Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.030
Filter
2.
Skelet Muscle ; 14(1): 11, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38769542

ABSTRACT

BACKGROUND: Myotonic Dystrophy type I (DM1) is the most common muscular dystrophy in adults. Previous reports have highlighted that neuromuscular junctions (NMJs) deteriorate in skeletal muscle from DM1 patients and mouse models thereof. However, the underlying pathomechanisms and their contribution to muscle dysfunction remain unknown. METHODS: We compared changes in NMJs and activity-dependent signalling pathways in HSALR and Mbnl1ΔE3/ΔE3 mice, two established mouse models of DM1. RESULTS: Muscle from DM1 mouse models showed major deregulation of calcium/calmodulin-dependent protein kinases II (CaMKIIs), which are key activity sensors regulating synaptic gene expression and acetylcholine receptor (AChR) recycling at the NMJ. Both mouse models exhibited increased fragmentation of the endplate, which preceded muscle degeneration. Endplate fragmentation was not accompanied by changes in AChR turnover at the NMJ. However, the expression of synaptic genes was up-regulated in mutant innervated muscle, together with an abnormal accumulation of histone deacetylase 4 (HDAC4), a known target of CaMKII. Interestingly, denervation-induced increase in synaptic gene expression and AChR turnover was hampered in DM1 muscle. Importantly, CaMKIIß/ßM overexpression normalized endplate fragmentation and synaptic gene expression in innervated Mbnl1ΔE3/ΔE3 muscle, but it did not restore denervation-induced synaptic gene up-regulation. CONCLUSIONS: Our results indicate that CaMKIIß-dependent and -independent mechanisms perturb synaptic gene regulation and muscle response to denervation in DM1 mouse models. Changes in these signalling pathways may contribute to NMJ destabilization and muscle dysfunction in DM1 patients.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Disease Models, Animal , Muscle, Skeletal , Myotonic Dystrophy , Neuromuscular Junction , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/physiopathology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Neuromuscular Junction/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Mice , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Receptors, Cholinergic/metabolism , Receptors, Cholinergic/genetics , Male , Mice, Inbred C57BL
3.
Biol Direct ; 19(1): 37, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734627

ABSTRACT

BACKGROUND: Clear cell renal cell carcinoma (RCC) is the most common kidney tumor. The analysis from medical database showed that Scm-like with four MBT domains protein 2 (SFMBT2) was decreased in advanced clear cell RCC cases, and its downregulation was associated with the poor prognosis. This study aims to investigate the role of SFMBT2 in clear cell RCC. METHODS: The expression of SFMBT2 in clear cell RCC specimens were determined by immunohistochemistry staining and western blot. The overexpression and knockdown of SFMBT2 was realized by infection of lentivirus loaded with SFMBT2 coding sequence or silencing fragment in 786-O and 769-P cells, and its effects on proliferation and metastasis were assessed by MTT, colony formation, flow cytometry, wound healing, transwell assay, xenograft and metastasis experiments in nude mice. The interaction of SFMBT2 with histone deacetylase 3 (HDAC3) and seven in absentia homolog 1 (SIAH1) was confirmed by co-immunoprecipitation. RESULTS: In our study, SFMBT2 exhibited lower expression in clear cell RCC specimens with advanced stages than those with early stages. Overexpression of SFMBT2 inhibited the growth and metastasis of clear cell RCC cells, 786-O and 769-P, in vitro and in vivo, and its silencing displayed opposites effects. HDAC3 led to deacetylation of SFMBT2, and the HDAC3 inhibitor-induced acetylation prevented SFMBT2 from SIAH1-mediated ubiquitination modification and proteasome degradation. K687 in SFMBT2 protein molecule may be the key site for acetylation and ubiquitination. CONCLUSIONS: SFMBT2 exerted an anti-tumor role in clear cell RCC cells, and HDAC3-mediated deacetylation promoted SIAH1-controlled ubiquitination of SFMBT2. SFMBT2 may be considered as a novel clinical diagnostic marker and/or therapeutic target of clear cell RCC, and crosstalk between its post-translational modifications may provide novel insights for agent development.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Mice, Nude , Ubiquitination , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Humans , Acetylation , Kidney Neoplasms/metabolism , Kidney Neoplasms/genetics , Animals , Mice , Cell Line, Tumor , Cell Proliferation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Gene Expression Regulation, Neoplastic
4.
Appl Microbiol Biotechnol ; 108(1): 332, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734756

ABSTRACT

Histone acetylation modifications in filamentous fungi play a crucial role in epigenetic gene regulation and are closely linked to the transcription of secondary metabolite (SM) biosynthetic gene clusters (BGCs). Histone deacetylases (HDACs) play a pivotal role in determining the extent of histone acetylation modifications and act as triggers for the expression activity of target BGCs. The genus Chaetomium is widely recognized as a rich source of novel and bioactive SMs. Deletion of a class I HDAC gene of Chaetomium olivaceum SD-80A, g7489, induces a substantial pleiotropic effect on the expression of SM BGCs. The C. olivaceum SD-80A ∆g7489 strain exhibited significant changes in morphology, sporulation ability, and secondary metabolic profile, resulting in the emergence of new compound peaks. Notably, three polyketides (A1-A3) and one asterriquinone (A4) were isolated from this mutant strain. Furthermore, our study explored the BGCs of A1-A4, confirming the function of two polyketide synthases (PKSs). Collectively, our findings highlight the promising potential of molecular epigenetic approaches for the elucidation of novel active compounds and their biosynthetic elements in Chaetomium species. This finding holds great significance for the exploration and utilization of Chaetomium resources. KEY POINTS: • Deletion of a class I histone deacetylase activated secondary metabolite gene clusters. • Three polyketides and one asterriquinone were isolated from HDAC deleted strain. • Two different PKSs were reported in C. olivaceum SD-80A.


Subject(s)
Chaetomium , Histone Deacetylases , Multigene Family , Polyketides , Secondary Metabolism , Chaetomium/genetics , Chaetomium/enzymology , Chaetomium/metabolism , Secondary Metabolism/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Polyketides/metabolism , Gene Deletion , Gene Expression Regulation, Fungal , Polyketide Synthases/genetics , Polyketide Synthases/metabolism , Biosynthetic Pathways/genetics , Epigenesis, Genetic
5.
FASEB J ; 38(10): e23659, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38733301

ABSTRACT

HDAC3 inhibition has been shown to improve memory and reduce amyloid-ß (Aß) in Alzheimer's disease (AD) models, but the underlying mechanisms are unclear. We investigated the molecular effects of HDAC3 inhibition on AD pathology, using in vitro and ex vivo models of AD, based on our finding that HDAC3 expression is increased in AD brains. For this purpose, N2a mouse neuroblastoma cells as well as organotypic brain cultures (OBCSs) of 5XFAD and wild-type mice were incubated with various concentrations of the HDAC3 selective inhibitor RGFP966 (0.1-10 µM) for 24 h. Treatment with RGFP966 or HDAC3 knockdown in N2a cells was associated with an increase on amyloid precursor protein (APP) and mRNA expressions, without alterations in Aß42 secretion. In vitro chromatin immunoprecipitation analysis revealed enriched HDAC3 binding at APP promoter regions. The increase in APP expression was also detected in OBCSs from 5XFAD mice incubated with 1 µM RGFP966, without changes in Aß. In addition, HDAC3 inhibition resulted in a reduction of activated Iba-1-positive microglia and astrocytes in 5XFAD slices, which was not observed in OBCSs from wild-type mice. mRNA sequencing analysis revealed that HDAC3 inhibition modulated neuronal regenerative pathways related to neurogenesis, differentiation, axonogenesis, and dendritic spine density in OBCSs. Our findings highlight the complexity and diversity of the effects of HDAC3 inhibition on AD models and suggest that HDAC3 may have multiple roles in the regulation of APP expression and processing, as well as in the modulation of neuroinflammatory and neuroprotective genes.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Disease Models, Animal , Histone Deacetylases , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Mice , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice, Transgenic , Brain/metabolism , Brain/pathology , Amyloid beta-Peptides/metabolism , Cell Line, Tumor , Male , Mice, Inbred C57BL , Microglia/metabolism , Phenylenediamines/pharmacology , Acrylamides
6.
Nat Commun ; 15(1): 4128, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750015

ABSTRACT

Mechanisms of functional cross-talk between global transcriptional repression and efficient DNA damage repair during genotoxic stress are poorly known. In this study, using human AF9 as representative of Super Elongation Complex (SEC) components, we delineate detailed mechanisms of these processes. Mechanistically, we describe that Poly-Serine domain-mediated oligomerization is pre-requisite for AF9 YEATS domain-mediated TFIID interaction-dependent SEC recruitment at the promoter-proximal region for release of paused RNA polymerase II. Interestingly, during genotoxic stress, CaMKII-mediated phosphorylation-dependent nuclear export of AF9-specific deacetylase HDAC5 enhances concomitant PCAF-mediated acetylation of K339 residue. This causes monomerization of AF9 and reduces TFIID interaction for transcriptional downregulation. Furthermore, the K339 acetylation-dependent enhanced AF9-DNA-PKc interaction leads to phosphorylation at S395 residue which reduces AF9-SEC interaction resulting in transcriptional downregulation and efficient repair of DNA damage. After repair, nuclear re-entry of HDAC5 reduces AF9 acetylation and restores its TFIID and SEC interaction to restart transcription.


Subject(s)
DNA Damage , DNA Repair , Histone Deacetylases , Protein Processing, Post-Translational , Transcription, Genetic , Humans , Acetylation , Phosphorylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , RNA Polymerase II/metabolism , Transcription Factor TFIID/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/chemistry , Protein Multimerization , HEK293 Cells , HeLa Cells , Transcriptional Elongation Factors/metabolism , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/chemistry
7.
Plant Signal Behav ; 19(1): 2353536, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38771929

ABSTRACT

Cellular behavior, cell differentiation and ontogenetic development in eukaryotes result from complex interactions between epigenetic and classic molecular genetic mechanisms, with many of these interactions still to be elucidated. Histone deacetylase enzymes (HDACs) promote the interaction of histones with DNA by compacting the nucleosome, thus causing transcriptional repression. MADS-domain transcription factors are highly conserved in eukaryotes and participate in controlling diverse developmental processes in animals and plants, as well as regulating stress responses in plants. In this work, we focused on finding out putative interactions of Arabidopsis thaliana HDACs and MADS-domain proteins using an evolutionary perspective combined with bioinformatics analyses and testing the more promising predicted interactions through classic molecular biology tools. Through bioinformatic analyses, we found similarities between HDACs proteins from different organisms, which allowed us to predict a putative protein-protein interaction between the Arabidopsis thaliana deacetylase HDA15 and the MADS-domain protein XAANTAL1 (XAL1). The results of two-hybrid and Bimolecular Fluorescence Complementation analysis demonstrated in vitro and in vivo HDA15-XAL1 interaction in the nucleus. Likely, this interaction might regulate developmental processes in plants as is the case for this type of interaction in animals.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Histone Deacetylases , MADS Domain Proteins , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , MADS Domain Proteins/metabolism , MADS Domain Proteins/genetics , Protein Binding , Two-Hybrid System Techniques
8.
Int J Biol Macromol ; 268(Pt 2): 131938, 2024 May.
Article in English | MEDLINE | ID: mdl-38692539

ABSTRACT

ING proteins display a high level of evolutionary conservation across various species, and play a crucial role in modulating histone acetylation levels, thus regulating various important biological processes in yeast and humans. Filamentous fungi possess distinct biological characteristics that differentiate them from yeasts and humans, and the specific roles of ING proteins in filamentous fungi remain largely unexplored. In this study, an ING protein, Fng2, orthologous to the yeast Pho23, has been identified in the wheat head blight fungus Fusarium graminearum. The deletion of the FNG2 gene resulted in defects in vegetative growth, conidiation, sexual reproduction, plant infection, and deoxynivalenol (DON) biosynthesis. Acting as a global regulator, Fng2 exerts negative control over histone H4 acetylation and governs the expression of over 4000 genes. Moreover, almost half of the differentially expressed genes in the fng3 mutant were found to be co-regulated by Fng2, emphasizing the functional association between these two ING proteins. Notably, the fng2 fng3 double mutant exhibits significantly increased H4 acetylation and severe defects in both fungal development and pathogenesis. Furthermore, Fng2 localizes within the nucleus and associates with the FgRpd3 histone deacetylase (HDAC) to modulate gene expression. Overall, Fng2's interaction with FgRpd3, along with its functional association with Fng3, underscores its crucial involvement in governing gene expression, thereby significantly influencing fungal growth, asexual and sexual development, pathogenicity, and secondary metabolism.


Subject(s)
Fungal Proteins , Fusarium , Gene Expression Regulation, Fungal , Histone Deacetylases , Plant Diseases , Triticum , Fusarium/pathogenicity , Fusarium/genetics , Triticum/microbiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Acetylation , Plant Diseases/microbiology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histones/metabolism , Trichothecenes/metabolism , Mutation , Protein Binding
9.
Int Immunopharmacol ; 134: 112246, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759372

ABSTRACT

BACKGROUND: A wide array of histone deacetylase (HDAC) inhibitors and aryl hydrocarbon receptor (AHR) agonists commonly arrest experimental autoimmune encephalomyelitis (EAE). However, it is not known whether HDAC inhibition is linked to the AHR signaling pathway in EAE. METHODS: We investigated how the pan-HDAC inhibitor SB939 (pracinostat) exerted immunoregulatory action in the myelin oligodendrocyte glycoprotein 35-55 (MOG35-55)-induced EAE mouse model by evaluating changes in of signal transducer and activator of transcription 3 (STAT3) acetylation and the expression of indoleamine 2,3-dioxygenase 1 (IDO1) and AHR in inflamed spinal cords during EAE evolution. We proved the involvement of IDO1 and the AHR in SB939-mediated immunosuppression using Ido1-/- and Ahr-/- mice. RESULTS: Administration with SB939 halted EAE progression, which depended upon IDO1 expression in neurons of the central nervous system (CNS). Our in vitro and in vivo studies demonstrated that SB939 sustained the interleukin-6-induced acetylation of STAT3, resulting in the stable transcriptional activation of Ido1. The therapeutic effect of SB939 also required the AHR, which is expressed mainly in CD4+ T cells and macrophages in CNS disease lesions. Finally, SB939 was shown to markedly reduce the proliferation of CD4+ T cells in inflamed neuronal tissues but not in the spleen or draining lymph nodes. CONCLUSIONS: Overall, our results suggest that IDO1 tryptophan metabolites produced by neuronal cells may act on AHR in pathogenic CD4+ T cells in a paracrine fashion in the CNS and that the specific induction of IDO1 expression in neurons at disease-afflicted sites can be considered a therapeutic approach to block the progression of multiple sclerosis without affecting systemic immunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Histone Deacetylase Inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase , Mice, Inbred C57BL , Mice, Knockout , Neurons , STAT3 Transcription Factor , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Mice , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Female , Spinal Cord/pathology , Spinal Cord/metabolism , Spinal Cord/immunology , Spinal Cord/drug effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Central Nervous System/immunology , Central Nervous System/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Disease Progression , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Peptide Fragments/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Interleukin-6/metabolism , Interleukin-6/genetics
10.
Int J Mol Sci ; 25(10)2024 May 18.
Article in English | MEDLINE | ID: mdl-38791568

ABSTRACT

Toxoplasma gondii is an intracellular parasite that is important in medicine and veterinary science and undergoes distinct developmental transitions in its intermediate and definitive hosts. The switch between stages of T. gondii is meticulously regulated by a variety of factors. Previous studies have explored the role of the microrchidia (MORC) protein complex as a transcriptional suppressor of sexual commitment. By utilizing immunoprecipitation and mass spectrometry, constituents of this protein complex have been identified, including MORC, Histone Deacetylase 3 (HDAC3), and several ApiAP2 transcription factors. Conditional knockout of MORC or inhibition of HDAC3 results in upregulation of a set of genes associated with schizogony and sexual stages in T. gondii tachyzoites. Here, our focus extends to two primary ApiAP2s (AP2XII-1 and AP2XI-2), demonstrating their significant impact on the fitness of asexual tachyzoites and their target genes. Notably, the targeted disruption of AP2XII-1 and AP2XI-2 resulted in a profound alteration in merozoite-specific genes targeted by the MORC-HDAC3 complex. Additionally, considerable overlap was observed in downstream gene profiles between AP2XII-1 and AP2XI-2, with AP2XII-1 specifically binding to a subset of ApiAP2 transcription factors, including AP2XI-2. These findings reveal an intricate cascade of ApiAP2 regulatory networks involved in T. gondii schizogony development, orchestrated by AP2XII-1 and AP2XI-2. This study provides valuable insights into the transcriptional regulation of T. gondii growth and development, shedding light on the intricate life cycle of this parasitic pathogen.


Subject(s)
Histone Deacetylases , Protozoan Proteins , Toxoplasma , Toxoplasma/genetics , Toxoplasma/metabolism , Toxoplasma/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Animals , Gene Expression Regulation , Toxoplasmosis/parasitology , Toxoplasmosis/genetics , Toxoplasmosis/metabolism
11.
Nat Commun ; 15(1): 4166, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755146

ABSTRACT

Failure of proper ventricular trabeculation is often associated with congenital heart disease. Support from endocardial cells, including the secretion of extracellular matrix and growth factors is critical for trabeculation. However, it is poorly understood how the secretion of extracellular matrix and growth factors is initiated and regulated by endocardial cells. We find that genetic knockout of histone deacetylase 3 in the endocardium in mice results in early embryo lethality and ventricular hypotrabeculation. Single cell RNA sequencing identifies significant downregulation of extracellular matrix components in histone deacetylase 3 knockout endocardial cells. Secretome from cultured histone deacetylase 3 knockout mouse cardiac endothelial cells lacks transforming growth factor ß3 and shows significantly reduced capacity in stimulating cultured cardiomyocyte proliferation, which is remarkably rescued by transforming growth factor ß3 supplementation. Mechanistically, we identify that histone deacetylase 3 knockout induces transforming growth factor ß3 expression through repressing microRNA-129-5p. Our findings provide insights into the pathogenesis of congenital heart disease and conceptual strategies to promote myocardial regeneration.


Subject(s)
Endocardium , Histone Deacetylases , Mice, Knockout , MicroRNAs , Myocytes, Cardiac , Animals , Endocardium/metabolism , Mice , MicroRNAs/metabolism , MicroRNAs/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Myocytes, Cardiac/metabolism , Transforming Growth Factor beta3/metabolism , Transforming Growth Factor beta3/genetics , Cell Proliferation , Myocardium/metabolism , Endothelial Cells/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Extracellular Matrix/metabolism , Female
12.
Bone Res ; 12(1): 32, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789434

ABSTRACT

Extracellular matrix (ECM) stiffening is a typical characteristic of cartilage aging, which is a quintessential feature of knee osteoarthritis (KOA). However, little is known about how ECM stiffening affects chondrocytes and other molecules downstream. This study mimicked the physiological and pathological stiffness of human cartilage using polydimethylsiloxane (PDMS) substrates. It demonstrated that epigenetic Parkin regulation by histone deacetylase 3 (HDAC3) represents a new mechanosensitive mechanism by which the stiffness matrix affected chondrocyte physiology. We found that ECM stiffening accelerated cultured chondrocyte senescence in vitro, while the stiffness ECM downregulated HDAC3, prompting Parkin acetylation to activate excessive mitophagy and accelerating chondrocyte senescence and osteoarthritis (OA) in mice. Contrarily, intra-articular injection with an HDAC3-expressing adeno-associated virus restored the young phenotype of the aged chondrocytes stimulated by ECM stiffening and alleviated OA in mice. The findings indicated that changes in the mechanical ECM properties initiated pathogenic mechanotransduction signals, promoted the Parkin acetylation and hyperactivated mitophagy, and damaged chondrocyte health. These results may provide new insights into chondrocyte regulation by the mechanical properties of ECM, suggesting that the modification of the physical ECM properties may be a potential OA treatment strategy.


Subject(s)
Cellular Senescence , Chondrocytes , Down-Regulation , Extracellular Matrix , Histone Deacetylases , Osteoarthritis , Animals , Chondrocytes/metabolism , Chondrocytes/pathology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Extracellular Matrix/metabolism , Osteoarthritis/pathology , Humans , Mice , Cellular Senescence/drug effects , Mice, Inbred C57BL , Mitophagy/drug effects , Male , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Acetylation , Cells, Cultured
13.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 40-47, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814236

ABSTRACT

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.


Subject(s)
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
14.
Nat Commun ; 15(1): 4322, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773107

ABSTRACT

Heterochromatin is generally associated with the nuclear periphery, but how the spatial organization of heterochromatin is regulated to ensure epigenetic silencing remains unclear. Here we found that Sad1, an inner nuclear membrane SUN-family protein in fission yeast, interacts with histone H2A-H2B but not H3-H4. We solved the crystal structure of the histone binding motif (HBM) of Sad1 in complex with H2A-H2B, revealing the intimate contacts between Sad1HBM and H2A-H2B. Structure-based mutagenesis studies revealed that the H2A-H2B-binding activity of Sad1 is required for the dynamic distribution of Sad1 throughout the nuclear envelope (NE). The Sad1-H2A-H2B complex mediates tethering telomeres and the mating-type locus to the NE. This complex is also important for heterochromatin silencing. Mechanistically, H2A-H2B enhances the interaction between Sad1 and HDACs, including Clr3 and Sir2, to maintain epigenetic identity of heterochromatin. Interestingly, our results suggest that Sad1 exhibits the histone-enhanced liquid-liquid phase separation property, which helps recruit heterochromatin factors to the NE. Our results uncover an unexpected role of SUN-family proteins in heterochromatin regulation and suggest a nucleosome-independent role of H2A-H2B in regulating Sad1's functionality.


Subject(s)
Heterochromatin , Histones , Protein Binding , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , Heterochromatin/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/chemistry , Histones/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces/genetics , Telomere/metabolism , Telomere/genetics , Nuclear Envelope/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Crystallography, X-Ray
15.
Arthritis Res Ther ; 26(1): 96, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711064

ABSTRACT

BACKGROUND: Gout is caused by monosodium urate (MSU) crystals deposition to trigger immune response. A recent study suggested that inhibition of Class I Histone deacetylases (HDACs) can significantly reduce MSU crystals-induced inflammation. However, which one of HDACs members in response to MSU crystals was still unknown. Here, we investigated the roles of HDAC3 in MSU crystals-induced gouty inflammation. METHODS: Macrophage specific HDAC3 knockout (KO) mice were used to investigate inflammatory profiles of gout in mouse models in vivo, including ankle arthritis, foot pad arthritis and subcutaneous air pouch model. In the in vitro experiments, bone marrow-derived macrophages (BMDMs) from mice were treated with MSU crystals to assess cytokines, potential target gene and protein. RESULTS: Deficiency of HDAC3 in macrophage not only reduced MSU-induced foot pad and ankle joint swelling but also decreased neutrophils trafficking and IL-1ß release in air pouch models. In addition, the levels of inflammatory genes related to TLR2/4/NF-κB/IL-6/STAT3 signaling pathway were significantly decreased in BMDMs from HDAC3 KO mice after MSU treatment. Moreover, RGFP966, selective inhibitor of HDAC3, inhibited IL-6 and TNF-α production in BMDMs treated with MSU crystals. Besides, HDAC3 deficiency shifted gene expression from pro-inflammatory macrophage (M1) to anti-inflammatory macrophage (M2) in BMDMs after MSU challenge. CONCLUSIONS: Deficiency of HDAC3 in macrophage alleviates MSU crystals-induced gouty inflammation through inhibition of TLR2/4 driven IL-6/STAT3 signaling pathway, suggesting that HDAC3 could contribute to a potential therapeutic target of gout.


Subject(s)
Acrylamides , Gout , Histone Deacetylases , Macrophages , Mice, Inbred C57BL , Mice, Knockout , Phenylenediamines , Uric Acid , Animals , Uric Acid/toxicity , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/deficiency , Macrophages/metabolism , Macrophages/drug effects , Gout/metabolism , Gout/pathology , Mice , Inflammation/metabolism , Inflammation/chemically induced , Male , Arthritis, Gouty/chemically induced , Arthritis, Gouty/metabolism , Arthritis, Gouty/pathology , Disease Models, Animal , Signal Transduction/drug effects
16.
Nat Commun ; 15(1): 4450, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789411

ABSTRACT

Histone deacetylases (HDACs) play a crucial role in transcriptional regulation and are implicated in various diseases, including cancer. They are involved in histone tail deacetylation and canonically linked to transcriptional repression. Previous studies suggested that HDAC recruitment to cell-cycle gene promoters via the retinoblastoma (RB) protein or the DREAM complex through SIN3B is essential for G1/S and G2/M gene repression during cell-cycle arrest and exit. Here we investigate the interplay among DREAM, RB, SIN3 proteins, and HDACs in the context of cell-cycle gene repression. Knockout of SIN3B does not globally derepress cell-cycle genes in non-proliferating HCT116 and C2C12 cells. Loss of SIN3A/B moderately upregulates several cell-cycle genes in HCT116 cells but does so independently of DREAM/RB. HDAC inhibition does not induce general upregulation of RB/DREAM target genes in arrested transformed or non-transformed cells. Our findings suggest that E2F:RB and DREAM complexes can repress cell-cycle genes without relying on HDAC activity.


Subject(s)
E2F Transcription Factors , Histone Deacetylases , Repressor Proteins , Retinoblastoma Protein , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , HCT116 Cells , Repressor Proteins/metabolism , Repressor Proteins/genetics , E2F Transcription Factors/metabolism , E2F Transcription Factors/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/genetics , Mice , Animals , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/genetics , Kv Channel-Interacting Proteins/metabolism , Kv Channel-Interacting Proteins/genetics , Cell Cycle/genetics , Promoter Regions, Genetic/genetics , Gene Expression Regulation , Genes, cdc
17.
Proc Natl Acad Sci U S A ; 121(18): e2312111121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38657041

ABSTRACT

Class II histone deacetylases (HDACs) are important in regulation of gene transcription during T cell development. However, our understanding of their cell-specific functions is limited. In this study, we reveal that class IIa Hdac4 and Hdac7 (Hdac4/7) are selectively induced in transcription, guiding the lineage-specific differentiation of mouse T-helper 17 (Th17) cells from naive CD4+ T cells. Importantly, Hdac4/7 are functionally dispensable in other Th subtypes. Mechanistically, Hdac4 interacts with the transcription factor (TF) JunB, facilitating the transcriptional activation of Th17 signature genes such as Il17a/f. Conversely, Hdac7 collaborates with the TF Aiolos and Smrt/Ncor1-Hdac3 corepressors to repress transcription of Th17 negative regulators, including Il2, in Th17 cell differentiation. Inhibiting Hdac4/7 through pharmacological or genetic methods effectively mitigates Th17 cell-mediated intestinal inflammation in a colitis mouse model. Our study uncovers molecular mechanisms where HDAC4 and HDAC7 function distinctively yet cooperatively in regulating ordered gene transcription during Th17 cell differentiation. These findings suggest a potential therapeutic strategy of targeting HDAC4/7 for treating Th17-related inflammatory diseases, such as ulcerative colitis.


Subject(s)
Cell Differentiation , Colitis , Histone Deacetylases , Nuclear Receptor Co-Repressor 1 , Th17 Cells , Animals , Th17 Cells/cytology , Th17 Cells/metabolism , Th17 Cells/immunology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice , Colitis/genetics , Colitis/metabolism , Colitis/immunology , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Interleukin-17/metabolism , Gene Expression Regulation , Mice, Inbred C57BL , Humans , Repressor Proteins/metabolism , Repressor Proteins/genetics , Interleukin-2/metabolism
18.
Nat Commun ; 15(1): 3635, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688903

ABSTRACT

Although intratumoral heterogeneity has been established in pediatric central nervous system tumors, epigenomic alterations at the cell type level have largely remained unresolved. To identify cell type-specific alterations to cytosine modifications in pediatric central nervous system tumors, we utilize a multi-omic approach that integrated bulk DNA cytosine modification data (methylation and hydroxymethylation) with both bulk and single-cell RNA-sequencing data. We demonstrate a large reduction in the scope of significantly differentially modified cytosines in tumors when accounting for tumor cell type composition. In the progenitor-like cell types of tumors, we identify a preponderance differential Cytosine-phosphate-Guanine site hydroxymethylation rather than methylation. Genes with differential hydroxymethylation, like histone deacetylase 4 and insulin-like growth factor 1 receptor, are associated with cell type-specific changes in gene expression in tumors. Our results highlight the importance of epigenomic alterations in the progenitor-like cell types and its role in cell type-specific transcriptional regulation in pediatric central nervous system tumors.


Subject(s)
Central Nervous System Neoplasms , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Child , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Epigenomics/methods , Repressor Proteins/metabolism , Repressor Proteins/genetics , Single-Cell Analysis , Transcription, Genetic , Cytosine/metabolism
19.
Exp Cell Res ; 438(2): 114050, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38663474

ABSTRACT

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.


Subject(s)
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
20.
Biomolecules ; 14(4)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38672510

ABSTRACT

Histone deacetylase (HDAC) 9 is a negative regulator of adipogenic differentiation, which is required for maintenance of healthy adipose tissues. We reported that HDAC9 expression is upregulated in adipose tissues during obesity, in conjunction with impaired adipogenic differentiation, adipocyte hypertrophy, insulin resistance, and hepatic steatosis, all of which were alleviated by global genetic deletion of Hdac9. Here, we developed a novel transgenic (TG) mouse model to test whether overexpression of Hdac9 is sufficient to induce adipocyte hypertrophy, insulin resistance, and hepatic steatosis in the absence of obesity. HDAC9 TG mice gained less body weight than wild-type (WT) mice when fed a standard laboratory diet for up to 40 weeks, which was attributed to reduced fat mass (primarily inguinal adipose tissue). There was no difference in insulin sensitivity or glucose tolerance in 18-week-old WT and HDAC9 TG mice; however, at 40 weeks of age, HDAC9 TG mice exhibited impaired insulin sensitivity and glucose intolerance. Tissue histology demonstrated adipocyte hypertrophy, along with reduced numbers of mature adipocytes and stromovascular cells, in the HDAC9 TG mouse adipose tissue. Moreover, increased lipids were detected in the livers of aging HDAC9 TG mice, as evaluated by oil red O staining. In conclusion, the experimental aging HDAC9 TG mice developed adipocyte hypertrophy, insulin resistance, and hepatic steatosis, independent of obesity. This novel mouse model may be useful in the investigation of the impact of Hdac9 overexpression associated with metabolic and aging-related diseases.


Subject(s)
Adipocytes , Fatty Liver , Histone Deacetylases , Insulin Resistance , Animals , Mice , Adipocytes/metabolism , Adipocytes/pathology , Aging/genetics , Aging/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Hypertrophy/genetics , Hypertrophy/metabolism , Insulin Resistance/genetics , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...