Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Int Immunopharmacol ; 139: 112745, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39059099

RÉSUMÉ

Acute kidney injury (AKI) manifests as a clinical syndrome characterised by the rapid accumulation of metabolic wastes, such as blood creatinine and urea nitrogen, leading to a sudden decline in renal function. Currently, there is a lack of specific therapeutic drugs for AKI. Previously, we identified gastrin-releasing peptide receptor (GRPR) as a pathogenic factor in AKI. In this study, we investigated the therapeutic potential of a novel Chinese medicine monomer, aurantiamide (AA), which exhibits structural similarities to our previously reported GRPR antagonist, RH-1402. We compared the therapeutic efficacy of AA with RH-1402 both in vitro and in vivo using various AKI models. Our results demonstrated that, in vitro, AA attenuated injury, necroptosis, and inflammatory responses in human renal tubular epithelial cells subjected to repeated hypoxia/reoxygenation and lipopolysaccharide stimulation. In vivo, AA ameliorated renal tubular injury and inflammation in mouse models of ischemia/reperfusion and cecum ligation puncture-induced AKI, surpassing the efficacy of RH-1402. Furthermore, molecular docking and cellular thermal shift assay confirmed GRPR as a direct target of AA, which was further validated in primary cells. Notably, in GRPR-silenced HK-2 cells and GRPR systemic knockout mice, AA failed to mitigate renal inflammation and injury, underscoring the importance of GRPR in AA's mechanism of action. In conclusion, our study has demonstrated that AA serve as a novel antagonist of GRPR and a promising clinical candidate for AKI treatment.


Sujet(s)
Atteinte rénale aigüe , Souris de lignée C57BL , Souris knockout , Nécroptose , Récepteur bombésine , Animaux , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/anatomopathologie , Humains , Nécroptose/effets des médicaments et des substances chimiques , Souris , Mâle , Lignée cellulaire , Récepteur bombésine/métabolisme , Récepteur bombésine/antagonistes et inhibiteurs , Inflammation/traitement médicamenteux , Modèles animaux de maladie humaine , Rein/anatomopathologie , Rein/effets des médicaments et des substances chimiques , Rein/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique
2.
Acta Pharmacol Sin ; 45(8): 1673-1685, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38641746

RÉSUMÉ

Acute kidney injury (AKI) is defined as sudden loss of renal function characterized by increased serum creatinine levels and reduced urinary output with a duration of 7 days. Ferroptosis, an iron-dependent regulated necrotic pathway, has been implicated in the progression of AKI, while ferrostatin-1 (Fer-1), a selective inhibitor of ferroptosis, inhibited renal damage, oxidative stress and tubular cell death in AKI mouse models. However, the clinical translation of Fer-1 is limited due to its lack of efficacy and metabolic instability. In this study we designed and synthesized four Fer-1 analogs (Cpd-A1, Cpd-B1, Cpd-B2, Cpd-B3) with superior plasma stability, and evaluated their therapeutic potential in the treatment of AKI. Compared with Fer-1, all the four analogs displayed a higher distribution in mouse renal tissue in a pharmacokinetic assay and a more effective ferroptosis inhibition in erastin-treated mouse tubular epithelial cells (mTECs) with Cpd-A1 (N-methyl-substituted-tetrazole-Fer-1 analog) being the most efficacious one. In hypoxia/reoxygenation (H/R)- or LPS-treated mTECs, treatment with Cpd-A1 (0.25 µM) effectively attenuated cell damage, reduced inflammatory responses, and inhibited ferroptosis. In ischemia/reperfusion (I/R)- or cecal ligation and puncture (CLP)-induced AKI mouse models, pre-injection of Cpd-A1 (1.25, 2.5, 5 mg·kg-1·d-1, i.p.) dose-dependently improved kidney function, mitigated renal tubular injury, and abrogated inflammation. We conclude that Cpd-A1 may serve as a promising therapeutic agent for the treatment of AKI.


Sujet(s)
Atteinte rénale aigüe , Ferroptose , Souris de lignée C57BL , Phénylènediamines , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/métabolisme , Souris , Mâle , Phénylènediamines/pharmacologie , Phénylènediamines/usage thérapeutique , Cyclohexylamines/pharmacologie , Cyclohexylamines/usage thérapeutique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme
3.
Int J Biol Macromol ; 248: 125811, 2023 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-37467831

RÉSUMÉ

Circular RNA (circRNA) has been implicated in liver fibrosis and modulated by multiple elusive molecular mechanisms, while the effects of N6-methyladenosine (m6A) modification on circRNA are still elusive. Herein, we identify circIRF2 from our circRNA sequencing data, which decreased in liver fibrogenesis stage and restored in resolution stage, indicating that dysregulated circIRF2 may be closely associated with liver fibrosis. Gain/loss-of-function analysis was performed to evaluate the effects of circIRF2 on liver fibrosis at both the fibrogenesis and resolution in vivo. Ectopic expression of circIRF2 attenuated liver fibrogenesis and HSCs activation at the fibrogenesis stage, whereas downregulation of circIRF2 impaired mouse liver injury repair and inflammation resolution. Mechanistically, YTHDF2 recognized m6A-modified circIRF2 and diminished circIRF2 stability, partly accounting for the decreased circIRF2 in liver fibrosis. Microarray was applied to investigate miRNAs regulated by circIRF2, our data elucidate cytoplasmic circIRF2 may directly harbor miR-29b-1-5p and competitively relieve its inhibitory effect on FOXO3, inducing FOXO3 nuclear translocation and accumulation. Clinically, circIRF2 downregulation was prevalent in liver fibrosis patients compared with healthy individuals. In summary, our findings offer a novel insight into m6A modification-mediated regulation of circRNA and suggest that circIRF2 may be an exploitable prognostic marker and/or therapeutic target for liver fibrosis.


Sujet(s)
microARN , ARN circulaire , Souris , Animaux , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Cellules étoilées du foie/métabolisme , Cirrhose du foie/anatomopathologie , microARN/génétique , microARN/métabolisme , Facteurs de transcription/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Protéines de liaison à l'ARN/métabolisme
4.
Br J Pharmacol ; 180(20): 2641-2660, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37248964

RÉSUMÉ

BACKGROUND AND PURPOSE: Necroptosis plays an essential role in acute kidney injury and is mediated by receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and mixed lineage kinase domain-like pseudokinase (MLKL). A novel RIPK3 inhibitor, compound 42 (Cpd-42) alleviates the systemic inflammatory response. The current study was designed to investigate whether Cpd-42 exhibits protective effects on acute kidney injury and reveal the underlying mechanisms. EXPERIMENTAL APPROACH: The effects of Cpd-42 were determined in vivo through cisplatin- and ischaemia/reperfusion (I/R)-induced acute kidney injury and in vitro through cisplatin- and hypoxia/re-oxygenation (H/R)-induced cell damage. Transmission electron microscopy and periodic acid-Schiff staining were used to identify renal pathology. Cellular thermal shift assay and RIPK3-knockout mouse renal tubule epithelial cells were used to explore the relationship between Cpd-42 and RIPK3. Molecular docking and site-directed mutagenesis were used to determine the binding site of RIPK3 with Cpd-42. KEY RESULTS: Cpd-42 reduced human proximal tubule epithelial cell line (HK-2) cell damage, necroptosis and inflammatory responses in vitro. Furthermore, in vivo, cisplatin- and I/R-induced acute kidney injury was alleviated by Cpd-42 treatment. Cpd-42 inhibited necroptosis by interacting with two key hydrogen bonds of RIPK3 at Thr94 and Ser146, which further blocked the phosphorylation of RIPK3 and mitigated acute kidney injury. CONCLUSION AND IMPLICATIONS: Acting as a novel RIPK3 inhibitor, Cpd-42 reduced kidney damage, inflammatory response and necroptosis in acute kidney injury by binding to sites Thr94 and Ser146 on RIPK3. Cpd-42 could be a promising treatment for acute kidney injury.


Sujet(s)
Atteinte rénale aigüe , Cisplatine , Souris , Animaux , Humains , Cisplatine/pharmacologie , Nécroptose , Simulation de docking moléculaire , Atteinte rénale aigüe/métabolisme , Protein kinases/métabolisme , Souris knockout , Apoptose , Receptor-Interacting Protein Serine-Threonine Kinases
5.
Int Immunopharmacol ; 112: 109262, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36166972

RÉSUMÉ

Acute kidney injury (AKI) is a clinical syndrome that is defined as a sudden decline in renal function and characterized by inflammation and programmed cell death of renal tubular epithelial cells. Necroptosis is a form of regulated cell death that requires activation of receptor interacting protein kinase 3 (RIPK3) and its phosphorylation of the substrate MLKL. RIPK3 plays an important role in acute kidney injury, and hence developing its inhibitors is considered as one of the promising strategies aimed at prevention and treatment of AKI. Recently, we discovered AZD5423 as a novel potent RIPK3 inhibitor using a computer-aided hybrid virtual screening strategy according to three-dimensional structure of RIPK3. Our findings revealed that AZD5423 strongly inhibits activation of RIPK3, and MLKL phosphorylation upon cisplatin-, hypoxia/reoxygenation (H/R)- and TNF-α stimuli as compared with GSK872, which is a previously identified RIPK3 inhibitor. Importantly, AZD5423 exerts effective protection against cisplatin- and ischemia/reperfusion (I/R)-induced AKI mouse model. The results of cellular thermal shift assay and experiments in RIPK3 knockout cells indicated that AZD5423 could directly target RIPK3 to inhibit RIPK3 kinase activity. Mechanistically, the docking of AZD5423 and RIPK3 suggested that the kinase domain of RIPK3 for Lys50, Arg313, Lys29, Arg37 might form hydrogen bonds with AZD5423. Site-directed mutagenesis further revealed that AZD5423 reduces injury response via interacting with the key RIPK3 amino acid residues of Lys50 and Arg313. In conclusion, our study has demonstrated that AZD5423 may serve as a potent inhibitor of RIPK3 kinase and a promising clinical candidate for AKI treatment.


Sujet(s)
Atteinte rénale aigüe , Nécroptose , Souris , Animaux , Cisplatine/usage thérapeutique , Facteur de nécrose tumorale alpha/métabolisme , Souris de lignée C57BL , Receptor-Interacting Protein Serine-Threonine Kinases/métabolisme , Atteinte rénale aigüe/induit chimiquement , Inflammation/métabolisme , Acides aminés
6.
Int Immunopharmacol ; 110: 109034, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35834952

RÉSUMÉ

Hepatic fibrosis is an essential pathology of multiple chronicliverdiseases. The aim of this study was to investigate the role of miR-301a-3p in hepatic fibrosis. We found that miR-301a-3p was upregulated in hepatic fibrosis patients and in culture-activated human hepatic stellate cells (HSCs). Interestingly, miR-301a-3p expression was increased in hepatic fibrosis progression mice while decreased in hepatic fibrosis recovery mice, indicating that miR-301a-3p may participate in the hepatic fibrosis pathology. Functionally, the effects of miR-301a-3p both on hepatic fibrosis progression and regression were assessed in vivo. Inhibiting miR-301a-3p amelioratedmouse liver fibrogenesis and collagen deposition and suppressed HSC activation and fibrogenic factor expression. Whereas, in hepatic fibrosis regression, upregulating miR-301a-3p impaired mouse hepatic fibrosis recovery by inducing HSC activation and triggering inflammation. Consistently, gain-of-function and loss-of-function analysis of miR-301a-3p were performed to evaluate its effects on human HSCs LX-2 cell. We found that suppressing miR-301a-3p inhibited LX-2 cell activation and proliferation, and induced LX-2 cell apoptosis, accompaniedby decreased fibrotic mediators expression. Collectively, these findings suggest miR-301a-3p drives liver fibrogenesis and HSC activation in hepatic fibrosis. Mechanistically, we demonstrated miR-301a-3p binds directly to phosphatase and tensin homolog (PTEN) by luciferase reporter analysis, pull-down, and RIP assay. Indicating that miR-301a-3p plays a critical rolein promotingliverfibrogenesis viamodulating the PTEN/platelet derived growth factor ß (PDGFR-ß) pathway. In conclusion, our findings demonstrate that miR-301a-3p expression is closely correlated with hepatic fibrosis pathology, and that enhancing miR-301a-3p maintains the HSC profibrogenic phenotype, triggers inflammatoryresponses, promotes fibrogenic factor production, and further exacerbates liver fibrogenesis. These findings suggest that miR-301a-3p may serve as a promising diagnostic and prognosis biomarker for hepatic fibrosis treatment.


Sujet(s)
Cellules étoilées du foie , microARN/métabolisme , Animaux , Prolifération cellulaire , Cellules étoilées du foie/anatomopathologie , Humains , Cirrhose du foie/métabolisme , Souris , microARN/génétique , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Protéines proto-oncogènes c-sis/pharmacologie , Transduction du signal
7.
Eur J Pharmacol ; 929: 175116, 2022 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-35780825

RÉSUMÉ

Renal fibrosis, a common feature of chronic kidney disease, causes the progressive loss of renal function, in which TGF-ß1 plays a critical role. In this study, we found that expression levels of TGF-ß1 and its receptor 1 (TGF-ßR1) were both significantly increased in obstructive fibrosis kidneys. AZ12601011 is a small molecular inhibitor of TGF-ßR1; however, its therapeutic potential for renal fibrosis remains unclear. During the experiments, AZ12601011 was applied to various models of renal fibrosis followed by unilateral ureteral obstruction (UUO) and ischemia/reperfusion (I/R) in vivo, in addition to renal tubular epithelial cells (TECs) challenged by hypoxia/reoxygenation (H/R) and TGF-ß1in vitro. Our results revealed that AZ12601011 ameliorated renal injuries and fibrosis shown by PAS, HE, and Masson staining, which was consistent with the decrease in Col-1 and α-SMA expression in the kidneys from UUO and I/R mice. Similarly, in vitro data showed that AZ12601011 inhibited the induction of Col-1 and α-SMA in both TECs treated with TGF-ß1 and H/R. In addition, the results of cellular thermal shift assay (CETSA), molecular docking, and western bolt indicated that AZ12601011 could directly bind to TGF-ßR1 and block activation of the downstream Smad3. Taken together, our findings suggest that AZ12601011 can attenuate renal fibrosis by blocking the TGF-ß/Smad3 signaling pathway and it might serve as a promising clinical candidate in the fight against fibrotic kidney diseases.


Sujet(s)
Maladies du rein , Insuffisance rénale chronique , Obstruction urétérale , Animaux , Fibrose , Rein/anatomopathologie , Maladies du rein/traitement médicamenteux , Maladies du rein/métabolisme , Maladies du rein/prévention et contrôle , Souris , Simulation de docking moléculaire , Récepteur de type I du facteur de croissance transformant bêta/métabolisme , Insuffisance rénale chronique/anatomopathologie , Facteur de croissance transformant bêta-1/métabolisme , Obstruction urétérale/complications , Obstruction urétérale/traitement médicamenteux
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE