Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.220
Filtrer
1.
Clin Epigenetics ; 16(1): 102, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097736

RÉSUMÉ

BACKGROUND: Oesophageal cancer remains a challenging disease with high mortality rates and few therapeutic options. In view of these difficulties, epigenetic drugs have emerged as potential alternatives for patient care. The goal of this study was to evaluate the effect and biological consequences of Panobinostat treatment, an HDAC (histone deacetylase) inhibitor already approved for treatment of patients with multiple myeloma, in oesophageal cell lines of normal and malignant origin, with the latter being representative of the two main histological subtypes: adenocarcinoma and squamous cell carcinoma. RESULTS: Panobinostat treatment inhibited growth and hindered proliferation, colony formation and invasion of oesophageal cancer cells. Considering HDAC tissue expression, HDAC1 was significantly upregulated in normal oesophageal epithelium in comparison with tumour tissue, whereas HDAC3 was overexpressed in oesophageal cancer compared to non-malignant mucosa. No differences between normal and tumour tissue were observed for HDAC2 and HDAC8 expression. CONCLUSIONS: Panobinostat exposure effectively impaired malignant features of oesophageal cancer cells. Because HDAC3 was shown to be overexpressed in oesophageal tumour samples, this epigenetic drug may represent an alternative therapeutic option for oesophageal cancer patients.


Sujet(s)
Adénocarcinome , Carcinome épidermoïde , Prolifération cellulaire , Tumeurs de l'oesophage , Inhibiteurs de désacétylase d'histone , Histone deacetylases , Panobinostat , Humains , Panobinostat/pharmacologie , Panobinostat/usage thérapeutique , Panobinostat/administration et posologie , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/anatomopathologie , Lignée cellulaire tumorale , Adénocarcinome/traitement médicamenteux , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Histone deacetylases/génétique , Histone deacetylases/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/génétique , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Histone Deacetylase 1/génétique , Histone Deacetylase 2/génétique , Protéines de répression/génétique , Acides hydroxamiques/pharmacologie , Acides hydroxamiques/usage thérapeutique , Épigenèse génétique/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Indoles/pharmacologie , Indoles/usage thérapeutique , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/anatomopathologie
2.
Clinics (Sao Paulo) ; 79: 100413, 2024.
Article de Anglais | MEDLINE | ID: mdl-39024795

RÉSUMÉ

OBJECTIVE: Atherosclerosis (AS) is a common pathogenesis of cardiovascular diseases. Puerarin (Pue) is a Chinese herbal remedy used to prevent and treat AS. Here, this research investigated the effect of Pue on AS progression. METHODS: ApoE-/- mice were induced with acrolein. Body weight, blood lipid index, inflammatory factors, mitochondrial oxidative stress, and lipid deposition were detected. IL-6 and TNF-α were detected by ELISA. Oil red staining and H&E staining were used to observe the aortic sinus plaque lesions. Serum expressions of inflammatory factors IL-6, TNF-a, SOD, GSH and MDA were detected by ELISA, the mRNA expression levels of HDAC1 in the aorta were detected by RT-qPCR, and IL-6 and TNF-α in the aorta were detected by immunohistochemistry. JNK, p-JNK, OPA-1, and HDAC1 were detected by Western blotting. RESULTS: Pue administration can effectively reduce lipid accumulation in AS mice induced by acrolein. Pue promoted the activity of SOD, GSH and MDA, and inhibited the formation of atherosclerotic plaques and the process of aortic histological changes. Pue reduced IL-6 and TNF-α. HDAC1 expression was down-regulated and p-JNK-1 and JNK protein expression was up-regulated. CONCLUSION: Pue reduces inflammation and alleviates AS induced by acrolein by mediating the JNK pathway to inhibit HDAC1-mediated oxidative stress disorder.


Sujet(s)
Acroléine , Athérosclérose , Histone Deacetylase 1 , Isoflavones , Stress oxydatif , Animaux , Athérosclérose/induit chimiquement , Athérosclérose/métabolisme , Athérosclérose/traitement médicamenteux , Stress oxydatif/effets des médicaments et des substances chimiques , Histone Deacetylase 1/métabolisme , Isoflavones/pharmacologie , Isoflavones/usage thérapeutique , Acroléine/pharmacologie , Mâle , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-6/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Modèles animaux de maladie humaine , Test ELISA , Technique de Western , Aorte/effets des médicaments et des substances chimiques , Aorte/anatomopathologie
3.
Cell Commun Signal ; 22(1): 361, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010083

RÉSUMÉ

BACKGROUND: Breast cancer is one of the most lethal cancers in women. Despite significant advances in the diagnosis and treatment of breast cancer, many patients still succumb to this disease, and thus, novel effective treatments are urgently needed. Natural product coumarin has been broadly investigated since it reveals various biological properties in the medicinal field. Accumulating evidence indicates that histone deacetylase inhibitors (HDACIs) are promising novel anti-breast cancer agents. However, most current HDACIs exhibit only moderate effects against solid tumors and are associated with severe side effects. Thus, to develop more effective HDACIs for breast cancer therapy, hydroxamate of HDACIs was linked to coumarin core, and coumarin-hydroxamate hybrids were designed and synthesized. METHODS: A substituted coumarin moiety was incorporated into the classic hydroxamate HDACIs by the pharmacophore fusion strategy. ZN444B was identified by using the HDACI screening kit and cell viability assay. Molecular docking was performed to explore the binding mode of ZN444B with HDAC1. Western blot, immunofluorescent staining, cell viability, colony formation and cell migration and flow cytometry assays were used to analyze the anti-breast cancer effects of ZN444B in vitro. Orthotopic studies in mouse models were applied for preclinical evaluation of efficacy and toxicity in vivo. Proteomic analysis, dual-luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation, immunofluorescent staining assays along with immunohistochemical (IHC) analysis were used to elucidate the molecular basis of the actions of ZN444B. RESULTS: We synthesized and identified a novel coumarin-hydroxamate conjugate, ZN444B which possesses promising anti-breast cancer activity both in vitro and in vivo. A molecular docking model showed that ZN444B binds to HDAC1 with high affinity. Further mechanistic studies revealed that ZN444B specifically decreases FOS-like antigen 2 (FOSL2) mRNA levels by inhibiting the deacetylase activity of HDAC1 on Sp1 at K703 and abrogates the binding ability of Sp1 to the FOSL2 promoter. Furthermore, FOSL2 expression positively correlates with breast cancer progression and metastasis. Silencing FOSL2 expression decreases the sensitivity of breast cancer cells to ZN444B treatment. In addition, ZN444B shows no systemic toxicity in mice. CONCLUSIONS: Our findings highlight the potential of FOSL2 as a new biomarker and therapeutic target for breast cancer and that targeting the HDAC1-Sp1-FOSL2 signaling axis with ZN444B may be a promising therapeutic strategy for breast cancer.


Sujet(s)
Tumeurs du sein , Coumarines , Histone Deacetylase 1 , Acides hydroxamiques , Transduction du signal , Coumarines/composition chimique , Coumarines/pharmacologie , Humains , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/génétique , Femelle , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Acides hydroxamiques/pharmacologie , Acides hydroxamiques/composition chimique , Acides hydroxamiques/usage thérapeutique , Facteur de transcription Sp1/métabolisme , Souris , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Lignée cellulaire tumorale , Simulation de docking moléculaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris nude , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Souris de lignée BALB C , Mouvement cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/usage thérapeutique , Découverte de médicament
4.
PLoS One ; 19(7): e0306168, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008483

RÉSUMÉ

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.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , Tumeurs du sein , Prolifération cellulaire , Inhibiteurs de désacétylase d'histone , Humains , Inhibiteurs de désacétylase d'histone/pharmacologie , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/antagonistes et inhibiteurs , Femelle , Animaux , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Rats , Prolifération cellulaire/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Lignée cellulaire tumorale , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/métabolisme , Cellules MCF-7 , Récepteur alpha des oestrogènes/métabolisme , Récepteurs des oestrogènes/métabolisme
5.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062806

RÉSUMÉ

Histone arginine residue methylation is crucial for individual development and gene regulation. However, the dynamics of histone arginine methylation in response to cellular stress remains largely unexplored. In addition, the interplay and regulatory mechanisms between this and other histone modifications are important scientific questions that require further investigation. This study aimed to investigate the changes in histone arginine methylation in response to DNA damage. We report a global decrease in histone H3R26 symmetric dimethylation (H3R26me2s) and hypoacetylation at the H3K27 site in response to DNA damage. Notably, H3R26me2s exhibits a distribution pattern similar to that of H3K27ac across the genome, both of which are antagonistic to H3K27me3. Additionally, histone deacetylase 1 (HDAC1) may be recruited to the H3R26me2s demethylation region to mediate H3K27 deacetylation. These findings suggest crosstalk between H3R26me2s and H3K27ac in regulating gene expression.


Sujet(s)
Arginine , Histone Deacetylase 1 , Histone , Histone/métabolisme , Arginine/métabolisme , Méthylation , Humains , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Altération de l'ADN , Acétylation , Maturation post-traductionnelle des protéines , Stress physiologique/génétique
6.
J Transl Med ; 22(1): 689, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39075515

RÉSUMÉ

BACKGROUND: Lung cancer stands as the second most prevalent malignant neoplasm worldwide. Addressing the underlying mechanisms propelling the progression of non-small cell lung cancer is of paramount importance. In this study, we have elucidated the pivotal role of PHF12 in this context. MATERIALS AND METHODS: We harnessed clinical lung cancer tissue samples and non-small cell lung cancer cell lines to discern the expression pattern of PHF12. In vitro assays probing cell proliferation were conducted to substantiate the functional impact of PHF12. Furthermore, an in vivo Xenograft model was employed to dissect the role of PHF12. Employing ChIP assays and qRT-PCR, we delved into the intricate binding dynamics between PHF12 and HDAC1. Mechanistic insights into the PHF12-HDAC1 axis in lung cancer progression were pursued via RNA-seq and GSEA analyses. RESULTS: Notably, PHF12 exhibited a substantial upregulation within tumor tissue, concomitant with its correlation to HDAC1. The trilogy of cell proliferation assays, transwell assays, and the Xenograft model collectively underscored the promoting influence of PHF12 on lung cancer proliferation, both in vitro and in vivo. The ChIP assay unveiled the transcriptional regulatory role of PHF12 in governing HDAC1 expression. This correlation extended to both mRNA and protein levels. PHF12 promotes NSCLC progression through regulating HDCA1 expression. Intriguingly, the rescue of function within NSCLC cell lines post PHF12 knockdown was achievable through HDAC1 overexpression. Additionally, our findings unveiled the capacity of the PHF12-HDAC1 axis to activate the EGFR/AKT signaling pathway, thereby further corroborating its significance in lung cancer progression. CONCLUSION: Our study identified PHF12 as an oncogenic role in lung cancer proliferation and migration for the first time. PHF12 transcriptionally regulate HDAC1 and activate EGFR/AKT signaling pathway in NSCLC progression. PHF12 may serve as an important target in lung cancer therapy.


Sujet(s)
Carcinogenèse , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Récepteurs ErbB , Régulation de l'expression des gènes tumoraux , Histone Deacetylase 1 , Tumeurs du poumon , Protéines proto-oncogènes c-akt , Transduction du signal , Facteurs de transcription , Humains , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Animaux , Lignée cellulaire tumorale , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Souris nude , Souris , Femelle
7.
Nat Commun ; 15(1): 6417, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39080280

RÉSUMÉ

The splicing factor RNA-binding motif protein 10 (RBM10) is frequently mutated in lung adenocarcinoma (LUAD) (9-25%). Most RBM10 cancer mutations are loss-of-function, correlating with increased tumorigenesis and limiting the efficacy of current LUAD targeted therapies. Remarkably, therapeutic strategies leveraging RBM10 deficiency remain unexplored. Here, we conduct a CRISPR-Cas9 synthetic lethality (SL) screen and identify ~60 RBM10 SL genes, including WEE1 kinase. WEE1 inhibition sensitizes RBM10-deficient LUAD cells in-vitro and in-vivo. Mechanistically, we identify a splicing-independent role of RBM10 in regulating DNA replication fork progression and replication stress response, which underpins RBM10-WEE1 SL. Additionally, RBM10 interacts with active DNA replication forks, relying on DNA Primase Subunit 1 (PRIM1) that synthesizes Okazaki RNA primers. Functionally, we demonstrate that RBM10 serves as an anchor for recruiting Histone Deacetylase 1 (HDAC1) to facilitate H4K16 deacetylation and R-loop homeostasis to maintain replication fork stability. Collectively, our data reveal a role of RBM10 in fine-tuning DNA replication and provide therapeutic arsenal for targeting RBM10-deficient tumors.


Sujet(s)
Adénocarcinome pulmonaire , Réplication de l'ADN , Tumeurs du poumon , Protéines de liaison à l'ARN , Humains , Réplication de l'ADN/génétique , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Animaux , Lignée cellulaire tumorale , Souris , Systèmes CRISPR-Cas , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/génétique , Structures en boucle R
8.
J Biomol Struct Dyn ; 42(11): 5642-5656, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38870352

RÉSUMÉ

Histone deacetylase 1 (HDAC1), a class I HDAC enzyme, is crucial for histone modification. Currently, it is emerged as one of the important biological targets for designing small molecule drugs through cancer epigenetics. Along with synthetic inhibitors different natural inhibitors are showing potential HDAC1 inhibitions. In order to gain insights into the relationship between the molecular structures of the natural inhibitors and HDAC1, different molecular modelling techniques (Bayesian classification, recursive partitioning, molecular docking and molecular dynamics simulations) have been applied on a dataset of 155 HDAC1 nature-inspired inhibitors with diverse scaffolds. The Bayesian study showed acceptable ROC values for both the training set and test sets. The Recursive partitioning study produced decision tree 1 with 6 leaves. Further, molecular docking study was processed for generating the protein ligand complex which identified some potential amino acid residues such as F205, H28, L271, P29, F150, Y204 for the binding interactions in case of natural inhibitors. Stability of these HDAC1-natutal inhibitors complexes has been also evaluated by molecular dynamics simulation study. The current modelling study is an attempt to get a deep insight into the different important structural fingerprints among different natural compounds modulating HDAC1 inhibition.Communicated by Ramaswamy H. Sarma.


Sujet(s)
Découverte de médicament , Épigenèse génétique , Histone Deacetylase 1 , Inhibiteurs de désacétylase d'histone , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Tumeurs , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone Deacetylase 1/composition chimique , Histone Deacetylase 1/métabolisme , Inhibiteurs de désacétylase d'histone/composition chimique , Inhibiteurs de désacétylase d'histone/pharmacologie , Découverte de médicament/méthodes , Humains , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/enzymologie , Liaison aux protéines , Produits biologiques/composition chimique , Produits biologiques/pharmacologie , Ligands , Théorème de Bayes , Relation structure-activité , Sites de fixation
9.
Mar Drugs ; 22(6)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38921561

RÉSUMÉ

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.


Sujet(s)
Antinéoplasiques , Apoptose , Composés hétérocycliques bicycliques , Synergie des médicaments , Inhibiteurs de désacétylase d'histone , Leucémie aigüe myéloïde , Sulfonamides , Humains , Sulfonamides/pharmacologie , Sulfonamides/composition chimique , Leucémie aigüe myéloïde/traitement médicamenteux , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/composition chimique , Composés hétérocycliques bicycliques/pharmacologie , Composés hétérocycliques bicycliques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Histone Deacetylase 1/antagonistes et inhibiteurs , Histone deacetylases/métabolisme , Animaux , Caspase-3/métabolisme , Protéine Mcl-1/antagonistes et inhibiteurs
10.
Ann Hematol ; 103(8): 3015-3027, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38847852

RÉSUMÉ

Bone marrow stromal cells (BMSCs) can promote the growth of Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Histone deacetylases (HDACs) play essential roles in the proliferation and apoptosis resistance of Ph + ALL cells. In our previous study, inhibiting histone deacetylase 1 (HDAC1) decreases the proliferation of Ph + ALL cells. However, little is known regarding how HDAC1 in BMSCs of Ph + ALL patients affects the imatinib (IM) resistance. Therefore, the present work examined the roles of HDAC1 in BMSCs. Overexpression of HDAC1 was found in BMSCs of Ph + ALL patients with IM resistance. In addition, the Ph + ALL cell line SUP-B15 was co-cultured with BMSCs after lentivirus transfection for regulating HDAC1 expression. Knockdown of HDAC1 within BMSCs elevated the IM-mediated SUP-B15 cell apoptosis, while increasing HDAC1 expression had an opposite effect. IL-6 in BMSCs, which is an important factor for the microenvironment-associated chemoresistance, showed evident up-regulation in HDAC1-upregulated BMSCs and down-regulation in HDAC1-downregulated BMSCs. While recombinant IL-6 (rIL-6) can reversed the sensitivity of SUP-B15 cells to IM induced by downregulating HDAC1 expression in BMSCs. HDAC1 showed positive regulation on IL-6 transcription and secretion. Moreover, IL-6 secretion induced by HDAC1 in BMSCs might enhance IM resistance in Ph + ALL cells. With regard to the underlying molecular mechanism, NF-κB, an important signal responsible for IL-6 transcription in BMSCs, mediated the HDAC1-regulated IL-6 expression. Collectively, this study facilitated to develop HDAC1 inhibitors based not only the corresponding direct anti-Ph + ALL activity but also the regulation of bone marrow microenvironment.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Histone Deacetylase 1 , Mésilate d'imatinib , Interleukine-6 , Cellules souches mésenchymateuses , Leucémie-lymphome lymphoblastique à précurseurs B et T , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Humains , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Mésilate d'imatinib/pharmacologie , Mésilate d'imatinib/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Interleukine-6/métabolisme , Interleukine-6/génétique , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Mâle , Femelle , Lignée cellulaire tumorale , Adulte , Apoptose/effets des médicaments et des substances chimiques , Enfant , Adolescent , Chromosome Philadelphie , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes dans la leucémie/effets des médicaments et des substances chimiques
11.
BMB Rep ; 57(6): 299-304, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38835116

RÉSUMÉ

Upregulation of PRAME (preferentially expressed antigen of melanoma) has been implicated in the progression of a variety of cancers, including melanoma. The tumor suppressor p53 is a transcriptional regulator that mediates cell cycle arrest and apoptosis in response to stress signals. Here, we report that PRAME is a novel repressive target of p53. This was supported by analysis of melanoma cell lines carrying wild-type p53 and human melanoma databases. mRNA expression of PRAME was downregulated by p53 overexpression and activation using DNA-damaging agents, but upregulated by p53 depletion. We identified a p53-responsive element (p53RE) in the promoter region of PRAME. Luciferase and ChIP assays showed that p53 represses the transcriptional activity of the PRAME promoter and is recruited to the p53RE together with HDAC1 upon etoposide treatment. The functional significance of p53 activationmediated PRAME downregulation was demonstrated by measuring colony formation and p27 expression in melanoma cells. These data suggest that p53 activation, which leads to PRAME downregulation, could be a therapeutic strategy in melanoma cells. [BMB Reports 2024; 57(6): 299-304].


Sujet(s)
Antigènes néoplasiques , Mélanome , Régions promotrices (génétique) , Protéine p53 suppresseur de tumeur , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Mélanome/métabolisme , Mélanome/génétique , Mélanome/anatomopathologie , Antigènes néoplasiques/métabolisme , Antigènes néoplasiques/génétique , Lignée cellulaire tumorale , Régions promotrices (génétique)/génétique , Régulation de l'expression des gènes tumoraux , Étoposide/pharmacologie , Histone Deacetylase 1/métabolisme , Régulation négative/effets des médicaments et des substances chimiques
12.
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
13.
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
14.
PLoS One ; 19(6): e0304530, 2024.
Article de Anglais | MEDLINE | ID: mdl-38829908

RÉSUMÉ

Rheumatoid arthritis (RA) is a systemic immune-mediated disease characterized by joint inflammation and destruction. The disease typically affects small joints in the hands and feet, later progressing to involve larger joints such as the knees, shoulders, and hips. While the reasons for these joint-specific differences are unclear, distinct epigenetic patterns associated with joint location have been reported. In this study, we evaluated the unique epigenetic landscapes of fibroblast-like synoviocytes (FLS) from hip and knee synovium in RA patients, focusing on the expression and regulation of Homeobox (HOX) transcription factors. These highly conserved genes play a critical role in embryonic development and are known to maintain distinct expression patterns in various adult tissues. We found that several HOX genes, especially HOXD10, were differentially expressed in knee FLS compared with hip FLS. Epigenetic differences in chromatin accessibility and histone marks were observed in HOXD10 promoter between knee and hip FLS. Histone modification, particularly histone acetylation, was identified as an important regulator of HOXD10 expression. To understand the mechanism of differential HOXD10 expression, we inhibited histone deacetylases (HDACs) with small molecules and siRNA. We found that HDAC1 blockade or deficiency normalized the joint-specific HOXD10 expression patterns. These observations suggest that epigenetic differences, specifically histone acetylation related to increased HDAC1 expression, play a crucial role in joint-specific HOXD10 expression. Understanding these mechanisms could provide insights into the regional aspects of RA and potentially lead to therapeutic strategies targeting specific patterns of joint involvement during the course of disease.


Sujet(s)
Polyarthrite rhumatoïde , Épigenèse génétique , Fibroblastes , Protéines à homéodomaine , Cellules synoviales , Humains , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/génétique , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Régions promotrices (génétique) , Articulation du genou/anatomopathologie , Articulation du genou/métabolisme , Régulation de l'expression des gènes , Histone/métabolisme , Acétylation , Articulation de la hanche/anatomopathologie , Articulation de la hanche/métabolisme
15.
Sci Rep ; 14(1): 12091, 2024 05 27.
Article de Anglais | MEDLINE | ID: mdl-38802425

RÉSUMÉ

Estrogen receptor-negative [ER(-)] mammary cancer is the most aggressive type of breast cancer (BC) with higher rate of metastasis and recurrence. In recent years, dietary prevention of BC with epigenetically active phytochemicals has received increased attention due to its feasibility, effectiveness, and ease of implementation. In this regard, combinatorial phytochemical intervention enables more efficacious BC inhibition by simultaneously targeting multiple tumorigenic pathways. We, therefore, focused on investigation of the effect of sulforaphane (SFN)-rich broccoli sprouts (BSp) and withaferin A (WA)-rich Ashwagandha (Ash) combination on BC prevention in estrogen receptor-negative [ER(-)] mammary cancer using transgenic mice. Our results indicated that combinatorial BSp + Ash treatment significantly reduced tumor incidence and tumor growth (~ 75%) as well as delayed (~ 21%) tumor latency when compared to the control treatment and combinatorial BSp + Ash treatment was statistically more effective in suppressing BC compared to single BSp or Ash intervention. At the molecular level, the BSp and Ash combination upregulated tumor suppressors (p53, p57) along with apoptosis associated proteins (BAX, PUMA) and BAX:BCL-2 ratio. Furthermore, our result indicated an expressional decline of epigenetic machinery HDAC1 and DNMT3A in mammary tumor tissue because of combinatorial treatment. Interestingly, we have reported multiple synergistic interactions between BSp and Ash that have impacted both tumor phenotype and molecular expression due to combinatorial BSp and Ash treatment. Our RNA-seq analysis results also demonstrated a transcriptome-wide expressional reshuffling of genes associated with multiple cell-signaling pathways, transcription factor activity and epigenetic regulations due to combined BSp and Ash administration. In addition, we discovered an alteration of gut microbial composition change because of combinatorial treatment. Overall, combinatorial BSp and Ash supplementation can prevent ER(-) BC through enhanced tumor suppression, apoptosis induction and transcriptome-wide reshuffling of gene expression possibly influencing multiple cell signaling pathways, epigenetic regulation and reshaping gut microbiota.


Sujet(s)
Tumeurs du sein , Épigenèse génétique , Microbiome gastro-intestinal , Isothiocyanates , Sulfoxydes , Withanolides , Isothiocyanates/pharmacologie , Animaux , Withanolides/pharmacologie , Sulfoxydes/pharmacologie , Femelle , Souris , Épigenèse génétique/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/prévention et contrôle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Souris transgéniques , Extraits de plantes/pharmacologie , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Humains , Brassica/composition chimique , Histone Deacetylase 1/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Anticarcinogènes/pharmacologie
16.
Cancer Lett ; 594: 216962, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38768680

RÉSUMÉ

PA28γ overexpression is aberrant and accompanied by poor patient prognosis in various cancers, the precise regulatory mechanism of this crucial gene in the tumor microenvironment remains incompletely understood. In this study, using oral squamous cell carcinoma as a model, we demonstrated that PA28γ exhibits high expression in cancer-associated fibroblasts (CAFs), and its expression significantly correlates with the severity of clinical indicators of malignancy. Remarkably, we found that elevated levels of secreted IGF2 from PA28γ+ CAFs can enhance stemness maintenance and promote tumor cell aggressiveness through the activation of the MAPK/AKT pathway in a paracrine manner. Mechanistically, PA28γ upregulates IGF2 expression by stabilizing the E2F3 protein, a transcription factor of IGF2. Further mechanistic insights reveal that HDAC1 predominantly mediates the deacetylation and subsequent ubiquitination and degradation of E2F3. Notably, PA28γ interacts with HDAC1 and accelerates its degradation via a 20S proteasome-dependent pathway. Additionally, PA28γ+ CAFs exert an impact on the tumor immune microenvironment by secreting IGF2. Excitingly, our study suggests that targeting PA28γ+ CAFs or secreted IGF2 could increase the efficacy of PD-L1 therapy. Thus, our findings reveal the pivotal role of PA28γ in cell interactions in the tumor microenvironment and propose novel strategies for augmenting the effectiveness of immune checkpoint blockade in oral squamous cell carcinoma.


Sujet(s)
Fibroblastes associés au cancer , Facteur de transcription E2F3 , Histone Deacetylase 1 , Facteur de croissance IGF-II , Tumeurs de la bouche , Transduction du signal , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Humains , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Facteur de croissance IGF-II/métabolisme , Facteur de croissance IGF-II/génétique , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/génétique , Facteur de transcription E2F3/métabolisme , Facteur de transcription E2F3/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/génétique , Lignée cellulaire tumorale , Animaux , Souris , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Mâle , Femelle
17.
mBio ; 15(6): e0237723, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38709067

RÉSUMÉ

Plasmodium falciparum, the deadly protozoan parasite responsible for malaria, has a tightly regulated gene expression profile closely linked to its intraerythrocytic development cycle. Epigenetic modifiers of the histone acetylation code have been identified as key regulators of the parasite's transcriptome but require further investigation. In this study, we map the genomic distribution of Plasmodium falciparum histone deacetylase 1 (PfHDAC1) across the erythrocytic asexual development cycle and find it has a dynamic occupancy over a wide array of developmentally relevant genes. Overexpression of PfHDAC1 results in a progressive increment in parasite load over consecutive rounds of the asexual infection cycle and is associated with enhanced gene expression of multiple families of host cell invasion factors (merozoite surface proteins, rhoptry proteins, etc.) and with increased merozoite invasion efficiency. With the use of class-specific inhibitors, we demonstrate that PfHDAC1 activity in parasites is crucial for timely intraerythrocytic development. Interestingly, overexpression of PfHDAC1 results in decreased sensitivity to frontline-drug dihydroartemisinin in parasites. Furthermore, we identify that artemisinin exposure can interfere with PfHDAC1 abundance and chromatin occupancy, resulting in enrichment over genes implicated in response/resistance to artemisinin. Finally, we identify that dihydroartemisinin exposure can interrupt the in vitro catalytic deacetylase activity and post-translational phosphorylation of PfHDAC1, aspects that are crucial for its genomic function. Collectively, our results demonstrate PfHDAC1 to be a regulator of critical functions in asexual parasite development and host invasion, which is responsive to artemisinin exposure stress and deterministic of resistance to it. IMPORTANCE: Malaria is a major public health problem, with the parasite Plasmodium falciparum causing most of the malaria-associated mortality. It is spread by the bite of infected mosquitoes and results in symptoms such as cyclic fever, chills, and headache. However, if left untreated, it can quickly progress to a more severe and life-threatening form. The World Health Organization currently recommends the use of artemisinin combination therapy, and it has worked as a gold standard for many years. Unfortunately, certain countries in southeast Asia and Africa, burdened with a high prevalence of malaria, have reported cases of drug-resistant infections. One of the major problems in controlling malaria is the emergence of artemisinin resistance. Population genomic studies have identified mutations in the Kelch13 gene as a molecular marker for artemisinin resistance. However, several reports thereafter indicated that Kelch13 is not the main mediator but rather hinted at transcriptional deregulation as a major determinant of drug resistance. Earlier, we identified PfGCN5 as a global regulator of stress-responsive genes, which are known to play a central role in artemisinin resistance generation. In this study, we have identified PfHDAC1, a histone deacetylase as a cell cycle regulator, playing an important role in artemisinin resistance generation. Taken together, our study identified key transcriptional regulators that play an important role in artemisinin resistance generation.


Sujet(s)
Antipaludiques , Artémisinines , Histone Deacetylase 1 , Plasmodium falciparum , Plasmodium falciparum/génétique , Plasmodium falciparum/effets des médicaments et des substances chimiques , Plasmodium falciparum/croissance et développement , Artémisinines/pharmacologie , Antipaludiques/pharmacologie , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Protéines de protozoaire/génétique , Protéines de protozoaire/métabolisme , Humains , Érythrocytes/parasitologie , Paludisme à Plasmodium falciparum/parasitologie , Reproduction asexuée/génétique
18.
J Exp Clin Cancer Res ; 43(1): 152, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38812060

RÉSUMÉ

BACKGROUND: Intrahepatic cholangiocarcinoma (ICCA) is a heterogeneous group of malignant tumors characterized by high recurrence rate and poor prognosis. Heterochromatin Protein 1α (HP1α) is one of the most important nonhistone chromosomal proteins involved in transcriptional silencing via heterochromatin formation and structural maintenance. The effect of HP1α on the progression of ICCA remained unclear. METHODS: The effect on the proliferation of ICCA was detected by experiments in two cell lines and two ICCA mouse models. The interaction between HP1α and Histone Deacetylase 1 (HDAC1) was determined using Electrospray Ionization Mass Spectrometry (ESI-MS) and the binding mechanism was studied using immunoprecipitation assays (co-IP). The target gene was screened out by RNA sequencing (RNA-seq). The occupation of DNA binding proteins and histone modifications were predicted by bioinformatic methods and evaluated by Cleavage Under Targets and Tagmentation (CUT & Tag) and Chromatin immunoprecipitation (ChIP). RESULTS: HP1α was upregulated in intrahepatic cholangiocarcinoma (ICCA) tissues and regulated the proliferation of ICCA cells by inhibiting the interferon pathway in a Signal Transducer and Activator of Transcription 1 (STAT1)-dependent manner. Mechanistically, STAT1 is transcriptionally regulated by the HP1α-HDAC1 complex directly and epigenetically via promoter binding and changes in different histone modifications, as validated by high-throughput sequencing. Broad-spectrum HDAC inhibitor (HDACi) activates the interferon pathway and inhibits the proliferation of ICCA cells by downregulating HP1α and targeting the heterodimer. Broad-spectrum HDACi plus interferon preparation regimen was found to improve the antiproliferative effects and delay ICCA development in vivo and in vitro, which took advantage of basal activation as well as direct activation of the interferon pathway. HP1α participates in mediating the cellular resistance to both agents. CONCLUSIONS: HP1α-HDAC1 complex influences interferon pathway activation by directly and epigenetically regulating STAT1 in transcriptional level. The broad-spectrum HDACi plus interferon preparation regimen inhibits ICCA development, providing feasible strategies for ICCA treatment. Targeting the HP1α-HDAC1-STAT1 axis is a possible strategy for treating ICCA, especially HP1α-positive cases.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Homologue-5 de la protéine chromobox , Histone Deacetylase 1 , Facteur de transcription STAT-1 , Animaux , Femelle , Humains , Mâle , Souris , Tumeurs des canaux biliaires/métabolisme , Tumeurs des canaux biliaires/traitement médicamenteux , Tumeurs des canaux biliaires/anatomopathologie , Tumeurs des canaux biliaires/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Cholangiocarcinome/métabolisme , Cholangiocarcinome/traitement médicamenteux , Cholangiocarcinome/anatomopathologie , Cholangiocarcinome/génétique , Homologue-5 de la protéine chromobox/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Histone Deacetylase 1/métabolisme , Facteur de transcription STAT-1/métabolisme
19.
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
20.
Cell Rep ; 43(4): 114065, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38578828

RÉSUMÉ

Epigenetic modification shapes differentiation trajectory and regulates the exhaustion state of chimeric antigen receptor T (CAR-T) cells. Limited efficacy induced by terminal exhaustion closely ties with intrinsic transcriptional regulation. However, the comprehensive regulatory mechanisms remain largely elusive. Here, we identify class I histone deacetylase inhibitors (HDACi) as boosters of CAR-T cell function by high-throughput screening of chromatin-modifying drugs, in which M344 and chidamide enhance memory maintenance and resistance to exhaustion of CAR-T cells that induce sustained antitumor efficacy both in vitro and in vivo. Mechanistically, HDACi decrease HDAC1 expression and enhance H3K27ac activity. Multi-omics analyses from RNA-seq, ATAC-seq, and H3K27ac CUT&Tag-seq show that HDACi upregulate expression of TCF4, LEF1, and CTNNB1, which subsequently activate the canonical Wnt/ß-catenin pathway. Collectively, our findings elucidate the functional roles of class I HDACi in enhancing CAR-T cell function, which provides the basis and therapeutic targets for synergic combination of CAR-T cell therapy and HDACi treatment.


Sujet(s)
Aminopyridines , Inhibiteurs de désacétylase d'histone , Voie de signalisation Wnt , Inhibiteurs de désacétylase d'histone/pharmacologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Animaux , Humains , Souris , Benzamides/pharmacologie , Lignée cellulaire tumorale , Immunothérapie adoptive/méthodes , Récepteurs chimériques pour l'antigène/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Lymphocytes T/immunologie , Histone Deacetylase 1/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE