Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.147
Filter
1.
Int J Mol Sci ; 25(13)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-38999998

ABSTRACT

The information provided from the papers reviewed here about the role of epigenetics in chronic craniofacial neuropathic pain is critically important because epigenetic dysregulation during the development and maintenance of chronic neuropathic pain is not yet well characterized, particularly for craniofacial pain. We have noted that gene expression changes reported vary depending on the nerve injury model and the reported sample collection time point. At a truly chronic timepoint of 10 weeks in our model of chronic neuropathic pain, functional groupings of genes examined include those potentially contributing to anti-inflammation, nerve repair/regeneration, and nociception. Genes altered after treatment with the epigenetic modulator LMK235 are discussed. All of these differentials are key in working toward the development of diagnosis-targeted therapeutics and likely for the timing of when the treatment is provided. The emphasis on the relevance of time post-injury is reiterated here.


Subject(s)
Epigenesis, Genetic , Histone Deacetylases , Neuralgia , Neuralgia/genetics , Animals , Humans , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Chronic Pain/genetics , Facial Pain/genetics
2.
Mol Biol Rep ; 51(1): 824, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39023688

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a global life-threatening problem and therapeutic interventions are still encountered. IQGAP genes are involved in HCC oncogenesis. The modulatory effect of statins on the expression of IQGAP genes is still unclear. This study aims to study the effect of free SV and chitosan (CS) decorated simvastatin (SV) loaded solid lipid nanoparticles (C-SV-SLNs) on HCC mortality. METHODS AND RESULTS: Plain, SV-SLN, and C-SV- SLN were prepared and characterized in terms of particle size (PS), zeta potential (ZP), and polydispersity index (PDI). The biosafety of different SLN was investigated using fresh erythrocytes, moreover, cytotoxicity was investigated using HepG2 cell lines. The effect of SLNs on IQGAPs gene expression as well as JNK, HDAC6, and HDAC8 activity was investigated using PCR and MOE-docking. The current results displayed that SV-SLNs have nanosized, negative ZP and are homogenous, CS decoration shifts the ZP of SLN into cationic ZP. Furthermore, all SLNs exhibited desirable biosafety in terms of no deleterious effect on erythrocyte integrity. SV solution and SV-SLN significantly increase the mortality of HepG2 compared to undertreated cells, however, the effect of SV-SLN is more pronounced compared to free SV. Remarkably, C-SV-SLN elicits high HepG2 cell mortality compared to free SV and SV-SLN. The treatment of HepG2 cells with SV solution, SV-SLN, or C-SV-SLN significantly upregulates the IQGAP2 gene with repression of IQGAP1 and IQGAP3 genes. MOE-docking studies revealed both SV and tenivastatin exhibit interactions with the active sites of JNK, HDAC6, and HDAC8. Moreover, tenivastatin exhibited greater interactions with magnesium and zinc compared to SV. CONCLUSIONS: This research provides novel insights into the therapeutic potential of SV, SV-SLN and C-SV-SLNs in HCC treatment, modulating critical signaling cascades involving IQGAPs, JNK, and HDAC. The development of C-SV-SLNs presents a promising strategy for effective HCC therapy.


Subject(s)
Carcinoma, Hepatocellular , Chitosan , Histone Deacetylases , Liver Neoplasms , Nanoparticles , ras GTPase-Activating Proteins , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Hep G2 Cells , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Chitosan/pharmacology , ras GTPase-Activating Proteins/genetics , ras GTPase-Activating Proteins/metabolism , Nanoparticles/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Particle Size , Liposomes , Repressor Proteins
3.
FASEB J ; 38(13): e23819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38984942

ABSTRACT

Peritoneal dialysis is a common treatment for end-stage renal disease, but complications often force its discontinuation. Preventive treatments for peritoneal inflammation and fibrosis are currently lacking. Cyclo(His-Pro) (CHP), a naturally occurring cyclic dipeptide, has demonstrated protective effects in various fibrotic diseases, yet its potential role in peritoneal fibrosis (PF) remains uncertain. In a mouse model of induced PF, CHP was administered, and quantitative proteomic analysis using liquid chromatography-tandem mass spectrometry was employed to identify PF-related protein signaling pathways. The results were further validated using human primary cultured mesothelial cells. This analysis revealed the involvement of histone deacetylase 3 (HDAC3) in the PF signaling pathway. CHP administration effectively mitigated PF in both peritoneal tissue and human primary cultured mesothelial cells, concurrently regulating fibrosis-related markers and HDAC3 expression. Moreover, CHP enhanced the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) while suppressing forkhead box protein M1 (FOXM1), known to inhibit Nrf2 transcription through its interaction with HDAC3. CHP also displayed an impact on spleen myeloid-derived suppressor cells, suggesting an immunomodulatory effect. Notably, CHP improved mitochondrial function in peritoneal tissue, resulting in increased mitochondrial membrane potential and adenosine triphosphate production. This study suggests that CHP can significantly prevent PF in peritoneal dialysis patients by modulating HDAC3 expression and associated signaling pathways, reducing fibrosis and inflammation markers, and improving mitochondrial function.


Subject(s)
Histone Deacetylases , Peritoneal Fibrosis , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/prevention & control , Peritoneal Fibrosis/pathology , Mice , Humans , Male , Mice, Inbred C57BL , Signal Transduction/drug effects , Peritoneal Dialysis/adverse effects , Peritoneum/pathology , Peritoneum/metabolism
4.
FASEB J ; 38(13): e23794, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38967258

ABSTRACT

Obesity is often associated with low-grade inflammation. The incidence of obesity has increased annually worldwide, which seriously affects human health. A previous study indicated that long noncoding RNA SNHG12 was downregulated in obesity. Nevertheless, the role of SNHG12 in obesity remains to be elucidated. In this study, qRT-PCR, western blot, and ELISA were utilized to examine the gene and protein expression. Flow cytometry was employed to investigate the M2 macrophage markers. RNA pull-down assay and RIP were utilized to confirm the interactions of SNHG12, hnRNPA1, and HDAC9. Eventually, a high-fat diet-fed mouse model was established for in vivo studies. SNHG12 overexpression suppressed adipocyte inflammation and insulin resistance and promoted M2 polarization of macrophages that was caused by TNF-α treatment. SNHG12 interacted with hnRNPA1 to downregulate HDAC9 expression, which activated the Nrf2 signaling pathway. HDAC9 overexpression reversed the effect of SNHG12 overexpression on inflammatory response, insulin resistance, and M2 phenotype polarization. Overexpression of SNHG12 improved high-fat diet-fed mouse tissue inflammation. This study revealed the protective effect of SNHG12 against adipocyte inflammation and insulin resistance. This result further provides a new therapeutic target for preventing inflammation and insulin resistance in obesity.


Subject(s)
Adipocytes , Diet, High-Fat , Histone Deacetylases , Inflammation , Insulin Resistance , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Obesity , RNA, Long Noncoding , Repressor Proteins , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Inflammation/metabolism , Inflammation/genetics , Adipocytes/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Diet, High-Fat/adverse effects , Male , Obesity/metabolism , Obesity/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction , Macrophages/metabolism
5.
Nutrients ; 16(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38999793

ABSTRACT

The epigenetic regulation of nuclear factor erythroid 2-related factor 2 (Nrf2), a pivotal redox transcription factor, plays a crucial role in maintaining cellular homeostasis. Recent research has underscored the significance of epigenetic modifications of Nrf2 in the pathogenesis of diabetic foot ulcers (DFUs). This study investigates the epigenetic reversal of Nrf2 by pterostilbene (PTS) in human endothelial cells in a hyperglycemic microenvironment (HGM). The activation potential of PTS on Nrf2 was evaluated through ARE-Luciferase reporter assays and nuclear translocation studies. Following 72 h of exposure to an HGM, mRNA expression and protein levels of Nrf2 and its downstream targets NAD(P)H quinone oxidoreductase 1 (NQO1), heme-oxygenase 1(HO-1), superoxide dismutase (SOD), and catalase (CAT) exhibited a decrease, which was mitigated in PTS-pretreated endothelial cells. Epigenetic markers, including histone deacetylases (HDACs class I-IV) and DNA methyltransferases (DNMTs 1/3A and 3B), were found to be downregulated under diabetic conditions. Specifically, Nrf2-associated HDACs, including HDAC1, HDAC2, HDAC3, and HDAC4, were upregulated in HGM-induced endothelial cells. This upregulation was reversed in PTS-pretreated cells, except for HDAC2, which exhibited elevated expression in endothelial cells treated with PTS in a hyperglycemic microenvironment. Additionally, PTS was observed to reverse the activity of the methyltransferase enzyme DNMT. Furthermore, CpG islands in the Nrf2 promoter were hypermethylated in cells exposed to an HGM, a phenomenon potentially counteracted by PTS pretreatment, as shown by methyl-sensitive restriction enzyme PCR (MSRE-qPCR) analysis. Collectively, our findings highlight the ability of PTS to epigenetically regulate Nrf2 expression under hyperglycemic conditions, suggesting its therapeutic potential in managing diabetic complications.


Subject(s)
Antioxidants , Endothelial Cells , Epigenesis, Genetic , Hyperglycemia , NF-E2-Related Factor 2 , Stilbenes , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Humans , Epigenesis, Genetic/drug effects , Stilbenes/pharmacology , Hyperglycemia/metabolism , Antioxidants/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cellular Microenvironment/drug effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Gene Silencing , Oxidative Stress/drug effects , DNA Methylation/drug effects
6.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000021

ABSTRACT

Retinoblastoma, a pediatric ocular malignancy, presents significant challenges in comprehending its molecular underpinnings and targeted therapeutic approaches. The dysregulated activity of histone deacetylases (HDACs) has been associated with retinoblastoma pathogenesis, influencing critical cellular processes like cell cycle regulation or retinal ganglion cell apoptosis. Through their deacetylase activity, HDACs exert control over key tumor suppressors and oncogenes, influencing the delicate equilibrium between proliferation and cell death. Furthermore, the interplay between HDACs and the retinoblastoma protein pathway, a pivotal aspect of retinoblastoma etiology, reveals a complex network of interactions influencing the tumor microenvironment. The examination of HDAC inhibitors, encompassing both established and novel compounds, offers insights into potential approaches to restore acetylation balance and impede retinoblastoma progression. Moreover, the identification of specific HDAC isoforms exhibiting varying expression in retinoblastoma provides avenues for personalized therapeutic strategies, allowing for interventions tailored to individual patient profiles. This review focuses on the intricate interrelationship between HDACs and retinoblastoma, shedding light on epigenetic mechanisms that control tumor development and progression. The exploration of HDAC-targeted therapies underscores the potential for innovative treatment modalities in the pursuit of more efficacious and personalized management strategies for this disease.


Subject(s)
Histone Deacetylase Inhibitors , Histone Deacetylases , Retinoblastoma , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma/pathology , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Animals , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Epigenesis, Genetic , Acetylation , Tumor Microenvironment , Gene Expression Regulation, Neoplastic , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/genetics
7.
Chem Biol Drug Des ; 104(1): e14581, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38997237

ABSTRACT

N-heterocyclic compounds are important molecular scaffolds in the search for new drugs, since most drugs contain heterocyclic moieties in their molecular structure, and some of these classes of heterocycles are able to provide ligands for two or more biological targets. Ketene dithioacetals are important building blocks in organic synthesis and are widely used in the synthesis of N-heterocyclic compounds. In this work, we used double vinylic substitution reactions on ketene dithioacetals to synthesize a small library of heterocyclic derivatives and evaluated their cytotoxic activity in breast and ovarian cancer cells, identifying two benzoxazoles with good potency and selectivity. In silico predictions indicate that the two most active derivatives exhibit physicochemical properties within the range of drug-like compounds and showed potential to interact with HDAC8 and ERK1 cancer-related targets.


Subject(s)
Antineoplastic Agents , Ethylenes , Heterocyclic Compounds , Ketones , Humans , Cell Line, Tumor , Ethylenes/chemistry , Ethylenes/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Ketones/chemistry , Ketones/pharmacology , Ketones/chemical synthesis , Structure-Activity Relationship , Histone Deacetylases/metabolism , Molecular Docking Simulation , Drug Screening Assays, Antitumor , Acetals/chemistry , Acetals/pharmacology , Acetals/chemical synthesis , Repressor Proteins
8.
PLoS One ; 19(7): e0306168, 2024.
Article in English | MEDLINE | ID: mdl-39008483

ABSTRACT

Dual-targeting chromatin regulation and DNA damage repair signaling presents a promising avenue for cancer therapy. Applying rational drug design, we synthesized a potent dual-targeting small molecule, SP-1-303. Here, we report SP-1-303 as a class I isoform selective histone deacetylase (HDAC) inhibitor and an activator of the ataxia-telangiectasia mutated protein (ATM). In vitro enzymatic assays demonstrated selective inhibition of HDAC1 and HDAC3. Cellular growth inhibition studies show that SP-1-303 differentially inhibits growth of estrogen receptor positive breast cancer (ER+ BC) cells with effective growth inhibition concentrations (EC50) for MCF-7 and T47D cells ranging from 0.32 to 0.34 µM, compared to 1.2-2.5 µM for triple negative breast cancer cells, and ~12 µM for normal breast epithelial cells. Western analysis reveals that SP-1-303 decreases estrogen receptor alpha (ER-α) expression and increases p53 protein expression, while inducing the phosphorylation of ATM and its substrates, BRCA1 and p53, in a time-dependent manner in ER+ BC cells. Pharmacokinetic evaluation demonstrates an area under the curve (AUC) of 5227.55 ng/ml × h with an elimination half-life of 1.26 h following intravenous administration in a rat model. Collectively, SP-1-303 emerges as a novel second generation class I (HDAC1 and HDAC3) selective HDAC inhibitor, and ATM activator, capable of modulating ER expression, and inhibiting growth of ER+ BC cells. Combined targeting of class I HDACs and ATM by SP-1-303 offers a promising therapeutic approach for treating ER+ breast cancers and supports further preclinical evaluation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Breast Neoplasms , Cell Proliferation , Histone Deacetylase Inhibitors , Humans , Histone Deacetylase Inhibitors/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Female , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Rats , Cell Proliferation/drug effects , Histone Deacetylases/metabolism , Cell Line, Tumor , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , MCF-7 Cells , Estrogen Receptor alpha/metabolism , Receptors, Estrogen/metabolism
9.
BMC Genomics ; 25(1): 657, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956453

ABSTRACT

BACKGROUND: Histone deacetylases (HDACs) and histone acetyltransferases (HATs) are involved in plant growth and development as well as in response to environmental changes, by dynamically regulating gene acetylation levels. Although there have been numerous reports on the identification and function of HDAC and HAT in herbaceous plants, there are fewer report related genes in woody plants under drought stress. RESULTS: In this study, we performed a genome-wide analysis of the HDAC and HAT families in Populus trichocarpa, including phylogenetic analysis, gene structure, conserved domains, and expression analysis. A total of 16 PtrHDACs and 12 PtrHATs were identified in P. trichocarpa genome. Analysis of cis-elements in the promoters of PtrHDACs and PtrHATs revealed that both gene families could respond to a variety of environmental signals, including hormones and drought. Furthermore, real time quantitative PCR indicated that PtrHDA906 and PtrHAG3 were significantly responsive to drought. PtrHDA906, PtrHAC1, PtrHAC3, PtrHAG2, PtrHAG6 and PtrHAF1 consistently responded to abscisic acid, methyl jasmonate and salicylic acid under drought conditions. CONCLUSIONS: Our study demonstrates that PtrHDACs and PtrHATs may respond to drought through hormone signaling pathways, which helps to reveal the hub of acetylation modification in hormone regulation of abiotic stress.


Subject(s)
Droughts , Gene Expression Regulation, Plant , Histone Acetyltransferases , Histone Deacetylases , Phylogeny , Populus , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Populus/genetics , Populus/enzymology , Stress, Physiological/genetics , Gene Expression Profiling , Promoter Regions, Genetic , Genome, Plant , Plant Proteins/genetics , Plant Proteins/metabolism
10.
Respir Res ; 25(1): 263, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956592

ABSTRACT

BACKGROUND: Aberrant activation of macrophages is associated with pathogenesis of acute lung injury (ALI). However, the potential pathogenesis has not been explored. OBJECTIVES: We aimed to identify whether histone deacetylase (HDAC) 10 is involved in lipopolysaccharide (LPS)-exposed ALI and reveal the underlying pathogenesis by which it promotes lung inflammation in LPS-exposed ALI via modifying P62 with deacetylation. METHODS: We constructed an ALI mice model stimulated with LPS to determine the positive effect of Hdac10 deficiency. Moreover, we cultured murine alveolar macrophage cell line (MH-S cells) and primary bone marrow-derived macrophages (BMDMs) to explore the pro-inflammatory activity and mechanism of HDAC10 after LPS challenge. RESULTS: HDAC10 expression was increased both in mice lung tissues and macrophage cell lines and promoted inflammatory cytokines production exposed to LPS. Hdac10 deficiency inhibited autophagy and inflammatory response after LPS stimulation. In vivo, Hdac10fl/fl-LysMCre mice considerably attenuated lung inflammation and inflammatory cytokines release exposed to LPS. Mechanistically, HDAC10 interacts with P62 and mediates P62 deacetylation at lysine 165 (K165), by which it promotes P62 expression and increases inflammatory cytokines production. Importantly, we identified that Salvianolic acid B (SAB), an HDAC10 inhibitor, reduces lung inflammatory response in LPS-stimulated ALI. CONCLUSION: These results uncover a previously unknown role for HDAC10 in regulating P62 deacetylation and aggravating lung inflammation in LPS-induced ALI, implicating that targeting HDAC10 is an effective therapy for LPS-exposed ALI.


Subject(s)
Acute Lung Injury , Histone Deacetylases , Lipopolysaccharides , Lysine , Mice, Inbred C57BL , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Acute Lung Injury/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Lipopolysaccharides/toxicity , Mice , Acetylation , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/deficiency , Lysine/metabolism , Mice, Knockout , Male , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics , Myeloid Cells/metabolism
11.
J Neuroinflammation ; 21(1): 170, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997746

ABSTRACT

Ischemia-induced retinopathy is a hallmark finding of common visual disorders including diabetic retinopathy (DR) and central retinal artery and vein occlusions. Treatments for ischemic retinopathies fail to improve clinical outcomes and the design of new therapies will depend on understanding the underlying disease mechanisms. Histone deacetylases (HDACs) are an enzyme class that removes acetyl groups from histone and non-histone proteins, thereby regulating gene expression and protein function. HDACs have been implicated in retinal neurovascular injury in preclinical studies in which nonspecific HDAC inhibitors mitigated retinal injury. Histone deacetylase 3 (HDAC3) is a class I histone deacetylase isoform that plays a central role in the macrophage inflammatory response. We recently reported that myeloid cells upregulate HDAC3 in a mouse model of retinal ischemia-reperfusion (IR) injury. However, whether this cellular event is an essential contributor to retinal IR injury is unknown. In this study, we explored the role of myeloid HDAC3 in ischemia-induced retinal neurovascular injury by subjecting myeloid-specific HDAC3 knockout (M-HDAC3 KO) and floxed control mice to retinal IR. The M-HDAC3 KO mice were protected from retinal IR injury as shown by the preservation of inner retinal neurons, vascular integrity, and retinal thickness. Electroretinography confirmed that this neurovascular protection translated to improved retinal function. The retinas of M-HDAC3 KO mice also showed less proliferation and infiltration of myeloid cells after injury. Interestingly, myeloid cells lacking HDAC3 more avidly engulfed apoptotic cells in vitro and after retinal IR injury in vivo compared to wild-type myeloid cells, suggesting that HDAC3 hinders the reparative phagocytosis of dead cells, a process known as efferocytosis. Further mechanistic studies indicated that although HDAC3 KO macrophages upregulate the reparative enzyme arginase 1 (A1) that enhances efferocytosis, the inhibitory effect of HDAC3 on efferocytosis is not solely dependent on A1. Finally, treatment of wild-type mice with the HDAC3 inhibitor RGFP966 ameliorated the retinal neurodegeneration and thinning caused by IR injury. Collectively, our data show that HDAC3 deletion enhances macrophage-mediated efferocytosis and protects against retinal IR injury, suggesting that inhibiting myeloid HDAC3 holds promise as a novel therapeutic strategy for preserving retinal integrity after ischemic insult.


Subject(s)
Histone Deacetylases , Mice, Inbred C57BL , Mice, Knockout , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice , Myeloid Cells/metabolism , Phagocytosis/drug effects , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Diseases/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retina/metabolism , Retina/pathology , Efferocytosis
12.
Chem Pharm Bull (Tokyo) ; 72(7): 630-637, 2024.
Article in English | MEDLINE | ID: mdl-38945939

ABSTRACT

Alzheimer's disease (AD) is the leading cause of senile dementia, and the rapid increase in the frequency of AD cases has been attributed to population aging. However, current drugs have difficulty adequately suppressing symptoms and there is still a medical need for symptomatic agents. On the other hand, it has recently become clear that epigenetic dysfunctions are deeply involved in the development of cognitive impairments. Therefore, epigenetics-related proteins have attracted much attention as drug targets for AD. Early-developed epigenetic inhibitors were inappropriate for AD treatment because of their limited potential for oral administration, blood-brain barrier penetration, high target selectivity, and sufficient dose-limiting toxicity which are essential properties for small molecule drugs targeting chronic neurodegenerative diseases such as AD. In recent years, drug discovery studies have been actively performed to overcome such problems and several novel inhibitors targeting the epigenetics-related proteins are of interest as promising AD therapeutic agents. Here, we review the small molecule inhibitors of histone deacetylase (HDAC), lysine-specific demethylase 1 (LSD1) or bromodomains and extra-terminal domain (BET) protein, that enable memory function improvement in AD model mice.


Subject(s)
Alzheimer Disease , Epigenesis, Genetic , Histone Deacetylase Inhibitors , Histone Demethylases , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Epigenesis, Genetic/drug effects , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Histone Deacetylases/metabolism
13.
Mar Drugs ; 22(6)2024 May 28.
Article in English | MEDLINE | ID: mdl-38921561

ABSTRACT

Acute myeloid leukemia (AML) is a hematologic malignancy characterized by infiltration of the blood and bone marrow, exhibiting a low remission rate and high recurrence rate. Current research has demonstrated that class I HDAC inhibitors can downregulate anti-apoptotic proteins, leading to apoptosis of AML cells. In the present investigation, we conducted structural modifications of marine cytotoxin Santacruzamate A (SCA), a compound known for its inhibitory activity towards HDACs, resulting in the development of a novel series of potent class I HDACs hydrazide inhibitors. Representative hydrazide-based compound 25c exhibited concentration-dependent induction of apoptosis in AML cells as a single agent. Moreover, 25c exhibited a synergistic anti-AML effect when combined with Venetoclax, a clinical Bcl-2 inhibitor employed in AML therapy. This combination resulted in a more pronounced downregulation of anti-apoptotic proteins Mcl-1 and Bcl-xL, along with a significant upregulation of the pro-apoptotic protein cleaved-caspase3 and the DNA double-strand break biomarker γ-H2AX compared to monotherapy. These results highlighted the potential of 25c as a promising lead compound for AML treatment, particularly when used in combination with Venetoclax.


Subject(s)
Antineoplastic Agents , Apoptosis , Bridged Bicyclo Compounds, Heterocyclic , Drug Synergism , Histone Deacetylase Inhibitors , Leukemia, Myeloid, Acute , Sulfonamides , Humans , Sulfonamides/pharmacology , Sulfonamides/chemistry , Leukemia, Myeloid, Acute/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylases/metabolism , Animals , Caspase 3/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors
14.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928274

ABSTRACT

Epigenetic modulation, including histone modification, alters gene expression and controls cell fate. Histone deacetylases (HDACs) are identified as important regulators of dental pulp cell (DPC) mineralisation processes. Currently, there is a paucity of information regarding the nature of histone modification and HDAC expression in the dentine-pulp complex during dentinogenesis. The aim of this study was to investigate post-translational histone modulation and HDAC expression during DPC mineralisation and the expression of Class I/II HDACs during tooth development and in adult teeth. HDAC expression (isoforms -1 to -6) was analysed in mineralising primary rat DPCs using qRT-PCR and Western blot with mass spectrometry being used to analyse post-translational histone modifications. Maxillary molar teeth from postnatal and adult rats were analysed using immunohistochemical (IHC) staining for HDACs (1-6). HDAC-1, -2, and -4 protein expression increased until days 7 and 11, but decreased at days 14 and 21, while other HDAC expression increased continuously for 21 days. The Class II mineralisation-associated HDAC-4 was strongly expressed in postnatal sample odontoblasts and DPCs, but weakly in adult teeth, while other Class II HDACs (-5, -6) were relatively strongly expressed in postnatal DPCs and adult odontoblasts. Among Class I HDACs, HDAC-1 showed high expression in postnatal teeth, notably in ameloblasts and odontoblasts. HDAC-2 and -3 had extremely low expression in the rat dentine-pulp complex. Significant increases in acetylation were noted during DPC mineralisation processes, while trimethylation H3K9 and H3K27 marks decreased, and the HDAC-inhibitor suberoylanilide hydroxamic acid (SAHA) enhanced H3K27me3. These results highlight a dynamic alteration in histone acetylation during mineralisation and indicate the relevance of Class II HDAC expression in tooth development and regenerative processes.


Subject(s)
Dental Pulp , Dentin , Dentinogenesis , Histone Deacetylases , Animals , Acetylation , Rats , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Dentin/metabolism , Dental Pulp/metabolism , Dental Pulp/cytology , Dental Pulp/growth & development , Protein Processing, Post-Translational , Histones/metabolism , Molar/metabolism , Molar/growth & development , Odontoblasts/metabolism , Male
15.
Int J Mol Sci ; 25(12)2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38928463

ABSTRACT

The prevalence of dilated cardiomyopathy (DCM) is increasing globally, highlighting the need for innovative therapeutic approaches to prevent its onset. In this study, we examined the energetic and epigenetic distinctions between dilated and non-dilated human myocardium-derived mesenchymal stem/stromal cells (hmMSCs) and assessed the effects of class I and II HDAC inhibitors (HDACi) on these cells and their cardiomyogenic differentiation. Cells were isolated from myocardium biopsies using explant outgrowth methods. Mitochondrial and histone deacetylase activities, ATP levels, cardiac transcription factors, and structural proteins were assessed using flow cytometry, PCR, chemiluminescence, Western blotting, and immunohistochemistry. The data suggest that the tested HDAC inhibitors improved acetylation and enhanced the energetic status of both types of cells, with significant effects observed in dilated myocardium-derived hmMSCs. Additionally, the HDAC inhibitors activated the cardiac transcription factors Nkx2-5, HOPX, GATA4, and Mef2C, and upregulated structural proteins such as cardiac troponin T and alpha cardiac actin at both the protein and gene levels. In conclusion, our findings suggest that HDACi may serve as potential modulators of the energetic status and cardiomyogenic differentiation of human heart hmMSCs. This avenue of exploration could broaden the search for novel therapeutic interventions for dilated cardiomyopathy, ultimately leading to improvements in heart function.


Subject(s)
Cardiomyopathy, Dilated , Cell Differentiation , Histone Deacetylase Inhibitors , Mesenchymal Stem Cells , Humans , Histone Deacetylase Inhibitors/pharmacology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Differentiation/drug effects , Myocardium/cytology , Myocardium/metabolism , Myocardium/pathology , Histone Deacetylases/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , MEF2 Transcription Factors/metabolism , MEF2 Transcription Factors/genetics , Homeobox Protein Nkx-2.5/metabolism , Homeobox Protein Nkx-2.5/genetics , Acetylation/drug effects , Transcription Factors/metabolism , Transcription Factors/genetics , Cells, Cultured
16.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891780

ABSTRACT

The kinetics and mechanism of drug binding to its target are critical to pharmacological efficacy. A high throughput (HTS) screen often results in hundreds of hits, of which usually only simple IC50 values are determined during reconfirmation. However, kinetic parameters such as residence time for reversible inhibitors and the kinact/KI ratio, which is the critical measure for evaluating covalent inactivators, are early predictive measures to assess the chances of success of the hits in the clinic. Using the promising cancer target human histone deacetylase 8 as an example, we present a robust method that calculates concentration-dependent apparent rate constants for the inhibition or inactivation of HDAC8 from dose-response curves recorded after different pre-incubation times. With these data, hit compounds can be classified according to their mechanism of action, and the relevant kinetic parameters can be calculated in a highly parallel fashion. HDAC8 inhibitors with known modes of action were correctly assigned to their mechanism, and the binding mechanisms of some hits from an internal HDAC8 screening campaign were newly determined. The oxonitriles SVE04 and SVE27 were classified as fast reversible HDAC8 inhibitors with moderate time-constant IC50 values of 4.2 and 2.6 µM, respectively. The hit compound TJ-19-24 and SAH03 behave like slow two-step inactivators or reversible inhibitors, with a very low reverse isomerization rate.


Subject(s)
Histone Deacetylase Inhibitors , Histone Deacetylases , Repressor Proteins , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Kinetics , Repressor Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Protein Binding , High-Throughput Screening Assays/methods
17.
Plant Physiol Biochem ; 213: 108841, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38879987

ABSTRACT

Epigenetic modifications, such as histone alterations, play crucial roles in regulating the flowering process in Arabidopsis, a typical long-day model plant. Histone modifications are notably involved in the intricate regulation of FLC, a key inhibitor of flowering. Although sirtuin-like protein and NAD+-dependent deacetylases play an important role in regulating energy metabolism, plant stress responses, and hormonal signal transduction, the mechanisms underlying their developmental transitions remain unclear. Thus, this study aimed to reveal how Arabidopsis NAD + -dependent deacetylase AtSRT1 affects flowering by regulating the expression of flowering integrators. Genetic and molecular evidence demonstrated that AtSRT1 mediates histone deacetylation by directly binding near the transcriptional start sites (TSS) of the flowering integrator genes FT and SOC1 and negatively regulating their expression by modulating the expression of the downstream gene LFY to inhibit flowering. Additionally, AtSRT1 directly down-regulates the expression of TOR, a glucose-driven central hub of energy signaling, which controls cell metabolism and growth in response to nutritional and environmental factors. This down-regulation occurs through binding near the TSS of TOR, facilitating the addition of H3K27me3 marks on FLC via the TOR-FIE-PRC2 pathway, further repressing flowering. These results uncover a multi-pathway regulatory network involving deacetylase AtSRT1 during the flowering process, highlighting its interaction with TOR as a hub for the coordinated regulation of energy metabolism and flowering initiation. These findings significantly enhance understanding of the complexity of histone modifications in the regulation of flowering.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Flowers , Gene Expression Regulation, Plant , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis/growth & development , Arabidopsis/physiology , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Flowers/genetics , Flowers/growth & development , Signal Transduction , MADS Domain Proteins/metabolism , MADS Domain Proteins/genetics , Histones/metabolism , Energy Metabolism/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics
18.
Drug Discov Today ; 29(7): 104052, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38830501

ABSTRACT

Current treatment options for Alzheimer's disease (AD) focus on symptom relief rather than halting disease progression. In this context, targeting histone deacetylation emerges as a promising therapeutic alternative. Dysregulation of histone deacetylase (HDAC) activity is present in AD, contributing to cognitive decline. Pharmacological HDAC inhibition has shown benefits in preclinical models, namely reduced amyloid beta plaque formation, lower phosphorylation and aggregation of tau protein, greater microtubule stability, less neuroinflammation, and improved metabolic homeostasis and cell survival. Nonetheless, clinical trials evidenced limitations such as insufficient selectivity or blood-brain barrier penetration. Hence, future innovative strategies are required to enhance their efficacy/safety.


Subject(s)
Alzheimer Disease , Epigenome , Histone Deacetylase Inhibitors , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Humans , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Animals , Histone Deacetylases/metabolism
19.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892038

ABSTRACT

The effects of the enzyme N-acetylgalactosamine-4-sulfatase (Arylsulfatase B, ARSB), which removes the 4-sulfate group at the non-reducing end of chondroitin 4-sulfate, on the expression of PD-L1 were determined, and the underlying mechanism of PD-L1 expression was elucidated. Initial experiments in human melanoma cells (A375) showed that PD-L1 expression increased from 357 ± 31 to 796 ± 50 pg/mg protein (p < 10-11) when ARSB was silenced in A375 cells. In subcutaneous B16F10 murine melanomas, PD-L1 declined from 1227 ± 189 to 583 ± 110 pg/mg protein (p = 1.67 × 10-7), a decline of 52%, following treatment with exogenous, bioactive recombinant ARSB. This decline occurred in association with reduced tumor growth and prolongation of survival, as previously reported. The mechanism of regulation of PD-L1 expression by ARSB is attributed to ARSB-mediated alteration in chondroitin 4-sulfation, leading to changes in free galectin-3, c-Jun nuclear localization, HDAC3 expression, and effects of acetyl-H3 on the PD-L1 promoter. These findings indicate that changes in ARSB contribute to the expression of PD-L1 in melanoma and can thereby affect the immune checkpoint response. Exogenous ARSB acted on melanoma cells and normal melanocytes through the IGF2 receptor. The decline in PD-L1 expression by exogenous ARSB may contribute to the impact of ARSB on melanoma progression.


Subject(s)
B7-H1 Antigen , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Histone Deacetylases , Melanoma, Experimental , Melanoma , N-Acetylgalactosamine-4-Sulfatase , Animals , Humans , Mice , N-Acetylgalactosamine-4-Sulfatase/metabolism , N-Acetylgalactosamine-4-Sulfatase/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Cell Line, Tumor , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/genetics , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Galectin 3/metabolism , Galectin 3/genetics , Promoter Regions, Genetic , Blood Proteins , Galectins
20.
Front Immunol ; 15: 1369406, 2024.
Article in English | MEDLINE | ID: mdl-38835760

ABSTRACT

Epigenetic mechanisms are involved in several cellular functions, and their role in the immune system is of prime importance. Histone deacetylases (HDACs) are an important set of enzymes that regulate and catalyze the deacetylation process. HDACs have been proven beneficial targets for improving the efficacy of immunotherapies. HDAC11 is an enzyme involved in the negative regulation of T cell functions. Here, we investigated the potential of HDAC11 downregulation using RNA interference in CAR-T cells to improve immunotherapeutic outcomes against prostate cancer. We designed and tested four distinct short hairpin RNA (shRNA) sequences targeting HDAC11 to identify the most effective one for subsequent analyses. HDAC11-deficient CAR-T cells (shD-NKG2D-CAR-T) displayed better cytotoxicity than wild-type CAR-T cells against prostate cancer cell lines. This effect was attributed to enhanced activation, degranulation, and cytokine release ability of shD-NKG2D-CAR-T when co-cultured with prostate cancer cell lines. Our findings reveal that HDAC11 interference significantly enhances CAR-T cell proliferation, diminishes exhaustion markers PD-1 and TIM3, and promotes the formation of T central memory TCM populations. Further exploration into the underlying molecular mechanisms reveals increased expression of transcription factor Eomes, providing insight into the regulation of CAR-T cell differentiation. Finally, the shD-NKG2D-CAR-T cells provided efficient tumor control leading to improved survival of tumor-bearing mice in vivo as compared to their wild-type counterparts. The current study highlights the potential of HDAC11 downregulation in improving CAR-T cell therapy. The study will pave the way for further investigations focused on understanding and exploiting epigenetic mechanisms for immunotherapeutic outcomes.


Subject(s)
Histone Deacetylases , Immunotherapy, Adoptive , Prostatic Neoplasms , RNA, Small Interfering , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Prostatic Neoplasms/immunology , Humans , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Animals , Mice , RNA, Small Interfering/genetics , Cell Line, Tumor , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Gene Silencing , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL