Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
EMBO Rep ; 25(9): 3970-3989, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39090319

ABSTRACT

The tandem Tudor-like domain-containing protein Spindlin1 (SPIN1) is a transcriptional coactivator with critical functions in embryonic development and emerging roles in cancer. However, the involvement of SPIN1 in DNA damage repair has remained unclear. Our study shows that SPIN1 is recruited to DNA lesions through its N-terminal disordered region that binds to Poly-ADP-ribose (PAR), and facilitates homologous recombination (HR)-mediated DNA damage repair. SPIN1 promotes H3K9me3 accumulation at DNA damage sites and enhances the interaction between H3K9me3 and Tip60, thereby promoting the activation of ATM and HR repair. We also show that SPIN1 increases chemoresistance. These findings reveal a novel role for SPIN1 in the activation of H3K9me3-dependent DNA repair pathways, and suggest that SPIN1 may contribute to cancer chemoresistance by modulating the efficiency of double-strand break (DSB) repair.


Subject(s)
Cell Cycle Proteins , Drug Resistance, Neoplasm , Histones , Lysine Acetyltransferase 5 , Phosphoproteins , Protein Binding , Humans , Drug Resistance, Neoplasm/genetics , Histones/metabolism , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferase 5/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , DNA Breaks, Double-Stranded , Recombinational DNA Repair , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Poly Adenosine Diphosphate Ribose/metabolism , Cell Line, Tumor , DNA Damage , DNA Repair , Microtubule-Associated Proteins
2.
Nat Commun ; 15(1): 7092, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154037

ABSTRACT

Mammalian TIP60 is a multi-functional enzyme with histone acetylation and histone dimer exchange activities. It plays roles in diverse cellular processes including transcription, DNA repair, cell cycle control, and embryonic development. Here we report the cryo-electron microscopy structures of the human TIP60 complex with the core subcomplex and TRRAP module refined to 3.2-Å resolution. The structures show that EP400 acts as a backbone integrating the motor module, the ARP module, and the TRRAP module. The RUVBL1-RUVBL2 hexamer serves as a rigid core for the assembly of EP400 ATPase and YL1 in the motor module. In the ARP module, an ACTL6A-ACTB heterodimer and an extra ACTL6A make hydrophobic contacts with EP400 HSA helix, buttressed by network interactions among DMAP1, EPC1, and EP400. The ARP module stably associates with the motor module but is flexibly tethered to the TRRAP module, exhibiting a unique feature of human TIP60. The architecture of the nucleosome-bound human TIP60 reveals an unengaged nucleosome that is located between the core subcomplex and the TRRAP module. Our work illustrates the molecular architecture of human TIP60 and provides architectural insights into how this complex is bound by the nucleosome.


Subject(s)
Cryoelectron Microscopy , Lysine Acetyltransferase 5 , Humans , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferase 5/chemistry , Lysine Acetyltransferase 5/genetics , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Nucleosomes/chemistry , DNA Helicases/metabolism , DNA Helicases/chemistry , Models, Molecular , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/genetics , Carrier Proteins/metabolism , Carrier Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Protein Binding , Protein Multimerization , Bromodomain Containing Proteins , Adaptor Proteins, Signal Transducing
3.
Cancer Sci ; 115(9): 2862-2870, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38965933

ABSTRACT

The bromodomain is a conserved protein-protein interaction module that functions exclusively to recognize acetylated lysine residues on histones and other proteins. It is noteworthy that bromodomain-containing proteins are involved in transcriptional modulation by recruiting various transcription factors and/or protein complexes such as ATP-dependent chromatin remodelers and acetyltransferases. Bromodomain-containing protein 8 (BRD8), a molecule initially recognized as skeletal muscle abundant protein and thyroid hormone receptor coactivating protein of 120 kDa (TrCP120), was shown to be a subunit of the NuA4/TIP60-histone acetyltransferase complex. BRD8 has been reported to be upregulated in a subset of cancers and implicated in the regulation of cell proliferation as well as in the response to cytotoxic agents. However, little is still known about the underlying molecular mechanisms. In recent years, it has become increasingly clear that the bromodomain of BRD8 recognizes acetylated and/or nonacetylated histones H4 and H2AZ, and that BRD8 is associated with cancer development in both a NuA4/TIP60 complex-dependent and -independent manner. In this review, we will provide an overview of the current knowledge on the molecular function of BRD8, focusing on the biological role of the bromodomain of BRD8 in cancer cells.


Subject(s)
Carcinogenesis , Neoplasms , Transcription Factors , Humans , Transcription Factors/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Carcinogenesis/metabolism , Histones/metabolism , Histone Acetyltransferases/metabolism , Acetylation , Lysine Acetyltransferase 5/metabolism , Animals , Gene Expression Regulation, Neoplastic , Cell Proliferation , Bromodomain Containing Proteins
4.
EMBO Rep ; 25(9): 3925-3943, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39043961

ABSTRACT

HDAC8, a member of class I HDACs, plays a pivotal role in cell cycle regulation by deacetylating the cohesin subunit SMC3. While cyclins and CDKs are well-established cell cycle regulators, our knowledge of other regulators remains limited. Here we reveal the acetylation of K202 in HDAC8 as a key cell cycle regulator responsive to stress. K202 acetylation in HDAC8, primarily catalyzed by Tip60, restricts HDAC8 activity, leading to increased SMC3 acetylation and cell cycle arrest. Furthermore, cells expressing the mutant form of HDAC8 mimicking K202 acetylation display significant alterations in gene expression, potentially linked to changes in 3D genome structure, including enhanced chromatid loop interactions. K202 acetylation impairs cell cycle progression by disrupting the expression of cell cycle-related genes and sister chromatid cohesion, resulting in G2/M phase arrest. These findings indicate the reversible acetylation of HDAC8 as a cell cycle regulator, expanding our understanding of stress-responsive cell cycle dynamics.


Subject(s)
Cell Cycle Proteins , Cell Cycle , Chromosomal Proteins, Non-Histone , Histone Deacetylases , Repressor Proteins , Acetylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Humans , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Cell Cycle/genetics , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferase 5/genetics , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Chondroitin Sulfate Proteoglycans
5.
Nature ; 631(8021): 663-669, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961290

ABSTRACT

The Warburg effect is a hallmark of cancer that refers to the preference of cancer cells to metabolize glucose anaerobically rather than aerobically1,2. This results in substantial accumulation of lacate, the end product of anaerobic glycolysis, in cancer cells3. However, how cancer metabolism affects chemotherapy response and DNA repair in general remains incompletely understood. Here we report that lactate-driven lactylation of NBS1 promotes homologous recombination (HR)-mediated DNA repair. Lactylation of NBS1 at lysine 388 (K388) is essential for MRE11-RAD50-NBS1 (MRN) complex formation and the accumulation of HR repair proteins at the sites of DNA double-strand breaks. Furthermore, we identify TIP60 as the NBS1 lysine lactyltransferase and the 'writer' of NBS1 K388 lactylation, and HDAC3 as the NBS1 de-lactylase. High levels of NBS1 K388 lactylation predict poor patient outcome of neoadjuvant chemotherapy, and lactate reduction using either genetic depletion of lactate dehydrogenase A (LDHA) or stiripentol, a lactate dehydrogenase A inhibitor used clinically for anti-epileptic treatment, inhibited NBS1 K388 lactylation, decreased DNA repair efficacy and overcame resistance to chemotherapy. In summary, our work identifies NBS1 lactylation as a critical mechanism for genome stability that contributes to chemotherapy resistance and identifies inhibition of lactate production as a promising therapeutic cancer strategy.


Subject(s)
Cell Cycle Proteins , Drug Resistance, Neoplasm , Lactic Acid , Nuclear Proteins , Recombinational DNA Repair , Animals , Female , Humans , Male , Mice , Acid Anhydride Hydrolases/metabolism , Anaerobiosis , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Genomic Instability , Lactic Acid/metabolism , Lysine/chemistry , Lysine/metabolism , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferase 5/genetics , MRE11 Homologue Protein/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Organoids , Glycolysis , Neoadjuvant Therapy , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/deficiency , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Anticonvulsants/pharmacology
6.
Genes Genomics ; 46(7): 871-879, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805168

ABSTRACT

BACKGROUND: Colon cancer is the third most common cancer globally. The expression of histone deacetylase 3 (HDAC3) is upregulated, whereas the expression of tat interactive protein, 60 kDa (TIP60) is downregulated in colon cancer. However, the relationship between HDAC3 and TIP60 in colon cancer has not been clearly elucidated. OBJECTIVE: We investigated whether TIP60 could regulate the expression of HDAC3 and suppress colon cancer cell proliferation. METHODS: RNA sequencing data (GSE108834) showed that HDAC3 expression was regulated by TIP60. Subsequently, we generated TIP60-knockdown HCT116 cells and examined the expression of HDAC3 by western blotting and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We examined the expression pattern of HDAC3 in various cancers using publicly available datasets. The promoter activity of HDAC3 was validated using a dual-luciferase assay, and transcription factors binding to HDAC3 were identified using GeneCards and Promo databases, followed by validation using chromatin immunoprecipitation-quantitative polymerase chain reaction. Cell proliferation and apoptosis were assessed using colony formation assays and fluorescence-activated cell sorting analysis of HCT116 cell lines. RESULTS: In response to TIP60 knockdown, the expression level and promoter activity of HDAC3 increased. Conversely, when HDAC3 was downregulated by overexpression of TIP60, proliferation of HCT116 cells was inhibited and apoptosis was promoted. CONCLUSION: TIP60 plays a crucial role in the regulation of HDAC3 transcription, thereby influencing cell proliferation and apoptosis in colon cancer. Consequently, TIP60 may function as a tumor suppressor by inhibiting HDAC3 expression in colon cancer cells.


Subject(s)
Cell Proliferation , Colonic Neoplasms , Gene Expression Regulation, Neoplastic , Histone Deacetylases , Lysine Acetyltransferase 5 , Humans , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Cell Proliferation/genetics , HCT116 Cells , Apoptosis/genetics , Promoter Regions, Genetic
7.
Immunol Res ; 72(4): 741-753, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38630408

ABSTRACT

Massive evidence shows that intestinal tryptophan metabolites affected by intestinal flora can modulate the progression of rheumatoid arthritis (RA). However, the effects and mechanisms of intestinal tryptophan metabolites on RA are not yet detailed. Herein, we investigated the protective effects of intestinal tryptophan metabolites on RA and its detailed mechanisms. In this study, the collagen-induced arthritis (CIA) rat model was established. Based on metabolomics analysis, the contents of ß-indole-3-acetic acid (IAA), indolylpropionic acid, and indole-3-ß-acrylic acid in the sera of CIA rats were significantly less compared with those of the normal rats. Under the condition of Treg or Th17 cell differentiation, IAA significantly promoted the differentiation and activation of Treg cells instead of Th17 cells. Intestinal tryptophan metabolites are well-known endogenic ligands of aryl hydrocarbon receptor (AhR). Not surprisingly, IAA increased the level of Foxp3 through activating the AhR pathway. Interestingly, IAA had little impact on the level of Foxp3 mRNA, but reducing the ubiquitination and degradation of Foxp3. Mechanically, IAA reduced the expression of the transcriptional coactivator TAZ, which was almost completely reversed by either AhR antagonist CH223191 or siRNA. In vitro, IAA decreased the combination of TAZ and the histone acetyltransferase Tip60, while it increased the combination of Tip60 and Foxp3. In CIA rats, oral administration of IAA increased the number of Treg cells and relieved the inflammation. A combined use with CH223191 almost abolished the effect of IAA. Taken together, IAA attenuated CIA by promoting the differentiation of Treg cells through reducing the ubiquitination of Foxp3 via the AhR-TAZ-Tip60 pathway.


Subject(s)
Arthritis, Experimental , Forkhead Transcription Factors , Indoleacetic Acids , Lysine Acetyltransferase 5 , Receptors, Aryl Hydrocarbon , Signal Transduction , T-Lymphocytes, Regulatory , Th17 Cells , Ubiquitination , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Forkhead Transcription Factors/metabolism , Rats , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Signal Transduction/drug effects , Indoleacetic Acids/metabolism , Lysine Acetyltransferase 5/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Humans , Male , Cell Differentiation , Disease Models, Animal
8.
PLoS One ; 19(4): e0300732, 2024.
Article in English | MEDLINE | ID: mdl-38662722

ABSTRACT

KAT5 (S. pombe Mst1, human TIP60) is a MYST family histone acetyltransferase conserved from yeast to humans that is involved in multiple cellular activities. This family is characterized in part by containing a chromodomain, a motif associated with binding methylated histones. We show that a chromodomain mutation in the S. pombe Kat5, mst1-W66R, has defects in pericentromere silencing. mst1-W66R is sensitive to camptothecin (CPT) but only at an increased temperature of 36°C, although it is proficient for growth at this temperature. We also describe a de-silencing effect at the pericentromere by CPT that is independent of RNAi and methylation machinery. We also show that mst1-W66R disrupts recruitment of proteins to repair foci in response to camptothecin-induced DNA damage. Our data suggest a function of Mst1 chromodomain in centromere heterochromatin formation and a separate role in genome-wide damage repair in CPT.


Subject(s)
Centromere , DNA Repair , Mutation , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Schizosaccharomyces/genetics , Schizosaccharomyces/drug effects , Schizosaccharomyces/metabolism , Centromere/metabolism , Centromere/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Camptothecin/pharmacology , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferase 5/genetics , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , DNA Damage , Heterochromatin/metabolism , Heterochromatin/genetics , Humans
9.
Commun Biol ; 7(1): 396, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561411

ABSTRACT

Myocardial ischemia-reperfusion injury (MIRI) is involved in the pathogenesis of multiple cardiovascular diseases. This study elucidated the biological function of lysine acetyltransferase 5 (KAT5) in cardiomyocyte pyroptosis during MIRI. Oxygen-glucose deprivation/reoxygenation and left anterior descending coronary artery ligation were used to establish MIRI models. Here we show, KAT5 and STIP1 homology and U-box-containing protein 1 (STUB1) were downregulated, while large tumor suppressor kinase 2 (LATS2) was upregulated in MIRI models. KAT5/STUB1 overexpression or LATS2 silencing repressed cardiomyocyte pyroptosis. Mechanistically, KAT5 promoted STUB1 transcription via acetylation modulation, and subsequently caused ubiquitination and degradation of LATS2, which activated YAP/ß-catenin pathway. Notably, the inhibitory effect of STUB1 overexpression on cardiomyocyte pyroptosis was abolished by LATS2 overexpression or KAT5 depletion. Our findings suggest that KAT5 overexpression inhibits NLRP3-mediated cardiomyocyte pyroptosis to relieve MIRI through modulation of STUB1/LATS2/YAP/ß-catenin axis, providing a potential therapeutic target for MIRI.


Subject(s)
Myocardial Reperfusion Injury , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Pyroptosis , Ubiquitination , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Lysine Acetyltransferase 5/metabolism
10.
J Virol ; 98(3): e0169523, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38349085

ABSTRACT

Histone modifications function in both cellular and viral gene expression. However, the roles of acetyltransferases and histone acetylation in parvoviral infection remain poorly understood. In the current study, we found the histone deacetylase (HDAC) inhibitor, trichostatin A (TSA), promoted the replication and transcription of parvovirus minute virus of canines (MVC). Notably, the expression of host acetyltransferases KAT5, GTF3C4, and KAT2A was increased in MVC infection, as well as H4 acetylation (H4K12ac). KAT5 is not only responsible for H4K12ac but also crucial for viral replication and transcription. The viral nonstructural protein NS1 interacted with KAT5 and enhanced its expression. Further study showed that Y44 in KAT5, which may be tyrosine-phosphorylated, is indispensable for NS1-mediated enhancement of KAT5 and efficient MVC replication. The data demonstrated that NS1 interacted with KAT5, which resulted in an enhanced H4K12ac level to promote viral replication and transcription, implying the epigenetic addition of H4K12ac in viral chromatin-like structure by KAT5 is vital for MVC replication.IMPORTANCEParvoviral genomes are chromatinized with host histones. Therefore, histone acetylation and related acetyltransferases are required for the virus to modify histones and open densely packed chromatin structures. This study illustrated that histone acetylation status is important for MVC replication and transcription and revealed a novel mechanism that the viral nonstructural protein NS1 hijacks the host acetyltransferase KAT5 to enhance histone acetylation of H4K12ac, which relies on a potential tyrosine phosphorylation site, Y44 in KAT5. Other parvoviruses share a similar genome organization and coding potential and may adapt a similar strategy for efficient viral replication and transcription.


Subject(s)
Lysine Acetyltransferase 5 , Parvoviridae Infections , Animals , Dogs , Acetylation , Acetyltransferases/metabolism , Chromatin , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Parvoviridae Infections/metabolism , Parvoviridae Infections/veterinary , Parvoviridae Infections/virology , Tyrosine/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Cell Line , Dog Diseases/metabolism , Dog Diseases/virology , Lysine Acetyltransferase 5/metabolism
11.
J Mol Biol ; 436(7): 168414, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38141874

ABSTRACT

The lysine acetyltransferase KAT5 is a pivotal enzyme responsible for catalyzing histone H4 acetylation in cells. In addition to its indispensable HAT domain, KAT5 also encompasses a conserved Tudor-knot domain at its N-terminus. However, the function of this domain remains elusive, with conflicting findings regarding its role as a histone reader. In our study, we have employed a CRISPR tiling array approach and unveiled the Tudor-knot motif as an essential domain for cell survival. The Tudor-knot domain does not bind to histone tails and is not required for KAT5's chromatin occupancy. However, its absence leads to a global reduction in histone acetylation, accompanied with genome-wide alterations in gene expression that consequently result in diminished cell viability. Mechanistically, we find that the Tudor-knot domain regulates KAT5's HAT activity on nucleosomes by fine-tuning substrate accessibility. In summary, our study uncovers the Tudor-knot motif as an essential domain for cell survival and reveals its critical role in modulating KAT5's catalytic efficiency on nucleosome and KAT5-dependent transcriptional programs critical for cell viability.


Subject(s)
Histones , Lysine Acetyltransferase 5 , Nucleosomes , Tudor Domain , Acetylation , Chromatin/metabolism , Histones/genetics , Histones/metabolism , Nucleosomes/metabolism , Lysine Acetyltransferase 5/chemistry , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Humans
12.
Mol Immunol ; 163: 147-162, 2023 11.
Article in English | MEDLINE | ID: mdl-37793204

ABSTRACT

BACKGROUND: Aryl hydrocarbon receptor (AhR) plays an important role in the occurrence and development of ulcerative colitis (UC). In this study, the effect and mechanism of 3, 3'-diindolylmethane (DIM), the classical AhR agonist, on UC was investigated from the angle of recovering the balance of Th17/Treg. METHODS: The in vivo colitis model was established in mice by using dextran sulfate sodium, and CD4+ T cells were used to simulate the in vitro differentiation of Treg and Th17 cells. The proportions and related factors of Th17 and Treg cells were measured using flow cytometry, Q-PCR and western blotting. The glycolysis was evaluated by examining the glucose uptake, glucose consumption and lactate production using kits or immunofluorescence. The activation of AhR was detected by western blotting and the XRE-luciferase reporter gene. The co-immunoprecipitation, transfection or other methods were selected to investigate and identify the signaling molecular pathway. RESULTS: DIM significantly attenuated symptoms of colitis mice by rebuilding the balance of Th17/Treg in anoxic colons. In hypoxia, a more potent promotion of Treg differentiation was showed by DIM relative to normoxia, and siFoxp3 prevented DIM-suppressed Th17 differentiation. DIM repressed the excessive glycolysis in hypoxia evidenced by down-regulated glucose uptake, lactate production, Glut1 and HK2 levels. Interestingly, IL-10, the function-related factor of Treg cells, showed the feedback effect of DIM-suppressed glycolysis. Besides, 2-deoxy-D-glucose, HK2 plasmid and IL-10 antibody prevented increase of DIM on the expression of Foxp3 at the transcriptional level and subsequent Treg differentiation through the lactate-STAT3 pathway, and reasons for the direct improvement of DIM on Foxp3 protein was attributed to promoting the formation of HIF-1α/TIP60 complexes as well as subsequent acetylation and protein stability. Finally, AhR dependence and mechanisms for DIM-improved Treg differentiation in vitro and in vivo were well confirmed by using plasmids or inhibitors. CONCLUSIONS: DIM enhances activation of AhR and subsequent "glycolysis-lactate-STAT3″ and TIP60 signals-mediated Treg differentiation.


Subject(s)
Colitis, Ulcerative , Colitis , Receptors, Aryl Hydrocarbon , Animals , Mice , Cell Differentiation , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis, Ulcerative/drug therapy , Forkhead Transcription Factors/metabolism , Glucose/metabolism , Glycolysis , Hypoxia/metabolism , Interleukin-10/metabolism , Lactic Acid/metabolism , Lactic Acid/pharmacology , Receptors, Aryl Hydrocarbon/agonists , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Th17 Cells , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Lysine Acetyltransferase 5/drug effects , Lysine Acetyltransferase 5/metabolism
13.
Nature ; 614(7949): 732-741, 2023 02.
Article in English | MEDLINE | ID: mdl-36792830

ABSTRACT

Neuronal activity is crucial for adaptive circuit remodelling but poses an inherent risk to the stability of the genome across the long lifespan of postmitotic neurons1-5. Whether neurons have acquired specialized genome protection mechanisms that enable them to withstand decades of potentially damaging stimuli during periods of heightened activity is unknown. Here we identify an activity-dependent DNA repair mechanism in which a new form of the NuA4-TIP60 chromatin modifier assembles in activated neurons around the inducible, neuronal-specific transcription factor NPAS4. We purify this complex from the brain and demonstrate its functions in eliciting activity-dependent changes to neuronal transcriptomes and circuitry. By characterizing the landscape of activity-induced DNA double-strand breaks in the brain, we show that NPAS4-NuA4 binds to recurrently damaged regulatory elements and recruits additional DNA repair machinery to stimulate their repair. Gene regulatory elements bound by NPAS4-NuA4 are partially protected against age-dependent accumulation of somatic mutations. Impaired NPAS4-NuA4 signalling leads to a cascade of cellular defects, including dysregulated activity-dependent transcriptional responses, loss of control over neuronal inhibition and genome instability, which all culminate to reduce organismal lifespan. In addition, mutations in several components of the NuA4 complex are reported to lead to neurodevelopmental and autism spectrum disorders. Together, these findings identify a neuronal-specific complex that couples neuronal activity directly to genome preservation, the disruption of which may contribute to developmental disorders, neurodegeneration and ageing.


Subject(s)
Brain , DNA Repair , Multiprotein Complexes , Neurons , Synapses , Basic Helix-Loop-Helix Transcription Factors , Brain/metabolism , DNA Breaks, Double-Stranded , Gene Expression Regulation , Lysine Acetyltransferase 5/metabolism , Multiprotein Complexes/metabolism , Neurons/metabolism , Synapses/metabolism , Mutation , Longevity/genetics , Genome , Aging/genetics , Neurodegenerative Diseases
14.
J Cell Biochem ; 124(1): 103-117, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36377816

ABSTRACT

Tat-interactive protein 60 kDa (TIP60, also known as lysine acetyltransferase 5 [KAT5]) is a member of the MYST protein family with histone acetyltransferase activity. Recent studies have reported that TIP60 has multiple functions in many signal transduction mechanisms, especially p53-mediated apoptosis. Although the activation of apoptosis signaling pathways requires the presence of cellular reactive oxygen species (ROS) at a certain level, an imbalance between the production and consumption of ROS in cells results in oxidative stress (OS). In this study, we investigated for the first time how the absence of the Tip60 gene in the liver affects gene expression, enzyme activity, and protein expression of the hepatic antioxidant members localized in the cytoplasm, including superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR), glutathione peroxidase (GPx), and glutathione S-transferase (GST). First, we successfully generated liver-specific Tip60 knockout mice (mutants) using Cre/LoxP recombination. The reduced glutathione level and nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) expression, a marker of OS, increased significantly in the Tip60 mutant liver. Gene expression, activity, and protein expression of the enzymatic antioxidant system, including SOD, CAT, GR, GPx, and GST were investigated in mutants and control groups. Despite a significant correlation between the gene, enzyme activity, and protein content for CAT and GR, this was not true for SOD and GPx. The overall results suggest that TIP60 acts on the hepatic antioxidant system both at the gene and protein levels, but the actual effect of the deletion of Tip60 is observed at the protein level, especially for SOD and GPx.


Subject(s)
Antioxidants , Liver , Lysine Acetyltransferase 5 , Oxidative Stress , Trans-Activators , Animals , Mice , Antioxidants/metabolism , Catalase/genetics , Catalase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Reductase/genetics , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Liver/enzymology
15.
J Mol Cell Biol ; 14(8)2022 12 26.
Article in English | MEDLINE | ID: mdl-36190325

ABSTRACT

Stable transmission of genetic information during cell division requires faithful mitotic spindle assembly and chromosome segregation. In eukaryotic cells, nuclear envelope breakdown (NEBD) is required for proper chromosome segregation. Although a list of mitotic kinases has been implicated in NEBD, how they coordinate their activity to dissolve the nuclear envelope and protein machinery such as nuclear pore complexes was unclear. Here, we identified a regulatory mechanism in which Nup62 is acetylated by TIP60 in human cell division. Nup62 is a novel substrate of TIP60, and the acetylation of Lys432 by TIP60 dissolves nucleoporin Nup62-Nup58-Nup54 complex during entry into mitosis. Importantly, this acetylation-elicited remodeling of nucleoporin complex promotes the distribution of Nup62 to the mitotic spindle, which is indispensable for orchestrating correct spindle orientation. Moreover, suppression of Nup62 perturbs accurate chromosome segregation during mitosis. These results establish a previously uncharacterized regulatory mechanism in which TIP60-elicited nucleoporin dynamics promotes chromosome segregation in mitosis.


Subject(s)
Chromosome Segregation , Lysine Acetyltransferase 5 , Nuclear Pore Complex Proteins , Humans , Acetylation , Mitosis , Nuclear Envelope/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Lysine Acetyltransferase 5/metabolism
16.
Genes Genomics ; 44(11): 1353-1361, 2022 11.
Article in English | MEDLINE | ID: mdl-35951156

ABSTRACT

BACKGROUND: Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is upregulated in colon cancer cells and associated with silencing tumor suppressor genes (TSGs) to promote colon cancer cell proliferation. OBJECTIVE: To investigate epigenetic modification of UHRF1 by TIP60. Whether UHRF1 acetylation by TIP60 can induce cell proliferation in colon cancer cells. METHODS: Acetylation sites of UHRF1 by TIP60 was predicted by ASEB (Acetylation Set Enrichment Based) method and identified by immunoprecipitation assay using anti-pan-acetyl lysine antibody and in vitro acetylation assay. Based on this method, UHRF1 acetylation-deficient mimic 4KR (K644R, K646R, K648R, K650R) mutant was generated to investigate effects of UHRF1 acetylation by TIP60. shRNA system was used to generate stable knockdown cell line of UHRF1. With transient transfection of UHRF1 WT and 4KR, the effects of UHRF1 4KR mutant on Jun dimerization protein 2 (JDP2) gene expression, cell proliferation and cell cycle were investigated by RT-qPCR and FACS analysis in shUHRF1 colon cancer cell line. RESULTS: Downregulation of TIP60-mediated UHRF1 acetylation is correlated with suppressed cell cycle progression. Acetylation-deficient mimic of UHRF1 showed poor cell growth through increased expression of JDP2 gene. CONCLUSIONS: Acetylation of UHRF1 4K residues by TIP60 is important for colon cancer cell growth. Furthermore, upregulated JDP2 expression by acetylation-deficient mutant of UHRF1 might be an important epigenetic target for colon cancer cell proliferation.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Colonic Neoplasms , Lysine Acetyltransferase 5 , Ubiquitin-Protein Ligases , Acetylation , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Proliferation/genetics , Colonic Neoplasms/genetics , DNA Methylation , Humans , Lysine/genetics , Lysine/metabolism , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , RNA, Small Interfering , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism
17.
Nat Commun ; 13(1): 4355, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906200

ABSTRACT

Transcriptional regulation by RNA polymerase II is associated with changes in chromatin structure. Activated and promoter-bound heat shock transcription factor 1 (HSF1) recruits transcriptional co-activators, including histone-modifying enzymes; however, the mechanisms underlying chromatin opening remain unclear. Here, we demonstrate that HSF1 recruits the TRRAP-TIP60 acetyltransferase complex in HSP72 promoter during heat shock in a manner dependent on phosphorylation of HSF1-S419. TRIM33, a bromodomain-containing ubiquitin ligase, is then recruited to the promoter by interactions with HSF1 and a TIP60-mediated acetylation mark, and cooperates with the related factor TRIM24 for mono-ubiquitination of histone H2B on K120. These changes in histone modifications are triggered by phosphorylation of HSF1-S419 via PLK1, and stabilize the HSF1-transcription complex in HSP72 promoter. Furthermore, HSF1-S419 phosphorylation is constitutively enhanced in and promotes proliferation of melanoma cells. Our results provide mechanisms for HSF1 phosphorylation-dependent establishment of an active chromatin status, which is important for tumorigenesis.


Subject(s)
Chromatin , Histones , Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/genetics , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Histones/metabolism , Humans , Lysine Acetyltransferase 5/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Transcription Factors/genetics
18.
EMBO Rep ; 23(9): e54128, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35876654

ABSTRACT

The long noncoding RNA LINC00839 has been shown to be involved in the progression of some cancer types, such as bladder cancer, prostate cancer, breast cancer, and neuroblastoma. However, if LINC00839 has roles in colorectal cancer (CRC), it has not been elucidated so far. Here, we focus on the biological role and involved mechanisms of LINC00839 in CRC. We show that LINC00839 is selectively upregulated in CRC and locates to the nucleus. High expression of LINC00839 is associated with poor outcomes in CRC patients. Functional experiments show that LINC00839 promotes CRC proliferation, invasion, and metastasis in vitro and in vivo. Mechanistically, LINC00839 recruits Ruvb1 to the Tip60 complex and increases its acetylase activity. LINC00839 guides the complex to the NRF1 promoter and promotes acetylation of lysines 5 and 8 of histones H4, thereby upregulating the expression of NRF1. Subsequently, NRF1 activates mitochondrial metabolism and biogenesis, thereby promoting CRC progression. In summary, our study reports on a mechanism by which LINC00839 positively regulates NRF1, thus promoting mitochondrial metabolism and biogenesis, as well as CRC progression.


Subject(s)
Colorectal Neoplasms , RNA, Long Noncoding , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Lysine Acetyltransferase 5/metabolism , Male , Nuclear Respiratory Factor 1/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
19.
Cell Death Dis ; 13(7): 627, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35853868

ABSTRACT

Histone acetylation is essential for initiating and maintaining a permissive chromatin conformation and gene transcription. Dysregulation of histone acetylation can contribute to tumorigenesis and metastasis. Using inducible cre-recombinase and CRISPR/Cas9-mediated deletion, we investigated the roles of the histone lysine acetyltransferase TIP60 (KAT5/HTATIP) in human cells, mouse cells, and mouse embryos. We found that loss of TIP60 caused complete cell growth arrest. In the absence of TIP60, chromosomes failed to align in a metaphase plate during mitosis. In some TIP60 deleted cells, endoreplication occurred instead. In contrast, cell survival was not affected. Remarkably, the cell growth arrest caused by loss of TIP60 was independent of the tumor suppressors p53, INK4A and ARF. TIP60 was found to be essential for the acetylation of H2AZ, specifically at lysine 7. The mRNA levels of 6236 human and 8238 mouse genes, including many metabolism genes, were dependent on TIP60. Among the top 50 differentially expressed genes, over 90% were downregulated in cells lacking TIP60, supporting a role for TIP60 as a key co-activator of transcription. We propose a primary role of TIP60 in H2AZ lysine 7 acetylation and transcriptional activation, and that this fundamental role is essential for cell proliferation. Growth arrest independent of major tumor suppressors suggests TIP60 as a potential anti-cancer drug target.


Subject(s)
Histones , Lysine Acetyltransferase 5 , Lysine , Tumor Suppressor Protein p53 , Acetylation , Animals , Cell Cycle Checkpoints/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Lysine Acetyltransferase 5/deficiency , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Cell Rep ; 39(11): 110947, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705031

ABSTRACT

A recurrent chromosomal translocation found in acute myeloid leukemia leads to an in-frame fusion of the transcription repressor ZMYND11 to MBTD1, a subunit of the NuA4/TIP60 histone acetyltransferase complex. To understand the abnormal molecular events that ZMYND11-MBTD1 expression can create, we perform a biochemical and functional characterization comparison to each individual fusion partner. ZMYND11-MBTD1 is stably incorporated into the endogenous NuA4/TIP60 complex, leading to its mislocalization on the body of genes normally bound by ZMYND11. This can be correlated to increased chromatin acetylation and altered gene transcription, most notably on the MYC oncogene, and alternative splicing. Importantly, ZMYND11-MBTD1 expression favors Myc-driven pluripotency during embryonic stem cell differentiation and self-renewal of hematopoietic stem/progenitor cells. Altogether, these results indicate that the ZMYND11-MBTD1 fusion functions primarily by mistargeting the NuA4/TIP60 complex to the body of genes, altering normal transcription of specific genes, likely driving oncogenesis in part through the Myc regulatory network.


Subject(s)
Chromatin , Histone Acetyltransferases , Oncogene Proteins, Fusion , Open Reading Frames , Acetylation , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Open Reading Frames/genetics , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL