Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.197
Filtrer
1.
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
2.
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
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.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Nucleic Acids Res ; 52(10): 5698-5719, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38587186

RÉSUMÉ

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.


Sujet(s)
Chromatine , Réparation de l'ADN , Protéines de liaison à l'ADN , Immunité , Facteurs de transcription , Humains , Lignée cellulaire tumorale , Chromatine/métabolisme , Assemblage et désassemblage de la chromatine/génétique , Cassures double-brin de l'ADN , Réparation de l'ADN/génétique , Protéines de liaison à l'ADN/déficit , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Histone Deacetylase 1/génétique , Histone Deacetylase 1/métabolisme , Recombinaison homologue/génétique , Immunité/génétique , Tumeurs/diagnostic , Tumeurs/génétique , Tumeurs/immunologie , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Nucleotidyltransferases/génétique , Nucleotidyltransferases/métabolisme , Transactivateurs , Facteurs de transcription/déficit , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
13.
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
14.
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
15.
Brain Res Bull ; 211: 110944, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38604377

RÉSUMÉ

Ischemic stroke is a serious cerebrovascular condition. Isobavachalcone (ISO) has been documented to exhibit an anti-inflammatory effect across a variety of diseases; however, its protective impact on ischemic stroke remains unexplored. In this study, we evaluated the influence of ISO in both transient middle cerebral artery occlusion/reperfusion (tMCAO/R) rat models and oxygen-glucose deprivation/reperfusion (OGD/R) cell models. We observed that pretreatment with 50 mg/kg ISO diminished the volume of brain infarction, reduced brain edema, and ameliorated neurological deficits in rats. A reduction in Nissl bodies was noted in the tMCAO/R group, which was reversed following treatment with 50 mg/kg ISO. TUNEL/NeuN double staining revealed a decrease in TUNEL-positive cells in tMCAO/R rats treated with ISO. Furthermore, ISO treatment suppressed the expression of cleaved caspase-3 and BAX, while elevating the expression of BCL-2 in tMCAO/R rats. The levels of CD86 and iNOS were elevated in tMCAO/R rats; conversely, ISO treatment enhanced the expression of CD206 and Arg-1. Additionally, the expression of TNF-α, IL-6, and IL-1ß was elevated in tMCAO/R rats, whereas ISO treatment counteracted this effect. ISO treatment also increased the expression of TGF-ß and IL-10 in the ischemic penumbra of tMCAO/R rats. It was found that ISO treatment hindered microglial M1 polarization and favored M2 polarization. Histone Deacetylase 1 (HDAC1) is the downstream target protein of ISO, with ISO treatment resulting in decreased HDAC1 expression in both tMCAO/R rats and OGD/R-induced cells. Overexpression of HDAC1 was shown to promote microglial M1 polarization and inhibit M2 polarization in OGD/R+ISO cells. Overall, ISO treatment mitigated brain damage following ischemic stroke by promoting M2 polarization and attenuated ischemic injury by repressing HDAC1 expression.


Sujet(s)
Chalcones , Histone Deacetylase 1 , Accident vasculaire cérébral ischémique , Rat Sprague-Dawley , Animaux , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/métabolisme , Accident vasculaire cérébral ischémique/anatomopathologie , Mâle , Rats , Histone Deacetylase 1/métabolisme , Chalcones/pharmacologie , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Neuroprotecteurs/pharmacologie , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/métabolisme , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Modèles animaux de maladie humaine
16.
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
17.
Biomed Pharmacother ; 174: 116537, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38579402

RÉSUMÉ

Chronic Lymphocytic Leukemia (CLL) patients have a defective expression of the proapoptotic protein p66Shc and of its transcriptional factor STAT4, which evoke molecular abnormalities, impairing apoptosis and worsening disease prognosis and severity. p66Shc expression is epigenetically controlled and transcriptionally modulated by STAT4; epigenetic modifiers are deregulated in CLL cells and specific histone deacetylases (HDACs) like HDAC1, are overexpressed. Reactivation of STAT4/p66Shc expression may represent an attractive and challenging strategy to reverse CLL apoptosis defects. New selective class I HDAC inhibitors (HDACis, 6a-g) were developed with increased potency over existing agents and preferentially interfering with the CLL-relevant isoform HDAC1, to unveil the role of class I HDACs in the upregulation of STAT4 expression, which upregulates p66Shc expression and hence normalizes CLL cell apoptosis. 6c (chlopynostat) was identified as a potent HDAC1i with a superior profile over entinostat. 6c induces marked apoptosis of CLL cells compared with SAHA, which was associated with an upregulation of STAT4/p66Shc protein expression. The role of HDAC1, but not HDAC3, in the epigenetic upregulation of STAT4/p66Shc was demonstrated for the first time in CLL cells and was validated in siRNA-induced HDAC1/HDAC3 knock-down EBV-B cells. To sum up, HDAC1 inhibition is necessary to reactivate STAT4/p66Shc expression in patients with CLL. 6c is one of the most potent HDAC1is known to date and represents a novel pharmacological tool for reversing the impairment of the STAT4/p66Shc apoptotic machinery.


Sujet(s)
Apoptose , Lymphocytes B , Inhibiteurs de désacétylase d'histone , Leucémie chronique lymphocytaire à cellules B , Facteur de transcription STAT-4 , Protéine transformante-1 contenant un domaine d'homologie-2 de Src , Humains , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Leucémie chronique lymphocytaire à cellules B/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie , Protéine transformante-1 contenant un domaine d'homologie-2 de Src/métabolisme , Protéine transformante-1 contenant un domaine d'homologie-2 de Src/génétique , Facteur de transcription STAT-4/métabolisme , Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes B/métabolisme , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/antagonistes et inhibiteurs , Benzamides/pharmacologie , Mâle , Sujet âgé , Femelle , Adulte d'âge moyen
18.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167162, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38604490

RÉSUMÉ

The molecular mechanism underlying the promotion of fracture healing by mechanical stimuli remains unclear. The present study aimed to investigate the role of zinc finger protein 36 like 2 (ZFP36L2)-histone deacetylase 1 (HDAC1) axis on the osteogenic responses to moderate mechanical stimulation. Appropriate stimulation of fluid shear stress (FSS) was performed on MC3T3-E1 cells transduced with ZFP36L2 and HDAC1 recombinant adenoviruses, aiming to validate the influence of mechanical stress on the expression of ZFP36L2-HDAC1 and the osteogenic differentiation and mineralization. The results showed that moderate FSS stimulation significantly upregulated the expression of ZFP36L2 in MC3T3-E1 cells (p < 0.01). The overexpression of ZFP36L1 markedly enhanced the levels of osteogenic differentiation markers, including bone morphogenetic protein 2 (BMP2), runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), Osterix, and collagen type I alpha 1 (COL1A1) (p < 0.01). ZFP36L2 accelerated the degradation of HDAC1 by specifically binding to its 3' UTR region, thereby fulfilling its function at the post-transcriptional regulatory gene level and promoting the osteogenic differentiation and mineralization fate of cells. Mechanical unloading notably diminished/elevated the expression of ZFP36L2/HDAC1, decreased bone mineral density and bone volume fraction, hindered the release of osteogenic-related factors and vascular endothelial growth factor in callus tissue (p < 0.01), and was detrimental to fracture healing. Collectively, proper stress stimulation plays a crucial role in facilitating osteogenesis through the promotion of ZFP36L2 and subsequent degradation of HDAC1. Targeting ZFP36L2-HDAC1 axis may provide promising insights to enhance bone defect healing.


Sujet(s)
Différenciation cellulaire , Histone Deacetylase 1 , Ostéogenèse , Contrainte mécanique , Animaux , Souris , Os et tissu osseux/métabolisme , Lignée cellulaire , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Ostéoblastes/métabolisme , Ostéogenèse/physiologie
19.
Cell Death Dis ; 15(4): 289, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38653973

RÉSUMÉ

GATA-binding protein 4 (GATA4) is recognized for its significant roles in embryogenesis and various cancers. Through bioinformatics and clinical data, it appears that GATA4 plays a role in breast cancer development. Yet, the specific roles and mechanisms of GATA4 in breast cancer progression remain elusive. In this study, we identify GATA4 as a tumor suppressor in the invasion and migration of breast cancer. Functionally, GATA4 significantly reduces the transcription of MMP9. On a mechanistic level, GATA4 diminishes MMP9 transcription by interacting with p65 at the NF-κB binding site on the MMP9 promoter. Additionally, GATA4 promotes the recruitment of HDAC1, amplifying the bond between p65 and HDAC1. This leads to decreased acetylation of p65, thus inhibiting p65's transcriptional activity on the MMP9 promoter. Moreover, GATA4 hampers the metastasis of breast cancer in vivo mouse model. In summary, our research unveils a novel mechanism wherein GATA4 curtails breast cancer cell metastasis by downregulating MMP9 expression, suggesting a potential therapeutic avenue for breast cancer metastasis.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Facteur de transcription GATA-4 , Régulation de l'expression des gènes tumoraux , Histone Deacetylase 1 , Matrix metalloproteinase 9 , Invasion tumorale , Humains , Facteur de transcription GATA-4/métabolisme , Facteur de transcription GATA-4/génétique , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Femelle , Mouvement cellulaire/génétique , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Animaux , Acétylation , Lignée cellulaire tumorale , Souris , Facteur de transcription RelA/métabolisme , Transcription génétique , Régions promotrices (génétique)/génétique , Souris nude , Souris de lignée BALB C
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE