Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.471
1.
Cell Commun Signal ; 22(1): 306, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38831454

BACKGROUND: Dysregulation in histone acetylation, a significant epigenetic alteration closely associated with major pathologies including cancer, promotes tumorigenesis, inactivating tumor-suppressor genes and activating oncogenic pathways. AMP-activated protein kinase (AMPK) is a cellular energy sensor that regulates a multitude of biological processes. Although a number of studies have identified the mechanisms by which AMPK regulates cancer growth, the underlying epigenetic mechanisms remain unknown. METHODS: The impact of metformin, an AMPK activator, on cervical cancer was evaluated through assessments of cell viability, tumor xenograft model, pan-acetylation analysis, and the role of the AMPK-PCAF-H3K9ac signaling pathway. Using label-free quantitative acetylproteomics and chromatin immunoprecipitation-sequencing (ChIP) technology, the activation of AMPK-induced H3K9 acetylation was further investigated. RESULTS: In this study, we found that metformin, acting as an AMPK agonist, activates AMPK, thereby inhibiting the proliferation of cervical cancer both in vitro and in vivo. Mechanistically, AMPK activation induces H3K9 acetylation at epigenetic level, leading to chromatin remodeling in cervical cancer. This also enhances the binding of H3K9ac to the promoter regions of multiple tumor suppressor genes, thereby promoting their transcriptional activation. Furthermore, the absence of PCAF renders AMPK activation incapable of inducing H3K9 acetylation. CONCLUSIONS: In conclusion, our findings demonstrate that AMPK mediates the inhibition of cervical cancer growth through PCAF-dependent H3K9 acetylation. This discovery not only facilitates the clinical application of metformin but also underscores the essential role of PCAF in AMPK activation-induced H3K9 hyperacetylation.


AMP-Activated Protein Kinases , Cell Proliferation , Histones , Metformin , Uterine Cervical Neoplasms , p300-CBP Transcription Factors , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/genetics , Humans , Acetylation/drug effects , Female , Histones/metabolism , AMP-Activated Protein Kinases/metabolism , Cell Proliferation/drug effects , Animals , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/genetics , Metformin/pharmacology , Mice , Mice, Nude , Cell Line, Tumor , Enzyme Activation/drug effects
2.
Molecules ; 29(10)2024 May 20.
Article En | MEDLINE | ID: mdl-38792269

Quercetin, a flavonoid polyphenol found in many plants, has garnered significant attention due to its potential cancer chemoprevention. Our previous studies have shown that acetyl modification of the hydroxyl group of quercetin altered its antitumor effects in HepG2 cells. However, the antitumor effect in other cancer cells with different gene mutants remains unknown. In this study, we investigated the antitumor effect of quercetin and its methylated derivative 3,3',4',7-O-tetramethylquercetin (4Me-Q) and acetylated derivative 3,3',4',7-O-tetraacetylquercetin (4Ac-Q) on two human breast cancer cells, MCF-7 (wt-p53, caspase-3-ve) and MDA-MB-231 (mt-p53, caspase-3+ve). The results demonstrated that 4Ac-Q exhibited significant cell proliferation inhibition and apoptosis induction in both MCF-7 and MDA-MB-231 cells. Conversely, methylation of quercetin was found to lose the activity. The human apoptosis antibody array revealed that 4Ac-Q might induce apoptosis in MCF-7 cells via a p53-dependent pathway, while in MDA-MB-231 cells, it was induced via a caspase-3-dependent pathway. Furthermore, an evaluation using a superoxide inhibitor, MnTBAP, revealed 4Ac-Q-induced apoptosis in MCF-7 cells in a superoxide-independent manner. These findings provide valuable insights into the potential of acetylated quercetin as a new approach in cancer chemoprevention and offer new avenues for health product development.


Apoptosis , Breast Neoplasms , Cell Proliferation , Quercetin , Humans , Quercetin/pharmacology , Quercetin/analogs & derivatives , Quercetin/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Acetylation/drug effects , Apoptosis/drug effects , Methylation , Female , Cell Proliferation/drug effects , MCF-7 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Tumor Suppressor Protein p53/metabolism , Caspase 3/metabolism
3.
Biochem Biophys Res Commun ; 717: 150061, 2024 Jul 12.
Article En | MEDLINE | ID: mdl-38718570

Epithelial mesenchymal transition (EMT) is a critical process implicated in the pathogenesis of retinal fibrosis and the exacerbation of diabetic retinopathy (DR) within retinal pigment epithelium (RPE) cells. Apigenin (AP), a potential dietary supplement for managing diabetes and its associated complications, has demonstrated inhibitory effects on EMT in various diseases. However, the specific impact and underlying mechanisms of AP on EMT in RPE cells remain poorly understood. In this study, we have successfully validated the inhibitory effects of AP on high glucose-induced EMT in ARPE-19 cells and diabetic db/db mice. Notably, our findings have identified CBP/p300 as a potential therapeutic target for EMT in RPE cells and have further substantiated that AP effectively downregulates the expression of EMT-related genes by attenuating the activity of CBP/p300, consequently reducing histone acetylation alterations within the promoter region of these genes. Taken together, our results provide novel evidence supporting the inhibitory effect of AP on EMT in RPE cells, and highlight the potential of specifically targeting CBP/p300 as a strategy for inhibiting retinal fibrosis in the context of DR.


Apigenin , Epithelial-Mesenchymal Transition , Glucose , Histones , Retinal Pigment Epithelium , Epithelial-Mesenchymal Transition/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Animals , Apigenin/pharmacology , Acetylation/drug effects , Humans , Glucose/metabolism , Glucose/toxicity , Histones/metabolism , Cell Line , Mice , p300-CBP Transcription Factors/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , Mice, Inbred C57BL , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Diabetic Retinopathy/drug therapy , E1A-Associated p300 Protein/metabolism , Male , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , CREB-Binding Protein/metabolism , CREB-Binding Protein/genetics
4.
PLoS One ; 19(5): e0298032, 2024.
Article En | MEDLINE | ID: mdl-38820384

The FA/BRCA pathway safeguards DNA replication by repairing interstrand crosslinks (ICL) and maintaining replication fork stability. Chromatin structure, which is in part regulated by histones posttranslational modifications (PTMs), has a role in maintaining genomic integrity through stabilization of the DNA replication fork and promotion of DNA repair. An appropriate balance of PTMs, especially acetylation of histones H4 in nascent chromatin, is required to preserve a stable DNA replication fork. To evaluate the acetylation status of histone H4 at the replication fork of FANCA deficient cells, we compared histone acetylation status at the DNA replication fork of isogenic FANCA deficient and FANCA proficient cell lines by using accelerated native immunoprecipitation of nascent DNA (aniPOND) and in situ protein interactions in the replication fork (SIRF) assays. We found basal hypoacetylation of multiple residues of histone H4 in FA replication forks, together with increased levels of Histone Deacetylase 1 (HDAC1). Interestingly, high-dose short-term treatment with mitomycin C (MMC) had no effect over H4 acetylation abundance at the replication fork. However, chemical inhibition of histone deacetylases (HDAC) with Suberoylanilide hydroxamic acid (SAHA) induced acetylation of the FANCA deficient DNA replication forks to levels comparable to their isogenic control counterparts. This forced permanence of acetylation impacted FA cells homeostasis by inducing DNA damage and promoting G2 cell cycle arrest. Altogether, this caused reduced RAD51 foci formation and increased markers of replication stress, including phospho-RPA-S33. Hypoacetylation of the FANCA deficient replication fork, is part of the cellular phenotype, the perturbation of this feature by agents that prevent deacetylation, such as SAHA, have a deleterious effect over the delicate equilibrium they have reached to perdure despite a defective FA/BRCA pathway.


DNA Damage , DNA Replication , Fanconi Anemia Complementation Group A Protein , Histones , Histones/metabolism , Humans , DNA Replication/drug effects , Acetylation/drug effects , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group A Protein/genetics , Mitomycin/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Vorinostat/pharmacology , Hydroxamic Acids/pharmacology
5.
Biol Res ; 57(1): 33, 2024 May 27.
Article En | MEDLINE | ID: mdl-38802872

BACKGROUND: There is a need for novel treatments for neuroblastoma, despite the emergence of new biological and immune treatments, since refractory pediatric neuroblastoma is still a medical challenge. Phyto cannabinoids and their hemisynthetic derivatives have shown evidence supporting their anticancer potential. The aim of this research was to examine Phytocannabinoids or hemisynthetic cannabinoids, which reduce the SHSY-5Y, neuroblastoma cell line's viability. METHODS: Hexane and acetyl acetate extracts were produced starting with Cannabis sativa L. as raw material, then, 9-tetrahidrocannabinol, its acid counterpart and CBN were isolated. In addition, acetylated derivatives of THC and CBN were synthesized. The identification and purity of the chemicals was determined by High Performance Liquid Chromatography and 1H y 13C Magnetic Nuclear Resonance. Then, the capacity to affect the viability of SHSY-5Y, a neuroblastoma cell line, was examined using the resazurin method. Finally, to gain insight into the mechanism of action of the extracts, phytocannabinoids and acetylated derivatives on the examined cells, a caspase 3/7 determination was performed on cells exposed to these compounds. RESULTS: The structure and purity of the isolated compounds was demonstrated. The extracts, the phytocannabinoids and their acetylated counterparts inhibited the viability of the SHSY 5Y cells, being CBN the most potent of all the tested molecules with an inhibitory concentration of 50 percent of 9.5 µM. CONCLUSION: Each of the evaluated molecules exhibited the capacity to activate caspases 3/7, indicating that at least in part, the cytotoxicity of the tested phytocannabinoids and their hemi-synthetic derivatives is mediated by apoptosis.


Cannabinoids , Cannabis , Caspase 3 , Cell Survival , Neuroblastoma , Plant Extracts , Humans , Cannabis/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Line, Tumor , Neuroblastoma/drug therapy , Cell Survival/drug effects , Caspase 3/metabolism , Caspase 3/drug effects , Cannabinoids/pharmacology , Cannabinoids/chemistry , Caspase 7/metabolism , Apoptosis/drug effects , Acetylation/drug effects , Chromatography, High Pressure Liquid
6.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 40-47, 2024 May 27.
Article En | MEDLINE | ID: mdl-38814236

Periodontal ligament stem cells (PDLSCs) show plasticity towards the adipogenic lineage; however, little has been done on the participation of epigenetic mechanisms. Histone acetylation is a dynamic process, though balanced by histone acetyltransferases (HATs) and histone deacetylases (HDACs) activities. This process can be halted by HDACs inhibitors, such as trichostatin A (TSA) and valproic acid (VPA). This study aimed to determine the role of HDACs class I in adipogenic differentiation of PDL cells. PDLSCs were treated with TSA at concentrations of 100, 200, and 250 nM, or VPA at 1, 4 and 8 mM. Cell viability was assessed using MTT assays. Gene expression of pluripotency markers (NANOG, OCT4, SOX2), HAT genes (p300, GCN5), and HDACs genes (HDAC1-3) was analyzed by RT-qPCR. Adipogenic differentiation was evaluated via oil red O staining, and acetylation of histone H3 lysine 9 (H3K9ac) was examined by Western blot. VPA treatment resulted in a 60% reduction in cell proliferation, compared to a 50% when using TSA. Cell viability was not affected by either inhibitor. Furthermore, both TSA and VPA induced adipogenic differentiation, through an increase in the deposition of lipid droplets and in GCN5 and p300 expression were observed. Western blot analysis showed that TSA increased H3K9ac levels on adipogenic differentiation of PDLSCs. These findings highlight the potential of HDAC inhibitors as a tool for modulating H3K9 acetylation status and thus influencing adipogenic differentiation of PDLCs.


Adipogenesis , Cell Differentiation , Cell Survival , Histone Deacetylase Inhibitors , Periodontal Ligament , Valproic Acid , Humans , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Histone Deacetylase Inhibitors/pharmacology , Adipogenesis/drug effects , Adipogenesis/genetics , Valproic Acid/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Acetylation/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Cells, Cultured , Histones/metabolism , Cell Proliferation/drug effects , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism
7.
Chem Biol Interact ; 396: 111035, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38703807

Early life stress (ELS) can cause long-term changes by epigenetic factors, especially histone acetylation modification, playing a crucial role, affect normal cognition, mood, and behavior, and increase susceptibility to post-traumatic stress disorder (PTSD) in adulthood. It has been found that paeoniflorin (PF) can cross the blood-brain barrier to exert anti-PTSD effects on adult PTSD rats. However, whether PF can alleviate the harmful effects caused by ELS in adulthood has not yet been reported. Therefore, to explore the relationship between ELS and PTSD susceptibility in adulthood and its mechanism, in this study, SPS was used as a stressor of ELS, and the mathematical tool Z-normalization was employed as an evaluation criterion of behavioral resilience susceptibility. To investigate the regulatory mechanism of PF on histone acetylation in the hippocampus and amygdala of ELS rats in adulthood, using changes in HATs/HDACs as the entry point, meanwhile, the epigenetic marks (H3K9 and H4K12) in the key brain regions of ELS (hippocampus and amygdala) were evaluated, and the effects of PF on behavioral representation and PTSD susceptibility were observed. This study found that ELS lead to a series of PTSD-like behaviors in adulthood and caused imbalance of HATs/HDACs ratio in the hippocampus and amygdala, which confirms that ELS is an important risk factor for the development of PTSD in adulthood. In addition, paeoniflorin may improve ELS-induced PTSD-like behaviors and reduce the susceptibility of ELS rats to develop PTSD in adulthood by modulating the HATs/HDACs ratio in the hippocampus and amygdala.


Amygdala , Glucosides , Hippocampus , Histones , Monoterpenes , Stress Disorders, Post-Traumatic , Stress, Psychological , Animals , Glucosides/pharmacology , Glucosides/therapeutic use , Monoterpenes/pharmacology , Monoterpenes/therapeutic use , Hippocampus/metabolism , Hippocampus/drug effects , Acetylation/drug effects , Amygdala/metabolism , Amygdala/drug effects , Histones/metabolism , Rats , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Male , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Rats, Sprague-Dawley , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism
8.
BMC Oral Health ; 24(1): 510, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38689229

BACKGROUND: Periodontitis is a chronic osteolytic inflammatory disease, where anti-inflammatory intervention is critical for restricting periodontal damage and regenerating alveolar bone. Ropinirole, a dopamine D2 receptor agonist, has previously shown therapeutic potential for periodontitis but the underlying mechanism is still unclear. METHODS: Human gingival fibroblasts (HGFs) treated with LPS were considered to mimic periodontitis in vitro. The dosage of Ropinirole was selected through the cell viability of HGFs evaluation. The protective effects of Ropinirole on HGFs were evaluated by detecting cell viability, cell apoptosis, and pro-inflammatory factor levels. The molecular docking between NAT10 and Ropinirole was performed. The interaction relationship between NAT10 and KLF6 was verified by ac4C Acetylated RNA Immunoprecipitation followed by qPCR (acRIP-qPCR) and dual-luciferase reporter assay. RESULTS: Ropinirole alleviates LPS-induced damage of HGFs by promoting cell viability, inhibiting cell apoptosis and the levels of IL-1ß, IL-18, and TNF-α. Overexpression of NAT10 weakens the effects of Ropinirole on protecting HGFs. Meanwhile, NAT10-mediated ac4C RNA acetylation promotes KLF6 mRNA stability. Upregulation of KLF6 reversed the effects of NAT10 inhibition on HGFs. CONCLUSIONS: Taken together, Ropinirole protected HGFs through inhibiting the NAT10 ac4C RNA acetylation to decrease the KLF6 mRNA stability from LPS injury. The discovery of this pharmacological and molecular mechanism of Ropinirole further strengthens its therapeutic potential for periodontitis.


Fibroblasts , Indoles , Kruppel-Like Factor 6 , N-Terminal Acetyltransferases , Periodontitis , Humans , Acetylation/drug effects , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Gingiva/drug effects , Gingiva/metabolism , Indoles/pharmacology , Indoles/therapeutic use , Kruppel-Like Factor 6/metabolism , Lipopolysaccharides , Molecular Docking Simulation , Periodontitis/drug therapy , Periodontitis/metabolism , N-Terminal Acetyltransferases/antagonists & inhibitors
9.
Int J Biochem Cell Biol ; 171: 106583, 2024 Jun.
Article En | MEDLINE | ID: mdl-38657899

Protein crotonylation plays a role in regulating cellular metabolism, gene expression, and other biological processes. NDUFA9 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 9) is closely associated with the activity and function of mitochondrial respiratory chain complex I. Mitochondrial function and respiratory chain are closely related to browning of white adipocytes, it's speculated that NDUFA9 and its crotonylation are associated with browning of white adipocytes. Firstly, the effect of NDUFA9 on white adipose tissue was verified in white fat browning model mice, and it was found that NDUFA9 promoted mitochondrial respiration, thermogenesis, and browning of white adipose tissue. Secondly, in cellular studies, it was discovered that NDUFA9 facilitated browning of white adipocytes by enhancing mitochondrial function, mitochondrial complex I activity, ATP synthesis, and mitochondrial respiration. Again, the level of NDUFA9 crotonylation was increased by treating cells with vorinostat (SAHA)+sodium crotonate (NaCr) and overexpressing NDUFA9, it was found that NDUFA9 crotonylation promoted browning of white adipocytes. Meanwhile, the acetylation level of NDUFA9 was increased by treating cells with SAHA+sodium acetate (NaAc) and overexpressing NDUFA9, the assay revealed that NDUFA9 acetylation inhibited white adipocytes browning. Finally, combined with the competitive relationship between acetylation and crotonylation, it was also demonstrated that NDUFA9 crotonylation promoted browning of white adipocytes. Above results indicate that NDUFA9 and its crotonylation modification promote mitochondrial function, which in turn promotes browning of white adipocytes. This study establishes a theoretical foundation for the management and intervention of obesity, which is crucial in addressing obesity and related medical conditions in the future.


Adipocytes, White , Mitochondria , Animals , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Adipocytes, White/metabolism , Adipocytes, White/drug effects , Adipocytes, White/cytology , Male , Mice, Inbred C57BL , Thermogenesis/drug effects , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , 3T3-L1 Cells , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , Acetylation/drug effects
10.
Cancer Res Commun ; 4(5): 1307-1320, 2024 May 20.
Article En | MEDLINE | ID: mdl-38669046

Anticancer drug-tolerant persister (DTP) cells at an early phase of chemotherapy reshape refractory tumors. Aldehyde dehydrogenase 1 family member A3 (ALDH1A3) is commonly upregulated by various anticancer drugs in gastric cancer patient-derived cells (PDC) and promotes tumor growth. However, the mechanism underlying the generation of ALDH1A3-positive DTP cells remains elusive. Here, we investigated the mechanism of ALDH1A3 expression and a combination therapy targeting gastric cancer DTP cells. We found that gastric cancer tissues treated with neoadjuvant chemotherapy showed high ALDH1A3 expression. Chromatin immunoprecipitation (ChIP)-PCR and ChIP sequencing analyses revealed that histone H3 lysine 27 acetylation was enriched in the ALDH1A3 promoter in 5-fluorouracil (5-FU)-tolerant persister PDCs. By chemical library screening, we found that the bromodomain and extraterminal (BET) inhibitors OTX015/birabresib and I-BET-762/molibresib suppressed DTP-related ALDH1A3 expression and preferentially inhibited DTP cell growth. In DTP cells, BRD4, but not BRD2/3, was recruited to the ALDH1A3 promoter and BRD4 knockdown decreased drug-induced ALDH1A3 upregulation. Combination therapy with 5-FU and OTX015 significantly suppressed in vivo tumor growth. These observations suggest that BET inhibitors are efficient DTP cell-targeting agents for gastric cancer treatment. SIGNIFICANCE: Drug resistance hampers the cure of patients with cancer. To prevent stable drug resistance, DTP cancer cells are rational therapeutic targets that emerge during the early phase of chemotherapy. This study proposes that the epigenetic regulation by BET inhibitors may be a rational therapeutic strategy to eliminate DTP cells.


Drug Resistance, Neoplasm , Fluorouracil , Histones , Stomach Neoplasms , Transcription Factors , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Humans , Animals , Histones/metabolism , Mice , Acetylation/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Xenograft Model Antitumor Assays , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Mice, Nude , Aldehyde Oxidoreductases/metabolism , Aldehyde Oxidoreductases/genetics , Cell Proliferation/drug effects , Male , Female , Antineoplastic Agents/pharmacology , Promoter Regions, Genetic/drug effects , Mice, Inbred BALB C , Bromodomain Containing Proteins
11.
Molecules ; 29(8)2024 Apr 15.
Article En | MEDLINE | ID: mdl-38675608

Increased oxidative stress is one of the critical pathologies inducing age-related macular degeneration (AMD), characterized by retinal pigment epithelial (RPE) cell damage and death. The unbalanced acetylation and deacetylation of histones have been implicated in AMD pathogenesis or hydrogen peroxide (H2O2)-induced cell damage. Therefore, strategies aimed at controlling the balance between acetylation and deacetylation may effectively protect RPE cells from oxidative damage. Artemisinin is an antimalarial lactone drug derived from Artemisia annua, with antioxidant activity known to modulate histone acetylation in the brain, but its effect on the retina is unknown. In this study, we aimed to investigate whether Artemisinin exerts a cytoprotective effect on oxidative stress-induced apoptosis in RPE cells by regulating histone acetylation. We hypothesized that Artemisinin confers cytoprotection toward H2O2-induced apoptosis in RPE cells through this mechanism. In the present study, we found that Artemisinin at a sub-clinic dosage of 20 µM inhibited the H2O2-induced cell viability decrease and B-cell lymphoma 2 (Bcl-2) protein level decrease and attenuated the H2O2-induced decrease in the histone H4 lysine (Lys) 8 acetylation [Acetyl-H4 (Lys 8)] level in the retinal RPE cell line D407. As expected, histone deacetylase inhibitor Trichostatin A at the concentration of 250 nM increased the Acetyl-H4 (Lys 8) level in D407 cells and attenuated the H2O2-induced cell viability decrease and apoptosis. Similar findings were obtained using adult RPE (ARPE)19 cells, another human RPE cell line, and primary human RPE cell cultures. In conclusion, these results confirmed our hypothesis and indicated that Artemisinin attenuated H2O2-induced apoptosis in apparent correlation with the increase in the Acetyl-H4 (Lys 8) level, which is associated with gene transcription and cell survival. By modulating histone acetylation, Artemisinin may restore the balance between acetylation and deacetylation and enhance the resistance and survival of RPE cells under oxidative stress. Our study provides novel mechanistic insights into the effect of Artemisinin on histone acetylation and apoptosis in RPE cells and supports the potential application of Artemisinin in the prevention and/or treatment of AMD.


Apoptosis , Artemisinins , Cell Survival , Histones , Hydrogen Peroxide , Lysine , Oxidative Stress , Retinal Pigment Epithelium , Humans , Histones/metabolism , Apoptosis/drug effects , Acetylation/drug effects , Hydrogen Peroxide/pharmacology , Artemisinins/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/cytology , Lysine/metabolism , Cell Survival/drug effects , Oxidative Stress/drug effects , Cell Line , Cytoprotection/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism
12.
Exp Cell Res ; 438(2): 114050, 2024 May 15.
Article En | MEDLINE | ID: mdl-38663474

Myocardial infarction (MI) is a potentially fatal disease that causes a significant number of deaths worldwide. The strategy of increasing fatty acid oxidation in myocytes is considered a therapeutic avenue to accelerate metabolism to meet energy demands. We conducted the study aiming to investigate the effect of KN-93, which induces histone deacetylase (HDAC)4 shuttling to the nucleus, on fatty acid oxidation and the expression of related genes. A mouse model of myocardial infarction was induced by isoprenaline administration. Heart damage was assessed by the detection of cardiac injury markers. The level of fatty acid oxidation level was evaluated by testing the expression of related genes. Both immunofluorescence and immunoblotting in the cytosol or nucleus were utilized to observe the distribution of HDAC4. The interaction between HDAC4 and specificity protein (SP)1 was confirmed by co-immunoprecipitation. The acetylation level of SP1 was tested after KN-93 treatment and HDAC4 inhibitor. Oxygen consumption rate and immunoblotting experiments were used to determine whether the effect of KN-93 on increasing fatty acid oxidation is through HDAC4 and SP1. Administration of KN-93 significantly reduced cardiac injury in myocardial infarction and promoted fatty acid oxidation both in vitro and in vivo. KN-93 was shown to mediate nuclear translocation of HDAC4. HDAC4 was found to interact with SP1 and reduce SP1 acetylation. HDAC4 or SP1 inhibitors attenuated the effect of KN-93 on fatty acid oxidation. In conclusion, KN-93 promotes HDAC4 translocation to the nucleus, thereby potentially enhancing fatty acid oxidation by SP1.


Cell Nucleus , Fatty Acids , Histone Deacetylases , Myocardial Infarction , Oxidation-Reduction , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Fatty Acids/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice , Oxidation-Reduction/drug effects , Cell Nucleus/metabolism , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Mice, Inbred C57BL , Humans , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Acetylation/drug effects , Active Transport, Cell Nucleus/drug effects
13.
Adv Sci (Weinh) ; 11(19): e2306378, 2024 May.
Article En | MEDLINE | ID: mdl-38482749

Aspirin, also named acetylsalicylate, can directly acetylate the side-chain of lysine in protein, which leads to the possibility of unexplained drug effects. Here, the study used isotopic-labeling aspirin-d3 with mass spectrometry analysis to discover that aspirin directly acetylates 10 HDACs proteins, including SIRT1, the most studied NAD+-dependent deacetylase. SIRT1 is also acetylated by aspirin in vitro. It is also identified that aspirin directly acetylates lysine 408 of SIRT1, which abolishes SIRT1 deacetylation activity by impairing the substrates binding affinity. Interestingly, the lysine 408 of SIRT1 can be acetylated by CBP acetyltransferase in cells without aspirin supplement. Aspirin can inhibit SIRT1 to increase the levels of acetylated p53 and promote p53-dependent apoptosis. Moreover, the knock-in mice of the acetylation-mimic mutant of SIRT1 show the decreased production of pro-inflammatory cytokines and maintain intestinal immune homeostasis. The study indicates the importance of the acetylated internal functional site of SIRT1 in maintaining intestinal immune homeostasis.


Aspirin , Homeostasis , Sirtuin 1 , Sirtuin 1/metabolism , Sirtuin 1/genetics , Animals , Aspirin/pharmacology , Acetylation/drug effects , Mice , Homeostasis/drug effects , Humans , Intestines/drug effects , Mice, Inbred C57BL
14.
FEBS J ; 291(9): 1909-1924, 2024 May.
Article En | MEDLINE | ID: mdl-38380720

Breast cancer is often treated with chemotherapy. However, the development of chemoresistance results in treatment failure. Long non-coding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has been shown to contribute to chemoresistance in breast cancer cells. In studying the transcriptional regulation of NEAT1 using multi-omics approaches, we showed that NEAT1 is up-regulated by 5-fluorouracil in breast cancer cells with wild-type cellular tumor antigen p53 but not in mutant-p53-expressing breast cancer cells. The regulation of NEAT1 involves mediator complex subunit 12 (MED12)-mediated repression of histone acetylation marks at the promoter region of NEAT1. Knockdown of MED12 but not coactivator-associated arginine methyltransferase 1 (CARM1) induced histone acetylation at the NEAT1 promoter, leading to elevated NEAT1 mRNAs, resulting in a chemoresistant phenotype. The MED12-dependent regulation of NEAT1 differs between wild-type and mutant p53-expressing cells. MED12 depletion led to increased expression of NEAT1 in a wild-type p53 cell line, but decreased expression in a mutant p53 cell line. Chemoresistance caused by MED12 depletion can be partially rescued by NEAT1 knockdown in p53 wild-type cells. Collectively, our study reveals a novel mechanism of chemoresistance dependent on MED12 transcriptional regulation of NEAT1 in p53 wild-type breast cancer cells.


Breast Neoplasms , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Mediator Complex , RNA, Long Noncoding , Tumor Suppressor Protein p53 , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Drug Resistance, Neoplasm/genetics , Female , Mediator Complex/genetics , Mediator Complex/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Fluorouracil/pharmacology , Cell Line, Tumor , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Acetylation/drug effects , Histones/metabolism , Histones/genetics
15.
Circ Heart Fail ; 15(5): e008547, 2022 05.
Article En | MEDLINE | ID: mdl-35418250

BACKGROUND: High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS: Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS: Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS: Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.


Cardiomyopathy, Dilated , Doxorubicin , Oxidative Stress , Sirtuin 3 , Acetylation/drug effects , Animals , Cardiomyopathy, Dilated/chemically induced , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/prevention & control , Doxorubicin/adverse effects , Doxorubicin/pharmacology , Heart Failure/metabolism , Humans , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Rats , Sirtuin 3/genetics , Sirtuin 3/metabolism
16.
Oxid Med Cell Longev ; 2022: 4592170, 2022.
Article En | MEDLINE | ID: mdl-35251473

Lysine ß-hydroxybutyrylation (Kbhb) is a newly identified protein posttranslational modification (PTM) derived from ß-hydroxybutyrate (BHB), a product of ketone body metabolism in liver. BHB could serve as an energy source and play a role in the suppression of oxidative stress. The plasma concentration of BHB could increase up to 20 mM during starvation and in pathological conditions. Despite the progress, how the cells derived from extrahepatic tissues respond to elevated environmental BHB remains largely unknown. Given that BHB can significantly drive Kbhb, we characterized the BHB-induced lysine ß-hydroxybutyrylome and acetylome by quantitative proteomics. A total of 840 unique Kbhb sites on 429 proteins were identified, with 42 sites on 39 proteins increased by more than 50% in response to BHB. The results showed that the upregulated Kbhb induced by BHB was involved in aminoacyl-tRNA biosynthesis, 2-oxocarboxylic acid metabolism, citrate cycle, glycolysis/gluconeogenesis, and pyruvate metabolism pathways. Moreover, some BHB-induced Kbhb substrates were significantly involved in diseases such as cancer. Taken together, we investigate the dynamics of lysine ß-hydroxybutyrylome and acetylome induced by environmental BHB, which reveals the roles of Kbhb in regulating various biological processes and expands the biological functions of BHB.


3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Lysine/metabolism , Protein Processing, Post-Translational/drug effects , Proteome/drug effects , Proteomics/methods , Signal Transduction/drug effects , Acetylation/drug effects , Animals , Cells, Cultured , Mice , Protein Binding/drug effects , Proteome/metabolism , Up-Regulation/drug effects
17.
Clin Transl Med ; 12(2): e699, 2022 02.
Article En | MEDLINE | ID: mdl-35184403

BACKGROUND: Persistent hyperglycemia decreases the sensitivity of insulin-sensitive organs to insulin, owing to which cells fail to take up and utilize glucose, which exacerbates the progression of type 2 diabetes mellitus (T2DM). lncRNAs' abnormal expression is reported to be associated with the progression of diabetes and plays a significant role in glucose metabolism. Herein, we study the detailed mechanism underlying the functions of lncRNA EPB41L4A-AS1in T2DM. METHODS: Data from GEO datasets were used to analyze the expression of EPB41L4A-AS1 between insulin resistance or type 2 diabetes patients and the healthy people. Gene expression was evaluated by qRT-PCR and western blotting. Glucose uptake was measured by Glucose Uptake Fluorometric Assay Kit. Glucose tolerance of mice was detected by Intraperitoneal glucose tolerance tests. Cell viability was assessed by CCK-8 assay. The interaction between EPB41L4A-AS1 and GCN5 was explored by RNA immunoprecipitation, RNA pull-down and RNA-FISH combined immunofluorescence. Oxygen consumption rate was tested by Seahorse XF Mito Stress Test. RESULTS: EPB41L4A-AS1 was abnormally increased in the liver of patients with T2DM and upregulated in the muscle cells of patients with insulin resistance and in T2DM cell models. The upregulation was associated with increased TP53 expression and reduced glucose uptake. Mechanistically, through interaction with GCN5, EPB41L4A-AS1 regulated histone H3K27 crotonylation in the GLUT4 promoter region and nonhistone PGC1ß acetylation, which inhibited GLUT4 transcription and suppressed glucose uptake by muscle cells. In contrast, EPB41L4A-AS1 binding to GCN5 enhanced H3K27 and H3K14 acetylation in the TXNIP promoter region, which activated transcription by promoting the recruitment of the transcriptional activator MLXIP. This enhanced GLUT4/2 endocytosis and further suppressed glucose uptake. CONCLUSION: Our study first showed that the EPB41L4A-AS1/GCN5 complex repressed glucose uptake via targeting GLUT4/2 and TXNIP by regulating histone and nonhistone acetylation or crotonylation. Since a weaker glucose uptake ability is one of the major clinical features of T2DM, the inhibition of EPB41L4A-AS1 expression seems to be a potentially effective strategy for drug development in T2DM treatment.


Glucose Intolerance/etiology , RNA, Long Noncoding/pharmacology , p300-CBP Transcription Factors/pharmacology , Acetylation/drug effects , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression/genetics , Glucose Intolerance/physiopathology , Histones/drug effects , Histones/genetics , Histones/metabolism , Humans , RNA, Long Noncoding/therapeutic use , p300-CBP Transcription Factors/metabolism
18.
Cancer Control ; 29: 10732748221074051, 2022.
Article En | MEDLINE | ID: mdl-35067084

INTRODUCTION: The prototype DNA hypomethylating agents 5-azacytidine (5AC) and decitabine (DAC) are currently FDA-approved for treatment of blood and bone marrow disorders like myelodysplastic syndrome. 5AC and DAC are considered similar drugs and were shown to induce histone modifications that modulate gene expression. The aim of this study is to compare the effect of both drugs on histone acetylation and methylation at multiple histone amino acids residues. METHODS: Mass spectrometry was used to compare the effect of both drugs on 95 different histone posttranslational modifications (PTMs) in leukemia cells. ChIP-Seq analysis was used to compare the impact of both drugs on the genome-wide acetylation of the H3K9 mark using primary leukemia cells from six de-identified AML patients. RESULTS: Both DAC and 5AC induced histone PTMs in different histone isoforms like H1.4, H2A, H3, H3.1, and H4. Changes in both histone methylation and acetylation were observed with both drugs; however, there were distinct differences in the histone modifications induced by the two drugs. Since both drugs were shown to increase the activity of the HDAC SIRT6 previously, we tested the effect of 5AC on the acetylation of H3K9, the physiological substrate SIRT6, using ChIP-Seq analysis and compared it to the previously published DAC-induced changes. Significant H3K9 acetylation changes (P< .05) were detected at 925 genes after 5AC treatment vs only 182 genes after DAC treatment. Nevertheless, the gene set modified by 5AC was different from that modified by DAC with only ten similar genes modulated by both drugs. CONCLUSION: Despite similarity in chemical structure and DNA hypomethylating activity, 5AC and DAC induced widely different histone PTMs and considering them interchangeable should be carefully evaluated. The mechanism of these histone PTM changes is not clear and may involve modulation of the activity or the expression of the enzymes inducing histone PTMs.


Acetylation/drug effects , Azacitidine/pharmacology , DNA Methylation/drug effects , Decitabine/pharmacology , Histones/drug effects , Cell Line, Tumor , Humans , Leukemia/drug therapy , Protein Processing, Post-Translational/drug effects
19.
Toxicol Appl Pharmacol ; 436: 115882, 2022 02 01.
Article En | MEDLINE | ID: mdl-35016910

Oocyte maturation is essential for fertilization and early embryo development, and proper organelle functions guarantee this process to maintain high-quality oocytes. The type B trichothecene nivalenol (NIV) is a mycotoxin produced by Fusarium oxysporum and is commonly found in contaminated food. NIV intake affect growth, the immune system, and the female reproductive system. Here, we investigated NIV toxicity on mouse oocyte quality. Transcriptome analysis results showed that NIV exposure altered the expression of multiple genes involved in spindle formation and organelle function in mouse oocytes, indicating its toxicity on mouse oocyte maturation. Further analysis indicated that NIV exposure disrupted spindle structure and chromosome alignment, possibly through tubulin acetylation. NIV exposure induced aberrant mitochondria distribution and reduced mitochondria number, mitochondria membrane potential (MMP), and ATP levels. In addition, NIV caused the abnormal distribution of the Golgi apparatus and altered the expression of the vesicle trafficking protein Rab11. ER distribution was also disturbed under NIV exposure, indicating the effects of NIV on protein modification and transport in oocytes. Thus, our results demonstrated that NIV exposure affected spindle structure and organelles function in mouse oocytes.


Embryonic Development/drug effects , Oocytes/drug effects , Organelles/drug effects , Spindle Apparatus/drug effects , Trichothecenes/adverse effects , Acetylation/drug effects , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Chromosomes/drug effects , Female , Meiosis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Mycotoxins/adverse effects , Oocytes/metabolism , Oogenesis/drug effects , Organelles/metabolism , Spindle Apparatus/metabolism , Transcriptome/drug effects , Tubulin/metabolism
20.
J Med Chem ; 65(3): 2208-2224, 2022 02 10.
Article En | MEDLINE | ID: mdl-35005974

Glioma treatment remains a challenge with a low survival rate due to the lack of effective therapeutics. Monoamine oxidase A (MAO A) plays a role in glioma development, and MAO A inhibitors reduce glioma growth. Histone deacetylase (HDAC) inhibition has emerged as a promising therapy for various malignancies including gliomas. We have synthesized and evaluated N-methylpropargylamine-conjugated hydroxamic acids as dual inhibitors of MAO A and HDAC. Compounds display potent MAO A inhibition with IC50 from 0.03 to <0.0001 µM and inhibit HDAC isoforms and cell growth in the micromolar to nanomolar IC50 range. These selective MAO A inhibitors increase histone H3 and α-tubulin acetylation and induce cell death via nonapoptotic mechanisms. Treatment with 15 reduced tumor size, reduced MAO A activity in brain and tumor tissues, and prolonged the survival. This first report on dual inhibitors of MAO A and HDAC establishes the basis of translational research for an improved treatment of glioma.


Enzyme Inhibitors/chemistry , Histone Deacetylases/chemistry , Hydroxamic Acids/chemistry , Monoamine Oxidase/chemistry , Acetylation/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Glioma/drug therapy , Glioma/mortality , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/metabolism , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase/metabolism , Pargyline/analogs & derivatives , Pargyline/chemistry , Propylamines/chemistry , Structure-Activity Relationship , Transplantation, Heterologous
...