Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.741
Filtrer
1.
Immunity ; 57(10): 2296-2309.e5, 2024 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-39353440

RÉSUMÉ

The surface of the skin is continually exposed to pro-inflammatory stimuli; however, it is unclear why it is not constantly inflamed due to this exposure. Here, we showed undifferentiated keratinocytes residing in the deep epidermis could trigger a strong inflammatory response due to their high expression of pattern recognition receptors (PRRs) that detect damage or pathogens. As keratinocytes differentiated, they migrated outward toward the surface of the skin and decreased their PRR expression, which led to dampened immune responses. ZNF750, a transcription factor expressed only in differentiated keratinocytes, recruited the histone demethylase KDM1A/LSD1 to silence genes coding for PRRs (TLR3, IFIH1/MDA5, and DDX58/RIG1). Loss of ZNF750 or KDM1A in human keratinocytes or mice resulted in sustained and excessive inflammation resembling psoriatic skin, which could be restored to homeostatic conditions upon silencing of TLR3. Our findings explain how the skin's surface prevents excessive inflammation through ZNF750- and KDM1A-mediated suppression of PRRs.


Sujet(s)
Histone Demethylases , Inflammation , Kératinocytes , Récepteurs de reconnaissance de motifs moléculaires , Peau , Facteurs de transcription , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Humains , Kératinocytes/métabolisme , Animaux , Souris , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Récepteurs de reconnaissance de motifs moléculaires/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme , Inflammation/immunologie , Différenciation cellulaire/immunologie , Psoriasis/immunologie , Psoriasis/génétique , Psoriasis/métabolisme , Souris knockout , Extinction de l'expression des gènes , Souris de lignée C57BL , Protéines suppresseurs de tumeurs
2.
Immunity ; 57(10): 2263-2265, 2024 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-39383842

RÉSUMÉ

It's crucial for skin to establish efficient defense strategies. Liu et al. reveal that the transcription factor ZNF750 recruits the histone demethylase KDM1A to silence pattern recognition receptors in the outer epidermis, making their expression limited to deeper, undifferentiated keratinocytes to address threats penetrating the skin.


Sujet(s)
Kératinocytes , Peau , Facteurs de transcription , Humains , Kératinocytes/métabolisme , Kératinocytes/immunologie , Peau/immunologie , Peau/métabolisme , Animaux , Facteurs de transcription/métabolisme , Histone Demethylases/métabolisme , Épiderme/métabolisme , Épiderme/immunologie , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , Protéines de liaison à l'ADN/métabolisme
3.
Sci Rep ; 14(1): 23450, 2024 10 08.
Article de Anglais | MEDLINE | ID: mdl-39379699

RÉSUMÉ

BACKGROUND: Histone deacetylase 4 (HDAC4) and histone deacetylase 5 (HDAC5) are two isoforms of class IIa HDACs, and LMK235 is an HDAC inhibitor with higher selectivity for HDAC4/5. This study aimed to explore the expression and subcellular localization of HDAC4/5 and determine the mechanisms underlying the impact of LMK235 on ventricular remodelling post-MI. METHODS: The MI model was established by left anterior descending branch (LAD) ligation, and LMK235 or vehicle was intraperitoneally injected daily for 21 days. Cardiac function was determined by echocardiography. Inflammation was evaluated by HE staining and measuring inflammatory cytokine expression, and fibrosis was evaluated by Masson staining and measuring fibrotic biomarker expression. RESULTS: We found that LMK235 ameliorated cardiac dysfunction post-MI by suppressing inflammation and fibrosis, and LMK235 inhibited upregulation of lysine-specific demethylase 1 (LSD1) expression post-MI. In macrophages, LMK235 attenuated lipopolysaccharide (LPS) - induced inflammatory cytokine expression and inhibited LSD1 expression, while overexpression of LSD1 abrogated the anti-inflammatory effect of LMK235. In cardiac fibroblasts, LMK235 attenuated transforming growth factor-ß1 (TGF-ß1) - induced fibrotic biomarker expression and inhibited LSD1 expression, while overexpression of LSD1 abrogated the antifibrotic effect of LMK235. CONCLUSION: LMK235 attenuates chronic inflammation and fibrosis post-MI, leading to improved cardiac function. The anti-inflammatory effect of LMK235 may result from inhibition of the LSD1-NF-κB pathway in macrophages. The antifibrotic effect of LMK235 may result from inhibition of the LSD1-Smad2/3 pathway in cardiac fibroblasts.


Sujet(s)
Fibrose , Histone Demethylases , Inflammation , Infarctus du myocarde , Animaux , Histone Demethylases/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Infarctus du myocarde/métabolisme , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Inflammation/traitement médicamenteux , Mâle , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Rats , Remodelage ventriculaire/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Rat Sprague-Dawley , Cytokines/métabolisme , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1620-1630, 2024 Aug 20.
Article de Chinois | MEDLINE | ID: mdl-39276059

RÉSUMÉ

OBJECTIVE: To explore the inhibitory effect ORY-1001, a lysine-specific histone demethylase 1 (LSD1) inhibitor, on growth of glioblastoma (GBM) and the underlying mechanism. METHODS: We analyzed LSD1 expressions in GBM and normal brain tissues based on data from TCGA and HPA databases. Female BALB/c mouse models bearing xenografts derived from U87 cells or cells with lentivirus-mediated LSD1 silencing or Notch overexpression were treated with saline or 400 µg/kg ORY-1001 by gavage every 7 days, and GBM formation and survival time of the mice were recorded. The effect of ORY-1001 on GBM cell viability was assessed, and its effect on LSD1 expression was analyzed with Western blotting. The genes and pathways associated with LSD1 were analyzed using bioinformatics methods. Western blotting and qRT-PCR were used to detect Notch/HES1 pathway expression after LSD1 silencing and ORY-1001 treatment. The impact of ORY-1001 on viability of U87 cells with Notch1 silencing or overexpression was assessed, and the regulatory effects of ORY-1001 on Notch/HES1 pathway were analyzed using chromatin immunoprecipitation assay. RESULTS: A high expression of LSD1 in GBM was negatively correlated with patient survival (P < 0.001). ORY-1001 and LSD1 silencing obviously reduced tumor burden and prolonged the survival time of GBM-bearing mice. ORY-1001 treatment significantly inhibited the viability and dose-dependently decreased LSD1 expression in GBM cells, and such inhibitory effect of ORY-1001 was attenuated by LSD1 silencing. The Notch pathway enriched the differential genes related to LSD1, and Notch/HES1 pathway expression was significantly down-regulated after LSD1 silencing and ORY-1001 treatment. Notch1 overexpression significantly attenuated the anti-tumor effect of ORY-1001 on GBM. Mechanistically, ORY-1001 disrupted the interaction between LSD1 and the Notch pathway target genes including Notch3, HES1 and CR2. CONCLUSION: ORY-1001 down-regulates the Notch/HES1 pathway by inhibiting LSD1 expression to suppress the growth of GBM in mice.


Sujet(s)
Prolifération cellulaire , Glioblastome , Histone Demethylases , Souris de lignée BALB C , Facteur de transcription HES-1 , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Animaux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/génétique , Souris , Lignée cellulaire tumorale , Femelle , Facteur de transcription HES-1/métabolisme , Facteur de transcription HES-1/génétique , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal , Récepteurs Notch/métabolisme , Régulation négative , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie
5.
Sci Rep ; 14(1): 20817, 2024 09 06.
Article de Anglais | MEDLINE | ID: mdl-39242625

RÉSUMÉ

Metastasis accounts for almost 90% of breast cancer-related fatalities, making it frequent malignancy and the main reason of tumor mortality globally among women. LSD1 is a histone demethylase, which plays an important role in breast cancer. In order to explore the effect of LSD1 on invasion and migration of breast cancer, we treated breast cancer cells with MCF7 and T47D exosomes knocked down by LSD1, and the invasion and migration of breast cancer cells were significantly enhanced. This phenomenon indicates that LSD1 can inhibit the invasion and migration of breast cancer cells. miR-1290 expression was downregulated in LSD1 knockdown MCF7 exosomes. By analyzing the database of miR-1290 target gene NAT1, we verified that miR-1290 could regulate the expression of NAT1. These data provide fresh insights into the biology of breast cancer therapy by demonstrating how the epigenetic factor LSD1 stimulates the breast cancer cells' invasion and migration via controlling exosomal miRNA.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Exosomes , Régulation de l'expression des gènes tumoraux , Histone Demethylases , microARN , Invasion tumorale , Humains , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Exosomes/métabolisme , Mouvement cellulaire/génétique , Femelle , microARN/génétique , microARN/métabolisme , Lignée cellulaire tumorale , Cellules MCF-7
6.
Nat Commun ; 15(1): 7758, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39237615

RÉSUMÉ

Lysine-specific histone demethylase 1 (LSD1), which demethylates mono- or di- methylated histone H3 on lysine 4 (H3K4me1/2), is essential for early embryogenesis and development. Here we show that LSD1 is dispensable for mouse embryonic stem cell (ESC) self-renewal but is required for mouse ESC growth and differentiation. Reintroduction of a catalytically-impaired LSD1 (LSD1MUT) recovers the proliferation capability of mouse ESCs, yet the enzymatic activity of LSD1 is essential to ensure proper differentiation. Indeed, increased H3K4me1 in Lsd1 knockout (KO) mouse ESCs does not lead to major changes in global gene expression programs related to stemness. However, ablation of LSD1 but not LSD1MUT results in decreased DNMT1 and UHRF1 proteins coupled to global hypomethylation. We show that both LSD1 and LSD1MUT control protein stability of UHRF1 and DNMT1 through interaction with HDAC1 and the ubiquitin-specific peptidase 7 (USP7), consequently, facilitating the deacetylation and deubiquitination of DNMT1 and UHRF1. Our studies elucidate a mechanism by which LSD1 controls DNA methylation in mouse ESCs, independently of its lysine demethylase activity.


Sujet(s)
Protéines liant les séquences stimulatrices de type CCAAT , Différenciation cellulaire , DNA (Cytosine-5-)-methyltransferase 1 , Méthylation de l'ADN , Histone Demethylases , Souris knockout , Cellules souches embryonnaires de souris , Ubiquitin-protein ligases , Animaux , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Souris , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , Cellules souches embryonnaires de souris/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Protéines liant les séquences stimulatrices de type CCAAT/génétique , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Histone/métabolisme , Prolifération cellulaire , Ubiquitination
7.
Int J Med Sci ; 21(11): 2158-2169, 2024.
Article de Anglais | MEDLINE | ID: mdl-39239542

RÉSUMÉ

Pancreatic cancer (PC) is a challenging and heterogeneous disease with a high mortality rate. Despite advancements in treatment, the prognosis for PC patients remains poor, with a high chance of disease recurrence. Biomarkers are crucial for diagnosing cancer, predicting patient prognosis and selecting treatments. However, the current lack of effective biomarkers for PC could contribute to the insufficiency of existing treatments. These findings underscore the urgent need to develop novel strategies to fight this disease. This study utilized multiple comprehensive bioinformatic analyses to identify potential therapeutic target genes in PC, focusing on histone lysine demethylases (KDMs). We found that high expression levels of KDM family genes, particularly KDM1A, KDM5A and KDM5B, were associated with improved overall survival in the cohort. Furthermore, the infiltration of various immune cells, including B cells, neutrophils, CD8+ T cells, dendritic cells, and macrophages, was positively correlated with KDM1A, KDM5A, and KDM5B expression. Moreover, MetaCore pathway analysis revealed interesting connections between KDM1A and the cell cycle and proliferation, between KDM5A and DNA damage and double-strand break repair through homologous recombination, and between KDM5B and WNT/ß-catenin signaling. These findings suggest that KDM1A, KDM5A and KDM5B may serve as promising biomarkers and therapeutic targets for PC, a disease of high importance due to its aggressive nature and urgent need for novel biomarkers to improve diagnosis and treatment.


Sujet(s)
Marqueurs biologiques tumoraux , Régulation de l'expression des gènes tumoraux , Jumonji Domain-Containing Histone Demethylases , Tumeurs du pancréas , Humains , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Pronostic , Biologie informatique , Protéines F-box/métabolisme , Protéines F-box/génétique , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Thérapie moléculaire ciblée/méthodes , Protéine-2 de liaison à la protéine du rétinoblastome/métabolisme , Protéine-2 de liaison à la protéine du rétinoblastome/génétique , Voie de signalisation Wnt/génétique , Prolifération cellulaire/génétique , Protéines nucléaires , Protéines de répression
8.
Cells ; 13(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39273051

RÉSUMÉ

Intervertebral disc disease (IDD) is a debilitating spine condition that can be caused by intervertebral disc (IVD) damage which progresses towards IVD degeneration and dysfunction. Recently, human pluripotent stem cells (hPSCs) were recognized as a valuable resource for cell-based regenerative medicine in skeletal diseases. Therefore, adult somatic cells reprogrammed into human induced pluripotent stem cells (hiPSCs) represent an attractive cell source for the derivation of notochordal-like cells (NCs) as a first step towards the development of a regenerative therapy for IDD. Utilizing a differentiation method involving treatment with a four-factor cocktail targeting the BMP, FGF, retinoic acid, and Wnt signaling pathways, we differentiate CRISPR/Cas9-generated mCherry-reporter knock-in hiPSCs into notochordal-like cells. Comprehensive analysis of transcriptomic changes throughout the differentiation process identified regulation of histone methylation as a pivotal driver facilitating the differentiation of hiPSCs into notochordal-like cells. We further provide evidence that specific inhibition of histone demethylases KDM2A and KDM7A/B enhanced the lineage commitment of hiPSCs towards notochordal-like cells. Our results suggest that inhibition of KDMs could be leveraged to alter the epigenetic landscape of hiPSCs to control notochord-specific gene expression. Thus, our study highlights the importance of epigenetic regulators in stem cell-based regenerative approaches for the treatment of disc degeneration.


Sujet(s)
Différenciation cellulaire , Cellules souches pluripotentes induites , Jumonji Domain-Containing Histone Demethylases , Chorde , Humains , Cellules souches pluripotentes induites/métabolisme , Cellules souches pluripotentes induites/cytologie , Chorde/métabolisme , Chorde/cytologie , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Protéines F-box
9.
Eur J Med Chem ; 278: 116801, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39241481

RÉSUMÉ

The rational installation of pharmacophores targeting HSP90 and LSD1 axes has achieved significant anti-cancer capacity in prostate and colorectal cancer. Among the series of hybrids, inhibitor 6 exhibited remarkable anti-proliferative activity against prostate cancer cell lines PC-3 and DU145, with GI50 values of 0.24 and 0.30 µM, respectively. It demonstrated notable efficacy in combinatorial attack and cell death initiation towards apoptosis. The cell death process was mediated by PARP induction and γH2AX signaling, and was also characterized as caspase-dependent and Bcl-xL/Bax-independent. Notably, no difference in eye size or morphology was observed in the zebrafish treated with compound 6 compared to the reference group (AUY922). The profound treatment response in docetaxel-resistant PC-3 cells highlighted the dual inhibitory ability in improving docetaxel sensitivity. Additionally, at a minimum concentration of 1.25 µM, compound 6 effectively inhibited the growth of patient-derived colorectal cancer (CRC) organoids for up to 10 days in vitro. Together, the designed HSP90/LSD1 inhibitors present a novel route and significant clinical value for anti-cancer drug therapy.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Tumeurs colorectales , Tests de criblage d'agents antitumoraux , Protéines du choc thermique HSP90 , Histone Demethylases , Organoïdes , Tumeurs de la prostate , Animaux , Humains , Mâle , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Relation dose-effet des médicaments , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/métabolisme , Structure moléculaire , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/anatomopathologie , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Relation structure-activité , Danio zébré
10.
FASEB J ; 38(18): e70072, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39323294

RÉSUMÉ

The inability to efficiently metabolize homocysteine (Hcy) due to nutritional and genetic deficiencies, leads to hyperhomocysteinemia (HHcy) and endothelial dysfunction, a hallmark of atherosclerosis which underpins cardiovascular disease (CVD). PHF8 is a histone demethylase that demethylates H4K20me1, which affects the mammalian target of rapamycin (mTOR) signaling and autophagy, processes that play important roles in CVD. PHF8 is regulated by microRNA (miR) such as miR-22-3p and miR-1229-3p. Biochemically, HHcy is characterized by elevated levels of Hcy, Hcy-thiolactone and N-Hcy-protein. Here, we examined the effects of these metabolites on miR-22-3p, miR-1229-3p, and their target PHF8, as well as on the downstream consequences of these effects on H4K20me1, mTOR-, and autophagy-related proteins and mRNAs expression in human umbilical vein endothelial cells (HUVEC). We found that treatments with N-Hcy-protein, Hcy-thiolactone, or Hcy upregulated miR-22-3p and miR-1229-3p, attenuated PHF8 expression, upregulated H4K20me1, mTOR, and phospho-mTOR. Autophagy-related proteins (BECN1, ATG5, ATG7, lipidated LC3-II, and LC3-II/LC3-I ratio) were significantly downregulated by at least one of these metabolites. We also found similar changes in the expression of miR-22-3p, Phf8, mTOR- and autophagy-related proteins/mRNAs in vivo in hearts of Cbs-/- mice, which show severe HHcy and endothelial dysfunction. Treatments with inhibitors of miR-22-3p or miR-1229-3p abrogated the effects of Hcy-thiolactone, N-Hcy-protein, and Hcy on miR expression and on PHF8, H4K20me1, mTOR-, and autophagy-related proteins/mRNAs in HUVEC. Taken together, these findings show that Hcy metabolites upregulate miR-22-3p and miR-1229-3p expression, which then dysregulate the PHF8/H4K20me1/mTOR/autophagy pathway, important for vascular homeostasis.


Sujet(s)
Autophagie , Histone Demethylases , Homocystéine , Cellules endothéliales de la veine ombilicale humaine , microARN , Sérine-thréonine kinases TOR , Facteurs de transcription , Animaux , Femelle , Humains , Souris , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Homocystéine/métabolisme , Homocystéine/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Régulation positive
11.
J Med Chem ; 67(18): 16165-16184, 2024 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-39264726

RÉSUMÉ

LSD1 (histone lysine-specific demethylase 1) has been gradually disclosed to act as an immunomodulator to enhance antitumor immune response. Despite the identification of numerous potent LSD1 inhibitors, there remains a lack of LSD1 inhibitors approved for marketing. Novel LSD1 inhibitors with different mechanisms are therefore needed. Herein, we reported a series of novel quinazoline-based LSD1 inhibitors. Among them, compound Z-1 exhibited the best LSD1 inhibitory activity (IC50 = 0.108 µM). Z-1 also acted as a selective and cellular active as an LSD1 inhibitor. Furthermore, Z-1 promoted response of gastric cancer cells to T-cell killing effect by decreasing PD-L1 expression and further attenuated the PD-1/PD-L1 interaction. In vivo, Z-1 exhibited significant suppression effect on the growth of gastric cancer cells without obvious toxicity. Therefore, Z-1 represents a potential novel immunomodulator that targets LSD1, providing a lead compound with new function mechanism for gastric cancer treatment.


Sujet(s)
Histone Demethylases , Tumeurs de l'estomac , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Humains , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/anatomopathologie , Animaux , Lignée cellulaire tumorale , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Quinazolines/pharmacologie , Quinazolines/composition chimique , Quinazolines/synthèse chimique , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/synthèse chimique , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Découverte de médicament , Simulation de docking moléculaire
12.
J Med Chem ; 67(19): 17207-17225, 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39320444

RÉSUMÉ

Precedential evidence ascertaining the overexpression of LSD1 and HDACs in colorectal cancer spurred us to design a series of dual LSD1-HDAC inhibitors. Capitalizing on the modular nature of the three-component HDAC inhibitory model, tranylcypromine as a surface recognition motif was appended to zinc-binding motifs via diverse linkers. A compendium of hydroxamic acids was generated and evaluated for in vitro cytotoxicity against HCT-116 cells (human colorectal cancer cell lines). The most potent cell growth inhibitor 2 (GI50 = 0.495 µMm HCT-116 cells) shows promising anticancer effects by reducing colony formation and inducing cell cycle arrest in HCT-116 cells. It exhibits preferential inhibition of HDAC6, along with potent inhibition of LSD1 compared to standard inhibitors. Moreover, Compound 2 upregulates acetyl-tubulin, acetyl-histone H3, and H3K4me2, indicative of LSD1 and HDAC inhibition. In vivo, it demonstrates significant antitumor activity against colorectal cancer, better than irinotecan, and effectively inhibits growth in patient-derived CRC organoids.


Sujet(s)
Antinéoplasiques , Tumeurs colorectales , Inhibiteurs de désacétylase d'histone , Histone Demethylases , Organoïdes , Humains , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/synthèse chimique , Inhibiteurs de désacétylase d'histone/composition chimique , Animaux , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation structure-activité , Souris , Cellules HCT116 , Tests de criblage d'agents antitumoraux , Acides hydroxamiques/pharmacologie , Acides hydroxamiques/synthèse chimique , Acides hydroxamiques/composition chimique , Souris nude , Histone deacetylases/métabolisme
13.
Proc Natl Acad Sci U S A ; 121(40): e2321182121, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39325426

RÉSUMÉ

Senescence is a cell fate driven by different types of stress that results in exit from the cell cycle and expression of an inflammatory senescence-associated secretory phenotype (SASP). Here, we demonstrate that stable overexpression of miR-96-5p was sufficient to induce cellular senescence in the absence of genotoxic stress, inducing expression of certain markers of early senescence including SASP factors while repressing markers of deep senescence including LINE-1 and type 1 interferons. Stable miR-96-5p overexpression led to genome-wide changes in heterochromatin followed by epigenetic activation of p16Ink4a, p21Cip1, and SASP expression, induction of a marker of DNA damage, and induction of a transcriptional signature similar to other senescent lung and endothelial cell types. Expression of miR-96-5p significantly increased following senescence induction in culture cells and with aging in tissues from naturally aged and Ercc1-/Δ progeroid mice. Mechanistically, miR-96-5p directly suppressed expression of SIN3B and SIN3 corepressor complex constituents KDM5A and MORF4L2, and siRNA-mediated knockdown of these transcriptional regulators recapitulated the senescent phenotype. In addition, pharmacologic inhibition of the SIN3 complex suppressed senescence and SASP markers. These results clearly demonstrate that a single microRNA is sufficient to drive early senescence in the absence of genotoxic stress through targeting epigenetic and transcriptional regulators, identifying novel targets for the development of senotherapeutics.


Sujet(s)
Vieillissement de la cellule , Altération de l'ADN , microARN , microARN/génétique , microARN/métabolisme , Animaux , Vieillissement de la cellule/génétique , Souris , Humains , Protéines de répression/métabolisme , Protéines de répression/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Phénotype sécrétoire associé à la sénescence/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/génétique , Hétérochromatine/métabolisme , Hétérochromatine/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Épigenèse génétique , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Régulation de l'expression des gènes , Endonucleases
14.
J Biochem Mol Toxicol ; 38(10): e23864, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39318028

RÉSUMÉ

Previous research has indicated the highly expressed lysine-specific histone demethylase 1A (KDM1A) in several human malignancies, including triple-negative breast cancer (TNBC). However, its detailed mechanisms in TNBC development remain poorly understood. The mRNA levels of KDM1A and Yin Yang 1 (YY1) were determined by RT-qPCR analysis. Western blot was performed to measure KDM1A and ubiquitin-specific protease 1 (USP1) protein expression. Cell proliferation, apoptosis, invasion, migration and stemness were evaluated by MTT assay, EdU assay, flow cytometry, transwell invasion assay, wound-healing assay and sphere-formation assay, respectively. ChIP and dual-luciferase reporter assays were conducted to determine the relationship between YY1 and KDM1A. Xenograft tumor experiment and IHC were carried out to investigate the roles of USP1 and KDM1A in TNBC development in vivo. The highly expressed KDM1A was demonstrated in TNBC tissues and cells, and KDM1A knockdown significantly promoted cell apoptosis, and hampered cell proliferation, invasion, migration, and stemness in TNBC cells. USP1 could increase the stability of KDM1A via deubiquitination, and USP1 depletion restrained the progression of TNBC cells through decreasing KDM1A expression. Moreover, YY1 transcriptionally activated KDM1A expression by directly binding to its promoter in TNBC cells. Additionally, USP1 inhibition reduced KDM1A expression to suppress tumor growth in TNBC mice in vivo. In conclusion, YY1 upregulation increased KDM1A expression via transcriptional activation. USP1 stabilized KDM1A through deubiquitination to promote TNBC progression.


Sujet(s)
Histone Demethylases , Tumeurs du sein triple-négatives , Ubiquitin-specific proteases , Ubiquitination , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Humains , Femelle , Animaux , Lignée cellulaire tumorale , Souris , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Évolution de la maladie , Prolifération cellulaire , Souris nude , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Apoptose , Mouvement cellulaire
15.
BMC Cancer ; 24(1): 971, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118085

RÉSUMÉ

BACKGROUND: Urinary bladder cancer, is the 10th most common global cancer, diagnosed in over 600,000 people causing 200,000 deaths annually. Artemisinin and its derivatives are safe compounds that have recently been proven to possess potent anti-tumor effects in vivo, through inhibition of cancer cell growth. The aim of this study is to assess the efficiency of artemisinin as a cancer treatment alone and as a pre-treatment fore cisplatin therapy for high grade urothelial carcinoma. METHODS: Sixty male albino mice were divided into six groups, and BBN was used to induce urinary bladder cancer. Blood samples were tested for renal functions and complete blood counts, kidney and urinary bladder tissues were harvested for histopathological examination. Total RNAs from urinary bladder tissues was collected, and gene expression of FGFR3, HRAS, P53, and KDM6A was quantified using qRT-PCR. RESULTS: Compared to the induced cancer group, the results revealed that FGFR3 expression levels were down-regulated in the induced cancer group treated by artemisinin only and the induced cancer group pre-treated with artemisinin prior to cisplatin by ~ 0.86-fold and 0.4-folds, respectively, aligning with HRAS down-regulation by ~ 9.54-fold and 9.05-fold, respectively. Whereas, P53 expression levels were up-regulated by ~ 0.68-fold and 0.84-fold, respectively, in parallel with KDM6A expression, which is up-regulated by ~ 0.95-folds and 5.27-folds, respectively. Also, serum creatinine and urea levels decreased significantly in the induced cancer group treated by artemisinin alone and the induced cancer group pre-treated with artemisinin prior to cisplatin, whereas the induced cancer group treated by cisplatin their levels increased significantly. Moreover, Hb, PLT, RBC, and WBC counts improved in both cancer groups treated by artemisinin alone and pre-treated with artemisinin prior to cisplatin. Histologically, in kidney tissues, artemisinin pre-treatment significantly reduced renal injury caused by cisplatin. While Artemisinin treatment for cancer in bladder tissues reverted invasive urothelial carcinoma to moderate urothelial dysplasia. CONCLUSIONS: This study indicates that artemisinin demonstrated a significant effect in reversal of the multi-step carcinogenesis process of high grade urothelial carcinoma and could enhance the effect of cisplatin therapy using artemisinin pre-treatment.


Sujet(s)
Artémisinines , Cisplatine , Régulation de l'expression des gènes tumoraux , Histone Demethylases , Récepteur de type 3 des facteurs de croissance fibroblastique , Protéine p53 suppresseur de tumeur , Tumeurs de la vessie urinaire , Animaux , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Mâle , Artémisinines/pharmacologie , Artémisinines/usage thérapeutique , Récepteur de type 3 des facteurs de croissance fibroblastique/génétique , Récepteur de type 3 des facteurs de croissance fibroblastique/métabolisme , Souris , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Protéines proto-oncogènes p21(ras)/génétique , Humains , Modèles animaux de maladie humaine , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
16.
Bioorg Chem ; 151: 107698, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39126869

RÉSUMÉ

Non-alcoholic fatty liver disease (NAFLD) is the leading chronic liver disease worldwide, facing increasing challenges in terms of prevention and treatment. The methylation of lysine and arginine residues on histone proteins is dynamically controlled by histone methyltransferases (HMTs) and histone demethylases (HDMs), regulating chromatin structure and gene transcription. Mutations, genetic translocations, and altered gene expression involving HMTs and HDMs are frequently observed in NAFLD. HMTs and HDMs are receiving increasing attention in regulating NALFD. Targeting specific HMTs and HDMs for drug development is becoming a new strategy for treating NAFLD. This review provides a comprehensive summary of the regulatory mechanism of histone methylation/demethylation in NAFLD. Additionally, we discuss the potential applications of HMTs and HDMs inhibitors in preventing NAFLD, which may provide a scientific basis for the treatment of NAFLD.


Sujet(s)
Histone , Stéatose hépatique non alcoolique , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Humains , Méthylation , Histone/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Déméthylation , Animaux , Antienzymes/pharmacologie , Antienzymes/composition chimique , Histone méthyltransférases/métabolisme , Histone méthyltransférases/antagonistes et inhibiteurs , Structure moléculaire
17.
Nat Commun ; 15(1): 7366, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39191730

RÉSUMÉ

The lysine-specific histone demethylase 1 A (LSD1) is involved in antitumor immunity; however, its role in shaping CD8 + T cell (CTL) differentiation and function remains largely unexplored. Here, we show that pharmacological inhibition of LSD1 (LSD1i) in CTL in the context of adoptive T cell therapy (ACT) elicits phenotypic and functional alterations, resulting in a robust antitumor immunity in preclinical models in female mice. In addition, the combination of anti-PDL1 treatment with LSD1i-based ACT eradicates the tumor and leads to long-lasting tumor-free survival in a melanoma model, complementing the limited efficacy of the immune or epigenetic therapy alone. Collectively, these results demonstrate that LSD1 modulation improves antitumoral responses generated by ACT and anti-PDL1 therapy, providing the foundation for their clinical evaluation.


Sujet(s)
Lymphocytes T CD8+ , Histone Demethylases , Immunothérapie adoptive , Souris de lignée C57BL , Animaux , Histone Demethylases/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Immunothérapie adoptive/méthodes , Souris , Femelle , Lymphocytes T CD8+/immunologie , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Lignée cellulaire tumorale , Antigène CD274/antagonistes et inhibiteurs , Antigène CD274/métabolisme , Antigène CD274/immunologie , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Humains , Mélanome/immunologie , Mélanome/thérapie
18.
Cancer Lett ; 600: 217182, 2024 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-39154703

RÉSUMÉ

Histone lysine-specific demethylase 1 (LSD1) is frequently overexpressed in triple negative breast cancer (TNBC), which is associated with worse clinical outcome in TNBC patients. However, the underlying mechanisms by which LSD1 promotes TNBC progression remain to be identified. We recently established a genetically engineered murine model by crossing mammary gland conditional LSD1 knockout mice with Brca1-deficient mice to explore the role of LSD1 in TNBC pathogenesis. Cre-mediated Brca1 loss led to higher incidence of tumor formation in mouse mammary glands, which was hindered by concurrent depletion of LSD1, indicating a critical role of LSD1 in promoting Brca1-deficient tumors. We also demonstrated that the silencing of a tumor suppressor gene, Tissue Factor Pathway Inhibitor 2 (TFPI2), is functionally associated with LSD1-mediated TNBC progression. Mouse Brca1-deficient tumors exhibited elevated LSD1 expression and decreased TFPI2 level compared to normal mammary tissues. Analysis of TCGA database revealed that TFPI2 expression is significantly lower in aggressive ER-negative or basal-like BC. Restoration of TFPI2 through LSD1 inhibition increased H3K4me2 enrichment at the TFPI2 promoter, suppressed tumor progression, and enhanced antitumor efficacy of chemotherapeutic agent. Induction of TFPI2 by LSD1 ablation downregulates activity of matrix metalloproteinases (MMPs) that in turn increases the level of cytotoxic T lymphocyte attracting chemokines in tumor environment, leading to enhanced tumor infiltration of CD8+ T cells. Moreover, induction of TFPI2 potentiates antitumor effect of LSD1 inhibitor and immune checkpoint blockade in poorly immunogenic TNBC. Together, our study identifies previously unrecognized roles of TFPI2 in LSD1-mediated TNBC progression, therapeutic response, and immunogenic effects.


Sujet(s)
Évolution de la maladie , Glycoprotéines , Histone Demethylases , Tumeurs du sein triple-négatives , Animaux , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Femelle , Souris , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Glycoprotéines/génétique , Glycoprotéines/métabolisme , Souris knockout , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Protéine BRCA1/génétique
19.
J Med Chem ; 67(17): 15586-15605, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39196854

RÉSUMÉ

As histone modification enzymes, EZH2 mediates H3K27 trimethylation (H3K27me3), whereas LSD1 removes methyl groups from H3K4me1/2 and H3K9me1/2. Synergistic anticancer effects of combining inhibitors of these two enzymes are observed in leukemia and prostate cancer. Thus, a series of EZH2/LSD1 dual inhibitors are designed and synthesized to evaluate their anticancer activity. After the structure-activity study, one of the best compounds, ML234, displayed excellent antiproliferative capacity against prostate cancer cell lines LNCAP, PC3, and 22RV1. Enzymatic assays ascertained that the anticancer potency of ML234 was mediated through coinhibition of EZH2 and LSD1. Moreover, the accumulation of H3K4me2 and H3K9me2 and the decrease of H3K27me3 induced by ML234 were verified by Western blot analysis. More importantly, the compound remarkably suppressed the tumor growth and enhanced the therapeutic efficacy of clinical drug enzalutamide in the 22RV1 xenograft mouse model, indicating that it may have potential as an anticancer agent in prostate cancer.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Conception de médicament , Protéine-2 homologue de l'activateur de Zeste , Histone Demethylases , Tumeurs de la prostate , Mâle , Humains , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme , Animaux , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Protéine-2 homologue de l'activateur de Zeste/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Relation structure-activité , Souris , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests de criblage d'agents antitumoraux , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Antienzymes/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude
20.
Int Immunopharmacol ; 141: 112896, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39146782

RÉSUMÉ

Colorectal cancer (CRC), as a fatal cancer, is one of the most common cancers worldwide. Although the standard treatment for colorectal cancer is well researched and established, long-term patient survival remains poor, and mortality remains high. Therefore, more and more effective treatment options are needed. To evaluate the efficacy of bevacizumab, the histone demethylase inhibitor IOX1, or their combination for the treatment of colorectal cancer, we examined the effects of IOX1, bevacizumab, and IOX1 combined with bevacizumab on cell activity, proliferation, and migration of colorectal cancer cell lines HCT116, RKO, and CT26 by CCK8, colony formation assay, wound healing assay, and transwell assay. The effects of the drugs alone as well as in combination on apoptosis in colorectal cancer cell lines were examined by flow cytometry and further validated by Western blotting for apoptosis-related proteins. The antitumor effects of treatment alone or in combination on colorectal cancer cells were examined in animal models. Mice were injected subcutaneously with CT26 cells and the growth and immune infiltration in tumor tissues were detected by IHC after drug treatment. We found that IOX1 could effectively inhibit the activity of CRC cells and had a significant inhibitory effect on the proliferation and migration of CRC cells. The apoptosis rate increased in a dose-dependent manner after IOX1 treatment on colorectal cancer cells, and the expression of apoptosis-related proteins changed accordingly. Further combination with bevacizumab revealed that the combination had a more significant effect on the proliferation, migration, and apoptosis of CRC cells than either IOX1 or bevacizumab alone. In vivo experiments have found that both alone and combination drugs can inhibit the growth of mouse tumors, but the effect of combination inhibition is the most obvious. Combination therapy significantly inhibited the expression of proliferative marker (Ki67) in tumor xenograft models, and increased content of antigen-specific CD4+, CD8+T cell growth, and granzymeB (GZMB), which is associated with T cell cytotoxicity, was detected in combination therapy. Immunoassays suppressed the expression of relevant PD-1 and decreased. The anticancer drug bevacizumab and the histone demethylase inhibitor IOX1 may inhibit colon cancer cell growth by regulating apoptosis. The inhibitory effect of combination therapy on tumor growth may be achieved, in part, through upregulation of infiltration-mediated tumor immunity by T lymphocytes. The combination of IOX1 and bevacizumab produced significant synergistic effects. This study aims to provide a new direction for CRC combination therapy.


Sujet(s)
Apoptose , Bévacizumab , Prolifération cellulaire , Tumeurs colorectales , Animaux , Bévacizumab/usage thérapeutique , Bévacizumab/pharmacologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Humains , Apoptose/effets des médicaments et des substances chimiques , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Mouvement cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Souris nude , Histone Demethylases/antagonistes et inhibiteurs , Histone Demethylases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE