Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.349
Filtrer
1.
Genome Biol ; 25(1): 165, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918851

RÉSUMÉ

BACKGROUND: Vascular endothelial growth factor (VEGF) is one of the most powerful proangiogenic factors and plays an important role in multiple diseases. Increased glycolytic rates and lactate accumulation are associated with pathological angiogenesis. RESULTS: Here, we show that a feedback loop between H3K9 lactylation (H3K9la) and histone deacetylase 2 (HDAC2) in endothelial cells drives VEGF-induced angiogenesis. We find that the H3K9la levels are upregulated in endothelial cells in response to VEGF stimulation. Pharmacological inhibition of glycolysis decreases H3K9 lactylation and attenuates neovascularization. CUT& Tag analysis reveals that H3K9la is enriched at the promoters of a set of angiogenic genes and promotes their transcription. Interestingly, we find that hyperlactylation of H3K9 inhibits expression of the lactylation eraser HDAC2, whereas overexpression of HDAC2 decreases H3K9 lactylation and suppresses angiogenesis. CONCLUSIONS: Collectively, our study illustrates that H3K9la is important for VEGF-induced angiogenesis, and interruption of the H3K9la/HDAC2 feedback loop may represent a novel therapeutic method for treating pathological neovascularization.


Sujet(s)
Rétrocontrôle physiologique , Histone Deacetylase 2 , Histone , Néovascularisation physiologique , Facteur de croissance endothéliale vasculaire de type A , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Histone/métabolisme , Humains , Animaux , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Souris , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Glycolyse , Néovascularisation pathologique/métabolisme ,
2.
Mol Cancer ; 23(1): 116, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38822351

RÉSUMÉ

BACKGROUND: Elevated evidence suggests that the SENPs family plays an important role in tumor progression. However, the role of SENPs in AML remains unclear. METHODS: We evaluated the expression pattern of SENP1 based on RNA sequencing data obtained from OHSU, TCGA, TARGET, and MILE datasets. Clinical samples were used to verify the expression of SENP1 in the AML cells. Lentiviral vectors shRNA and sgRNA were used to intervene in SENP1 expression in AML cells, and the effects of SENP1 on AML proliferation and anti-apoptosis were detected using in vitro and in vivo models. Chip-qPCR, MERIP-qPCR, CO-IP, RNA pulldown, and dual-luciferase reporter gene assays were used to explore the regulatory mechanisms of SNEP1 in AML. RESULTS: SENP1 was significantly upregulated in high-risk AML patients and closely related to poor prognosis. The AKT/mTOR signaling pathway is a key downstream pathway that mediates SENP1's regulation of AML proliferation and anti-apoptosis. Mechanistically, the CO-IP assay revealed binding between SENP1 and HDAC2. SUMO and Chip-qPCR assays suggested that SENP1 can desumoylate HDAC2, which enhances EGFR transcription and activates the AKT pathway. In addition, we found that IGF2BP3 expression was upregulated in high-risk AML patients and was positively correlated with SENP1 expression. MERIP-qPCR and RIP-qPCR showed that IGF2BP3 binds SENP1 3-UTR in an m6A manner, enhances SENP1 expression, and promotes AKT pathway conduction. CONCLUSIONS: Our findings reveal a distinct mechanism of SENP1-mediated HDAC2-AKT activation and establish the critical role of the IGF2BP3/SENP1signaling axis in AML development.


Sujet(s)
Adénosine , Prolifération cellulaire , Cysteine endopeptidases , Histone Deacetylase 2 , Leucémie aigüe myéloïde , Protéines proto-oncogènes c-akt , Protéines de liaison à l'ARN , Sumoylation , Animaux , Femelle , Humains , Mâle , Souris , Adénosine/analogues et dérivés , Adénosine/métabolisme , Apoptose , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Lignée cellulaire tumorale , Cysteine endopeptidases/métabolisme , Cysteine endopeptidases/génétique , Évolution de la maladie , Régulation de l'expression des gènes dans la leucémie , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Pronostic , Protéines proto-oncogènes c-akt/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Transduction du signal , Tests d'activité antitumorale sur modèle de xénogreffe
3.
FASEB J ; 38(12): e23736, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38865202

RÉSUMÉ

Subclinical hypothyroidism (SCH) in pregnancy is the most common form of thyroid dysfunction in pregnancy, which can affect fetal nervous system development and increase the risk of neurodevelopmental disorders after birth. However, the mechanism of the effect of maternal subclinical hypothyroidism on fetal brain development and behavioral phenotypes is still unclear and requires further study. In this study, we constructed a mouse model of maternal subclinical hypothyroidism by exposing dams to drinking water containing 50 ppm propylthiouracil (PTU) during pregnancy and found that its offspring were accompanied by severe cognitive deficits by behavioral testing. Mechanistically, gestational SCH resulted in the upregulation of protein expression and activity of HDAC1/2/3 in the hippocampus of the offspring. ChIP analysis revealed that H3K9ac on the neurogranin (Ng) promoter was reduced in the hippocampus of the offspring of SCH, with a significant reduction in Ng protein, leading to reduced expression levels of synaptic plasticity markers PSD95 (a membrane-associated protein in the postsynaptic density) and SYN (synaptophysin, a specific marker for presynaptic terminals), and impaired synaptic plasticity. In addition, administration of MS-275 (an HDAC1/2/3-specific inhibitor) to SCH offspring alleviated impaired synaptic plasticity and cognitive dysfunction in offspring. Thus, our study suggests that maternal subclinical hypothyroidism may mediate offspring cognitive dysfunction through the HDAC1/2/3-H3K9ac-Ng pathway. Our study contributes to the understanding of the signaling mechanisms underlying maternal subclinical hypothyroidism-mediated cognitive impairment in the offspring.


Sujet(s)
Dysfonctionnement cognitif , Histone Deacetylase 1 , Histone Deacetylase 2 , Hypothyroïdie , Neurogranine , Effets différés de l'exposition prénatale à des facteurs de risque , Animaux , Neurogranine/métabolisme , Neurogranine/génétique , Hypothyroïdie/métabolisme , Femelle , Grossesse , Souris , Dysfonctionnement cognitif/métabolisme , Dysfonctionnement cognitif/étiologie , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Régulation négative , Hippocampe/métabolisme , Mâle , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Souris de lignée C57BL , Plasticité neuronale
4.
Cell Rep ; 43(6): 114308, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38829740

RÉSUMÉ

Dendritic cell (DC) progenitors adapt their transcriptional program during development, generating different subsets. How chromatin modifications modulate these processes is unclear. Here, we investigate the impact of histone deacetylation on DCs by genetically deleting histone deacetylase 1 (HDAC1) or HDAC2 in hematopoietic progenitors and CD11c-expressing cells. While HDAC2 is not critical for DC development, HDAC1 deletion impairs pro-pDC and mature pDC generation and affects ESAM+cDC2 differentiation from tDCs and pre-cDC2s, whereas cDC1s are unchanged. HDAC1 knockdown in human hematopoietic cells also impairs cDC2 development, highlighting its crucial role across species. Multi-omics analyses reveal that HDAC1 controls expression, chromatin accessibility, and histone acetylation of the transcription factors IRF4, IRF8, and SPIB required for efficient development of cDC2 subsets. Without HDAC1, DCs switch immunologically, enhancing tumor surveillance through increased cDC1 maturation and interleukin-12 production, driving T helper 1-mediated immunity and CD8+ T cell recruitment. Our study reveals the importance of histone acetylation in DC development and anti-tumor immunity, suggesting DC-targeted therapeutic strategies for immuno-oncology.


Sujet(s)
Différenciation cellulaire , Cellules dendritiques , Histone Deacetylase 1 , Cellules dendritiques/métabolisme , Cellules dendritiques/immunologie , Histone Deacetylase 1/métabolisme , Animaux , Humains , Souris , Souris de lignée C57BL , Acétylation , Tumeurs/immunologie , Tumeurs/anatomopathologie , Histone/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Histone Deacetylase 2/métabolisme , Interleukine-12/métabolisme
5.
Hum Genet ; 143(6): 747-759, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38753158

RÉSUMÉ

Histone deacetylases (HDACs) are enzymes pivotal for histone modification (i.e. acetylation marks removal), chromatin accessibility and gene expression regulation. Class I HDACs (including HDAC1, 2, 3, 8) are ubiquitously expressed and they often participate in multi-molecular protein complexes. To date, three neurodevelopmental disorders caused by mutations in genes encoding for HDACs (HDAC4, HDAC6 and HDAC8) and thus belonging to the group of chromatinopathies, have been described. We performed whole exome sequencing (WES) for a patient (#249) clinically diagnosed with the chromatinopathy Rubinstein-Taybi syndrome (RSTS) but negative for mutations in RSTS genes, identifying a de novo frameshift variant in HDAC2 gene. We then investigated its molecular effects in lymphoblastoid cell lines (LCLs) derived from the patient compared to LCLs from healthy donors (HD). As the variant was predicted to be likely pathogenetic and to affect the sequence of nuclear localization signal, we performed immunocytochemistry and lysates fractionation, observing a nuclear mis-localization of HDAC2 compared to HD LCLs. In addition, HDAC2 total protein abundance resulted altered in patient, and we found that newly identified variant in HDAC2 affects also acetylation levels, with significant difference in acetylation pattern among patient #249, HD and RSTS cells and in expression of a known molecular target. Remarkably, RNA-seq performed on #249, HD and RSTS cells shows differentially expressed genes (DEGs) common to #249 and RSTS. Interestingly, our reported patient was clinically diagnosed with RSTS, a chromatinopathy which known causative genes encode for enzymes antagonizing HDACs. These results support the role of HDAC2 as causative gene for chromatinopathies, strengthening the genotype-phenotype correlations in this relevant group of disorders.


Sujet(s)
, Histone Deacetylase 2 , Humains , Histone Deacetylase 2/génétique , Histone Deacetylase 2/métabolisme , Acétylation , Syndrome de Rubinstein-Taybi/génétique , Syndrome de Rubinstein-Taybi/anatomopathologie , Chromatine/génétique , Chromatine/métabolisme , Mâle , Femelle , Mutation , Mutation avec décalage du cadre de lecture , Lignée cellulaire
6.
Eur J Pharmacol ; 975: 176643, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38754539

RÉSUMÉ

Chronic diabetes mellitus is reported to be associated with acute kidney injury. The enzyme histone deacetylase-2 (HDAC-2) was found to be upregulated in diabetes-related kidney damage. Alpha-cyperone (α-CYP) is one of the active ingredients of Cyperus rotundus that possesses antioxidant and anti-inflammatory effects. We evaluated the effect of α-CYP on improving oxidative stress and tissue inflammation following renal ischemia/reperfusion (I/R) injury in diabetic rats. The effect of α-CYP on HDAC-2 expression in renal homogenates and in the NRK-52 E cell line was evaluated following renal I/R injury and high glucose conditions, respectively. Molecular docking was used to investigate the binding of α-CYP with the HDAC-2 active site. Both renal function and oxidative stress were shown to be impaired in diabetic rats due to renal I/R injury. Significant improvements in kidney/body weight ratio, creatinine clearance, serum creatinine, blood urea nitrogen (BUN), and uric acid were observed in diabetic rats treated with α-CYP (50 mg/kg) two weeks prior to renal I/R injury. α-CYP treatment also improved histological alterations in renal tissue and lowered levels of malondialdehyde, myeloperoxidase, and hydroxyproline. Treatment with α-CYP suppressed the increased HDAC-2 expression in the renal tissue of diabetic rats and in the NRK-52 E cell line. The molecular docking reveals that α-CYP binds to HDAC-2 with good affinity, ascertained by molecular dynamics simulations and binding free energy analysis. Overall, our data suggest that α-CYP can effectively prevent renal injury in diabetic rats by regulating oxidative stress, tissue inflammation, fibrosis and inhibiting HDAC-2 activity.


Sujet(s)
Diabète expérimental , Histone Deacetylase 2 , Rein , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Lésion d'ischémie-reperfusion , Animaux , Histone Deacetylase 2/métabolisme , Mâle , Rats , Diabète expérimental/traitement médicamenteux , Diabète expérimental/complications , Diabète expérimental/métabolisme , Diabète expérimental/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Rein/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Lignée cellulaire , Néphropathies diabétiques/traitement médicamenteux , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/métabolisme , Rat Wistar
7.
ACS Chem Neurosci ; 15(11): 2099-2111, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38747979

RÉSUMÉ

Despite recent FDA approvals, Alzheimer's disease (AD) still represents an unmet medical need. Among the different available therapeutic approaches, the development of multitarget molecules represents one of the most widely pursued. In this work, we present a second generation of dual ligands directed toward highly networked targets that are deeply involved in the development of the disease, namely, Histone Deacetylases (HDACs) and Glycogen Synthase Kinase 3ß (GSK-3ß). The synthesized compounds are highly potent GSK-3ß, HDAC2, and HDAC6 inhibitors with IC50 values in the nanomolar range of concentrations. Among them, compound 4 inhibits histone H3 and tubulin acetylation at 0.1 µM concentration, blocks hyperphosphorylation of tau protein, and shows interesting immunomodulatory and neuroprotective properties. These features, together with its ability to cross the blood-brain barrier and its favorable physical-chemical properties, make compound 4 a promising hit for the development of innovative disease-modifying agents.


Sujet(s)
Maladie d'Alzheimer , Glycogen synthase kinase 3 beta , Inhibiteurs de désacétylase d'histone , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Humains , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/antagonistes et inhibiteurs , Histone deacetylase 6/antagonistes et inhibiteurs , Histone deacetylase 6/métabolisme , Animaux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Protéines tau/métabolisme , Histone deacetylases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Acétylation , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/antagonistes et inhibiteurs
8.
Clin Transl Med ; 14(6): e1692, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38804602

RÉSUMÉ

BACKGROUND: Although numerous studies have indicated that activated pyroptosis can enhance the efficacy of antitumour therapy in several tumours, the precise mechanism of pyroptosis in colorectal cancer (CRC) remains unclear. METHODS: Pyroptosis in CRC cells treated with antitumour agents was assessed using various techniques, including Western blotting, lactate dehydrogenase release assay and microscopy analysis. To uncover the epigenetic mechanisms that regulate NLRP3, chromatin changes and NLRP3 promoter histone modifications were assessed using Assay for Transposase-Accessible Chromatin using sequencing and RNA sequencing. Chromatin immunoprecipitation‒quantitative polymerase chain reaction was used to investigate the NLRP3 transcriptional regulatory mechanism. Additionally, xenograft and patient-derived xenograft models were constructed to validate the effects of the drug combinations. RESULTS: As the core molecule of the inflammasome, NLRP3 expression was silenced in CRC, thereby limiting gasdermin D (GSDMD)-mediated pyroptosis. Supplementation with NLRP3 can rescue pyroptosis induced by antitumour therapy. Overexpression of HDAC2 in CRC silences NLRP3 via epigenetic regulation. Mechanistically, HDAC2 suppressed chromatin accessibility by eliminating H3K27 acetylation. HDAC2 knockout promotes H3K27ac-mediated recruitment of the BRD4-p-P65 complex to enhance NLRP3 transcription. Inhibiting HDAC2 by Santacruzamate A in combination with classic antitumour agents (5-fluorouracil or regorafenib) in CRC xenograft-bearing animals markedly activated pyroptosis and achieved a significant therapeutic effect. Clinically, HDAC2 is inversely correlated with H3K27ac/p-P65/NLRP3 and is a prognostic factor for CRC patients. CONCLUSION: Collectively, our data revealed a crucial role for HDAC2 in inhibiting NLRP3/GSDMD-mediated pyroptosis in CRC cells and highlighted HDAC2 as a potential therapeutic target for antitumour therapy. HIGHLIGHTS: Silencing of NLRP3 limits the GSDMD-dependent pyroptosis in colorectal cancer. HDAC2-mediated histone deacetylation leads to epigenetic silencing of NLRP3. HDAC2 suppresses the NLRP3 transcription by inhibiting the formation of H3K27ac/BRD4/p-P65 complex. Targeting HDAC2 activates pyroptosis and enhances therapeutic effect.


Sujet(s)
Tumeurs colorectales , Histone Deacetylase 2 , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Pyroptose/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Souris , Animaux , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Gasdermines , Protéines de liaison aux phosphates
9.
Viruses ; 16(5)2024 04 25.
Article de Anglais | MEDLINE | ID: mdl-38793560

RÉSUMÉ

Porcine reproductive and respiratory syndrome virus (PRRSV), a member of the Arteriviridae family, represents a persistent menace to the global pig industry, causing reproductive failure and respiratory disease in pigs. In this study, we delved into the role of histone deacetylases (HDAC2) during PRRSV infection. Our findings revealed that HDAC2 expression is downregulated upon PRRSV infection. Notably, suppressing HDAC2 activity through specific small interfering RNA led to an increase in virus production, whereas overexpressing HDAC2 effectively inhibited PRRSV replication by boosting the expression of IFN-regulated antiviral molecules. Furthermore, we identified the virus's nonstructural protein 11 (nsp11) as a key player in reducing HDAC2 levels. Mutagenic analyses of PRRSV nsp11 revealed that its antagonistic effect on the antiviral activity of HDAC2 is dependent on its endonuclease activity. In summary, our research uncovered a novel immune evasion mechanism employed by PRRSV, providing crucial insights into the pathogenesis of this virus and guiding the development of innovative prevention strategies against PRRSV infection.


Sujet(s)
Endoribonucleases , Histone Deacetylase 2 , Échappement immunitaire , Immunité innée , Syndrome dysgénésique et respiratoire porcin , Virus du syndrome respiratoire et reproducteur porcin , Protéines virales non structurales , Réplication virale , Virus du syndrome respiratoire et reproducteur porcin/immunologie , Virus du syndrome respiratoire et reproducteur porcin/génétique , Animaux , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique , Suidae , Syndrome dysgénésique et respiratoire porcin/virologie , Syndrome dysgénésique et respiratoire porcin/immunologie , Endoribonucleases/métabolisme , Endoribonucleases/génétique , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Lignée cellulaire , Humains
10.
Adv Sci (Weinh) ; 11(24): e2306810, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38647380

RÉSUMÉ

Persistent transcription of HBV covalently closed circular DNA (cccDNA) is critical for chronic HBV infection. Silencing cccDNA transcription through epigenetic mechanisms offers an effective strategy to control HBV. Long non-coding RNAs (lncRNAs), as important epigenetic regulators, have an unclear role in cccDNA transcription regulation. In this study, lncRNA sequencing (lncRNA seq) is conducted on five pairs of HBV-positive and HBV-negative liver tissue. Through analysis, HOXA-AS2 (HOXA cluster antisense RNA 2) is identified as a significantly upregulated lncRNA in HBV-infected livers. Further experiments demonstrate that HBV DNA polymerase (DNA pol) induces HOXA-AS2 after establishing persistent high-level HBV replication. Functional studies reveal that HOXA-AS2 physically binds to cccDNA and significantly inhibits its transcription. Mechanistically, HOXA-AS2 recruits the MTA1-HDAC1/2 deacetylase complex to cccDNA minichromosome by physically interacting with metastasis associated 1 (MTA1) subunit, resulting in reduced acetylation of histone H3 at lysine 9 (H3K9ac) and lysine 27 (H3K27ac) associated with cccDNA and subsequently suppressing cccDNA transcription. Altogether, the study reveals a mechanism to self-limit HBV replication, wherein the upregulation of lncRNA HOXA-AS2, induced by HBV DNA pol, can epigenetically suppress cccDNA transcription.


Sujet(s)
ADN circulaire , Épigenèse génétique , Virus de l'hépatite B , ARN long non codant , Protéines de répression , Transactivateurs , Humains , Virus de l'hépatite B/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Transactivateurs/génétique , Transactivateurs/métabolisme , Épigenèse génétique/génétique , ADN circulaire/génétique , ADN circulaire/métabolisme , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Histone Deacetylase 2/génétique , Histone Deacetylase 2/métabolisme , Transcription génétique/génétique , Hépatite B chronique/génétique , Hépatite B chronique/métabolisme , Hépatite B chronique/virologie
11.
Ocul Surf ; 33: 39-49, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38679196

RÉSUMÉ

PURPOSE: To investigate the roles of HDAC1/2 and HDAC3 in adult Meibomian gland (MG) homeostasis. METHODS: HDAC1/2 or HDAC3 were inducibly deleted in MG epithelial cells of adult mice. The morphology of MG was examined. Proliferation, apoptosis, and expression of MG acinus and duct marker genes, meibocyte differentiation genes, and HDAC target genes, were analyzed via immunofluorescence, TUNEL assay, and RNA in situ hybridization. RESULTS: Co-deletion of HDAC1/2 in MG epithelium caused gradual loss of acini and formation of cyst-like structures in the central duct. These phenotypes required homozygous deletion of both HDAC1 and HDAC2, indicating that they function redundantly in the adult MG. Short-term deletion of HDAC1/2 in MG epithelium had little effect on meibocyte maturation but caused decreased proliferation of acinar basal cells, excessive DNA damage, ectopic apoptosis, and increased p53 acetylation and p16 expression in the MG. By contrast, HDAC3 deletion in MG epithelium caused dilation of central duct, atrophy of acini, defective meibocyte maturation, increased acinar basal cell proliferation, and ectopic apoptosis and DNA damage. Levels of p53 acetylation and p21 expression were elevated in HDAC3-deficient MGs, while the expression of the differentiation regulator PPARγ and the differentiation markers PLIN2 and FASN was downregulated. CONCLUSIONS: HDAC1 and HDAC2 function redundantly in adult Meibomian gland epithelial progenitor cells and are essential for their proliferation and survival, but not for acinar differentiation, while HDAC3 is required to limit acinar progenitor cell proliferation and permit differentiation. HDAC1/2 and HDAC3 have partially overlapping roles in maintaining survival of MG cells.


Sujet(s)
Apoptose , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone deacetylases , Homéostasie , Glandes de Meibomius , Animaux , Glandes de Meibomius/métabolisme , Glandes de Meibomius/anatomopathologie , Souris , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Homéostasie/physiologie , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Prolifération cellulaire/physiologie , Méthode TUNEL , Hybridation in situ , Différenciation cellulaire/physiologie
12.
Food Funct ; 15(9): 5103-5117, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38680105

RÉSUMÉ

Hydroxytyrosol (HT), a phenolic extra-virgin olive oil compound used as a food supplement, has been recognized to protect liver function and alleviate stress-induced depressive-like behaviors. However, its protective effects against stress-induced liver injury (SLI) remain unknown. Here, the anti-SLI effect of HT was evaluated in mice with chronic unpredictable mild stress-induced SLI. Network pharmacology combined with molecular docking was used to clarify the underlying mechanism of action of HT against SLI, followed by experimental verification. The results showed that accompanying with the alleviation of HT on stress-induced depressive-like behaviors, HT was confirmed to exert the protective effects against SLI, as represented by reduced serum corticosterone (CORT), aspartate aminotransferase and alanine aminotransferase activities, as well as repair of liver structure, inhibition of oxidative homeostasis collapse, and inflammation reaction in the liver. Furthermore, core genes including histone deacetylase 1 and 2 (HDAC1/2), were identified as potential targets of HT in SLI based on bioinformatic screening and simulation. Consistently, HT significantly inhibited HDAC1/2 expression to maintain mitochondrial dysfunction in an autophagy-dependent manner, which was confirmed in a CORT-induced AML-12 cell injury and SLI mice models combined with small molecule inhibitors. We provide the first evidence that HT inhibits HDAC1/2 to induce autophagy in hepatocytes for maintaining mitochondrial dysfunction, thus preventing inflammation and oxidative stress for exerting an anti-SLI effect. This constitutes a novel therapeutic modality to synchronously prevent stress-induced depression-like behaviors and liver injury, supporting the advantaged therapeutic potential of HT.


Sujet(s)
Autophagie , Histone Deacetylase 2 , Alcool phénéthylique , Alcool phénéthylique/analogues et dérivés , Animaux , Souris , Alcool phénéthylique/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Mâle , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Souris de lignée C57BL , Histone Deacetylase 1/métabolisme , Simulation de docking moléculaire , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Stress psychologique/traitement médicamenteux , Stress psychologique/complications
13.
J Cell Sci ; 137(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38682259

RÉSUMÉ

SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.


Sujet(s)
Antigènes d'histocompatibilité de classe II , Histone Deacetylase 2 , Protéines nucléaires , Régions promotrices (génétique) , SARS-CoV-2 , Transactivateurs , Humains , Présentation d'antigène/génétique , Cellules présentatrices d'antigène/métabolisme , Cellules présentatrices d'antigène/immunologie , COVID-19/virologie , COVID-19/immunologie , COVID-19/génétique , COVID-19/métabolisme , Cysteine endopeptidases/métabolisme , Cysteine endopeptidases/génétique , Régulation négative/génétique , Cellules HEK293 , Antigènes d'histocompatibilité de classe II/métabolisme , Antigènes d'histocompatibilité de classe II/génétique , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Facteur-3 de régulation d'interféron/métabolisme , Facteur-3 de régulation d'interféron/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Régions promotrices (génétique)/génétique , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , SARS-CoV-2/immunologie , Transactivateurs/métabolisme , Transactivateurs/génétique , Protéines virales non structurales/métabolisme , Protéines virales non structurales/génétique
14.
Sci Rep ; 14(1): 9897, 2024 04 30.
Article de Anglais | MEDLINE | ID: mdl-38688962

RÉSUMÉ

Alzheimer's disease (AD) is associated with cognitive deficits and epigenetic deacetylation that can be modulated by natural products. The role of natural oxyresveratrol-ß-cyclodextrin (ORV) on cognition and histone deacetylase activity in AD is unclear. Herein, in-silico docking and molecular dynamics simulation analysis determined that oxyresveratrol potentially targets histone deacetylase-2 (HDAC2). We therefore evaluated the in vivo ameliorative effect of ORV against cognitive deficit, cerebral and hippocampal expression of HDAC in experimental AD rats. Intracerebroventricular injection of STZ (3 mg/kg) induced experimental AD and the rats were treated with low dose (200 mg/kg), high dose (400 mg/kg) of ORV and donepezil (10 mg/kg) for 21 days. The STZ-induced AD caused cognitive and behavioural deficits demonstrated by considerable increases in acetylcholinesterase activity and escape latency compared to sham control. The levels of malondialdehyde (MDA) and HDAC activity were significantly increased in AD disease group comparison to the sham. Interestingly, the ORV reversed the cognitive-behavioural deficit and prominently reduced the MDA and HDAC levels comparable to the effect of the standard drug, donepezil. The findings suggest anti-AD role of ORV via antioxidant effect and inhibition of HDAC in the hippocampal and frontal cortical area of rats for AD.


Sujet(s)
Maladie d'Alzheimer , Dysfonctionnement cognitif , Modèles animaux de maladie humaine , Histone Deacetylase 2 , Extraits de plantes , Stilbènes , Streptozocine , Animaux , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Rats , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/métabolisme , Stilbènes/pharmacologie , Stilbènes/usage thérapeutique , Mâle , Histone Deacetylase 2/métabolisme , Cyclodextrines bêta/pharmacologie , Simulation de docking moléculaire , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Malonaldéhyde/métabolisme , Donépézil/pharmacologie , Donépézil/usage thérapeutique , Simulation de dynamique moléculaire , Rat Wistar
15.
Mol Cancer ; 23(1): 85, 2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38678233

RÉSUMÉ

Nuclear condensates have been shown to regulate cell fate control, but its role in oncogenic transformation remains largely unknown. Here we show acquisition of oncogenic potential by nuclear condensate remodeling. The proto-oncogene SS18 and its oncogenic fusion SS18-SSX1 can both form condensates, but with drastically different properties and impact on 3D genome architecture. The oncogenic condensates, not wild type ones, readily exclude HDAC1 and 2 complexes, thus, allowing aberrant accumulation of H3K27ac on chromatin loci, leading to oncogenic expression of key target genes. These results provide the first case for condensate remodeling as a transforming event to generate oncogene and such condensates can be targeted for therapy. One sentence summary: Expulsion of HDACs complexes leads to oncogenic transformation.


Sujet(s)
Histone Deacetylase 1 , Histone Deacetylase 2 , Proto-oncogène Mas , Humains , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Noyau de la cellule/métabolisme , Chromatine/métabolisme , Chromatine/génétique , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Histone/métabolisme , Animaux
16.
J Affect Disord ; 360: 305-313, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38395201

RÉSUMÉ

BACKGROUND: Depression and chronic pain frequent co-occur, exacerbating each other's symptoms and hindering treatment. Emerging studies have highlighted abnormal gut microbiota in both conditions. Previous studies have demonstrated the clinical effectiveness of electro-acupuncture (EA) in managing these conditions, yet the underlying mechanisms remain elusive. METHODS: Spared nerve injury (SNI) was employed to induce chronic pain and depression-like behavior. Rats were randomly assigned to sham SNI (SS), SNI, and EA groups. SNI surgery was performed on all rats, except those in SS group, which underwent sham SNI surgery. Then EA group received 5 weeks of EA treatment. Pain and depression-like behavior were assessed through paw withdrawal threshold, sucrose-preference test, and forced swim test. Gut microbiota composition was analyzed via 16S rDNA sequencing. Brain-Derived Neurotrophic Factor (BDNF) and acetylation-related proteins in the medial prefrontal cortex (mPFC) were evaluated through enzyme-linked immunosorbent assay and western blot. RESULTS: EA treatment significantly ameliorated pain and depression-like behavior. The 16S rDNA sequencing showed EA modulated gut microbiota composition, increased short-chain fatty acids (SCFAs)-producing bacteria, including Akkermansi, Ruminococcaceae and Lachnospiraceae family, particularly Akkermansia. Furthermore, EA increased BDNF, AcH3 and decreased HDAC2 in mPFC. Notably, SCFAs-producing bacteria exhibited a negative correlation with HDAC2 levels. LIMITATIONS: This study exclusively investigated microbiota differences resulting from EA stimulation, without delving into the functional variations brought about by these microbial distinctions. CONCLUSIONS: The therapeutic effects of EA on the comorbidity of chronic pain and depression may involve the modulation of the gut microbiota, resulting in histone acetylation changes and upregulation of BDNF.


Sujet(s)
Dépression , Modèles animaux de maladie humaine , Électroacupuncture , Microbiome gastro-intestinal , Histone Deacetylase 2 , Rat Sprague-Dawley , Animaux , Rats , Dépression/thérapie , Dépression/métabolisme , Mâle , Histone Deacetylase 2/métabolisme , Cortex préfrontal/métabolisme , Douleur chronique/thérapie , Facteur neurotrophique dérivé du cerveau/métabolisme , Comportement animal
17.
Cell Death Differ ; 31(4): 447-459, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38413797

RÉSUMÉ

Hypoxia is a hallmark of cancer development. However, the molecular mechanisms by which hypoxia promotes tumor metastasis are not fully understood. In this study, we demonstrate that hypoxia promotes breast cancer metastasis through suppression of ΔNp63α in a HIF1α-independent manner. We show that hypoxia-activated XBP1s forms a stable repressor protein complex with HDAC2 and EZH2 to suppress ΔNp63α transcription. Notably, H3K27ac is predominantly occupied on the ΔNp63 promoter under normoxia, while H3K27me3 on the promoter under hypoxia. We show that XBP1s binds to the ΔNp63 promoter to recruit HDAC2 and EZH2 in facilitating the switch of H3K27ac to H3K27me3. Pharmacological inhibition or the knockdown of either HDAC2 or EZH2 leads to increased H3K27ac, accompanied by the reduced H3K27me3 and restoration of ΔNp63α expression suppressed by hypoxia, resulting in inhibition of cell migration. Furthermore, the pharmacological inhibition of IRE1α, but not HIF1α, upregulates ΔNp63α expression in vitro and inhibits tumor metastasis in vivo. Clinical analyses reveal that reduced p63 expression is correlated with the elevated expression of XBP1, HDAC2, or EZH2, and is associated with poor overall survival in human breast cancer patients. Together, these results indicate that hypoxia-activated XBP1s modulates the epigenetic program in suppression of ΔNp63α to promote breast cancer metastasis independent of HIF1α and provides a molecular basis for targeting the XBP1s/HDAC2/EZH2-ΔNp63α axis as a putative strategy in the treatment of breast cancer metastasis.


Sujet(s)
Tumeurs du sein , Protéine-2 homologue de l'activateur de Zeste , Épigenèse génétique , Histone Deacetylase 2 , Sous-unité alpha du facteur-1 induit par l'hypoxie , Protéines suppresseurs de tumeurs , Protéine-1 liant la boite X , Humains , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Protéine-1 liant la boite X/métabolisme , Protéine-1 liant la boite X/génétique , Histone Deacetylase 2/métabolisme , Histone Deacetylase 2/génétique , Femelle , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Animaux , Lignée cellulaire tumorale , Métastase tumorale , Souris , Régulation de l'expression des gènes tumoraux , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Hypoxie cellulaire/génétique
18.
Epigenomics ; 16(5): 277-292, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38356395

RÉSUMÉ

Background: The objective of this research was to determine whether HDAC2 function is associated with gastric cancer progression. Methods: HDAC2 was knocked out in EPG85.257 cells using CRISPR/Cas9 and tumorigenesis pathways were evaluated. Results: Cell proliferation, colony formation, wound healing and transwell invasion were inhibited in ΔHDAC2:EPG85.257 cells. Quantitative analyses revealed a significant downregulation of MMP1, p53, Bax, MAPK1, MAPK3, pro-Caspase3, ERK1/2, p-ERK1/2, AKT1/2/3, p-AKT1/2/3, p-NF-κB (p65), Twist, Snail and p-FAK transcripts/proteins, while SIRT1, PTEN, p21 and Caspase3 were upregulated in ΔHDAC2:EPG85.257 cells. Conclusion: These results indicated that HDAC2 enhanced migration, colony formation and transmigration ability. HDAC2 inhibition may improve gastric cancer chemotherapy pathways.


DNA changes are the main causes of cancer. Therefore, finding easy ways to manipulate and correct DNA changes has been the biggest medical concern in cancer treatment. Researchers have introduced CRISPR/Cas9 as the newest technology for gene editing that precisely and easily changes the genome of any cell. In our study, histone deacetylase-2 was disrupted in gastric cancer cells using CRISPR technology. This modification reduced growth kinetics and invasion of cancer cells. On the other hand, cell death (also called apoptosis) was induced. Sensitization of the cancer cells to chemotherapeutic agents is noticeable in this research. This study needs to uncover more signaling pathways in vitro and in vivo.


Sujet(s)
Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Lignée cellulaire tumorale , Apoptose , Prolifération cellulaire , Histone deacetylases/génétique , Histone deacetylases/métabolisme , Épigenèse génétique , Histone Deacetylase 2/génétique , Histone Deacetylase 2/métabolisme
19.
FASEB J ; 38(4): e23470, 2024 Feb 29.
Article de Anglais | MEDLINE | ID: mdl-38354035

RÉSUMÉ

Vascular calcification is a major risk factor for cardiovascular disease mortality, with a significant prevalence in chronic kidney disease (CKD). Pharmacological inhibition of histone acetyltransferase has been proven to protect against from vascular calcification. However, the role of Histone Deacetylase 2 (HDAC2) and molecular mechanisms in vascular calcification of CKD remains unknown. An in vivo model of CKD was established using mouse fed with a high adenine and phosphate diet, and an in vitro model was produced using human aortic vascular smooth muscle cells (VSMCs) stimulated with ß-glycerophosphate (ß-GP). HDAC2 expression was found to be reduced in medial artery of CKD mice and ß-GP-induced VSMCs. Overexpression of HDAC2 attenuated OPN and OCN upregulation, α-SMA and SM22α downregulation, and calcium deposition in aortas of CKD. The in vitro results also demonstrated that ß-GP-induced osteogenic differentiation was inhibited by HDAC2. Furthermore, we found that HDAC2 overexpression caused an increase in LC3II/I, a decrease in p62, and an induction of autophagic flux. Inhibition of autophagy using its specific inhibitor 3-MA blocked HDAC2's protective effect on osteogenic differentiation in ß-GP-treated VSMCs. Taken together, these results suggest that HDAC2 may protect against vascular calcification by the activation of autophagy, laying out a novel insight for the molecular mechanism in vascular calcification of CKD.


Sujet(s)
Glycérophosphate , Insuffisance rénale chronique , Calcification vasculaire , Humains , Animaux , Souris , Histone Deacetylase 2/génétique , Ostéogenèse , Autophagie
20.
Dig Dis Sci ; 69(3): 835-850, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38240850

RÉSUMÉ

BACKGROUND: Increased SOX4 (SRY-related HMG-box) activity aids cellular transformation and metastasis. However, its specific functions and downstream targets remain to be completely elusive in colorectal cancer (CRC). AIMS: To investigate the role of SOX4 in CRC progression and the underlying mechanism. METHODS: In the current study, online available datasets of CRC patients were explored to check the expression status of SOX4. To investigate the further functions, SOX4 was overexpressed and knocked down in CRC cells. Colony formation assay, flowcytometry analysis, and MTT assay were used to check for proliferation and apoptosis. Acridine orange staining was done to check the role of SOX4 in autophagy induction. Furthermore, western blot, qRT-PCR, and bioinformatic analysis was done to elucidate the downstream molecular mechanism of SOX4. RESULTS: GEPIA database showed enhanced expression of SOX4 mRNA in CRC tumor, and the human protein atlas (HPA) showed strong staining of SOX4 protein in tumor when compared to the normal tissue. Ectopic expression of SOX4 enhanced colony formation ability as well as rescued cells from apoptosis. SOX4 overexpressed cells showed the formation of acidic vesicular organelles (AVOs) which indicated autophagy. Further results revealed the activation of p-AKT/MAPK molecules upon overexpression of SOX4. SOX4 expression was found to be positively correlated with histone deacetylase 2 (HDAC2). Knockdown of SOX4 or HDAC2 inhibition induced apoptosis, revealed by decrease in BCL2 and increase in BAX expression, and inactivated the p-AKT/MAPK signaling. CONCLUSION: The study uncovers that SOX4/HDAC2 axis improves cell survivability and reduces apoptosis via activation of the p-AKT/MAPK pathway.


Sujet(s)
Tumeurs colorectales , Histone Deacetylase 2 , Protéines proto-oncogènes c-akt , Facteurs de transcription SOX-C , Humains , Apoptose/génétique , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire/génétique , Tumeurs colorectales/anatomopathologie , Régulation de l'expression des gènes tumoraux , Histone Deacetylase 2/génétique , Histone Deacetylase 2/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Facteurs de transcription SOX-C/génétique , Facteurs de transcription SOX-C/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...