Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.111
Filter
1.
Nat Commun ; 15(1): 8044, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39271654

ABSTRACT

Cell-cell junctions, and specifically desmosomes, are crucial for robust intercellular adhesion. Desmosomal function is compromised in the autoimmune blistering skin disease pemphigus vulgaris. We combine whole-genome knockout screening and a promotor screen of the desmosomal gene desmoglein 3 in human keratinocytes to identify novel regulators of intercellular adhesion. Kruppel-like-factor 5 (KLF5) directly binds to the desmoglein 3 regulatory region and promotes adhesion. Reduced levels of KLF5 in patient tissue indicate a role in pemphigus vulgaris. Autoantibody fractions from patients impair intercellular adhesion and reduce KLF5 levels in in vitro and in vivo disease models. These effects were dependent on increased activity of histone deacetylase 3, leading to transcriptional repression of KLF5. Inhibiting histone deacetylase 3 increases KLF5 levels and protects against the deleterious effects of autoantibodies in murine and human pemphigus vulgaris models. Together, KLF5 and histone deacetylase 3 are regulators of desmoglein 3 gene expression and intercellular adhesion and represent potential therapeutic targets in pemphigus vulgaris.


Subject(s)
Cell Adhesion , Desmoglein 3 , Keratinocytes , Kruppel-Like Transcription Factors , Pemphigus , Humans , Pemphigus/metabolism , Pemphigus/pathology , Pemphigus/immunology , Desmoglein 3/metabolism , Desmoglein 3/genetics , Animals , Keratinocytes/metabolism , Mice , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Autoantibodies/immunology , Desmosomes/metabolism , Disease Models, Animal , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Gene Expression Regulation , Promoter Regions, Genetic/genetics , Male
2.
Diagn Pathol ; 19(1): 120, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237939

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) remains one of the most lethal urological malignancies even though a great number of improvements in diagnosis and management have achieved over the past few decades. Accumulated evidence revealed that histone deacetylases (HDACs) play vital role in cell proliferation, differentiation and apoptosis. Nevertheless, the biological functions of histone deacetylation modification related genes in ccRCC remains poorly understood. METHOD: Bulk transcriptomic data and clinical information of ccRCC patients were obtained from the TCGA database and collected from the Chinese PLA General Hospital. A total of 36 histone deacetylation genes were selected and studied in our research. Univariate cox regression analysis, least absolute shrinkage and selection operator (LASSO) regression, random forest (RF) analysis, and protein-protein interaction (PPI) network analysis were applied to identify key genes affecting the prognosis of ccRCC. The 'oncoPredict' algorithm was utilized for drug-sensitive analysis. Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was used to explore the potential biological function. The ssGSEA algorithm was used for tumor immune microenvironment analysis. The expression levels of HDAC10 were validated by RT-PCR and immunohistochemistry (IHC). 5-ethynyl-2'-deoxyuridine (EdU assay), CCK-8 assay, cell transwell migration and invasion assay and colony formation assay were performed to detect the proliferation and invasion ability of ccRCC cells. A nomogram incorporating HDAC10 and clinicopathological characteristics was established to predict the prognosis of ccRCC patients. RESULT: Two machine learning algorithms and PPI analysis identified four histone deacetylation genes that have a significant association with the prognosis of ccRCC, with HDAC10 being the key gene among them. HDAC10 is highly expressed in ccRCC and its high expression is associated with poor prognosis for ccRCC patients. Pathway enrichment and the experiments of EdU staining, CCK-8 assay, cell transwell migration and invasion assay and colony formation assay demonstrated that HDAC10 mediated the proliferation and metastasis of ccRCC cells and involved in reshaping the tumor microenvironment (TME) of ccRCC. A clinically reliable prognostic predictive model was established by incorporating HDAC10 and other clinicopathological characteristics ( https://nomogramhdac10.shinyapps.io/HDAC10_Nomogram/ ). CONCLUSION: Our study found the increased expression of HDAC10 was closely associated with poor prognosis of ccRCC patients. HDAC10 showed a pro-tumorigenic effect on ccRCC and promote the proliferation and metastasis of ccRCC, which may provide new light on targeted therapy for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Cell Proliferation , Histone Deacetylases , Kidney Neoplasms , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Cell Proliferation/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Male , Female , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Prognosis , Tumor Microenvironment/genetics , Cell Line, Tumor , Protein Interaction Maps , Oncogenes/genetics , Aged
3.
Protein Sci ; 33(10): e5170, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39276009

ABSTRACT

The intrinsically disordered protein MeCP2 is a global transcriptional regulator encoded by the MECP2 gene. Although the structured domains of MeCP2 have been the subject of multiple studies, its unstructured regions have not been that extensively characterized. In this work, we show that MeCP2 possesses properties akin to those of supercharged proteins. By utilizing its unstructured portions, MeCP2 can successfully transduce across cell membranes and localize to heterochromatic foci in the nuclei, displaying uptake levels a third lower than a MeCP2 construct fused to the cell-penetrating peptide TAT. MeCP2 uptake can further be enhanced by the addition of compounds that promote endosomal escape following cellular trafficking by means of macropinocytosis. Using a combination of in silico prediction algorithms and live-cell imaging experiments, we mapped the sequence in MeCP2 responsible for its cellular incorporation, which bears a striking resemblance to TAT itself. Transduced MeCP2 was shown to interact with HDAC3. These findings provide valuable insight into the properties of MeCP2 and may be beneficial for devising future protein-based treatment strategies.


Subject(s)
Cell Membrane , Histone Deacetylases , Methyl-CpG-Binding Protein 2 , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/chemistry , Humans , Cell Membrane/metabolism , Cell Membrane/chemistry , Histone Deacetylases/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , HEK293 Cells , Protein Transport , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics
4.
Food Res Int ; 195: 114961, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39277234

ABSTRACT

Monacolin K (MK), also known as lovastatin, is a polyketide compound with the ability to reduce plasma cholesterol levels and many other bio-activities. Red yeast rice (also named Hongqu) rich in MK derived from Monascus fermentation has attracted widespread attention due to its excellent performance in reducing blood lipids. However, industrial Monascus fermentation suffers from the limitations such as low yield of MK, long fermentation period, and susceptibility to contamination. In this study, we firstly blocked the competitive pathway of MK biosynthesis to create polyketide synthase gene pigA (the key gene responsible for the biosynthesis of Monascus azaphilone pigments) deficient strain A1. Then, based on the strategies to increase precursor supply for MK biosynthesis, acetyl-CoA carboxylase gene acc overexpression strains C1 and C2 were constructed with WT and A1 as the parent, respectively. Finally, histone deacetylase gene hos2 overexpression strain H1 was constructed by perturbation of histone acetylation modification. HPLC detection revealed all these four strains significantly increased their abilities to produce MK. After 14 days of solid-state fermentation, the MK yields of strains A1, C1, C2, and H1 reached 2.03 g/100 g, 1.81 g/100 g, 2.45 g/100 g and 2.52 g/100 g, which increased by 28.5 %, 14.7 %, 43.9 % and 36.1 % compared to WT, respectively. RT-qPCR results showed that overexpression of hos2 significantly increased the expression level of almost all genes responsible for MK biosynthesis after 5-day growth. Overall, the abilities of these strains to produce MK has been greatly improved, and MK production period has been shortened to 14 days from 20 days, providing new approaches for efficient production of Hongqu rich in MK.


Subject(s)
Fermentation , Histones , Lovastatin , Monascus , Monascus/metabolism , Monascus/genetics , Acetylation , Histones/metabolism , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/genetics , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Hypolipidemic Agents/pharmacology , Biological Products/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics
5.
Sci Rep ; 14(1): 21165, 2024 09 10.
Article in English | MEDLINE | ID: mdl-39256449

ABSTRACT

Diabetes mellitus (DM) is a well-documented risk factor of intervertebral disc degeneration (IVDD). The current study was aimed to clarify the effects and mechanisms of NADH: ubiquinone oxidoreductase subunit A3 (NDUFA3) in human nucleus pulposus cells (HNPCs) exposed to high glucose. NDUFA3 was overexpressed in HNPCs via lenti-virus transduction, which were co-treated with high glucose and rotenone (a mitochondrial complex I inhibitor) for 48 h. Cell activities were assessed for cell viability, cell apoptosis, reactive oxygen species (ROS) production, mitochondrial membrane potential (MMP) ratio, oxygen consumption rate (OCR) and mitochondrial complexes I activities. High glucose decreased cell viability, increased apoptotic cells, increased ROS production, decreased MMP levels and OCR values in HNPCs in a dose-dependent manner. Rotenone co-treatment augmented the high glucose-induced injuries on cell viability, apoptosis, ROS production and mitochondrial function. NDUFA3 overexpression counteracted the high glucose-induced injuries in HNPCs. HDAC/H3K27ac mechanism was involved in regulating NDUFA3 transcription. NDUFA3 knockdown decreased cell viability and increased apoptotic cells, which were reversed by ROS scavenger N-acetylcysteine. HDAC/H3K27ac-mediated transcription of NDUFA3 protects HNPCs against high glucose-induced injuries through suppressing cell apoptosis, eliminating ROS, improving mitochondrial function and oxidative phosphorylation. This study sheds light on candidate therapeutic targets and deepens the understanding of molecular mechanisms behind DM-induced IVDD.


Subject(s)
Apoptosis , Electron Transport Complex I , Glucose , Histones , Mitochondria , Nucleus Pulposus , Humans , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Glucose/pharmacology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Nucleus Pulposus/metabolism , Nucleus Pulposus/drug effects , Reactive Oxygen Species/metabolism , Rotenone/pharmacology , Transcription, Genetic/drug effects
6.
Mol Med Rep ; 30(6)2024 Dec.
Article in English | MEDLINE | ID: mdl-39329199

ABSTRACT

Graves' orbitopathy (GO), a manifestation of Graves' disease, is characterized by orbital fibroblast­induced inflammation, leading to fibrosis or adipogenesis. Histone deacetylase (HDAC) serves a central role in autoimmune diseases and fibrosis. The present study investigated HDAC inhibition in orbital fibroblasts from patients with GO to evaluate its potential as a therapeutic agent. Primary cultured orbital fibroblasts were treated with an HDAC inhibitor, panobinostat, under the stimulation of IL­1ß, TGF­ß or adipogenic medium. Inflammatory cytokines, and fibrosis­ and adipogenesis­related proteins were analyzed using western blotting. The effects of panobinostat on HDAC mRNA expression were measured in GO orbital fibroblasts, and specific HDACs were inhibited using small interfering RNA transfection. Panobinostat significantly reduced the IL­1ß­induced production of inflammatory cytokines and TGF­ß­induced production of fibrosis­related proteins. It also suppressed adipocyte differentiation and adipogenic transcription factor production. Furthermore, it significantly attenuated HDAC7 mRNA expression in GO orbital fibroblasts. In addition, the silencing of HDAC7 led to anti­inflammatory and anti­fibrotic effects. In conclusion, by inhibiting HDAC7 gene expression, panobinostat may suppress the production of inflammatory cytokines, profibrotic proteins and adipogenesis in GO orbital fibroblasts. The present in vitro study suggested that HDAC7 could be a potential therapeutic target for inhibiting the inflammatory, adipogenic and fibrotic mechanisms of GO.


Subject(s)
Fibroblasts , Graves Ophthalmopathy , Histone Deacetylase Inhibitors , Histone Deacetylases , Humans , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/drug therapy , Graves Ophthalmopathy/genetics , Graves Ophthalmopathy/pathology , Histone Deacetylase Inhibitors/pharmacology , Fibroblasts/metabolism , Fibroblasts/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Cells, Cultured , Panobinostat/pharmacology , Cytokines/metabolism , Adipogenesis/drug effects , Male , Female , Middle Aged , Adult , Transforming Growth Factor beta/metabolism , Cell Differentiation/drug effects , Interleukin-1beta/metabolism
7.
J Cell Mol Med ; 28(18): e70114, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39317961

ABSTRACT

Nephrotoxicity is a major side effect of platinum-based antineoplastic drugs, and there is currently no available therapeutic intervention. Our study suggests that targeting histone deacetylase 8 could be a potential treatment for cisplatin-induced acute kidney injury (AKI). In a murine model of AKI induced by cisplatin, the administration of PCI-34051, a selective inhibitor of HDAC8, resulted in significant improvement in renal function and reduction in renal tubular damage and apoptosis. Pharmacological inhibition of HDAC8 also decreased caspase-3 and PARP1 cleavage, attenuated Bax expression and preserved Bcl-2 levels in the injured kidney. In cultured murine renal epithelial cells (mRTECs) exposed to cisplatin, treatment with PCI-34051 or transfection with HDAC8 siRNA reduced apoptotic cell numbers and diminished expression of cleaved caspase-3 and PARP1; conversely, overexpression of HDAC8 intensified these changes. Additionally, PCI-34051 reduced p53 expression levels along with those for p21, p-CDK2 and γ-H2AX while preserving MRE11 expression in the injured kidney. Similarly, pharmacological and genetic inhibition of HDAC8 reduced γ-H2AX and enhanced MRE11 expression; conversely, HDAC8 overexpression exacerbated these changes in mRTECs exposed to cisplatin. These results support that HDAC8 inhibition attenuates cisplatin-induced AKI through a mechanism associated with reducing DNA damage and promoting its repair.


Subject(s)
Acute Kidney Injury , Apoptosis , Cisplatin , DNA Damage , Histone Deacetylase Inhibitors , Histone Deacetylases , Recombinational DNA Repair , Tumor Suppressor Protein p53 , Animals , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Cisplatin/adverse effects , Cisplatin/pharmacology , DNA Damage/drug effects , Mice , Recombinational DNA Repair/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Apoptosis/drug effects , Histone Deacetylase Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Male , Mice, Inbred C57BL , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Histones/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Caspase 3/metabolism , Repressor Proteins/metabolism , Repressor Proteins/genetics , MRE11 Homologue Protein/metabolism , MRE11 Homologue Protein/genetics , Disease Models, Animal , Hydroxamic Acids/pharmacology , Indoles
8.
Planta ; 260(4): 93, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39264431

ABSTRACT

MAIN CONCLUSION: This review focuses on HATs and HDACs that modify non-histone proteins, summarizes functional mechanisms of non-histone acetylation as well as the roles of HATs and HDACs in rice and Arabidopsis. The growth and development of plants, as well as their responses to biotic and abiotic stresses, are governed by intricate gene and protein regulatory networks, in which epigenetic modifying enzymes play a crucial role. Histone lysine acetylation levels, modulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), are well-studied in the realm of transcriptional regulation. However, the advent of advanced proteomics has unveiled that non-histone proteins also undergo acetylation, with its underlying mechanisms now being clarified. Indeed, non-histone acetylation influences protein functionality through diverse pathways, such as modulating protein stability, adjusting enzymatic activity, steering subcellular localization, influencing interactions with other post-translational modifications, and managing protein-protein and protein-DNA interactions. This review delves into the recent insights into the functional mechanisms of non-histone acetylation in plants. We also provide a summary of the roles of HATs and HDACs in rice and Arabidopsis, and explore their potential involvement in the regulation of non-histone proteins.


Subject(s)
Arabidopsis , Histone Acetyltransferases , Histone Deacetylases , Oryza , Plant Proteins , Protein Processing, Post-Translational , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Acetylation , Oryza/genetics , Oryza/metabolism , Oryza/enzymology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/enzymology , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Plant Proteins/metabolism , Plant Proteins/genetics , Gene Expression Regulation, Plant , Histones/metabolism
9.
J Cell Mol Med ; 28(17): e70081, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39261913

ABSTRACT

Diet-induced obesity is associated with enhanced systemic inflammation that limits bone regeneration. HDAC inhibitors are currently being explored as anti-inflammatory agents. Prior reports show that myeloid progenitor-directed Hdac3 ablation enhances intramembranous bone healing in female mice. In this study, we determined if Hdac3 ablation increased intramembranous bone regeneration in mice fed a high-fat/high-sugar (HFD) diet. Micro-CT analyses demonstrated that HFD-feeding enhanced the formation of periosteal reaction tissue of control littermates, reflective of suboptimal bone healing. We confirmed enhanced bone volume within the defect of Hdac3-ablated females and showed that Hdac3 ablation reduced the amount of periosteal reaction tissue following HFD feeding. Osteoblasts cultured in a conditioned medium derived from Hdac3-ablated cells exhibited a four-fold increase in mineralization and enhanced osteogenic gene expression. We found that Hdac3 ablation elevated the secretion of several chemokines, including CCL2. We then confirmed that Hdac3 deficiency increased the expression of Ccl2. Lastly, we show that the proportion of CCL2-positve cells within bone defects was significantly higher in Hdac3-deficient mice and was further enhanced by HFD. Overall, our studies demonstrate that Hdac3 deletion enhances intramembranous bone healing in a setting of diet-induced obesity, possibly through increased production of CCL2 by macrophages within the defect.


Subject(s)
Diet, Western , Histone Deacetylases , Osteogenesis , Animals , Female , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/deficiency , Mice , Diet, Western/adverse effects , Osteoblasts/metabolism , Diet, High-Fat/adverse effects , Periosteum/metabolism , Periosteum/pathology , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Bone Regeneration , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Obesity/etiology , Obesity/pathology
10.
Plant Mol Biol ; 114(5): 101, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39312030

ABSTRACT

Histone deacetylation, one of most important types of post-translational modification, plays multiple indispensable roles in plant growth and development and abiotic stress responses. However, little information about the roles of histone deacetylase in regulating inflorescence architecture, fruit yield, and stress responses is available in tomato. Functional characterization revealed that SlHDT1 participated in the control of inflorescence architecture and fruit yield by regulating auxin signalling, and influenced tolerance to drought and salt stresses by governing abscisic acid (ABA) signalling. More inflorescence branches and higher fruit yield, which were influenced by auxin signalling, were observed in SlHDT1-RNAi transgenic plants. Moreover, tolerance to drought and salt stresses was decreased in SlHDT1-RNAi transgenic lines compared with the wild type (WT). Changes in parameters related to the stress response, including decreases in survival rate, chlorophyll content, relative water content (RWC), proline content, catalase (CAT) activity and ABA content and an increase in malonaldehyde (MDA) content, were observed in SlHDT1-RNAi transgenic lines. In addition, the RNA-seq analysis revealed varying degrees of downregulation for genes such as the stress-related genes SlABCC10 and SlGAME6 and the pathogenesis-related protein P450 gene SlCYP71A1, and upregulation of the pathogenesis-related protein P450 genes SlCYP94B1, SlCYP734A7 and SlCYP94A2 in SlHDT1-RNAi transgenic plants, indicating that SlHDT1 plays an important role in the response to biotic and abiotic stresses by mediating stress-related gene expression. In summary, the data suggest that SlHDT1 plays essential roles in the regulation of inflorescence architecture and fruit yield and in the response to drought and salt stresses.


Subject(s)
Abscisic Acid , Droughts , Fruit , Gene Expression Regulation, Plant , Plant Proteins , Plants, Genetically Modified , Salt Tolerance , Solanum lycopersicum , Solanum lycopersicum/genetics , Solanum lycopersicum/physiology , Solanum lycopersicum/growth & development , Salt Tolerance/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Abscisic Acid/metabolism , Fruit/genetics , Fruit/growth & development , Fruit/metabolism , Stress, Physiological/genetics , Indoleacetic Acids/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism
11.
Aging (Albany NY) ; 16(17): 12239-12251, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39248711

ABSTRACT

The relationship between metastasis-associated protein 2 (MTA2) overexpression and tumor growth and metastasis has been extensively studied in a variety of tumor cells but not in human osteosarcoma cells. This study aims to elucidate the clinical significance, underlying molecular mechanisms, and biological functions of MTA2 in human osteosarcoma in vitro and in vivo. Our results show that MTA2 was elevated in osteosarcoma cell lines and osteosarcoma tissues and was associated with tumor stage and overall survival of osteosarcoma patients. Knockdown of MTA2 inhibited osteosarcoma cell migration and invasion by reducing the expression of urokinase-type plasminogen activator (uPA). Bioinformatic analysis demonstrated that high levels of uPA in human osteosarcoma tissues correlated positively with MTA2 expression. Furthermore, treatment with recombinant human uPA (Rh-uPA) caused significant restoration of OS cell migration and invasion in MTA2 knockdown osteosarcoma cells. We found that ERK1/2 depletion increased the expression of uPA, facilitating osteosarcoma cell migration and invasion. Finally, MTA2 depletion significantly reduced tumor metastasis and the formation of lung nodules in vivo. Overall, our study suggests that MTA2 knockdown suppresses osteosarcoma cell metastasis by decreasing uPA expression via ERK signaling. This finding provides new insight into potential treatment strategies against osteosarcoma metastasis by targeting MTA2.


Subject(s)
Bone Neoplasms , Cell Movement , Gene Knockdown Techniques , Histone Deacetylases , Osteosarcoma , Repressor Proteins , Urokinase-Type Plasminogen Activator , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , Humans , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Repressor Proteins/genetics , Repressor Proteins/metabolism , Cell Movement/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Male , Female , Mice , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Mice, Nude , MAP Kinase Signaling System/genetics
12.
Appl Microbiol Biotechnol ; 108(1): 467, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39292268

ABSTRACT

Epigenetic regulation plays a central role in the regulation of a number of cellular processes such as proliferation, differentiation, cell cycle, and apoptosis. In particular, small molecule epigenetic modulators are key elements that can effectively influence gene expression by precisely regulating the epigenetic state of cells. To identify useful small-molecule regulators that enhance the expression of recombinant proteins in Chinese hamster ovary (CHO) cells, we examined a novel dual-HDAC/LSD1 inhibitor I-4 as a supplement for recombinant CHO cells. Treatment with 2 µM I-4 was most effective in increasing monoclonal antibody production. Despite cell cycle arrest at the G1/G0 phase, which inhibits cell growth, the addition of the inhibitor at 2 µM to monoclonal antibody-expressing CHO cell cultures resulted in a 1.94-fold increase in the maximal monoclonal antibody titer and a 2.43-fold increase in specific monoclonal antibody production. In addition, I-4 significantly increased the messenger RNA levels of the monoclonal antibody and histone H3 acetylation and methylation levels. We also investigated the effect on HDAC-related isoforms and found that interference with the HDAC5 gene increased the monoclonal antibody titer by 1.64-fold. The results of this work provide an effective method of using epigenetic regulatory strategies to enhance the expression of recombinant proteins in CHO cells. KEY POINTS: • HDAC/LSD1 dual-target small molecule inhibitor can increase the expression level of recombinant monoclonal antibodies in CHO cells. • By affecting the acetylation and methylation levels of histones in CHO cells and downregulating HDAC5, the production of recombinant monoclonal antibodies increased. • It provides an effective pathway for applying epigenetic regulation strategies to enhance the expression of recombinant proteins.


Subject(s)
Antibodies, Monoclonal , Cricetulus , Epigenesis, Genetic , Recombinant Proteins , CHO Cells , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Histones/genetics , Acetylation , Cricetinae , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Methylation
13.
Mol Cell ; 84(17): 3175-3191.e8, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39096900

ABSTRACT

Heterochromatin enforces transcriptional gene silencing and can be epigenetically inherited, but the underlying mechanisms remain unclear. Here, we show that histone deacetylation, a conserved feature of heterochromatin domains, blocks SWI/SNF subfamily remodelers involved in chromatin unraveling, thereby stabilizing modified nucleosomes that preserve gene silencing. Histone hyperacetylation, resulting from either the loss of histone deacetylase (HDAC) activity or the direct targeting of a histone acetyltransferase to heterochromatin, permits remodeler access, leading to silencing defects. The requirement for HDAC in heterochromatin silencing can be bypassed by impeding SWI/SNF activity. Highlighting the crucial role of remodelers, merely targeting SWI/SNF to heterochromatin, even in cells with functional HDAC, increases nucleosome turnover, causing defective gene silencing and compromised epigenetic inheritance. This study elucidates a fundamental mechanism whereby histone hypoacetylation, maintained by high HDAC levels in heterochromatic regions, ensures stable gene silencing and epigenetic inheritance, providing insights into genome regulatory mechanisms relevant to human diseases.


Subject(s)
Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Silencing , Heterochromatin , Histone Deacetylases , Histones , Nucleosomes , Heterochromatin/metabolism , Heterochromatin/genetics , Nucleosomes/metabolism , Nucleosomes/genetics , Histones/metabolism , Histones/genetics , Acetylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Humans , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Animals
14.
Food Chem Toxicol ; 192: 114940, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39151879

ABSTRACT

Infertility caused by lipopolysaccharide (LPS) exposure due to infection is endangering male fertility worldwide, but the mechanism remains unclear. The blood-testis barrier (BTB) is essential for maintaining spermatogenesis and male fertility. In the present study, we showed that LPS (5.0 mg/kg) treatment markedly down-regulated the expression of BTB-related proteins, expanded the biotin penetration distance and caused histopathological injury in seminiferous tubules in mouse testes. Notably, testicular macrophage M1 polarization induced by LPS seems to be related to BTB damage, which was well confirmed by co-culture of RAW264.7 and TM4 cells in vitro. Interestingly, a low-dose LPS (0.1 mg/kg) pretreatment attenuated down-regulation of BTB-related proteins expression and histopathological injury and shorten biotin penetration distance in seminiferous tubules caused by LPS. Correspondingly, a low-dose LPS pretreatment suppresses testicular macrophage M1 polarization induced by LPS in mouse testes. Further experiments revealed that histone deacetylase 5 (HDAC5) was markedly down-regulated at 2 h and slightly down-regulated at 8 h, but up-regulated at 24 h in mouse testes after LPS treatment. Additionally, low-dose LPS pretreatment against the down-regulation of HDAC5 protein caused by LPS treatment. Notably, the suppressed testicular macrophage M1 polarization by low-dose LPS pretreatment was broken by BRD4354, a specific inhibitor of HDAC5 in vitro. These results suggest suppressed testicular macrophage M1 polarization by HDAC5 enforces insensitivity to LPS-elicited BTB damage.


Subject(s)
Blood-Testis Barrier , Histone Deacetylases , Lipopolysaccharides , Macrophages , Animals , Male , Lipopolysaccharides/toxicity , Blood-Testis Barrier/drug effects , Mice , Macrophages/drug effects , Macrophages/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Testis/drug effects , Testis/metabolism , RAW 264.7 Cells
15.
Methods ; 230: 147-157, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191338

ABSTRACT

Epigenetics involves reversible modifications in gene expression without altering the genetic code itself. Among these modifications, histone deacetylases (HDACs) play a key role by removing acetyl groups from lysine residues on histones. Overexpression of HDACs is linked to the proliferation and survival of tumor cells. To combat this, HDAC inhibitors (HDACi) are commonly used in cancer treatments. However, pan-HDAC inhibition can lead to numerous side effects. Therefore, isoform-selective HDAC inhibitors, such as HDAC3i, could be advantageous for treating various medical conditions while minimizing off-target effects. To date, computational approaches that use only the SMILES notation without any experimental evidence have become increasingly popular and necessary for the initial discovery of novel potential therapeutic drugs. In this study, we develop an innovative and high-precision stacked-ensemble framework, called Stack-HDAC3i, which can directly identify HDAC3i using only the SMILES notation. Using an up-to-date benchmark dataset, we first employed both molecular descriptors and Mol2Vec embeddings to generate feature representations that cover multi-view information embedded in HDAC3i, such as structural and contextual information. Subsequently, these feature representations were used to train baseline models using nine popular ML algorithms. Finally, the probabilistic features derived from the selected baseline models were fused to construct the final stacked model. Both cross-validation and independent tests showed that Stack-HDAC3i is a high-accuracy prediction model with great generalization ability for identifying HDAC3i. Furthermore, in the independent test, Stack-HDAC3i achieved an accuracy of 0.926 and Matthew's correlation coefficient of 0.850, which are 0.44-6.11% and 0.83-11.90% higher than its constituent baseline models, respectively.


Subject(s)
Histone Deacetylase Inhibitors , Histone Deacetylases , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/chemistry , Humans , Machine Learning , Drug Discovery/methods
16.
Elife ; 132024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093942

ABSTRACT

Diffuse midline gliomas (DMGs) are aggressive and fatal pediatric tumors of the central nervous system that are highly resistant to treatments. Lysine to methionine substitution of residue 27 on histone H3 (H3-K27M) is a driver mutation in DMGs, reshaping the epigenetic landscape of these cells to promote tumorigenesis. H3-K27M gliomas are characterized by deregulation of histone acetylation and methylation pathways, as well as the oncogenic MYC pathway. In search of effective treatment, we examined the therapeutic potential of dual targeting of histone deacetylases (HDACs) and MYC in these tumors. Treatment of H3-K27M patient-derived cells with Sulfopin, an inhibitor shown to block MYC-driven tumors in vivo, in combination with the HDAC inhibitor Vorinostat, resulted in substantial decrease in cell viability. Moreover, transcriptome and epigenome profiling revealed synergistic effect of this drug combination in downregulation of prominent oncogenic pathways such as mTOR. Finally, in vivo studies of patient-derived orthotopic xenograft models showed significant tumor growth reduction in mice treated with the drug combination. These results highlight the combined treatment with PIN1 and HDAC inhibitors as a promising therapeutic approach for these aggressive tumors.


Diffuse midline gliomas (DMGs) are among the most aggressive and fatal brain cancers in children. They are often associated with changes in histones, the proteins that control gene activity and give chromosomes their structure. Most children with DMGs, for example, share the same anomaly in their histone H3 protein (referred to as the H3-K27M mutation). This change affects how small chemical tags called methyl and acetyl groups can be added onto histone 3, which in turn alters the way the protein can switch genes on and off. As a result, tumours start to develop. One potential therapeutic strategy against DMGs is to use histone deacetylase inhibitors (HDACi), a promising type of drugs which inhibits the enzymes that remove acetyl groups from histones. Patients can develop resistance to HDACi, however, highlighting the need to explore other approaches. One possibility is to treat patients with several types of drugs, each usually targeting a distinct biological process that contributes to the emergence of cancer. This combined approach can have multiple benefits; the drugs potentially amplify each other's effect, for example, and it is also less likely for cells to become resistant to more than one compound at the time. In addition, each drug in the combination can be used in a lower dose to reduce side effects and benefit patients. DMG tumour cells often feature higher activity levels of a protein known as MYC, which can contribute to the growth of the tumour. Algranati, Oren et al. therefore set out to test whether combining an HDACi known as Vorinostat with a drug that blocks MYC activity (Sulfopin) can act as an effective treatment for this cancer. Tumour samples from eight DMG patients were treated with either Sulfopin alone, or Sulfopin in association with Vorinostat. Cells exposed to both drugs were less likely to survive, and additional genetic experiments showed that the combined treatment had resulted in pathways that promote tumour development being blocked. When both Sulfopin and Vorinostat were administered to mice made to grow human DMG tumors, the animals showed a greater reduction in tumor growth. Treatment options for DMG are usually limited, with chemotherapy often being ineffective and surgery impossible. The work by Algranati, Oren et al. suggests that combining HDACi and drugs targeting the MYC pathway is a strategy that should be examined further to determine whether clinical applications are possible.


Subject(s)
Glioma , Histone Deacetylase Inhibitors , Histone Deacetylases , Histones , Glioma/drug therapy , Glioma/genetics , Glioma/pathology , Humans , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Histones/genetics , Histone Deacetylase Inhibitors/pharmacology , Mice , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Vorinostat/pharmacology , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cell Line, Tumor , Child , Disease Models, Animal , Xenograft Model Antitumor Assays
17.
Proc Natl Acad Sci U S A ; 121(35): e2401861121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39167603

ABSTRACT

Insect developmental transitions are precisely coordinated by ecdysone and juvenile hormone (JH). We previously revealed that accumulated H3K27 trimethylation (H3K27me3) at the locus encoding JH signal transducer Hairy is involved in the larval-pupal transition in insects, but the underlying mechanism remains to be fully defined. Here, we show in Drosophila and Bombyx that Rpd3-mediated H3K27 deacetylation in the prothoracic gland during the last larval instar promotes ecdysone biosynthesis and the larval-pupal transition by enabling H3K27me3 accumulation at the Hairy locus to induce its transcriptional repression. Importantly, we find that the homeodomain transcription factor Schlank acts to switch active H3K27 acetylation (H3K27ac) to repressive H3K27me3 at the Hairy locus by directly binding to the Hairy promoter and then recruiting the histone deacetylase Rpd3 and the histone methyltransferase PRC2 component Su(z)12 through physical interactions. Moreover, Schlank inhibits Hairy transcription to facilitate the larval-pupal transition, and the Schlank signaling cascade is suppressed by JH but regulated in a positive feedback manner by ecdysone. Together, our data uncover that Schlank mediates epigenetic reprogramming of H3K27 modifications in hormone actions during insect developmental transition.


Subject(s)
Drosophila Proteins , Ecdysone , Gene Expression Regulation, Developmental , Histones , Larva , Animals , Histones/metabolism , Acetylation , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Ecdysone/metabolism , Larva/metabolism , Larva/growth & development , Larva/genetics , Bombyx/metabolism , Bombyx/genetics , Bombyx/growth & development , Juvenile Hormones/metabolism , Methylation , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Signal Transduction , Pupa/metabolism , Pupa/growth & development , Pupa/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Insect Proteins/metabolism , Insect Proteins/genetics , Repressor Proteins , Basic Helix-Loop-Helix Transcription Factors
18.
J Physiol Investig ; 67(4): 174-186, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39133038

ABSTRACT

ABSTRACT: Sepsis is a syndrome of systemic inflammatory response resulting from infection, which can lead to severe lung injury. Histone deacetylase 4 (HDAC4) is a key protein known to regulate a wide range of cellular processes. This study was designed to investigate the role of HDAC4 in lipopolysaccharide (LPS)-induced alveolar epithelial cell injury as well as to disclose its potential molecular mechanisms. The alveolar epithelial cell injury model was established by inducing A549 cells with LPS. A549 cell viability was detected by cell counting kit-8 assay and the transfection efficiency of small interfering RNA targeting HDAC4 was appraised utilizing Western blot. The levels of inflammatory cytokines and oxidative stress markers were detected using corresponding assay kits. Dichloro-dihydro-fluorescein diacetate assay was used for the measurement of reactive oxygen species (ROS) content. Flow cytometry, 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolyl-carbocyanine iodide-1 staining, adenosine triphosphate (ATP) assay kits, and MitoSOX Red assay kits were employed to estimate cell apoptosis, mitochondrial membrane potential, ATP level, and mitochondrial ROS level, respectively. The oxygen consumption rate of A549 cells was evaluated with XF96 extracellular flux analyzer. Western blot was applied for the evaluation of HDAC4, apoptosis- and c-Jun N-terminal kinase (JNK)/activating protein-1 (AP-1) signaling pathway-related proteins. HDAC4 expression was found to be increased in LPS-induced A549 cells and HDAC4 silence inhibited inflammatory damage, repressed oxidative stress, alleviated cell apoptosis, improved mitochondrial function, and blocked JNK/AP-1 signaling in A549 cells stimulated by LPS, which were all reversed by JNK activator anisomycin. Collectively, the interference with HDAC4 could ameliorate LPS-induced alveolar epithelial cell injury, and such protective effect may be potentially mediated through the JNK/AP-1 signaling pathway.


Subject(s)
Alveolar Epithelial Cells , Histone Deacetylases , Lipopolysaccharides , Sepsis , Humans , A549 Cells , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Sepsis/metabolism , Signal Transduction/drug effects , Transcription Factor AP-1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Repressor Proteins/metabolism , Repressor Proteins/genetics , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
19.
J Hazard Mater ; 477: 135093, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39088948

ABSTRACT

Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases , Particulate Matter , Up-Regulation , Animals , Particulate Matter/toxicity , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Epigenesis, Genetic/drug effects , Up-Regulation/drug effects , Mice , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Inflammation , Nanoparticles/chemistry , Nanoparticles/toxicity , Mice, Inbred C57BL , Cell Line , MAP Kinase Signaling System/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Male
20.
Am J Physiol Renal Physiol ; 327(4): F623-F636, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39116350

ABSTRACT

The chemotherapeutic agent cisplatin accumulates in the kidneys, leading to acute kidney injury (AKI). Preclinical and clinical studies have demonstrated sex-dependent outcomes of cisplatin-AKI. Deranged histone deacetylase (HDAC) activity is hypothesized to promote the pathogenesis of male murine cisplatin-AKI; however, it is unknown whether there are sex differences in the kidney HDACs. We hypothesized that there would be sex-specific Hdac expression, localization, or enzymatic activity, which may explain sexual dimorphic responses to cisplatin-AKI. In normal human kidney RNA samples, HDAC10 was significantly greater in the kidneys of women compared with men, whereas HDAC1, HDAC6, HDAC10, and HDAC11 were differentially expressed between the kidney cortex and medulla, regardless of sex. In a murine model of cisplatin-AKI (3 days after a 15 mg/kg injection), we found few sex- or cisplatin-related differences in Hdac kidney transcripts among the mice. Although Hdac9 was significantly greater in female mice compared with male mice, HDAC9 protein localization did not differ. Hdac7 transcripts were greater in the inner medulla of cisplatin-AKI mice, regardless of sex, and this agreed with a greater HDAC7 abundance. HDAC activity within the cortex, outer medulla, and inner medulla was significantly lower in cisplatin-AKI mice but did not differ between the sexes. In agreement with these findings, a class I HDAC inhibitor did not improve kidney injury or function. In conclusion, even though cisplatin-AKI was evident and there were transcript level differences among the different kidney regions in this model, there were few sex- or cisplatin-dependent effects on kidney HDAC localization or activity.NEW & NOTEWORTHY Kidney histone deacetylases (HDACs) are abundant in male and female mice, and the inner medulla has the greatest HDAC activity. A low dose of cisplatin caused acute kidney injury (AKI) in these mice, but there were few changes in kidney HDACs at the RNA/protein/activity level. A class I HDAC inhibitor failed to improve AKI outcomes. Defining the HDAC isoform, cellular source, and interventional timing is necessary to determine whether HDAC inhibition is a therapeutic strategy to prevent cisplatin-AKI in both sexes.


Subject(s)
Acute Kidney Injury , Cisplatin , Histone Deacetylases , Mice, Inbred C57BL , Animals , Cisplatin/toxicity , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Acute Kidney Injury/enzymology , Acute Kidney Injury/pathology , Female , Male , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Humans , Sex Factors , Mice , Histone Deacetylase Inhibitors/pharmacology , Disease Models, Animal , Kidney/drug effects , Kidney/metabolism , Kidney/enzymology , Kidney/pathology , Antineoplastic Agents/toxicity , Kidney Cortex/metabolism , Kidney Cortex/drug effects , Kidney Cortex/enzymology , Sex Characteristics
SELECTION OF CITATIONS
SEARCH DETAIL