Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 21.450
Filtrer
1.
Exp Dermatol ; 33(7): e15128, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38973249

RÉSUMÉ

Dry skin is common to many pruritic diseases and is difficult to improve with oral traditional antihistamines. Recently, increasing evidence indicated that histamine H4 receptor (H4R) plays an important role in the occurrence and development of pruritus. Extracellular signal-regulated kinase (ERK) phosphorylation activation in the spinal cord mediates histamine-induced acute and choric itch. However, whether the histamine H4 receptor regulates ERK activation in the dry skin itch remains unclear. In the study, we explore the role of the histamine H4 receptor and p-ERK in the spinal cord in a dry skin mouse model induced by acetone-ether-water (AEW). q-PCR, Western blot, pharmacology and immunofluorescence  were applied in the study. We established a dry skin itch model by repeated application of AEW on the nape of neck in mice. The AEW mice showed typically dry skin histological change and persistent spontaneous scratching behaviour. Histamine H4 receptor, instead of histamine H1 receptor, mediated spontaneous scratching behaviour in AEW mice. Moreover, c-Fos and p-ERK expression in the spinal cord neurons were increased and co-labelled with GRPR-positive neurons in AEW mice. Furthermore, H4R agonist 4-methyhistamine dihydrochloride (4-MH)induced itch. Both 4-MH-induced itch and the spontaneous itch in AEW mice were blocked by p-ERK inhibitor U0126. Finally, intrathecal H4R receptor antagonist JNJ7777120 inhibited spinal p-ERK expression in AEW mice. Our results indicated that spinal H4R mediates itch via ERK activation in the AEW-induced dry skin mice.


Sujet(s)
Acétone , Extracellular Signal-Regulated MAP Kinases , Prurit , Récepteur histaminergique H4 , Moelle spinale , Animaux , Prurit/induit chimiquement , Prurit/métabolisme , Récepteur histaminergique H4/métabolisme , Souris , Moelle spinale/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , Mâle , Acétone/pharmacologie , Eau , Oxyde de diéthyle , Modèles animaux de maladie humaine , Phosphorylation , Indoles/pharmacologie , Butadiènes/pharmacologie , Pipérazines/pharmacologie , Nitriles/pharmacologie , Peau/métabolisme , Maladie chronique , Méthylhistamines , Protéines proto-oncogènes c-fos/métabolisme , Souris de lignée C57BL
2.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(6): 1135-1140, 2024 Jun 20.
Article de Chinois | MEDLINE | ID: mdl-38977343

RÉSUMÉ

OBJECTIVE: To investigate the protective effect of dexmedetomidine (DEX) against erastin-induced ferroptosis in human renal tubular epithelial cells (HK-2 cells) and explore the underlying mechanism. METHODS: HK-2 cells were treated with erastin alone or in combination with different concentrations (2.5, 5.0 and 10 µmol/L) of DEX, and the changes in cell viability were observed using CCK-8 assay. To explore the mechanism by which DEX inhibits erastin-induced ferroptosis, HK-2 cells were treated with erastin, erastin+10 µmol/L DEX, or erastin+10 µmol/L DEX+ML385 (a Nrf2 inhibitor), after which the cell viability was assessed. The level of intracellular Fe2+ was detected by cell ferrous iron colorimetric assay kit, and flow cytometry was performed to detect reactive oxygen species (ROS); MDA and reduced glutathione assay kits were used to detect the contents of MDA and GSH in the cells; The expressions of Nrf2, HO-1 and GPX4 proteins were detected by Western blotting. RESULTS: Erastin treatment significantly inhibited the viability of the cells, decreased GSH content, and increased intracellular levels of Fe2+, ROS and MDA. The combined treatment with 10 µmol/L DEX markedly increased the viability of the cells, increased GSH content, reduced the levels of Fe2+, ROS and MDA, and upregulated the protein expressions of Nrf2, HO-1 and GPX4 in the cells. The application of ML385 obviously blocked the protective effect of DEX and caused significant inhibition of the Nrf2/HO-1/GPX4 pathway, decreased the cell viability and GSH content, and increased the levels of Fe2+, ROS and MDA in HK-2 cells. CONCLUSION: The protective effect of DEX against erastin-induced ferroptosis of HK-2 cells is probably mediated by activation of the Nrf2/HO-1/GPX4 pathway to inhibit oxidative stress.


Sujet(s)
Survie cellulaire , Dexmédétomidine , Cellules épithéliales , Ferroptose , Heme oxygenase-1 , Tubules rénaux , Facteur-2 apparenté à NF-E2 , Phospholipid hydroperoxide glutathione peroxidase , Espèces réactives de l'oxygène , Humains , Ferroptose/effets des médicaments et des substances chimiques , Facteur-2 apparenté à NF-E2/métabolisme , Dexmédétomidine/pharmacologie , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Tubules rénaux/cytologie , Tubules rénaux/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Heme oxygenase-1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie
3.
J Cell Mol Med ; 28(13): e18496, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38984939

RÉSUMÉ

Hepatocellular carcinoma (HCC), a prevalent malignancy worldwide, poses significant challenges in terms of prognosis, necessitating innovative therapeutic approaches. Ferroptosis offers notable advantages over apoptosis, holding promise as a novel therapeutic approach for HCC complexities. Moreover, while the interaction between long non-coding RNAs (lncRNAs) and mRNAs is pivotal in various physiological and pathological processes, their involvement in ferroptosis remains relatively unexplored. In this study, we constructed a ferroptosis-related lncRNA-mRNA correlation network in HCC using Pearson correlation analysis. Notably, the SLC7A11-AS1/SLC7A11 pair, exhibiting high correlation, was identified. Bioinformatics analysis revealed a significant correlation between the expression levels of this pair and key clinical characteristics of HCC patients, including gender, pathology, Ishak scores and tumour size. And poor prognosis was associated with high expression of this pair. Functional experiments demonstrated that SLC7A11-AS1, by binding to the 3'UTR region of SLC7A11 mRNA, enhanced its stability, thereby promoting HCC cell growth and resistance to erastin- induced ferroptosis. Additionally, in vivo studies confirmed that SLC7A11-AS1 knockdown potentiated the inhibitory effects of erastin on tumour growth. Overall, our findings suggest that targeting the SLC7A11-AS1/SLC7A11 pair holds promise as a potential therapeutic strategy for HCC patients.


Sujet(s)
Système y+ de transport d'acides aminés , Carcinome hépatocellulaire , Ferroptose , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , ARN long non codant , Ferroptose/génétique , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/traitement médicamenteux , Système y+ de transport d'acides aminés/génétique , Système y+ de transport d'acides aminés/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Animaux , Lignée cellulaire tumorale , Mâle , Femelle , Souris , Pronostic , Prolifération cellulaire/génétique , Souris nude , Adulte d'âge moyen , ARN messager/génétique , ARN messager/métabolisme , Pipérazines/pharmacologie
4.
Chem Biol Drug Des ; 104(1): e14584, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38997239

RÉSUMÉ

Transient receptor potential vanilloid 1 (TRPV1) is a non-selective cation channel, which is considered a highly validated target for pain perception. Repeated activation with agonists to desensitize receptors or use the antagonists can both exert analgesic effects. In this work, two series of novel phenylpiperazine derivatives were designed, synthesized, and evaluated for the in vitro receptor inhibitory activity and in vivo analgesic activity. Among them, L-21 containing sulfonylurea group was identified with potent TRPV1 antagonistic activity and analgesic activity in various pain models. At the same time, L-21 exhibited low risk of hyperthermia side effect. These results indicated that L-21 is a promising candidate for further development of novel TRPV1 antagonist to treat pain.


Sujet(s)
Analgésiques , Pipérazines , Canaux cationiques TRPV , Canaux cationiques TRPV/antagonistes et inhibiteurs , Canaux cationiques TRPV/métabolisme , Pipérazines/composition chimique , Pipérazines/pharmacologie , Pipérazines/synthèse chimique , Animaux , Souris , Analgésiques/pharmacologie , Analgésiques/composition chimique , Analgésiques/synthèse chimique , Humains , Douleur/traitement médicamenteux , Relation structure-activité , Mâle , Cellules HEK293 , Rats
5.
Proc Natl Acad Sci U S A ; 121(30): e2319574121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39024113

RÉSUMÉ

Regulated cell cycle progression ensures homeostasis and prevents cancer. In proliferating cells, premature S phase entry is avoided by the E3 ubiquitin ligase anaphasepromoting complex/cyclosome (APC/C), although the APC/C substrates whose degradation restrains G1-S progression are not fully known. The APC/C is also active in arrested cells that exited the cell cycle, but it is not clear whether APC/C maintains all types of arrest. Here, by expressing the APC/C inhibitor, EMI1, we show that APC/C activity is essential to prevent S phase entry in cells arrested by pharmacological cyclin-dependent kinases 4 and 6 (CDK4/6) inhibition (Palbociclib). Thus, active protein degradation is required for arrest alongside repressed cell cycle gene expression. The mechanism of rapid and robust arrest bypass from inhibiting APC/C involves CDKs acting in an atypical order to inactivate retinoblastoma-mediated E2F repression. Inactivating APC/C first causes mitotic cyclin B accumulation which then promotes cyclin A expression. We propose that cyclin A is the key substrate for maintaining arrest because APC/C-resistant cyclin A, but not cyclin B, is sufficient to induce S phase entry. Cells bypassing arrest from CDK4/6 inhibition initiate DNA replication with severely reduced origin licensing. The simultaneous accumulation of S phase licensing inhibitors, such as cyclin A and geminin, with G1 licensing activators disrupts the normal order of G1-S progression. As a result, DNA synthesis and cell proliferation are profoundly impaired. Our findings predict that cancers with elevated EMI1 expression will tend to escape CDK4/6 inhibition into a premature, underlicensed S phase and suffer enhanced genome instability.


Sujet(s)
Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Humains , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Complexe promoteur de l'anaphase/métabolisme , Complexe promoteur de l'anaphase/génétique , Lignée cellulaire tumorale , Phase S/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pipérazines/pharmacologie , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Facteurs de transcription E2F/métabolisme , Facteurs de transcription E2F/génétique , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Cyclines/métabolisme , Cyclines/génétique , Protéines F-box
6.
Neuropharmacology ; 257: 110057, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38964596

RÉSUMÉ

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by alterations and imbalances in multiple brain neurochemical systems, particularly the serotonergic neurotransmission. This includes changes in serotonin (5-HT) levels, aberrations in 5-HT transporter activity, and decreased synthesis and expression of 5-HT receptors (5-HT7Rs). The exact role of the brain 5-HT system in the development of ASD remains unclear, with conflicting evidence on its involvement. Recently, we have reported research has shown a significant decrease in serotonergic neurons originating from the raphe nuclei and projecting to the CA1 region of the dorsal hippocampus in autistic-like rats. Additionally, we have shown that chronic activation of 5-HT7Rs reverses the effects of autism induction on synaptic plasticity. However, the functional significance of 5-HT7Rs at the cellular level is still not fully understood. This study presents new evidence indicating an upregulation of 5-HT7R in the CA1 subregion of the hippocampus following the induction of autism. The present account also demonstrates that activation of 5-HT7R with its agonist LP-211 can reverse electrophysiological abnormalities in hippocampal pyramidal neurons in a rat model of autism induced by prenatal exposure to VPA. Additionally, in vivo administration of LP-211 resulted in improvements in motor coordination, novel object recognition, and a reduction in stereotypic behaviors in autistic-like offspring. The findings suggest that dysregulated expression of 5-HT7Rs may play a role in the pathophysiology of ASD, and that agonists like LP-211 could potentially be explored as a pharmacological treatment for autism spectrum disorder.


Sujet(s)
Modèles animaux de maladie humaine , Effets différés de l'exposition prénatale à des facteurs de risque , Récepteurs sérotoninergiques , Régulation positive , Acide valproïque , Animaux , Récepteurs sérotoninergiques/métabolisme , Acide valproïque/pharmacologie , Grossesse , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/induit chimiquement , Femelle , Régulation positive/effets des médicaments et des substances chimiques , Mâle , Région CA1 de l'hippocampe/métabolisme , Région CA1 de l'hippocampe/effets des médicaments et des substances chimiques , Rats , Pipérazines/pharmacologie , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/traitement médicamenteux , Rat Wistar , Trouble autistique/métabolisme , Trouble autistique/traitement médicamenteux
7.
Molecules ; 29(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38998996

RÉSUMÉ

Diabetes mellitus is a severe endocrine disease that affects more and more people every year. Modern medical chemistry sets itself the task of finding effective and safe drugs against diabetes. This review provides an overview of potential antidiabetic drugs based on three heterocyclic compounds, namely morpholine, piperazine, and piperidine. Studies have shown that compounds containing their moieties can be quite effective in vitro and in vivo for the treatment of diabetes and its consequences.


Sujet(s)
Hypoglycémiants , Morpholines , Pipérazine , Pipéridines , Humains , Pipéridines/composition chimique , Pipéridines/pharmacologie , Pipéridines/usage thérapeutique , Hypoglycémiants/composition chimique , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Morpholines/composition chimique , Morpholines/pharmacologie , Morpholines/usage thérapeutique , Pipérazine/composition chimique , Pipérazine/pharmacologie , Animaux , Pipérazines/composition chimique , Pipérazines/pharmacologie , Pipérazines/synthèse chimique , Pipérazines/usage thérapeutique , Diabète/traitement médicamenteux , Relation structure-activité
8.
Nat Commun ; 15(1): 5597, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961064

RÉSUMÉ

Cyclin-dependent kinases 4 and 6 (CDK4/6) play a pivotal role in cell cycle and cancer development. Targeting CDK4/6 has demonstrated promising effects against breast cancer. However, resistance to CDK4/6 inhibitors (CDK4/6i), such as palbociclib, remains a substantial challenge in clinical settings. Using high-throughput combinatorial drug screening and genomic sequencing, we find that the microphthalmia-associated transcription factor (MITF) is activated via O-GlcNAcylation by O-GlcNAc transferase (OGT) in palbociclib-resistant breast cancer cells and tumors. Mechanistically, O-GlcNAcylation of MITF at Serine 49 enhances its interaction with importin α/ß, thus promoting its translocation to nuclei, where it suppresses palbociclib-induced senescence. Inhibition of MITF or its O-GlcNAcylation re-sensitizes resistant cells to palbociclib. Moreover, clinical studies confirm the activation of MITF in tumors from patients who are palbociclib-resistant or undergoing palbociclib treatment. Collectively, our studies shed light on the mechanism regulating palbociclib resistance and present clinical evidence for developing therapeutic approaches to treat CDK4/6i-resistant breast cancer patients.


Sujet(s)
Tumeurs du sein , Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Résistance aux médicaments antinéoplasiques , Facteur de transcription associé à la microphtalmie , N-acetylglucosaminyltransferase , Pipérazines , Pyridines , Humains , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Facteur de transcription associé à la microphtalmie/métabolisme , Facteur de transcription associé à la microphtalmie/génétique , Femelle , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pyridines/pharmacologie , Lignée cellulaire tumorale , N-acetylglucosaminyltransferase/métabolisme , N-acetylglucosaminyltransferase/antagonistes et inhibiteurs , N-acetylglucosaminyltransferase/génétique , Animaux , Souris , Inhibiteurs de protéines kinases/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Sci Rep ; 14(1): 16030, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992220

RÉSUMÉ

This study examines the biological effects of palbociclib and ribociclib in hormone receptor-positive breast cancer, pivotal to the HARMONIA prospective phase III clinical trial. We explore the downstream impacts of these CDK4/6 inhibitors, focusing on cell lines and patient-derived tumor samples. We treated HR+ breast cancer cell lines (T47D, MCF7, and BT474) with palbociclib or ribociclib (100 nM or 500 nM), alone or combined with fulvestrant (1 nM), over periods of 24, 72, or 144 h. Our assessments included PAM50 gene expression, RB1 phosphorylation, Lamin-B1 protein levels, and senescence-associated ß-galactosidase activity. We further analyzed PAM50 gene signatures from the CORALLEEN and NeoPalAna phase II trials. Both CDK4/6 inhibitors similarly inhibited proliferation across the cell lines. At 100 nM, both drugs partially reduced p-RB1, with further decreases at 500 nM over 144 h. Treatment led to reduced Lamin-B1 expression and increased senescence-associated ß-galactosidase activity. Both drugs enhanced Luminal A and reduced Luminal B and proliferation signatures at both doses. However, the HER2-enriched signature significantly diminished only at the higher dose of 500 nM. Corresponding changes were observed in tumor samples from the CORALLEEN and NeoPalAna studies. At 2 weeks of treatment, both drugs significantly reduced the HER2-enriched signature, but at surgery, this reduction was consistent only with ribociclib. Our findings suggest that while both CDK4/6 inhibitors effectively modulate key biological pathways in HR+/HER2- breast cancer, nuances in their impact, particularly on the HER2-enriched signature, are dose-dependent, influenced by the addition of fulvestrant and warrant further investigation.


Sujet(s)
Aminopyridines , Tumeurs du sein , Prolifération cellulaire , Pipérazines , Purines , Pyridines , Humains , Aminopyridines/pharmacologie , Pipérazines/pharmacologie , Purines/pharmacologie , Pyridines/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteurs des oestrogènes/métabolisme , Fulvestrant/pharmacologie , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Kinase-4 cycline-dépendante/métabolisme , Récepteurs à la progestérone/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Kinase-6 cycline-dépendante/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
10.
Mol Med ; 30(1): 102, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009982

RÉSUMÉ

BACKGROUND: Acute monocytic leukemia-M5 (AML-M5) remains a challenging disease due to its high morbidity and poor prognosis. In addition to the evidence mentioned earlier, several studies have shown that programmed cell death (PCD) serves a critical function in treatment of AML-M5. However, the role and relationship between ferroptosis and necroptosis in AML-M5 remains unclear. METHODS: THP-1 cells were mainly treated with Erastin and IMP-366. The changes of ferroptosis and necroptosis levels were detected by CCK-8, western blot, quantitative real-time PCR, and electron microscopy. Flow cytometry was applied to detect the ROS and lipid ROS levels. MDA, 4-HNE, GSH and GSSG were assessed by ELISA kits. Intracellular distribution of FSP1 was studied by immunofluorescent staining and western blot. RESULTS: The addition of the myristoylation inhibitor IMP-366 to erastin-treated acute monocytic leukemia cell line THP-1 cell not only resulted in greater susceptibility to ferroptosis characterized by lipid peroxidation, glutathione (GSH) depletion and mitochondrial shrinkage, as the FSP1 position on membrane was inhibited, but also increased p-RIPK1 and p-MLKL protein expression, as well as a decrease in caspase-8 expression, and triggered the characteristic necroptosis phenomena, including cytoplasmic translucency, mitochondrial swelling, membranous fractures by FSP1 migration into the nucleus via binding importin α2. It is interesting to note that ferroptosis inhibitor fer-1 reversed necroptosis. CONCLUSION: We demonstrated that inhibition of myristoylation by IMP-366 is capable of switching ferroptosis and ferroptosis-dependent necroptosis in THP-1 cells. In these findings, FSP1-mediated ferroptosis and necroptosis are described as alternative mechanisms of PCD of THP-1 cells, providing potential therapeutic strategies and targets for AML-M5.


Sujet(s)
Ferroptose , Nécroptose , Humains , Cellules THP-1 , Membrane cellulaire/métabolisme , Noyau de la cellule/métabolisme , Espèces réactives de l'oxygène/métabolisme , Apoptose , Pipérazines/pharmacologie , Acrylamides , Sulfonamides , Protéines de liaison à l'ARN , Complexe protéique du pore nucléaire
11.
Proc Natl Acad Sci U S A ; 121(28): e2320655121, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38959043

RÉSUMÉ

SLC7A11 is a cystine transporter and ferroptosis inhibitor. How the stability of SLC7A11 is coordinately regulated in response to environmental cystine by which E3 ligase and deubiquitylase (DUB) remains elusive. Here, we report that neddylation inhibitor MLN4924 increases cystine uptake by causing SLC7A11 accumulation, via inactivating Cullin-RING ligase-3 (CRL-3). We identified KCTD10 as the substrate-recognizing subunit of CRL-3 for SLC7A11 ubiquitylation, and USP18 as SLC7A11 deubiquitylase. Upon cystine deprivation, the protein levels of KCTD10 or USP18 are decreased or increased, respectively, contributing to SLC7A11 accumulation. By destabilizing or stabilizing SLC7A11, KCTD10, or USP18 inversely regulates the cystine uptake and ferroptosis. Biologically, MLN4924 combination with SLC7A11 inhibitor Imidazole Ketone Erastin (IKE) enhanced suppression of tumor growth. In human breast tumor tissues, SLC7A11 levels were negatively or positively correlated with KCTD10 or USP18, respectively. Collectively, our study defines how SLC7A11 and ferroptosis is coordinately regulated by the CRL3KCTD10/E3-USP18/DUB axis, and provides a sound rationale of drug combination to enhance anticancer efficacy.


Sujet(s)
Système y+ de transport d'acides aminés , Cystine , Ferroptose , Pyrimidines , Ubiquitin thiolesterase , Humains , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Pyrimidines/pharmacologie , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Animaux , Cystine/métabolisme , Cyclopentanes/métabolisme , Cyclopentanes/pharmacologie , Lignée cellulaire tumorale , Ubiquitination , Femelle , Souris , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Pipérazines/pharmacologie , Cellules HEK293
12.
FASEB J ; 38(13): e23790, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38982638

RÉSUMÉ

Integrase strand transfer inhibitors (INSTIs) based antiretroviral therapy (ART) is currently used as first-line regimen to treat HIV infection. Despite its high efficacy and barrier to resistance, ART-associated neuropsychiatric adverse effects remain a major concern. Recent studies have identified a potential interaction between the INSTI, dolutegravir (DTG), and folate transport pathways at the placental barrier. We hypothesized that such interactions could also occur at the two major blood-brain interfaces: blood-cerebrospinal fluid barrier (BCSFB) and blood-brain barrier (BBB). To address this question, we evaluated the effect of two INSTIs, DTG and bictegravir (BTG), on folate transporters and receptor expression at the mouse BCSFB and the BBB in vitro, ex vivo and in vivo. We demonstrated that DTG but not BTG significantly downregulated the mRNA and/or protein expression of folate transporters (RFC/SLC19A1, PCFT/SLC46A1) in human and mouse BBB models in vitro, and mouse brain capillaries ex vivo. Our in vivo study further revealed a significant downregulation in Slc19a1 and Slc46a1 mRNA expression at the BCSFB and the BBB following a 14-day DTG oral treatment in C57BL/6 mice. However, despite the observed downregulatory effect of DTG in folate transporters/receptor at both brain barriers, a 14-day oral treatment of DTG-based ART did not significantly alter the brain folate level in animals. Interestingly, DTG treatment robustly elevated the mRNA and/or protein expression of pro-inflammatory cytokines and chemokines (Cxcl1, Cxcl2, Cxcl3, Il6, Il23, Il12) in primary cultures of mouse brain microvascular endothelial cells (BBB). DTG oral treatment also significantly upregulated proinflammatory cytokines and chemokine (Il6, Il1ß, Tnfα, Ccl2) at the BCSFB in mice. We additionally observed a downregulated mRNA expression of drug efflux transporters (Abcc1, Abcc4, and Abcb1a) and tight junction protein (Cldn3) at the CP isolated from mice treated with DTG. Despite the structural similarities, BTG only elicited minor effects on the markers of interest at both the BBB and BCSFB. In summary, our current data demonstrates that DTG but not BTG strongly induced inflammatory responses in a rodent BBB and BCSFB model. Together, these data provide valuable insights into the mechanism of DTG-induced brain toxicity, which may contribute to the pathogenesis of DTG-associated neuropsychiatric adverse effect.


Sujet(s)
Barrière hémato-encéphalique , Composés hétérocycliques 3 noyaux , Oxazines , Pipérazines , Pyridones , Animaux , Souris , Pipérazines/pharmacologie , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Composés hétérocycliques 3 noyaux/pharmacologie , Humains , Oxazines/pharmacologie , Inflammation/induit chimiquement , Inflammation/métabolisme , Souris de lignée C57BL , Femelle , Inhibiteurs de l'intégrase du VIH/pharmacologie , Inhibiteurs de l'intégrase du VIH/effets indésirables , Infections à VIH/traitement médicamenteux , Infections à VIH/métabolisme , Mâle , Antirétroviraux/effets indésirables , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques
13.
Oncol Res ; 32(7): 1197-1207, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948022

RÉSUMÉ

Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL's inhibitory effects on breast cancer using both in vitro (MCF7 and MDA-MB-468 cells) and in vivo (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL's anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and in vivo studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.


Sujet(s)
Autophagie , Tumeurs du sein , Pipérazines , Pyridines , Pyridones , Pyrimidinones , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Autophagie/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Femelle , Pyrimidinones/pharmacologie , Pyrimidinones/usage thérapeutique , Souris , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Souris nude , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules MCF-7
14.
Drug Dev Res ; 85(5): e22230, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38967729

RÉSUMÉ

The CDK4/CDK6 inhibitor palbociclib has shown the encouraging promise in the treatment of glioma. Here, we elucidated how palbociclib exerts suppressive functions in the M2 polarization of glioma-related microglia and the progression of glioma. Xenograft experiments were used to evaluate the function in vivo. The mRNA levels of transcription factor 12 (TCF12) and VSIG4 were detected by RT-qPCR, and their protein levels were assessed by immunoblotting. Cell migration was tested by wound-healing assay. Cell cycle distribution and M1/M2 microglia phenotype analysis were performed by flow cytometry. The levels of IFN-γ, TNF-α, IL-6,and TGF-ß were measured by ELISA. The TCF12/VSIG4 association was verified by luciferase reporter and chromatin immunoprecipitation (ChIP) assays. In U251 and LN229 glioma cells, TCF12 and VSIG4 were overexpressed, and palbociclib reduced their expression levels. TCF12 upregulation enhanced the proliferation and migration of glioma cells and the M2 polarization of glioma-associated microglia in vitro as well as the tumorigenicity of U251 glioma cells in vivo, which could be reversed by palbociclib. Mechanistically, TCF12 could enhance VSIG4 transcription and expression by binding to the VSIG4 promoter. TCF12 deficiency led to repression in glioma cell proliferation and migration as well as microglia M2 polarization, which could be abolished by increased VSIG4 expression. Our study reveals the novel TCF12/VSIG4 axis responsible for the efficacy of palbociclib in combating glioma, offering a rationale for the application of palbociclib in glioma treatment.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Gliome , Microglie , Pipérazines , Pyridines , Humains , Gliome/traitement médicamenteux , Gliome/métabolisme , Gliome/anatomopathologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pyridines/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Animaux , Lignée cellulaire tumorale , Souris , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Souris nude , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Souris de lignée BALB C , Antinéoplasiques/pharmacologie , Facteurs de transcription à motif basique hélice-boucle-hélice
15.
PLoS One ; 19(7): e0305612, 2024.
Article de Anglais | MEDLINE | ID: mdl-38990915

RÉSUMÉ

Breast cancer (BC) cells have a high risk of metastasis due to epithelial-mesenchymal transition (EMT). Palbociclib (CDK4/6 inhibitor) is an approved drug for BC treatment. However, the drug resistance and metastasis can impair the treatment outcome of Palbociclib. Understanding the mechanisms of EMT and Palbociclib drug resistance in BC is conducive to the formulation of novel therapeutic strategy. Here, we investigated the role of circHIAT1/miR-19a-3p/CADM2 axis in modulating EMT and Palbociclib resistance in BC. circHIAT1 and CADM2 were down-regulated in BC tissues and cell lines, and miR-19a-3p showed an up-regulation. circHIAT1 could interact with miR-19a-3p and suppress its activity, while miR-19a-3p functioned to negatively regulate CADM2. Forced over-expression of circHIAT1 could impaired the EMT status and migratory ability of BC cells, and this effect was inhibited by miR-19a-3p mimic. In addition, we also generated Palbociclib resistant BC cells, and showed that circHIAT1 and CADM2 were down-regulated in the resistant BC cells while miR-19a-3p showed an up-regulation. Forced circHIAT1 over-expression re-sensitized BC cells to Palbociclib treatment. Quercetin, a bioactive flavonoid, could suppressed the migration and invasion of BC cells, and re-sensitized BC cells to Palbociclib. The anti-cancer effect of quercetin could be attributed to its regulatory effect on circHIAT1/miR-19a-3p/CADM2 axis. In vivo tumorigenesis experiment further revealed that quercetin administration enhanced the anti-cancer effect of Palbociclib, an effect was dependent on the up-regulation of circHIAT1 by quercetin. In summary, this study identified quercetin as a potential anti-cancer compound to reverse Palbociclib resistance and impair EMT in BC cells by targeting circHIAT1/miR-19a-3p/CADM2 axis.


Sujet(s)
Tumeurs du sein , Kinase-6 cycline-dépendante , Résistance aux médicaments antinéoplasiques , Transition épithélio-mésenchymateuse , microARN , Pipérazines , Pyridines , Quercétine , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , microARN/génétique , microARN/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pipérazines/pharmacologie , Lignée cellulaire tumorale , Quercétine/pharmacologie , Animaux , Souris , Kinase-6 cycline-dépendante/métabolisme , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Kinase-4 cycline-dépendante/métabolisme , Kinase-4 cycline-dépendante/génétique , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Mouvement cellulaire/effets des médicaments et des substances chimiques , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe
16.
Anticancer Res ; 44(7): 2943-2952, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925834

RÉSUMÉ

BACKGROUND/AIM: Glioblastoma is an incurable cancer with limited treatment options and a low survival rate. Temozolomide is the standard marketed small-molecule agent for glioblastoma therapy; therefore, we aimed to find new drugs among the marketed medicines for brain diseases because of their cerebral migratory property and found lomerizine, used for the treatment of migraine. MATERIALS AND METHODS: We evaluated the effect of lomerizine and its metabolites against U251 glioblastoma cells and temozolomide-resistant cells, T98G and GB-1, caused by the expression of O(6)-methylguanine-DNA methyltransferase or P-glycoprotein, compared with temozolomide, and combined with it. The mechanism of action was investigated using inhibitors of necrosis or apoptosis. RESULTS: Lomerizine and its metabolite (M6) inhibited the proliferation of glioblastoma cells with greater potency and efficacy than temozolomide, including against temozolomide-resistant cells. The effects of lomerizine and M6 on glioblastoma were mainly attributed to the inhibition of proliferation because cells were not rescued by cell death inhibitors, such as necrosis or apoptosis inhibitors, although they were slightly rescued by necrostatin-1. Additionally, lomerizine and M6 combined with temozolomide were more effective at inhibiting the proliferation of U251 and GB-1 cells at some doses than single treatments. CONCLUSION: Lomerizine has been used for migraine treatment because of its brain-penetrating properties without serious side-effects; thus, it might potentially be expected to be used alone for glioblastoma, including temozolomide-resistant glioblastoma, or in combination with temozolomide.


Sujet(s)
Apoptose , Prolifération cellulaire , Glioblastome , Témozolomide , Humains , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Témozolomide/pharmacologie , Pipérazines/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Dacarbazine/pharmacologie , Dacarbazine/analogues et dérivés
17.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891871

RÉSUMÉ

Until the late 1800s, drug development was a chance finding based on observations and repeated trials and errors. Today, drug development must go through many iterations and tests to ensure it is safe, potent, and effective. This process is a long and costly endeavor, with many pitfalls and hurdles. The aim of the present review article is to explore what is needed for a molecule to move from the researcher bench to the patients' bedside, presented from an industry perspective through the development program of cariprazine. Cariprazine is a relatively novel antipsychotic medication, approved for the treatment of schizophrenia, bipolar mania, bipolar depression, and major depression as an add-on. It is a D3-preferring D3-D2 partial agonist with the highest binding to the D3 receptors compared to all other antipsychotics. Based on the example of cariprazine, there are several key factors that are needed for a molecule to move from the researcher bench to the patients' bedside, such as targeting an unmet medical need, having a novel mechanism of action, and a smart implementation of development plans.


Sujet(s)
Neuroleptiques , Pipérazines , Récepteur D3 de la dopamine , Humains , Neuroleptiques/usage thérapeutique , Neuroleptiques/pharmacologie , Pipérazines/usage thérapeutique , Pipérazines/pharmacologie , Récepteur D3 de la dopamine/métabolisme , Schizophrénie/traitement médicamenteux , Schizophrénie/métabolisme , Animaux , Trouble bipolaire/traitement médicamenteux , Trouble bipolaire/métabolisme , Développement de médicament
18.
Bioorg Med Chem ; 109: 117796, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38879996

RÉSUMÉ

The USFDA granted regular approval to Osimertinib (AZD9291) on March 2017, for treating individuals with metastatic Non-Small Cell Lung Cancer having EGFR T790M mutation. Clinically, Osimertinib stands at the forefront for the treatment of patients with Non-Small Cell Lung Cancer. Osimertinib forms a covalent bond with the Cys797 residue and predominantly spares binding to WT-EGFR, thereby reducing toxicity and enabling the administration of doses that effectively inhibit T790M. However, a high percentage of patients treated with Osimertinib (AZD9291) developed a tertiary cysteine797 to serine797 (C797S) mutation in the EGFR kinase domain, rendering resistance to it. This comprehensive review sheds light on the chemistry, computational aspects, structural features, and expansive spectrum of biological activities of Osimertinib and its analogues. The in-depth exploration of these facets serves as a valuable resource for medicinal chemists, empowering them to design better Osimertinib analogues. This exhaustive study not only provides insights into improving potency but also emphasizes considerations for mutant selectivity and optimizing pharmacokinetic properties. This review acts as a guiding beacon for the strategic design and development of next-generation Osimertinib analogues.


Sujet(s)
Acrylamides , Dérivés de l'aniline , Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Récepteurs ErbB , Tumeurs du poumon , Mutation , Inhibiteurs de protéines kinases , Acrylamides/composition chimique , Acrylamides/pharmacologie , Dérivés de l'aniline/composition chimique , Dérivés de l'aniline/pharmacologie , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Récepteurs ErbB/antagonistes et inhibiteurs , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/synthèse chimique , Relation structure-activité , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Structure moléculaire , Pipérazines/composition chimique , Pipérazines/pharmacologie , Indoles , Pyrimidines
19.
Toxicon ; 246: 107795, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-38849008

RÉSUMÉ

Amphotericin B (AmB) induced liver and kidney injury is often responsible for hepatic and renal dysfunction. Therefore, the protection strategy on liver and renal functions in patients treated with AmB should be emphasized. In this paper, diammonium glycyrrhizinate (DG) and piperazine ferulate (PF) were taken as the research object to study its hepatoprotective and neuroprotective effect on AmB-induced liver and kidney damage in vitro and in vivo. The microplate method and ELISA kits were employed for the biochemical detection in the serum and urine of mice. Flow cytometric analysis and western blotting analysis were conducted to study the mechanism of DG and PF. Our results confirmed the prevention capacity of DG and PF on AmB-induced liver and kidney injury through the alleviation of pathological changes and enzyme reducing action. Furthermore, DG and PF suppressed ROS-mediated mitochondrial apoptosis in AmB-treated mice and cells through Caspase pathway and Caspase-independent AIF pathway. In summary, DG and PF could protect AmB-induced hepatotoxicity and nephrotoxicity by disrupting oxidative stress and apoptosis.


Sujet(s)
Amphotéricine B , Apoptose , Lésions hépatiques dues aux substances , Acide glycyrrhizique , Neuroprotecteurs , Animaux , Apoptose/effets des médicaments et des substances chimiques , Souris , Acide glycyrrhizique/pharmacologie , Lésions hépatiques dues aux substances/prévention et contrôle , Lésions hépatiques dues aux substances/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Amphotéricine B/toxicité , Mâle , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pipérazine/pharmacologie , Agents protecteurs/pharmacologie
20.
Nat Commun ; 15(1): 5209, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890388

RÉSUMÉ

Despite the importance of spliceosome core components in cellular processes, their roles in cancer development, including hepatocellular carcinoma (HCC), remain poorly understood. In this study, we uncover a critical role for SmD2, a core component of the spliceosome machinery, in modulating DNA damage in HCC through its impact on BRCA1/FANC cassette exons and expression. Our findings reveal that SmD2 depletion sensitizes HCC cells to PARP inhibitors, expanding the potential therapeutic targets. We also demonstrate that SmD2 acetylation by p300 leads to its degradation, while HDAC2-mediated deacetylation stabilizes SmD2. Importantly, we show that the combination of Romidepsin and Olaparib exhibits significant therapeutic potential in multiple HCC models, highlighting the promise of targeting SmD2 acetylation and HDAC2 inhibition alongside PARP inhibitors for HCC treatment.


Sujet(s)
Carcinome hépatocellulaire , Exons , Tumeurs du foie , Phtalazines , Pipérazines , Inhibiteurs de poly(ADP-ribose) polymérases , Splicéosomes , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Acétylation , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Splicéosomes/métabolisme , Splicéosomes/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Phtalazines/pharmacologie , Exons/génétique , Pipérazines/pharmacologie , Animaux , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Depsipeptides/pharmacologie , Depsipeptides/usage thérapeutique , Souris , Altération de l'ADN/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE