Precursors/genetics , Genomic Instability , Animals , DNA Damage , RNA Processing, Post-Transcriptional , Chromatin Assembly and Disassembly/genetics
5.
Autophagy ; 20(9): 2041-2054, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38762759

ABSTRACT

Macroautophagy/autophagy is essential for the degradation and recycling of cytoplasmic materials. The initiation of this process is determined by phosphatidylinositol-3-kinase (PtdIns3K) complex, which is regulated by factor BECN1 (beclin 1). UFMylation is a novel ubiquitin-like modification that has been demonstrated to modulate several cellular activities. However, the role of UFMylation in regulating autophagy has not been fully elucidated. Here, we found that VCP/p97 is UFMylated on K109 by the E3 UFL1 (UFM1 specific ligase 1) and this modification promotes BECN1 stabilization and assembly of the PtdIns3K complex, suggesting a role for VCP/p97 UFMylation in autophagy initiation. Mechanistically, VCP/p97 UFMylation stabilizes BECN1 through ATXN3 (ataxin 3)-mediated deubiquitination. As a key component of the PtdIns3K complex, stabilized BECN1 facilitates assembly of this complex. Re-expression of VCP/p97, but not the UFMylation-defective mutant, rescued the VCP/p97 depletion-induced increase in MAP1LC3B/LC3B protein expression. We also showed that several pathogenic VCP/p97 mutations identified in a variety of neurological disorders and cancers were associated with reduced UFMylation, thus implicating VCP/p97 UFMylation as a potential therapeutic target for these diseases. Abbreviation: ATG14:autophagy related 14; Baf A1:bafilomycin A1;CMT2Y: Charcot-Marie-Toothdisease, axonal, 2Y; CYB5R3: cytochromeb5 reductase 3; DDRGK1: DDRGK domain containing 1; DMEM:Dulbecco'smodified Eagle's medium;ER:endoplasmic reticulum; FBS:fetalbovine serum;FTDALS6:frontotemporaldementia and/or amyotrophic lateral sclerosis 6; IBMPFD1:inclusion bodymyopathy with early-onset Paget disease with or withoutfrontotemporal dementia 1; LC-MS/MS:liquid chromatography tandem mass spectrometry; MAP1LC3B/LC3B:microtubule associated protein 1 light chain 3 beta; MS: massspectrometry; NPLOC4: NPL4 homolog, ubiquitin recognition factor;PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3;PIK3R4: phosphoinositide-3-kinase regulatory subunit 4; PtdIns3K:phosphatidylinositol 3-kinase; RPL26: ribosomal protein L26; RPN1:ribophorin I; SQSTM1/p62: sequestosome 1; UBA5: ubiquitin likemodifier activating enzyme 5; UFC1: ubiquitin-fold modifierconjugating enzyme 1; UFD1: ubiquitin recognition factor in ERassociated degradation 1; UFL1: UFM1 specific ligase 1; UFM1:ubiquitin fold modifier 1; UFSP2: UFM1 specific peptidase 2; UVRAG:UV radiation resistance associated; VCP/p97: valosin containingprotein; WT: wild-type.


Subject(s)
Autophagy , Beclin-1 , Ubiquitination , Valosin Containing Protein , Autophagy/physiology , Autophagy/genetics , Humans , Valosin Containing Protein/metabolism , Valosin Containing Protein/genetics , Beclin-1/metabolism , Ataxin-3/metabolism , Ataxin-3/genetics , Ubiquitin-Protein Ligases/metabolism , HeLa Cells , Phosphatidylinositol 3-Kinases/metabolism , Protein Stability , HEK293 Cells , Intracellular Signaling Peptides and Proteins
6.
Adv Sci (Weinh) ; 11(28): e2403485, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38803048

ABSTRACT

DNA damage plays a significant role in the tumorigenesis and progression of the disease. Abnormal DNA repair affects the therapy and prognosis of cancer. In this study, it is demonstrated that the deubiquitinase USP25 promotes non-homologous end joining (NHEJ), which in turn contributes to chemoresistance in cancer. It is shown that USP25 deubiquitinates SHLD2 at the K64 site, which enhances its binding with REV7 and promotes NHEJ. Furthermore, USP25 deficiency impairs NHEJ-mediated DNA repair and reduces class switch recombination (CSR) in USP25-deficient mice. USP25 is overexpressed in a subset of colon cancers. Depletion of USP25 sensitizes colon cancer cells to IR, 5-Fu, and cisplatin. TRIM25 is also identified, an E3 ligase, as the enzyme responsible for degrading USP25. Downregulation of TRIM25 leads to an increase in USP25 levels, which in turn induces chemoresistance in colon cancer cells. Finally, a peptide that disrupts the USP25-SHLD2 interaction is successfully identified, impairing NHEJ and increasing sensitivity to chemotherapy in PDX model. Overall, these findings reveal USP25 as a critical effector of SHLD2 in regulating the NHEJ repair pathway and suggest its potential as a therapeutic target for cancer therapy.


Subject(s)
DNA Breaks, Double-Stranded , Ubiquitin Thiolesterase , Animals , Mice , DNA Breaks, Double-Stranded/drug effects , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Humans , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Disease Models, Animal , DNA Repair/genetics , DNA End-Joining Repair/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/drug therapy , Transcription Factors/genetics , Transcription Factors/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
7.
J Biol Chem ; 300(6): 107312, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657865

ABSTRACT

Homologous-recombination deficiency due to breast cancer 1/2 (BRCA1/2) mutations or mimicking BRCA1/2 mutations confer synthetic lethality with poly-(ADP)-ribose polymerase 1/2 inhibitors. The chromatin regulator Pax2 transactivation domain interacting protein (PTIP) promotes stalled replication fork degradation in BRCA1-deficient cells, but the underlying mechanism by which PTIP regulates stalled replication fork stability is unclear. Here, we performed a series of in vitro analyses to dissect the function of UFMylation in regulating fork stabilization in BRCA1-deficient cells. By denaturing co-immunoprecipitation, we first found that replication stress can induce PTIP UFMylation. Interestingly, this post-translational modification promotes end resection and degradation of nascent DNA at stalled replication forks in BRCA1-deficient cells. By cell viability assay, we found that PTIP-depleted and UFL1-depleted BRCA1 knockdown cells are less sensitive to poly-(ADP)-ribose polymerase inhibitors than the siRNA targeting negative control BRCA1-deficient cells. These results identify a new mechanism by which PTIP UFMylation confers chemoresistance in BRCA1-deficient cells.


Subject(s)
BRCA1 Protein , DNA Replication , Humans , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Carrier Proteins/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Ubiquitination , Protein Processing, Post-Translational , DNA-Binding Proteins
8.
BMC Biol ; 22(1): 85, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627785

ABSTRACT

BACKGROUND: Inadequate DNA damage repair promotes aberrant differentiation of mammary epithelial cells. Mammary luminal cell fate is mainly determined by a few transcription factors including GATA3. We previously reported that GATA3 functions downstream of BRCA1 to suppress aberrant differentiation in breast cancer. How GATA3 impacts DNA damage repair preventing aberrant cell differentiation in breast cancer remains elusive. We previously demonstrated that loss of p18, a cell cycle inhibitor, in mice induces luminal-type mammary tumors, whereas depletion of either Brca1 or Gata3 in p18 null mice leads to basal-like breast cancers (BLBCs) with activation of epithelial-mesenchymal transition (EMT). We took advantage of these mutant mice to examine the role of Gata3 as well as the interaction of Gata3 and Brca1 in DNA damage repair in mammary tumorigenesis. RESULTS: Depletion of Gata3, like that of Brca1, promoted DNA damage accumulation in breast cancer cells in vitro and in basal-like breast cancers in vivo. Reconstitution of Gata3 improved DNA damage repair in Brca1-deficient mammary tumorigenesis. Overexpression of GATA3 promoted homologous recombination (HR)-mediated DNA damage repair and restored HR efficiency of BRCA1-deficient cells. Depletion of Gata3 sensitized tumor cells to PARP inhibitor (PARPi), and reconstitution of Gata3 enhanced resistance of Brca1-deficient tumor cells to PARP inhibitor. CONCLUSIONS: These results demonstrate that Gata3 functions downstream of BRCA1 to promote DNA damage repair and suppress dedifferentiation in mammary tumorigenesis and progression. Our findings suggest that PARP inhibitors are effective for the treatment of GATA3-deficient BLBCs.


Subject(s)
Mammary Neoplasms, Animal , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Mice , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Damage , DNA Repair , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
9.
Oncogene ; 43(23): 1769-1778, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38632437

ABSTRACT

Pyruvate kinase M2 (PKM2) is a central metabolic enzyme driving the Warburg effect in tumor growth. Previous investigations have demonstrated that PKM2 is subject to O-linked ß-N-acetylglucosamine (O-GlcNAc) modification, which is a nutrient-sensitive post-translational modification. Here we found that unc-51 like autophagy activating kinase 1 (ULK1), a glucose-sensitive kinase, interacts with PKM2 and phosphorylates PKM2 at Ser333. Ser333 phosphorylation antagonizes PKM2 O-GlcNAcylation, promotes its tetramer formation and enzymatic activity, and decreases its nuclear localization. As PKM2 is known to have a nuclear role in regulating c-Myc, we also show that PKM2-S333 phosphorylation inhibits c-Myc expression. By downregulating glucose consumption and lactate production, PKM2 pS333 attenuates the Warburg effect. Through mouse xenograft assays, we demonstrate that the phospho-deficient PKM2-S333A mutant promotes tumor growth in vivo. In conclusion, we identified a ULK1-PKM2-c-Myc axis in inhibiting breast cancer, and a glucose-sensitive phosphorylation of PKM2 in modulating the Warburg effect.


Subject(s)
Autophagy-Related Protein-1 Homolog , Breast Neoplasms , Carrier Proteins , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Warburg Effect, Oncologic , Humans , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Phosphorylation , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Female , Mice , Thyroid Hormones/metabolism , Thyroid Hormones/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Cell Line, Tumor , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Acetylglucosamine/metabolism
10.
Proc Natl Acad Sci U S A ; 121(18): e2322520121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38657044

ABSTRACT

The S-phase checkpoint involving CHK1 is essential for fork stability in response to fork stalling. PARP1 acts as a sensor of replication stress and is required for CHK1 activation. However, it is unclear how the activity of PARP1 is regulated. Here, we found that UFMylation is required for the efficient activation of CHK1 by UFMylating PARP1 at K548 during replication stress. Inactivation of UFL1, the E3 enzyme essential for UFMylation, delayed CHK1 activation and inhibits nascent DNA degradation during replication blockage as seen in PARP1-deficient cells. An in vitro study indicated that PARP1 is UFMylated at K548, which enhances its catalytic activity. Correspondingly, a PARP1 UFMylation-deficient mutant (K548R) and pathogenic mutant (F553L) compromised CHK1 activation, the restart of stalled replication forks following replication blockage, and chromosome stability. Defective PARP1 UFMylation also resulted in excessive nascent DNA degradation at stalled replication forks. Finally, we observed that PARP1 UFMylation-deficient knock-in mice exhibited increased sensitivity to replication stress caused by anticancer treatments. Thus, we demonstrate that PARP1 UFMylation promotes CHK1 activation and replication fork stability during replication stress, thus safeguarding genome integrity.


Subject(s)
Checkpoint Kinase 1 , DNA Replication , Poly (ADP-Ribose) Polymerase-1 , Animals , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/genetics , Mice , Humans , DNA Damage , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Cell Rep ; 43(2): 113779, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38358891

ABSTRACT

R-loops are three-stranded structures that can pose threats to genome stability. RNase H1 precisely recognizes R-loops to drive their resolution within the genome, but the underlying mechanism is unclear. Here, we report that ARID1A recognizes R-loops with high affinity in an ATM-dependent manner. ARID1A recruits METTL3 and METTL14 to the R-loop, leading to the m6A methylation of R-loop RNA. This m6A modification facilitates the recruitment of RNase H1 to the R-loop, driving its resolution and promoting DNA end resection at DSBs, thereby ensuring genome stability. Depletion of ARID1A, METTL3, or METTL14 leads to R-loop accumulation and reduced cell survival upon exposure to cytotoxic agents. Therefore, ARID1A, METTL3, and METTL14 function in a coordinated, temporal order at DSB sites to recruit RNase H1 and to ensure efficient R-loop resolution. Given the association of high ARID1A levels with resistance to genotoxic therapies in patients, these findings open avenues for exploring potential therapeutic strategies for cancers with ARID1A abnormalities.


Subject(s)
Adenine/analogs & derivatives , R-Loop Structures , RNA , Ribonuclease H , Humans , Genomic Instability , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Methyltransferases/genetics
12.
Cells ; 12(21)2023 10 29.
Article in English | MEDLINE | ID: mdl-37947621

ABSTRACT

Ubiquitin-fold modifier 1 (UFM1) is a newly identified ubiquitin-like protein that has been conserved during the evolution of multicellular organisms. In a similar manner to ubiquitin, UFM1 can become covalently linked to the lysine residue of a substrate via a dedicated enzymatic cascade. Although a limited number of substrates have been identified so far, UFM1 modification (UFMylation) has been demonstrated to play a vital role in a variety of cellular activities, including mammalian development, ribosome biogenesis, the DNA damage response, endoplasmic reticulum stress responses, immune responses, and tumorigenesis. In this review, we summarize what is known about the UFM1 enzymatic cascade and its biological functions, and discuss its recently identified substrates. We also explore the pathological role of UFMylation in human disease and the corresponding potential therapeutic targets and strategies.


Subject(s)
Protein Processing, Post-Translational , Proteins , Animals , Humans , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Ubiquitins/metabolism , Mammals/metabolism
13.
Biochim Biophys Acta Gene Regul Mech ; 1866(4): 194992, 2023 12.
Article in English | MEDLINE | ID: mdl-37797785

ABSTRACT

The forkhead box subfamily P (FOXP) of transcription factors, consisting of FOXP1, FOXP2, FOXP3, and FOXP4, is involved in the regulation of multisystemic functioning. Disruption of the transcriptional activity of FOXP proteins leads to neurodevelopmental disorders and immunological diseases, as well as the suppression or promotion of carcinogenesis. The transcriptional activities of FOXP proteins are directly or indirectly regulated by diverse post-translational modifications, including phosphorylation, ubiquitination, SUMOylation, acetylation, O-GlcNAcylation, and methylation. Here, we discuss how post-translational modifications modulate the multiple functions of FOXP proteins and examine the implications for tumorigenesis and cancer therapy.


Subject(s)
Protein Processing, Post-Translational , Transcription Factors
14.
Nucleic Acids Res ; 51(22): 12207-12223, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-37897354

ABSTRACT

Following a DNA double strand break (DSB), several nucleases and helicases coordinate to generate single-stranded DNA (ssDNA) with 3' free ends, facilitating precise DNA repair by homologous recombination (HR). The same nucleases can act on stalled replication forks, promoting nascent DNA degradation and fork instability. Interestingly, some HR factors, such as CtIP and BRCA1, have opposite regulatory effects on the two processes, promoting end resection at DSB but inhibiting the degradation of nascent DNA on stalled forks. However, the reason why nuclease actions are regulated by different mechanisms in two DNA metabolism is poorly understood. We show that human HELQ acts as a DNA end resection regulator, with opposing activities on DNA end resection at DSBs and on stalled forks as seen for other regulators. Mechanistically, HELQ helicase activity is required for EXO1-mediated DSB end resection, while ssDNA-binding capacity of HELQ is required for its recruitment to stalled forks, facilitating fork protection and preventing chromosome aberrations caused by replication stress. Here, HELQ synergizes with CtIP but not BRCA1 or BRCA2 to protect stalled forks. These findings reveal an unanticipated role of HELQ in regulating DNA end resection at DSB and stalled forks, which is important for maintaining genome stability.


Subject(s)
DNA Breaks, Double-Stranded , DNA Replication , Humans , DNA Helicases/genetics , DNA Repair , Homologous Recombination/genetics
15.
J Biol Chem ; 299(11): 105354, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37858678

ABSTRACT

O-linked N-acetylglucosamine (O-GlcNAc) glycosylation, a prevalent protein post-translational modification (PTM) that occurs intracellularly, has been shown to crosstalk with phosphorylation and ubiquitination. However, it is unclear whether it interplays with other PTMs. Here we studied its relationship with ADP-ribosylation, which involves decorating target proteins with the ADP-ribose moiety. We discovered that the poly(ADP-ribosyl)ation "eraser", ADP-ribose glycohydrolase (PARG), is O-GlcNAcylated at Ser26, which is in close proximity to its nuclear localization signal. O-GlcNAcylation of PARG promotes nuclear localization and chromatin association. Upon DNA damage, O-GlcNAcylation augments the recruitment of PARG to DNA damage sites and interacting with proliferating cell nuclear antigen (PCNA). In hepatocellular carcinoma (HCC) cells, PARG O-GlcNAcylation enhances the poly(ADP-ribosyl)ation of DNA damage-binding protein 1 (DDB1) and attenuates its auto-ubiquitination, thereby stabilizing DDB1 and allowing it to degrade its downstream targets, such as c-Myc. We further demonstrated that PARG-S26A, the O-GlcNAc-deficient mutant, promoted HCC in mouse xenograft models. Our findings thus reveal that PARG O-GlcNAcylation inhibits HCC, and we propose that O-GlcNAc glycosylation may crosstalk with many other PTMs.


Subject(s)
Carcinoma, Hepatocellular , Glycoside Hydrolases , Liver Neoplasms , Animals , Humans , Mice , Acetylglucosamine , ADP-Ribosylation , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Glycosylation , Protein Processing, Post-Translational
16.
Nat Struct Mol Biol ; 30(11): 1719-1734, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37735618

ABSTRACT

Chromatin relaxation is a prerequisite for the DNA repair machinery to access double-strand breaks (DSBs). Local histones around the DSBs then undergo prompt changes in acetylation status, but how the large demands of acetyl-CoA are met is unclear. Here, we report that pyruvate dehydrogenase 1α (PDHE1α) catalyzes pyruvate metabolism to rapidly provide acetyl-CoA in response to DNA damage. We show that PDHE1α is quickly recruited to chromatin in a polyADP-ribosylation-dependent manner, which drives acetyl-CoA generation to support local chromatin acetylation around DSBs. This process increases the formation of relaxed chromatin to facilitate repair-factor loading, genome stability and cancer cell resistance to DNA-damaging treatments in vitro and in vivo. Indeed, we demonstrate that blocking polyADP-ribosylation-based PDHE1α chromatin recruitment attenuates chromatin relaxation and DSB repair efficiency, resulting in genome instability and restored radiosensitivity. These findings support a mechanism in which chromatin-associated PDHE1α locally generates acetyl-CoA to remodel the chromatin environment adjacent to DSBs and promote their repair.


Subject(s)
Chromatin , DNA Breaks, Double-Stranded , Acetyl Coenzyme A/metabolism , Acetylation , DNA Repair , DNA Damage , Pyruvates
17.
Biosci Rep ; 43(10)2023 10 31.
Article in English | MEDLINE | ID: mdl-37728310

ABSTRACT

DNA replication forks are subject to intricate surveillance and strict regulation by sophisticated cellular machinery. Such close regulation is necessary to ensure the accurate duplication of genetic information and to tackle the diverse endogenous and exogenous stresses that impede this process. Stalled replication forks are vulnerable to collapse, which is a major cause of genomic instability and carcinogenesis. Replication stress responses, which are organized via a series of coordinated molecular events, stabilize stalled replication forks and carry out fork reversal and restoration. DNA damage tolerance and repair pathways such as homologous recombination and Fanconi anemia also contribute to replication fork stabilization. The signaling network that mediates the transduction and interplay of these pathways is regulated by a series of post-translational modifications, including ubiquitination, which affects the activity, stability, and interactome of substrates. In particular, the ubiquitination of replication protein A and proliferating cell nuclear antigen at stalled replication forks promotes the recruitment of downstream regulators. In this review, we describe the ubiquitination-mediated signaling cascades that regulate replication fork progression and stabilization. In addition, we discuss the targeting of replication fork stability and ubiquitination system components as a potential therapeutic approach for the treatment of cancer.


Subject(s)
DNA Replication , Neoplasms , Humans , DNA Replication/genetics , Ubiquitination , DNA Damage , Genomic Instability , Homologous Recombination , Neoplasms/genetics , Neoplasms/therapy
18.
Front Neurosci ; 17: 1242448, 2023.
Article in English | MEDLINE | ID: mdl-37599996

ABSTRACT

Primary microcephaly (MCPH), is a neurological disorder characterized by small brain size that results in numerous developmental problems, including intellectual disability, motor and speech delays, and seizures. Hitherto, over 30 MCPH causing genes (MCPHs) have been identified. Among these MCPHs, MCPH5, which encodes abnormal spindle-like microcephaly-associated protein (ASPM), is the most frequently mutated gene. ASPM regulates mitotic events, cell proliferation, replication stress response, DNA repair, and tumorigenesis. Moreover, using a data mining approach, we have confirmed that high levels of expression of ASPM correlate with poor prognosis in several types of tumors. Here, we summarize the neurological and non-neurological functions of ASPM and provide insight into its implications for the diagnosis and treatment of MCPH and cancer.

19.
Acta Biochim Biophys Sin (Shanghai) ; 55(12): 1864-1873, 2023 12 25.
Article in English | MEDLINE | ID: mdl-37559455

ABSTRACT

DNA double-strand break (DSB) repair by homologous recombination (HR) is crucial for the maintenance of genome stability and integrity. In this study, we aim to identify novel RNA binding proteins (RBPs) involved in HR repair because little is known about RBP function in HR. For this purpose, we carry out pulldown assays using a synthetic ssDNA/dsDNA structure coated with replication protein A (RPA) to mimic resected DNA, a crucial intermediate in HR-mediated DSB repair. Using this approach, we identify RNA-binding motif protein 14 (RBM14) as a potential binding partner. We further show that RBM14 interacts with an essential HR repair factor, CtIP. RBM14 is crucial for CtIP recruitment to DSB sites and for subsequent RPA coating and RAD51 replacement, facilitating efficient HR repair. Moreover, inhibition of RBM14 expression sensitizes cancer cells to X-ray irradiation. Together, our results demonstrate that RBM14 promotes DNA end resection to ensure HR repair and may serve as a potential target for cancer therapy.


Subject(s)
DNA Breaks, Double-Stranded , Recombinational DNA Repair , DNA Repair , Homologous Recombination , Replication Protein A/genetics , DNA/genetics , DNA End-Joining Repair
20.
Nucleic Acids Res ; 51(17): 9166-9182, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37503842

ABSTRACT

Histone deacetylase 6 (HDAC6) mediates DNA damage signaling by regulating the mismatch repair and nucleotide excision repair pathways. Whether HDAC6 also mediates DNA double-strand break (DSB) repair is unclear. Here, we report that HDAC6 negatively regulates DSB repair in an enzyme activity-independent manner. In unstressed cells, HDAC6 interacts with H2A/H2A.X to prevent its interaction with the E3 ligase RNF168. Upon sensing DSBs, RNF168 rapidly ubiquitinates HDAC6 at lysine 116, leading to HDAC6 proteasomal degradation and a restored interaction between RNF168 and H2A/H2A.X. H2A/H2A.X is ubiquitinated by RNF168, precipitating the recruitment of DSB repair factors (including 53BP1 and BRCA1) to chromatin and subsequent DNA repair. These findings reveal novel regulatory machinery based on an HDAC6-RNF168 axis that regulates the H2A/H2A.X ubiquitination status. Interfering with this axis might be leveraged to disrupt a key mechanism of cancer cell resistance to genotoxic damage and form a potential therapeutic strategy for cancer.


Subject(s)
DNA Repair , Humans , Cell Line, Tumor , DNA Damage , Histone Deacetylase 6/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL