Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 410
Filtrer
1.
Cell Biol Toxicol ; 40(1): 60, 2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39073694

RÉSUMÉ

Triptolide (TP) is a major active and toxic composition of the Chinese medicine Tripterygium wilfordii Hook. F. (TWHF), exhibiting various therapeutic bioactivities. Among the toxic effects, the hepatotoxicity of TP deserves serious attention. Previously, our research group proposed a new view of TP-related hepatotoxicity: hepatic hypersensitivity under lipopolysaccharide (LPS) stimulation. However, the mechanism of TP/LPS-induced hepatic hypersensitivity remains unclear. In this study, we investigated the mechanism underlying TP/LPS-induced hypersensitivity from the perspective of the inhibition of proteasome activity, activated endoplasmic reticulum stress (ERS)-related apoptosis, and the accumulation of reactive oxygen species (ROS). Our results showed that N-acetylcysteine (NAC), a common ROS inhibitor, decreased the expression of cleaved caspase-3 and cleaved PARP, which are associated with FLIP enhancement. Moreover, 4-phenylbutyric acid (4-PBA), an ERS inhibitor, was able to alleviate TP/LPS-induced hepatotoxicity by reducing ERS-related apoptosis protein expression (GRP78, p-eIF2α/eIF2α, ATF4, CHOP, cleaved caspase-3 and cleaved PARP) and ROS levels, with ATF4 being an indispensable mediator. In addition, the proteasome activity inhibitor MG-132 further aggravated ERS-related apoptosis, which indicated that the inhibition of proteasome activity also plays an important role in TP/LPS-related liver injuries. In summary, we propose that TP/LPS may upregulate the activation of ERS-associated apoptosis by inhibiting proteasome activity and enhancing ROS production through ATF4.


Sujet(s)
Acétylcystéine , Apoptose , Diterpènes , Chaperonne BiP du réticulum endoplasmique , Stress du réticulum endoplasmique , Composés époxy , Lipopolysaccharides , Phénanthrènes , Proteasome endopeptidase complex , Inhibiteurs du protéasome , Espèces réactives de l'oxygène , Phénanthrènes/pharmacologie , Phénanthrènes/toxicité , Diterpènes/pharmacologie , Diterpènes/toxicité , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lipopolysaccharides/toxicité , Composés époxy/toxicité , Composés époxy/pharmacologie , Animaux , Espèces réactives de l'oxygène/métabolisme , Proteasome endopeptidase complex/métabolisme , Inhibiteurs du protéasome/pharmacologie , Acétylcystéine/pharmacologie , Facteur de transcription ATF-4/métabolisme , Phénylbutyrates/pharmacologie , Souris , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/anatomopathologie , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme , Caspase-3/métabolisme , Mâle , Leupeptines
2.
J Appl Toxicol ; 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39030796

RÉSUMÉ

Bile acid homeostasis is crucial for the normal physiological functioning of the liver. Disruptions in bile acid profiles are closely linked to the occurrence of cholestatic liver injury. As part of our diagnostic and therapeutic approach, we aimed to investigate the disturbance in bile acid profiles during cholestasis and its correlation with cholestatic liver injury. Before the occurrence of liver injury, alterations in bile acid profiles were detected in both plasma and liver between 8 and 16 h, persisting up to 96 h. TCA, TCDCA, and TUDCA in the plasma, as well as TCA, TUDCA, TCDCA, TDCA, TLCA, and THDCA in the liver, emerged as early sensitive and potential markers for diagnosing ANIT-induced cholestasis at 8-16 h. The distinguishing features of ANIT-induced liver injury were as follows: T-BAs exceeding G-BAs and serum biochemical indicators surpassing free bile acids. Notably, plasma T-BAs, particularly TCA, exhibited higher sensitivity to cholestatic hepatotoxicity compared with serum enzyme activity and liver histopathology. Further investigation revealed that TCA exacerbated ANIT-induced liver injury by elevating liver function enzyme activity, inflammation, and bile duct proliferation and promoting the migration of bile duct epithelial cell. Nevertheless, no morphological changes or alterations in transaminase activity indicative of liver damage were observed in the rats treated with TCA alone. Additionally, there were no changes in bile acid profiles or inflammatory responses under physiological conditions with maintained bile acid homeostasis. In summary, our findings suggest that taurine-conjugated bile acids in both plasma and liver, particularly TCA, can serve as early and sensitive markers for predicting intrahepatic cholestatic drugs and can act as potent exacerbators of cholestatic liver injury progression. However, exogenous TCA does not induce liver injury under physiological conditions where bile acid homeostasis is maintained.

3.
Eur J Med Chem ; 276: 116705, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-39067439

RÉSUMÉ

Histologic spectrum studies in patients revealed fatty acid binding proteins 1 (FABP1) as a potential new target for the treatment of metabolic associated fatty liver disease. However, there is no FABP1 inhibitor has been reported except the first-in-class FABP1 inhibitor bearing acid moiety reported by our laboratory. Herein, we firstly report the structure-activity relationship of novel non-carboxylic acid FABP1 inhibitors, which resulted in the identification of the potent and selective FABP1 inhibitor 30. The IC50 value of compound 30 for subtype FABP4 in the same family was greater than 80 µM. Moreover, compound 30 significantly alleviated the hepatic steatosis in DIO mice, which is equivalent to that of clinical drug obeticholic acid. This study might be provided a promising probe for the development of FABP1 inhibitors and thus can help to further elucidate the pharmacology of FABP1.


Sujet(s)
Conception de médicament , Protéines de liaison aux acides gras , Protéines de liaison aux acides gras/antagonistes et inhibiteurs , Protéines de liaison aux acides gras/métabolisme , Animaux , Relation structure-activité , Souris , Humains , Structure moléculaire , Relation dose-effet des médicaments , Mâle , Souris de lignée C57BL
4.
Br J Cancer ; 131(1): 77-89, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38796598

RÉSUMÉ

BACKGROUND: Due to insufficient knowledge about key molecular events, Hepatocellular carcinoma (HCC) lacks effective treatment targets. Spliceosome-related genes were significantly altered in HCC. Oncofetal proteins are ideal tumor therapeutic targets. Screening of differentially expressed Spliceosome-related oncofetal protein in embryonic liver development and HCC helps discover effective therapeutic targets for HCC. METHODS: Differentially expressed spliceosome genes were analysis in fetal liver and HCC through bioinformatics analysis. Small nuclear ribonucleoprotein polypeptide E (SNRPE) expression was detected in fetal liver, adult liver and HCC tissues. The role of SNRPE in HCC was performed multiple assays in vitro and in vivo. SNRPE-regulated alternative splicing was recognized by RNA-Seq and confirmed by multiple assays. RESULTS: We herein identified SNRPE as a crucial oncofetal splicing factor, significantly associated with the adverse prognosis of HCC. SOX2 was identified as the activator for SNRPE reactivation. Efficient knockdown of SNRPE resulted in the complete cessation of HCC tumorigenesis and progression. Mechanistically, SNRPE knockdown reduced FGFR4 mRNA expression by triggering nonsense-mediated RNA decay. A partial inhibition of SNRPE-induced malignant progression of HCC cells was observed upon FGFR4 knockdown. CONCLUSIONS: Our findings highlight SNRPE as a novel oncofetal splicing factor and shed light on the intricate relationship between oncofetal splicing factors, splicing events, and carcinogenesis. Consequently, SNRPE emerges as a potential therapeutic target for HCC treatment. Model of oncofetal SNRPE promotes HCC tumorigenesis by regulating the AS of FGFR4 pre-mRNA.


Sujet(s)
Épissage alternatif , Carcinogenèse , Carcinome hépatocellulaire , Tumeurs du foie , Récepteur FGFR4 , Animaux , Humains , Souris , Carcinogenèse/génétique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Souris nude , Pronostic , Récepteur FGFR4/génétique , Récepteur FGFR4/métabolisme
5.
Int J Biochem Cell Biol ; 172: 106585, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38734232

RÉSUMÉ

Tamoxifen is an estrogen receptor modulator that has been reported to alleviate hepatic lipid accumulation in mice, but the mechanism is still unclear. Peroxisome fatty acid ß-oxidation is the main metabolic pathway for the overload of long-chain fatty acids. As long-chain fatty acids are a cause of hepatic lipid accumulation, the activation of peroxisome fatty acid ß-oxidation might be a novel therapeutic strategy for metabolic associated fatty liver disease. In this study, we investigated the mechanism of tamoxifen against hepatic lipid accumulation based on the activation of peroxisome fatty acid ß-oxidation. Tamoxifen reduced liver long-chain fatty acids and relieved hepatic lipid accumulation in high fat diet mice without sex difference. In vitro, tamoxifen protected primary hepatocytes against palmitic acid-induced lipotoxicity. Mechanistically, the RNA-sequence of hepatocytes isolated from the liver revealed that peroxisome fatty acid ß-oxidation was activated by tamoxifen. Protein and mRNA expression of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase were significantly increased in vivo and in vitro. Small interfering RNA enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase in primary hepatocytes abolished the therapeutic effects of tamoxifen in lipid accumulation. In conclusion, our results indicated that tamoxifen could relieve hepatic lipid accumulation in high fat diet mice based on the activation of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase-mediated peroxisome fatty acids ß-oxidation.


Sujet(s)
Énoyl-CoA hydratases , Hépatocytes , Métabolisme lipidique , Foie , Souris de lignée C57BL , Oxydoréduction , Péroxysomes , Tamoxifène , Animaux , Tamoxifène/pharmacologie , Souris , Métabolisme lipidique/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Oxydoréduction/effets des médicaments et des substances chimiques , Mâle , Péroxysomes/métabolisme , Péroxysomes/effets des médicaments et des substances chimiques , Énoyl-CoA hydratases/métabolisme , Énoyl-CoA hydratases/génétique , Régulation positive/effets des médicaments et des substances chimiques , Alimentation riche en graisse/effets indésirables , Femelle , Acides gras/métabolisme
6.
Eur J Med Chem ; 270: 116358, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38574638

RÉSUMÉ

The fatty acid-binding protein 1 (FABP1) is a fatty acid transporter protein that is considered as an emerging target for metabolic diseases. Despite forceful evidence that the inhibition of FABP1 is essential for ameliorating NASH, pharmacological control and validation of FABP1 are hindered by a lack of relevant inhibitors as pharmacological tool. Therefore, the development of effective FABP1 inhibitors is a current focus of research. Herein, we firstly reported the comprehensive structure-activity relationship (SAR) study of novel FABP1 inhibitors derived from high throughput screening of our in-house library, which resulting in the identification of the optimal compound 44 (IC50 = 4.46 ± 0.54 µM). Molecular docking studies revealed that 44 forms stable hydrogen bonds with amino acids around the active pocket of FABP1. Moreover, 44 alleviated the typical histological features of fatty liver in NASH mice, including steatosis, lobular inflammation, ballooning and fibrosis. Additionally, 44 has been demonstrated to have lipid metabolism regulating, anti-oxidative stress and hepatoprotective properties. This study might be provided a promising insight into the field of NASH and inspiration for the development of FABP1 inhibitors.


Sujet(s)
Stéatose hépatique non alcoolique , Souris , Animaux , Stéatose hépatique non alcoolique/traitement médicamenteux , Simulation de docking moléculaire , Métabolisme lipidique , Fibrose , Protéines de liaison aux acides gras/métabolisme , Foie/métabolisme
7.
Molecules ; 29(7)2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38611852

RÉSUMÉ

Moonlighting enzymes are multifunctional proteins that perform multiple functions beyond their primary role as catalytic enzymes. Extensive research and clinical practice have demonstrated their pivotal roles in the development and progression of cancer, making them promising targets for drug development. This article delves into multiple notable moonlighting enzymes, including GSK-3, GAPDH, and ENO1, and with a particular emphasis on an enigmatic phosphatase, PTP4A3. We scrutinize their distinct roles in cancer and the mechanisms that dictate their ability to switch roles. Lastly, we discuss the potential of an innovative approach to develop drugs targeting these moonlighting enzymes: target protein degradation. This strategy holds promise for effectively tackling moonlighting enzymes in the context of cancer therapy.


Sujet(s)
Glycogen Synthase Kinase 3 , Tumeurs , Humains , Phosphoric monoester hydrolases , Tumeurs/traitement médicamenteux , Catalyse , Développement de médicament , Protéines tumorales , Protein Tyrosine Phosphatases
8.
J Nanobiotechnology ; 22(1): 113, 2024 Mar 16.
Article de Anglais | MEDLINE | ID: mdl-38491372

RÉSUMÉ

Wounds are one of the most common health issues, and the cost of wound care and healing has continued to increase over the past decade. In recent years, there has been growing interest in developing innovative strategies to enhance the efficacy of wound healing. Tetrahedral framework nucleic acids (tFNAs) have emerged as a promising tool for wound healing applications due to their unique structural and functional properties. Therefore, it is of great significance to summarize the applications of tFNAs for wound healing. This review article provides a comprehensive overview of the potential of tFNAs as a novel therapeutic approach for wound healing. In this review, we discuss the possible mechanisms of tFNAs in wound healing and highlight the role of tFNAs in modulating key processes involved in wound healing, such as cell proliferation and migration, angiogenesis, and tissue regeneration. The targeted delivery and controlled release capabilities of tFNAs offer advantages in terms of localized and sustained delivery of therapeutic agents to the wound site. In addition, the latest research progress on tFNAs in wound healing is systematically introduced. We also discuss the biocompatibility and biosafety of tFNAs, along with their potential applications and future directions for research. Finally, the current challenges and prospects of tFNAs are briefly discussed to promote wider applications.


Sujet(s)
Acides nucléiques , Prolifération cellulaire , Cicatrisation de plaie
9.
J Colloid Interface Sci ; 661: 520-532, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38308892

RÉSUMÉ

Metal-organic frameworks (MOFs) have great potential for combating pathogenic bacterial infections and are expected to become an alternative to antibiotics. However, organic linkers obstruct and saturate the inorganic nodes of MOF structures, making it challenging to utilize the applied potential of metal centers. Here, we combined controlled ligand decarboxylation with noble metal nanoparticles to rationally remodel MIL-53, resulting in a hybrid nanozyme (AgAu@QMIL-53, AAQM) with excellent multiple enzyme-like activities that both eradicate bacteria and promote diabetic wound healing. Specifically, benefitting from oxidase (OXD)-like and peroxidase (POD)-like activities, AAQM converts oxygen (O2) and hydrogen peroxide (H2O2) into superoxide anion radicals (O2-) and hydroxyl radicals (OH) to eradicate bacteria. In in vitro antibacterial experiments, AAQM exhibited favorable killing efficacy against Pseudomonas aeruginosa (P. aeruginosa) and methicillin-resistant Staphylococcus aureus (MRSA) (>99 %). Notably, due to its superoxide (SOD)-like activity and outstanding reactive nitrogen species (RNS) elimination capacity, AAQM can produce adequate O2 and alleviate oxidative stress in diabetic wounds. Benefiting from the rational modification of MIL-53, the synthesized hybrid nanozyme can effectively kill bacteria while alleviating oxidative stress and ultimately promote infected diabetic wound healing. Overall, this biomimetic enzyme-catalyzed strategy will bring enlightenment to the design of self-antibacterial agents for efficient disinfection and wound healing simultaneously.


Sujet(s)
Diabète , Staphylococcus aureus résistant à la méticilline , Humains , Désinfection , Peroxyde d'hydrogène , Antibactériens/pharmacologie
10.
Ecotoxicol Environ Saf ; 272: 116072, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38342011

RÉSUMÉ

Triptolide (TP) is the major bioactive component of traditional Chinese medicine Tripterygium wilfordii Hook. F., a traditional Chinese medicinal plant categorized within the Tripterygium genus of the Celastraceae family. It is recognized for its therapeutic potential in addressing a multitude of diseases. Nonetheless, TP is known to exhibit multi-organ toxicity, notably hepatotoxicity, which poses a significant concern for the well-being of patients undergoing treatment. The precise mechanisms responsible for TP-induced hepatotoxicity remain unresolved. In our previous investigation, it was determined that TP induces heightened hepatic responsiveness to exogenous lipopolysaccharide (LPS). Additionally, natural killer (NK) cells were identified as a crucial effector responsible for mediating hepatocellular damage in this context. However, associated activating receptors and the underlying mechanisms governing NK cell represented innate lymphoid cell (ILC) activation remained subjects of inquiry and were not yet investigated. Herein, activating receptor Killer cell lectin like receptor K1 (NKG2D) of group 1 ILCs was specifically upregulated in TP- and LPS-induced acute liver failure (ALF), and in vivo blockade of NKG2D significantly reduced group 1 ILC mediated cytotoxicity and mitigated TP- and LPS-induced ALF. NKG2D ligand UL16-binding protein-like transcript 1 (MULT-1) was found upregulated in liver resident macrophages (LRMs) after TP administration, and LRMs did exhibit NK cell activating effect. Furthermore, M1 polarization of LRMs cells was observed, along with an elevation in intracellular tumor necrosis factor (TNF)-α levels. In vivo neutralization of TNF-α significantly alleviated TP- and LPS-induced ALF. In conclusion, the collaborative role of group 1 ILCs and LRMs in mediating hepatotoxicity was confirmed in TP- and LPS-induced ALF. TP-induced MULT-1 expression in LRMs was the crucial mechanism in the activation of group 1 ILCs via MULT-1-NKG2D signal upon LPS stimulation, emphasizing the importance of infection control after TP administration.


Sujet(s)
Lésions hépatiques dues aux substances , Diterpènes , Phénanthrènes , Animaux , Humains , Souris , Sous-famille K des récepteurs de cellules NK de type lectine , Lipopolysaccharides/toxicité , Immunité innée , Phénanthrènes/toxicité , Composés époxy/toxicité , Cellules tueuses naturelles , Macrophages , Lésions hépatiques dues aux substances/étiologie
11.
Chem Res Toxicol ; 37(2): 407-418, 2024 02 19.
Article de Anglais | MEDLINE | ID: mdl-38284557

RÉSUMÉ

Triptolide (TP) is a remarkable anti-inflammatory and immunosuppressive component separated from Tripterygium wilfordii Hook. F. However, its hepatotoxicity limits its application in the clinical. Our group has proposed a new perspective on TP-induced hepatotoxicity, in which TP enhances liver hypersensitivity upon lipopolysaccharide (LPS) stimulation. Because the cause of the disease is unknown, there is currently no uniform treatment available. In this study, we attempted to determine whether the GSK-3ß-JNK pathway affects liver damage and its regulatory mechanism in response to TP/LPS costimulation. In addition, we investigated the effect of CsA or the GSK 3ß inhibitor CHIR-98014 on TP/LPS-induced hepatotoxicity. The results showed that the TP/LPS cotreatment mice exhibited obvious hepatotoxicity, as indicated by a remarkable increase in the serum ALT and AST levels, glycogen depletion, GSK 3ß-JNK upregulation, and increased apoptosis. Instead of the specific knockdown of JNK1, the specific knockdown of JNK2 had a protective effect. Additionally, 40 mg/kg of CsA and 30 mg/kg of CHIR-98014 might provide protection. In summary, CHIR-98014 could protect against TP/LPS- or TP/TNF-α-induced activation of the GSK 3ß-JNK pathway and mitochondria-dependent apoptosis, improving the indirect hepatotoxicity induced by TP.


Sujet(s)
Aminopyridines , Lésions hépatiques dues aux substances , Diterpènes , Phénanthrènes , Pyrimidines , Souris , Animaux , Glycogen synthase kinase 3 beta/pharmacologie , Lipopolysaccharides/toxicité , Mitochondries , Apoptose , Diterpènes/pharmacologie , Phénanthrènes/pharmacologie , Composés époxy/toxicité , Lésions hépatiques dues aux substances/prévention et contrôle
12.
Bioorg Chem ; 143: 107071, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38199141

RÉSUMÉ

Farnesoid X receptor (FXR) was considered as a promising drug target in the treatment of cholestasis, drug-induced liver injury, and non-alcoholic steatohepatitis (NASH). However, the existing FXR agonists have shown different degrees of side effects in clinical trials without clear interpretation. MET-409 in clinical phase Ⅲ, has been proven significantly fewer side effects than that of other FXR agonists. This may be due to the completely different structure of FEX and other non-steroidal FXR agonists. Herein, the structure-based drug design was carried out based on FEX, and the more active FXR agonist LH10 (FEX EC50 = 0,3 µM; LH10 EC50 = 0.14 µM)) was screened out by the comprehensive SAR studies. Furthermore, LH10 exhibited robust hepatoprotective activity on the ANIT-induced cholestatic model and APAP-induced acute liver injury model, which was even better than positive control OCA. In the nonalcoholic steatohepatitis (NASH) model, LH10 significantly improved the pathological characteristics of NASH by regulating several major pathways including lipid metabolism, inflammation, oxidative stress, and fibrosis. With the above attractive results, LH10 is worthy of further evaluation as a novel agent for the treatment of liver disorders.


Sujet(s)
Cholestase , Stéatose hépatique non alcoolique , Humains , Stéatose hépatique non alcoolique/induit chimiquement , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Récepteurs cytoplasmiques et nucléaires , Foie/métabolisme , Dérivés du benzène/pharmacologie , Cholestase/métabolisme , Cholestase/anatomopathologie
13.
Basic Clin Pharmacol Toxicol ; 134(3): 315-324, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38048777

RÉSUMÉ

Bile acids are synthesized from cholesterol in the liver. Dysregulation of bile acid homeostasis, characterized by excessive accumulation in the liver, gallbladder and blood, can lead to hepatocellular damage and the development of cholestatic liver disease. Nuclear receptors play a crucial role in the control of bile acid metabolism by efficiently regulating bile acid synthesis and transport in the liver. Among these receptors, peroxisome proliferator-activated receptor (PPAR), a ligand-activated transcription factor belonging to the nuclear hormone receptor superfamily, controls the expression of genes involved in adipogenesis, lipid metabolism, inflammation and glucose homeostasis and has emerged as a potential therapeutic target for the treatment of the metabolic syndrome in the past two decades. Emerging evidence suggests that PPAR activation holds promise as a therapeutic target for cholestatic liver disease, as it affects both bile acid production and transport. This review provides a comprehensive overview of recent advances in elucidating the role of PPAR in the regulation of bile acid metabolism, highlighting the current position of PPAR agonists in the treatment of primary biliary cholangitis. By summarizing the specific regulatory effects of PPAR on bile acids, this review contributes to the exploration of novel therapeutic strategies for cholestatic liver diseases.


Sujet(s)
Maladies du foie , Récepteurs activés par les proliférateurs de peroxysomes , Humains , Récepteurs activés par les proliférateurs de peroxysomes/métabolisme , Récepteurs cytoplasmiques et nucléaires/métabolisme , Acides et sels biliaires , Métabolisme lipidique
14.
Int Immunopharmacol ; 125(Pt A): 111150, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37924700

RÉSUMÉ

Bile acid (BA) homeostasis throughout the enterohepatic circulation system is a guarantee of liver physiological functions. BA circulation disorders is one of the characteristic clinical manifestations of cholestasis, and have a closely relationship with intestinal barrier function, especially ileum. Here, our in vivo and in vitro studies showed that intestinal tight junctions (TJs) were disrupted by α-naphthylisothiocyanate (ANIT), which also down-regulated the protein expression of sphingosine-1-phosphate receptor 1 (S1PR1) in the top of villus of mice ileum. Activating S1PR1 by specific agonist SEW2871 could improve TJs via inhibiting ERK1/2/LKB1/AMPK signaling pathway in the ileum of ANIT-treated mice and ANIT-cultured Caco-2 cells. SEW2871 not only regained ileum TJs by activating S1PR1 in the epithelial cells of ileum mucosa, but also recovered ileum barrier function, which was further verified by the recovered BA homeostasis in mice ileum (content and tissue) by using of high-performance liquid chromatographytandem mass spectrometry (LC-MS/MS). Subsequently, the improved intestinal injury and inflammation further strengthened that SEW2871 modulated intestinal barrier function in ANIT-treated mice. Finally, our data revealed that along with the down-regulated levels of serum lipopolysaccharides (LPS), SEW2871 improved liver function and relieved hepatitis via blocking TLR4/MyD88/NF-kB signaling pathway in ANIT-treated mice. In conclusion, these results demonstrated that activating intestinal S1PR1 by SEW2871 could modulate intestinal barrier function, leading to the improvement of cholestatic hepatitis in ANIT-treated mice via the "gut-liver" axis.


Sujet(s)
Cholestase , Hépatite , Animaux , Humains , Souris , 1-Naphtyl-isothiocyanate/effets indésirables , 1-Naphtyl-isothiocyanate/métabolisme , 1-Naphtyl-isothiocyanate/toxicité , Cellules Caco-2 , Cholestase/métabolisme , Chromatographie en phase liquide , Hépatite/métabolisme , Foie/métabolisme , Récepteurs de la sphingosine-1-phosphate/métabolisme , Spectrométrie de masse en tandem
15.
Toxicol Lett ; 390: 25-32, 2023 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-37944651

RÉSUMÉ

Triptolide (TP) is extracted from the traditional Chinese medicine Tripterygium wilfordii Hook. F. (TWHF). Its severe toxic side effects, especially hepatotoxicity, have limited the clinical application of TP-related drugs. In this study, we investigated the mechanism of the hepatotoxic effects of TP from the perspective that TP inhibited the expression of the pro-survival protein X-linked inhibitor of apoptosis protein (XIAP) and enhanced FasL-mediated apoptosis of hepatocytes. TP and CD95/Fas antibody (Jo-2) were administered by gavage to C57BL/6 mice for 7 consecutive days. After co-administration of TP and Jo-2, mouse livers showed large areas of necrosis and apoptosis and significantly increased Caspase-3 activity. KEGG pathway enrichment analysis indicated that TP may cause the development of liver injury through the apoptotic signaling pathway. Proteinprotein interaction networks showed that XIAP played an essential role in this process. TP reduced the protein expression of XIAP after combination treatment with Jo-2/FasL in vivo/in vitro. TP and FasL co-stimulation significantly increased microRNA-137 (miR-137) levels in AML12 cells, while inhibition of miR-137 expression induced a rebound in XIAP protein expression. In conclusion, TP presensitizes hepatocytes and enhances the sensitivity of hepatocytes to the Fas/FasL pathway by inhibiting the protein expression of XIAP, leading to hepatocyte apoptosis.


Sujet(s)
microARN , Protéine inhibitrice de l'apoptose liée au chromosome X , Souris , Animaux , Protéine inhibitrice de l'apoptose liée au chromosome X/génétique , Protéine inhibitrice de l'apoptose liée au chromosome X/métabolisme , Protéine inhibitrice de l'apoptose liée au chromosome X/pharmacologie , Souris de lignée C57BL , Foie/métabolisme , Hépatocytes , Apoptose , microARN/métabolisme
16.
Xenobiotica ; 53(8-9): 559-571, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37885225

RÉSUMÉ

Cisplatin is a widely used chemotherapeutic agent to treat solid tumours in clinics. However, cisplatin-induced acute kidney injury (AKI) limits its clinical application. This study investigated the effect of hyperoside (a flavonol glycoside compound) on regulating AKI.The model of cisplatin-induced AKI was established, and hyperoside was preadministered to investigate its effect on improving kidney injury.Hyperoside ameliorated renal pathological damage, reduced the accumulation of SCr, BUN, Kim-1 and indoxyl sulphate in vivo, increased the excretion of indoxyl sulphate into the urine, and upregulated the expression of renal organic anion transporter 1 (Oat1). Moreover, evaluation of rat kidney slices demonstrated that hyperoside promoted the uptake of PAH (p-aminohippurate, the Oat1 substrate), which was confirmed by transient over-expression of OAT1 in HEK-293T cells. Additionally, hyperoside upregulated the mRNA expression of Oat1 upstream regulators hepatocyte nuclear factor-1α (HNF-1α) and pregnane X receptor (PXR).These findings indicated hyperoside could protect against cisplatin-induced AKI by promoting indoxyl sulphate excretion through regulating the expression and function of Oat1, suggesting hyperoside may offer a potential tactic for cisplatin-induced AKI treatment.


Sujet(s)
Atteinte rénale aigüe , Cisplatine , Rats , Animaux , Cisplatine/effets indésirables , Cisplatine/métabolisme , Transporteurs d'anions organiques sodium-indépendants/génétique , Transporteurs d'anions organiques sodium-indépendants/métabolisme , Protéine-1 de transport d'anions organiques/génétique , Protéine-1 de transport d'anions organiques/métabolisme , Indican/toxicité , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/traitement médicamenteux , Rein/métabolisme
17.
Dalton Trans ; 52(38): 13492-13496, 2023 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-37728022

RÉSUMÉ

The first hydroxyfluorooxoborate-nitrate mixed anion compound, K5[B3O3F4(OH)]2(NO3), was synthesized by the solution evaporation method. It displays a unique structure built by K+ cations, the hydroxylated and fluorinated six-membered ring [B3O3F4(OH)] and [NO3] groups. It possesses a band gap of 5.68 eV derived from the diffuse reflectance spectrum, which corresponds to an ultraviolet cutoff edge of 218 nm. First-principles calculations show that it has a large birefringence of 0.095 at 532 nm and the result of the response electron distribution anisotropy method indicates that all three anion groups contribute positively to the birefringence, verifying the synergic contributions from the multiple anion groups.

18.
Chin J Nat Med ; 21(8): 589-598, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37611977

RÉSUMÉ

Total glucosides of Rhizoma Smilacis Glabrae (RSG) are selective immunosuppressants that exhibit primary efficacy in the treatment of rheumatoid arthritis through targeted inhibition of activated T cells. In this study, we aimed to investigate the potential application of RSG in the treatment of psoriasis and elucidate its mechanism of action and material basis. Our findings revealed significant improvements upon administration of RSG in an imiquimod (IMQ)-induced psoriasis model. These improvements were characterized by a remarkable increase in the number of tail scales in mice and a substantial amelioration of skin erythema, ulceration, and flaking. By transcriptome sequencing and T-cell flow sorting assay, we identified notable effects of RSG on the modulation of various cellular processes. Specifically, RSG prominently down-regulated the Th17/Treg ratio in damaged skin tissues and reduced the proportion of G2 phase cells. Furthermore, RSG exhibited a stimulatory effect on the proliferation and differentiation of epithelial cells. Of particular interest, we discovered that ß-sitosterol, sitostenone, stigmasterol, smiglanin, and cinchonain Ib displayed potent inhibitory effects on the IL-17-mediated inflammatory response in HaCaT cells. In summary, our study highlights the therapeutic potential of RSG in the treatment of psoriasis, attributed to its ability to regulate the Th17/Treg balance. These findings contribute to the development of new indications for RSG and provide a solid theoretical foundation for further exploration in this field.


Sujet(s)
Polyarthrite rhumatoïde , Psoriasis , Animaux , Souris , Lymphocytes T régulateurs , Psoriasis/traitement médicamenteux , Dosage biologique , Glucosides/pharmacologie
19.
Biochem Pharmacol ; 215: 115742, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37567318

RÉSUMÉ

Human carboxylesterase 2 (hCES2) is an enzyme that metabolizes irinotecan to SN-38, a toxic metabolite considered a significant source of side effects (lethal delayed diarrhea). The hCES2 inhibitors could block the hydrolysis of irinotecan in the intestine and thus reduce the exposure of intestinal SN-38, which may alleviate irinotecan-associated diarrhea. However, existing hCES2 inhibitors (except loperamide) are not used in clinical applications due to lack of validity or acceptable safety. Therefore, developing more effective and safer drugs for treating delayed diarrhea is urgently needed. This study identified a lead compound 1 with a novel scaffold by high-throughput screening in our in-house library. After a comprehensive structure-activity relationship study, the optimal compound 24 was discovered as an efficient and highly selective hCES2 inhibitor (hCES2: IC50 = 6.72 µM; hCES1: IC50 > 100 µM). Further enzyme kinetics study indicated that compound 24 is a reversible inhibitor of hCES2 with competitive inhibition mode (Ki = 6.28 µM). The cell experiments showed that compound 24 could reduce the level of hCES2 in living cells (IC50 = 6.54 µM). The modeling study suggested that compound 24 fitted very well with the binding pocket of hCES2 by forming multiple interactions. Notably, compound 24 can effectively treat irinotecan-induced delayed diarrhea and DSS-induced ulcerative colitis, and its safety has also been verified in subtoxic studies. Based on the overall pharmacological and preliminary safety profiles, compound 24 is worthy of further evaluation as a novel agent for irinotecan-induced delayed diarrhea.


Sujet(s)
Rectocolite hémorragique , Humains , Irinotécan/effets indésirables , Rectocolite hémorragique/traitement médicamenteux , Carboxylesterase/métabolisme , Diarrhée/induit chimiquement , Diarrhée/traitement médicamenteux , Intestins , Relation structure-activité , Camptothécine/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE