Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 13.734
Filtrer
1.
Cell Mol Life Sci ; 81(1): 303, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39008099

RÉSUMÉ

Vitamin C (VC) serves as a pivotal nutrient for anti-oxidation process, metabolic responses, and stem cell differentiation. However, its precise contribution to placenta development and gestation remains obscure. Here, we demonstrated that physiological levels of VC act to stabilize Hand1, a key bHLH transcription factor vital for the development trajectory of trophoblast giant cell (TGC) lineages, thereby promoting the differentiation of trophoblast stem cells into TGC. Specifically, VC administration inactivated c-Jun N-terminal kinase (JNK) signaling, which directly phosphorylates Hand1 at Ser48, triggering the proteasomal degradation of Hand1. Conversely, a loss-of-function mutation at Ser48 on Hand1 not only significantly diminished both intrinsic and VC-induced stabilization of Hand1 but also underscored the indispensability of this residue. Noteworthy, the insufficiency of VC led to severe defects in the differentiation of diverse TGC subtypes and the formation of labyrinth's vascular network in rodent placentas, resulting in failure of maintenance of pregnancy. Importantly, VC deficiency, lentiviral knockdown of JNK or overexpression of Hand1 mutants in trophectoderm substantially affected the differentiation of primary and secondary TGC in E8.5 mouse placentas. Thus, these findings uncover the significance of JNK inactivation and consequential stabilization of Hand1 as a hitherto uncharacterized mechanism controlling VC-mediated placentation and perhaps maintenance of pregnancy.


Sujet(s)
Acide ascorbique , Facteurs de transcription à motif basique hélice-boucle-hélice , Différenciation cellulaire , JNK Mitogen-Activated Protein Kinases , Placentation , Trophoblastes , Animaux , Femelle , Grossesse , Acide ascorbique/pharmacologie , Acide ascorbique/métabolisme , Placentation/génétique , Souris , JNK Mitogen-Activated Protein Kinases/métabolisme , JNK Mitogen-Activated Protein Kinases/génétique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Trophoblastes/métabolisme , Trophoblastes/effets des médicaments et des substances chimiques , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Placenta/métabolisme , Phosphorylation , Humains , Souris de lignée C57BL
2.
J Cell Mol Med ; 28(14): e18534, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39031467

RÉSUMÉ

Intestinal dysbiosis is believed to play a role in the development of necrotizing enterocolitis (NEC). The efficacy of JNK-inhibitory peptide (CPJIP) in treating NEC was assessed. Treatment with CPJIP led to a notable reduction in p-JNK expression in IEC-6 cells and NEC mice. Following LPS stimulation, the expression of RNA and protein of claudin-1, claudin-3, claudin-4 and occludin was significantly decreased, with this decrease being reversed by CPJIP administration, except for claudin-3, which remained consistent in NEC mice. Moreover, the expression levels of the inflammatory factors TNF-α, IL-1ß and IL-6 were markedly elevated, a phenomenon that was effectively mitigated by the addition of CPJIP in both IEC-6 cells and NEC mice. CPJIP administration resulted in improved survival rates, ameliorated microscopic intestinal mucosal injury, and increased the total length of the intestines and colon in NEC mice. Additionally, CPJIP treatment led to a reduction in serum concentrations of FD-4, D-lactate and DAO. Furthermore, our results revealed that CPJIP effectively inhibited intestinal cell apoptosis and promoted cell proliferation in the intestine. This study represents the first documentation of CPJIP's ability to enhance the expression of tight junction components, suppress inflammatory responses, and rescue intestinal cell fate by inhibiting JNK activation, ultimately mitigating intestinal severity. These findings suggest that CPJIP has the potential to serve as a promising candidate for the treatment of NEC.


Sujet(s)
Apoptose , Entérocolite nécrosante , Inflammation , Muqueuse intestinale , Entérocolite nécrosante/traitement médicamenteux , Entérocolite nécrosante/métabolisme , Entérocolite nécrosante/anatomopathologie , Animaux , Souris , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Modèles animaux de maladie humaine , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Lignée cellulaire , Rats , JNK Mitogen-Activated Protein Kinases/métabolisme , Lipopolysaccharides ,
3.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38928105

RÉSUMÉ

Alcohol use disorder (AUD) is a chronic neurobehavioral condition characterized by a cycle of tolerance development, increased consumption, and reinstated craving and seeking behaviors during withdrawal. Understanding the intricate mechanisms of AUD necessitates reliable animal models reflecting its key features. Caenorhabditis elegans (C. elegans), with its conserved nervous system and genetic tractability, has emerged as a valuable model organism to study AUD. Here, we employ an ethanol vapor exposure model in Caenorhabditis elegans, recapitulating AUD features while maintaining high-throughput scalability. We demonstrate that ethanol vapor exposure induces intoxication-like behaviors, acute tolerance, and ethanol preference, akin to mammalian AUD traits. Leveraging this model, we elucidate the conserved role of c-jun N-terminal kinase (JNK) signaling in mediating acute ethanol tolerance. Mutants lacking JNK signaling components exhibit impaired tolerance development, highlighting JNK's positive regulation. Furthermore, we detect ethanol-induced JNK activation in C. elegans. Our findings underscore the utility of C. elegans with ethanol vapor exposure for studying AUD and offer novel insights into the molecular mechanisms underlying acute ethanol tolerance through JNK signaling.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Tolérance aux médicaments , Éthanol , Système de signalisation des MAP kinases , Animaux , Caenorhabditis elegans/génétique , Caenorhabditis elegans/effets des médicaments et des substances chimiques , Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , JNK Mitogen-Activated Protein Kinases/métabolisme , Alcoolisme/métabolisme , Alcoolisme/génétique , Modèles animaux de maladie humaine
4.
Stem Cell Res Ther ; 15(1): 179, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38902774

RÉSUMÉ

BACKGROUND: Adult skeletal muscle contains resident muscle stem cells (MuSC) with high myogenic and engraftment potentials, making them suitable for cell therapy and regenerative medicine approaches. However, purification process of MuSC remains a major hurdle to their use in the clinic. Indeed, muscle tissue enzymatic dissociation triggers a massive activation of stress signaling pathways, among which P38 and JNK MAPK, associated with a premature loss of MuSC quiescence. While the role of these pathways in the myogenic progression of MuSC is well established, the extent to which their dissociation-induced activation affects the functionality of these cells remains unexplored. METHODS: We assessed the effect of P38 and JNK MAPK induction on stemness marker expression and MuSC activation state during isolation by pharmacological approaches. MuSC functionality was evaluated by in vitro assays and in vivo transplantation experiments. We performed a comparative analysis of the transcriptome of human MuSC purified with pharmacological inhibitors of P38 and JNK MAPK (SB202190 and SP600125, respectively) versus available RNAseq resources. RESULTS: We monitored PAX7 protein levels in murine MuSC during muscle dissociation and demonstrated a two-step decline partly dependent on P38 and JNK MAPK activities. We showed that simultaneous inhibition of these pathways throughout the MuSC isolation process preserves the expression of stemness markers and limits their premature activation, leading to improved survival and amplification in vitro as well as increased engraftment in vivo. Through a comparative RNAseq analysis of freshly isolated human MuSC, we provide evidence that our findings in murine MuSC could be relevant to human MuSC. Based on these findings, we implemented a purification strategy, significantly improving the recovery yields of human MuSC. CONCLUSION: Our study highlights the pharmacological limitation of P38 and JNK MAPK activities as a suitable strategy to qualitatively and quantitatively ameliorate human MuSC purification process, which could be of great interest for cell-based therapies.


Sujet(s)
p38 Mitogen-Activated Protein Kinases , p38 Mitogen-Activated Protein Kinases/métabolisme , Animaux , Humains , Souris , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Muscles squelettiques/métabolisme , Muscles squelettiques/cytologie , Cellules souches/métabolisme , Cellules souches/cytologie , Cellules souches/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Mâle , Anthracènes/pharmacologie , JNK Mitogen-Activated Protein Kinases/métabolisme
5.
Yi Chuan ; 46(6): 490-501, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38886152

RÉSUMÉ

The JNK signaling pathway plays crucial roles in various physiological processes, including cell proliferation, differentiation, migration, apoptosis, and stress response. Dysregulation of this pathway is closely linked to the onset and progression of numerous major diseases, such as developmental defects and tumors. Identifying and characterizing novel components of the JNK signaling pathway to enhance and refine its network hold significant scientific and clinical importance for the prevention and treatment of associated cancers. This study utilized the model organism Drosophila and employed multidisciplinary approaches encompassing genetics, developmental biology, biochemistry, and molecular biology to investigate the interplay between Tip60 and the JNK signaling pathway, and elucidated its regulatory mechanisms. Our findings suggest that loss of Tip60 acetyltransferase activity results in JNK signaling pathway activation and subsequent induction of JNK-dependent apoptosis. Genetic epistasis analysis reveals that Tip60 acts downstream of JNK, paralleling with the transcription factor FOXO. The biochemical results confirm that Tip60 can bind to FOXO and acetylate it. Introduction of human Tip60 into Drosophila effectively mitigates apoptosis induced by JNK signaling activation, underscoring conserved regulatory role of Tip60 in the JNK signaling pathway from Drosophila to humans. This study further enhances our understanding of the regulatory network of the JNK signaling pathway. By revealing the role and mechanism of Tip60 in JNK-dependent apoptosis, it unveils new insights and potential therapeutic avenues for preventing and treating associated cancers.


Sujet(s)
Apoptose , Protéines de Drosophila , Facteurs de transcription Forkhead , Animaux , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Histone acetyltransferases/métabolisme , Histone acetyltransferases/génétique , Drosophila/génétique , Drosophila/métabolisme , Système de signalisation des MAP kinases , Humains , Transduction du signal , Drosophila melanogaster/génétique , Drosophila melanogaster/métabolisme , JNK Mitogen-Activated Protein Kinases/métabolisme , JNK Mitogen-Activated Protein Kinases/génétique
6.
Exp Cell Res ; 440(2): 114134, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38901790

RÉSUMÉ

Individuals with type 2 diabetes mellitus frequently display heightened levels of palmitic acid (PA) in their serum, which may lead to ß-cell damage. The involvement of ferroptosis, a form of oxidative cell death in lipotoxic ß-cell injury remains uncertain. Here, we have shown that PA induces intracellular lipid peroxidation, increases intracellular Fe2+ content and decreases intracellular glutathione peroxidase 4 (GPX4) expression. Furthermore, PA causes distinct changes in pancreatic islets and INS-1 cells, such as mitochondrial atrophy and increased membrane density. Furthermore, the presence of the ferroptosis inhibitor has a significant mitigating effect on PA-induced ß-cell damage. Mechanistically, PA increased ceramide content and c-Jun N-terminal kinase (JNK) phosphorylation. The ceramide synthase inhibitor effectively attenuated PA-induced ß-cell damage and GPX4/Fe2+ abnormalities, while inhibiting JNK phosphorylation. Additionally, the JNK inhibitor SP600125 improved PA-induced cell damage. In conclusion, by promoting ceramide synthesis, PA inhibited GPX4 expression and increased intracellular Fe2+ to induce ß-cell ferroptosis. Moreover, JNK may be a downstream mechanism of ceramide-triggered lipotoxic ferroptosis in ß-cells.


Sujet(s)
Céramides , Ferroptose , Cellules à insuline , Acide palmitique , Transduction du signal , Ferroptose/effets des médicaments et des substances chimiques , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Céramides/métabolisme , Acide palmitique/pharmacologie , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Rats , Peroxydation lipidique/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , JNK Mitogen-Activated Protein Kinases/métabolisme , Fer/métabolisme
7.
Int Immunopharmacol ; 137: 112522, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38908089

RÉSUMÉ

BACKGROUND: Periodontitis is a common oral disease with high prevalence worldwide. Neural epidermal growth factor-like 1 protein (Nell-1) has recently been reported to have anti-inflammation effects and may be a drug candidate for osteoarthritis. However, its immunotherapeutic effects in periodontitis remain unknown. Therefore, this study aimed to investigate the effects of Nell-1 on periodontitis in terms of macrophage polarization and analyze its possible underlying mechanism. METHODS: A rat ligation-induced experimental periodontitis model was established and locally injected with Nell-1 (n = 6/group). Periodontal tissue destruction and macrophage polarization in vivo were analyzed using micro-CT, histology analysis, and western blot. Enzyme-linked immunosorbent assay was used to evaluate serum inflammatory cytokines. Then, the RAW 264.7 macrophage cells were treated with lipopolysaccharide (LPS), Nell-1, and the c-Jun N-terminal kinases (JNK) inhibitor (SP600125). RT-PCR, western blot, and flow cytometry were performed to further analyze the effect of Nell-1 on macrophage polarization and the underlying mechanism in vitro. RESULTS: Local treatment with Nell-1 significantly alleviated the destruction of alveolar bone and fibers in periodontitis, and upregulated the ratio of M2/M1 macrophages in periodontal tissues (P < 0.05). In vitro, Nell-1 at the concentrations of 200 and 500 ng/mL could significantly inhibit the expression of M1-related inflammatory factors in LPS-stimulated macrophages, and increase the expression of M2-related markers, regulating the macrophage phenotype switch into M2 (P < 0.05). The mRNA of JNK and relative protein level of phospho-JNK/JNK were also upregulated by Nell-1 (P < 0.05). Additionally, the JNK inhibitor (SP600125) could reverse the effect of Nell-1 on macrophage polarization (P < 0.05). CONCLUSIONS: Nell-1 could modulate the ratio of M2/M1 macrophages possibly through the JNK/MAPK signaling pathway, subsequently attenuating the inflammation and destruction of periodontal tissues caused by periodontitis.


Sujet(s)
Macrophages , Parodontite , Animaux , Parodontite/traitement médicamenteux , Parodontite/immunologie , Parodontite/anatomopathologie , Parodontite/métabolisme , Souris , Macrophages/effets des médicaments et des substances chimiques , Macrophages/immunologie , Macrophages/métabolisme , Cellules RAW 264.7 , Rats , Mâle , Cytokines/métabolisme , Rat Sprague-Dawley , Lipopolysaccharides , Modèles animaux de maladie humaine , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , JNK Mitogen-Activated Protein Kinases/métabolisme , Activation des macrophages/effets des médicaments et des substances chimiques , Phénotype , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique
8.
Nat Commun ; 15(1): 5493, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38944657

RÉSUMÉ

JNK signaling is a critical regulator of inflammation and regeneration, but how it is controlled in specific tissue contexts remains unclear. Here we show that, in the Drosophila intestine, the TNF-type ligand, Eiger (Egr), is expressed exclusively by intestinal stem cells (ISCs) and enteroblasts (EBs), where it is induced by stress and during aging. Egr preferentially activates JNK signaling in a paracrine fashion in differentiated enterocytes (ECs) via its receptor, Grindelwald (Grnd). N-glycosylation genes (Alg3, Alg9) restrain this activation, and stress-induced downregulation of Alg3 and Alg9 correlates with JNK activation, suggesting a regulatory switch. JNK activity in ECs induces expression of the intermembrane protease Rhomboid (Rho), driving secretion of EGFR ligands Keren (Krn) and Spitz (Spi), which in turn activate EGFR signaling in progenitor cells (ISCs and EBs) to stimulate their growth and division, as well as to produce more Egr. This study uncovers an N-glycosylation-controlled, paracrine JNK-EGFR-JNK feedforward loop that sustains ISC proliferation during stress-induced gut regeneration.


Sujet(s)
Protéines de Drosophila , Récepteurs ErbB , Intestins , Système de signalisation des MAP kinases , Animaux , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Intestins/cytologie , Drosophila melanogaster/métabolisme , Drosophila melanogaster/génétique , Entérocytes/métabolisme , Entérocytes/cytologie , Cellules souches/métabolisme , Cellules souches/cytologie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/cytologie , Drosophila/métabolisme , Glycosylation , Récepteur peptidique invertébrés/métabolisme , Récepteur peptidique invertébrés/génétique , Prolifération cellulaire , JNK Mitogen-Activated Protein Kinases/métabolisme , Transduction du signal , Communication cellulaire , Différenciation cellulaire , Facteur de croissance épidermique , Protéines membranaires
9.
Mycotoxin Res ; 40(3): 457-466, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38913091

RÉSUMÉ

Aflatoxin B1 (AFB1) is classified as a Class I carcinogen and common pollutant in human and animal food products. Prolonged exposure to AFB1 can induce hepatocyte apoptosis and lead to hepatotoxicity. Therefore, preventing AFB1-induced hepatotoxicity remains a critical issue and is of great significance. Baicalin, a polyphenolic compound derived from Scutellaria baicalensis Georgi, has a variety of pharmacodynamic activities, such as antiapoptotic and anticancer activities. This study systematically investigated the alleviating effect of baicalin on AFB1-induced hepatotoxicity from the perspective of apoptosis and explored the possible molecular mechanism. In the normal human liver cell line L02, baicalin treatment significantly inhibited AFB1-induced c-Jun-N-terminal Kinase (JNK) activation and cell apoptosis. In addition, the in vitro mechanism study demonstrated that baicalin alleviates AFB1-induced hepatocyte apoptosis through suppressing the translocation of phosphorylated JNK to the nucleus and decreasing the phosphorylated c-Jun/c-Jun ratio and the Bax/Bcl2 ratio. Molecular docking and drug affinity responsive target stability assays demonstrated that baicalin has the potential to target JNK. This study provides a basis for the therapeutic effect of baicalin on hepatocyte apoptosis caused by AFB1, indicating that the development of baicalin and JNK pathway inhibitors has broad application prospects in the prevention of hepatotoxicity, especially hepatocyte apoptosis.


Sujet(s)
Aflatoxine B1 , Apoptose , Flavonoïdes , Hépatocytes , JNK Mitogen-Activated Protein Kinases , Flavonoïdes/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Aflatoxine B1/toxicité , Humains , JNK Mitogen-Activated Protein Kinases/métabolisme , Hépatocytes/effets des médicaments et des substances chimiques , Lignée cellulaire , Simulation de docking moléculaire , Scutellaria baicalensis/composition chimique
10.
Zhongguo Zhong Yao Za Zhi ; 49(10): 2745-2753, 2024 May.
Article de Chinois | MEDLINE | ID: mdl-38812175

RÉSUMÉ

This study investigated the protective effect of ginsenoside Rg_1(GRg_1) on oxygen and glucose deprivation/reoxygenation(OGD/R)-injured rat adrenal pheochromocytoma(PC12) cells and whether the underlying mechanism was related to the regulation of inositol-requiring enzyme 1(IRE1)-c-Jun N-terminal kinase(JNK)-C/EBP homologous protein(CHOP) signaling pathway. An OGD/R model was established in PC12 cells, and PC12 cells were randomly classified into control, model, OGD/R+GRg_1(0.1, 1, 10 µmol·L~(-1)), OGD/R+GRg_1+rapamycin(autophagy agonist), OGD/R+GRg_1+3-methyladenine(3-MA,autophagy inhibitor), OGD/R+GRg_1+tunicamycin(endoplasmic reticulum stress agonist), OGD/R+GRg_1+4-phenylbutyric acid(4-PBA, endoplasmic reticulum stress inhibitor), and OGD/R+GRg_1+3,5-dibromosalicylaldehyde(DBSA, IRE1 inhibitor) groups. Except the control group, the other groups were subjected to OGD/R treatment, i.e., oxygen and glucose deprivation for 6 h followed by reoxygenation for 6 h. Cell viability was detected by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium bromide(MTT) assay. Apoptosis was detected by Hoechst 33342 staining, and the fluorescence intensity of autophagosomes by the monodansylcadaverine(MDC) assay. Western blot was employed to determine the expression of autophagy-related proteins(Beclin1, LC3-Ⅱ, and p62) and the pathway-related proteins [IRE1, p-IRE1, JNK, p-JNK, glucose-regulated protein 78(GRP78), and CHOP]. The results showed that GRg_1 dose-dependently increased the viability of PC12 cells and down-regulated the expression of Beclin1, LC3-Ⅱ, p-IRE1, p-JNK, GRP78, and CHOP, compared with the model group. Furthermore, GRg_1 decreased the apoptosis rate and MDC fluorescence intensity and up-regulated the expression of p62 protein. Compared with the OGD/R+GRg_1(10 µmol·L~(-1)) group, OGD/R+GRg_1+rapamycin and OGD/R+GRg_1+tunicamycin groups showed increased apoptosis rate and MDC fluorescence intensity, up-regulated protein levels of Beclin1, LC3-Ⅱ, p-IRE1, p-JNK, GRP78, and CHOP, decreased relative cell survival rate, and down-regulated protein level of p62. The 3-MA, 4-PBA, and DBSA groups exerted the opposite effects. Taken together, GRg_1 may ameliorate OGD/R-induced PC12 cell injury by inhibiting autophagy via the IRE1-JNK-CHOP pathway.


Sujet(s)
Apoptose , Ginsénosides , Glucose , Protein-Serine-Threonine Kinases , Facteur de transcription CHOP , Animaux , Rats , Cellules PC12 , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Glucose/métabolisme , Ginsénosides/pharmacologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Apoptose/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Autophagie/effets des médicaments et des substances chimiques , Endoribonucleases/métabolisme , Endoribonucleases/génétique , JNK Mitogen-Activated Protein Kinases/métabolisme , JNK Mitogen-Activated Protein Kinases/génétique , Oxygène/métabolisme , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Complexes multienzymatiques
11.
Fitoterapia ; 176: 106015, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38762075

RÉSUMÉ

Five dihydrophenanthropyrans (1-5) were isolated from the pseudobulbs of Pholidota chinensis, among which 1,3-di(4'-hydroxybenzy)-imbricatin (3) was isolated from the nature for the first time. Their structures were elucidated and established through various spectroscopic methods. These compounds exhibited a potent inhibition effect on both N-formyl-methionyl-leucyl-phenylalanine (fMLF)-induced superoxide anion generation and elastase release with IC50 values ranging from 0.23 to 7.63 µM. Furthermore, dihydrophenanthropyrans (1-3) also demonstrated a dose-dependent reactive oxygen species (ROS) scavenging effect. In addition, dihydrophenanthropyrans (2-3) exhibited a dose-dependent reduction in the intracellular Ca2+ concentration ([Ca2+]i) in fMLF-activated human neutrophils. Moreover, dihydrophenanthropyrans (1-3) selectively inhibited the phosphorylation of c-Jun N-terminal kinases (JNKs) and p38, while only dihydrophenanthropyran (1) inhibited the phosphorylation of extracellular signal-regulated kinases (ERKs) in fMLF-activated human neutrophils. Notably, dihydrophenanthropyrans (1-3) did not affect protein kinase B (AKT) activity in these cells. These findings highlight the potent anti-inflammatory capabilities of dihydrophenanthropyrans, manifested through their ability to inhibit superoxide anion generation, suppress elastase release, and selectively modulate key signaling pathways in human neutrophils. This suggests that dihydrophenanthropyrans hold significant promise as therapeutic agents for conditions associated with neutrophil-mediated inflammation.


Sujet(s)
Calcium , Granulocytes neutrophiles , Superoxydes , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Humains , Structure moléculaire , Calcium/métabolisme , Superoxydes/métabolisme , Pancreatic elastase/métabolisme , Espèces réactives de l'oxygène/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/isolement et purification , Orchidaceae/composition chimique , Composés phytochimiques/pharmacologie , Composés phytochimiques/isolement et purification , Inflammation/traitement médicamenteux , Mitogen-Activated Protein Kinases/métabolisme , Chine , JNK Mitogen-Activated Protein Kinases/métabolisme
12.
Life Sci ; 350: 122750, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38801982

RÉSUMÉ

C-Jun-N-terminal-kinases (JNKs), members of the mitogen-activated-protein-kinase family, are significantly linked with neurological and neurodegenerative pathologies and cancer progression. However, JNKs serve key roles under physiological conditions, particularly within the central-nervous-system (CNS), where they are critical in governing neural proliferation and differentiation during both embryogenesis and adult stages. These processes control the development of CNS, avoiding neurodevelopment disorders. JNK are key to maintain the proper activity of neural-stem-cells (NSC) and neural-progenitors (NPC) that exist in adults, which keep the convenient brain plasticity and homeostasis. This review underscores how the interaction of JNK with upstream and downstream molecules acts as a regulatory mechanism to manage the self-renewal capacity and differentiation of NSC/NPC during CNS development and in adult neurogenic niches. Evidence suggests that JNK is reliant on non-canonical Wnt components, Fbw7-ubiquitin-ligase, and WDR62-scaffold-protein, regulating substrates such as transcription factors and cytoskeletal proteins. Therefore, understanding which pathways and molecules interact with JNK will bring knowledge on how JNK activation orchestrates neuronal processes that occur in CNS development and brain disorders.


Sujet(s)
Différenciation cellulaire , Cellules souches neurales , Neurogenèse , Humains , Animaux , Différenciation cellulaire/physiologie , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Neurogenèse/physiologie , Système de signalisation des MAP kinases/physiologie , JNK Mitogen-Activated Protein Kinases/métabolisme , Neurones/métabolisme , Neurones/cytologie
13.
Biomed Pharmacother ; 176: 116759, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788603

RÉSUMÉ

BACKGROUND: Sodium-glucose cotransporter-2 inhibitors, such as empagliflozin, are pivotal therapies for heart failure. However, the effect of empagliflozin on doxorubicin-related cardiac dysfunction remains unclear. METHODS: Human induced pluripotent stem cell- and embryonic stem cell-derived cardiomyocytes were used to investigate the direct effect of empagliflozin on human cardiomyocytes. Then, the c-Jun amino-terminal kinases (JNK) inhibitor SP600125 was administered to the doxorubicin cardiotoxicity model in vitro and in vivo to investigate the role of JNK in empagliflozin. RESULTS: In human stem cell-derived cardiomyocytes, pretreatment with empagliflozin attenuated doxorubicin-induced cleavage of caspase 3 and other apoptosis markers. Empagliflozin significantly attenuated doxorubicin-induced phosphorylation of JNK and p38. Inhibiting the phosphorylation of JNK (SP600125) or STAT3 attenuated doxorubicin-induced apoptosis, but inhibiting the phosphorylation of p38 did not. SP600125 inhibits the phosphorylation of STAT3 (S727), and a STAT3 (Y705) inhibitor also inhibits the phosphorylation of JNK. Empagliflozin and SP600125 attenuated doxorubicin-induced increases in reactive oxygen species (ROS) and decreases in oxidized nicotinamide adenine dinucleotide (NAD+). In animal studies, empagliflozin and SP600125 attenuated doxorubicin-induced cardiac dysfunction and fibrosis. CONCLUSIONS: Empagliflozin attenuated doxorubicin-induced apoptosis by inhibiting the phosphorylation of JNK and its downstream signaling pathways, including ROS and NAD+.


Sujet(s)
Apoptose , Composés benzhydryliques , Cardiotoxicité , Doxorubicine , Glucosides , Myocytes cardiaques , Glucosides/pharmacologie , Composés benzhydryliques/pharmacologie , Doxorubicine/toxicité , Doxorubicine/effets indésirables , Cardiotoxicité/traitement médicamenteux , Cardiotoxicité/prévention et contrôle , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Humains , Animaux , Apoptose/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Mâle , Espèces réactives de l'oxygène/métabolisme , Anthracènes/pharmacologie , JNK Mitogen-Activated Protein Kinases/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Souris , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Souris de lignée C57BL
14.
Food Chem Toxicol ; 188: 114633, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38608924

RÉSUMÉ

The cytotoxic mycotoxin deoxynivalenol (DON) reportedly has adverse effects on oocyte maturation and embryonic development in pigs. Recently, the interplay between cell apoptosis and endoplasmic reticulum (ER) stress has garnered increasing attention in embryogenesis. However, the involvement of the inositol-requiring enzyme 1 (IRE1)/c-jun N-terminal kinase (JNK)/C/EBP homologous protein (CHOP) pathways of unfolded protein response (UPR) signaling in DON-induced apoptosis in porcine embryos remains unknown. In this study, we revealed that exposure to DON (0.25 µM) substantially decreased cell viability until the blastocyst stage in porcine embryos, concomitant with initiation of cell apoptosis through the IRE1/JNK/CHOP pathways in response to ER stress. Quantitative PCR confirmed that UPR signaling-related transcription factors were upregulated in DON-treated porcine blastocysts. Western blot analysis showed that IRE1/JNK/CHOP signaling was activated in DON-exposed porcine embryos, indicating that ER stress-associated apoptosis was instigated. The ER stress inhibitor tauroursodeoxycholic acid protected against DON-induced ER stress in porcine embryos, indicating that the toxic effects of DON on early developmental competence of porcine embryos can be prevented. In conclusion, DON exposure impairs the developmental ability of porcine embryos by inducing ER stress-mediated apoptosis via IRE1/JNK/CHOP signaling.


Sujet(s)
Apoptose , Stress du réticulum endoplasmique , Facteur de transcription CHOP , Trichothécènes , Animaux , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Facteur de transcription CHOP/métabolisme , Facteur de transcription CHOP/génétique , Suidae , Trichothécènes/toxicité , JNK Mitogen-Activated Protein Kinases/métabolisme , JNK Mitogen-Activated Protein Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Embryon de mammifère/effets des médicaments et des substances chimiques , Réponse aux protéines mal repliées/effets des médicaments et des substances chimiques , Blastocyste/effets des médicaments et des substances chimiques , Blastocyste/métabolisme , Femelle
15.
Eur J Pharmacol ; 974: 176620, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38685305

RÉSUMÉ

The incidence and mortality of breast cancer, the most common malignant tumor among women in the world, are increasing year by year, which greatly threatens women's health. Ferroptosis is an iron and lipid reactive oxygen species (ROS)-dependent process, a novel form of cell death that is distinct from apoptosis and is closely related to the progression of breast cancer. Inducing the occurrence of ferroptosis in tumor cells can effectively block its malignant progress in vivo. Oridonin (ORI), the primary active ingredient extracted from the Chinese herbal medicine Rabdosia rubescens, has been shown to cause glutathione depletion and directly inhibit glutathione peroxidase 4 induced cell death by ferroptosis, but its mechanism of action in breast cancer remains inadequately elucidated. Therefore, we further investigated whether ORI could promote RSL3-induced ferroptosis in breast cancer cells by regulating the oxidative stress pathway JNK/Nrf2/HO-1. In our study, we assessed cell survival of RSL3 and ORI treatment by MTT assay, and found that co-treatment with RSL3 and ORI inhibited cell proliferation, as evidenced by the cloning assay. To investigate the ability of ORI to promote RSL3-induced ferroptosis in breast cancer cells, we measured levels of ROS, malondialdehyde, glutathione, superoxide dismutase, and Fe2+ content. Lipid peroxidation, ROS, and mitochondrial membrane potential levels induced by co-treatment of ORI with RSL3 were reversed by ferrostatin-1, further confirming that the cell death induced by RSL3 and ORI was ferroptosis rather than other programmed cell death modes. Moreover, RSL3 and ORI co-treatment regulated the JNK/Nrf2/HO-1 axis, as demonstrated by western blotting and target activator validation. Our results showed that ORI could enhance the inhibitory effect of RSL3 on breast cancer cells viability via the induction of ferroptosis. Mechanistically, it potentiated RSL3-induced ferroptosis in breast cancer cells by activating the JNK/Nrf2/HO-1 axis. This study provides a theoretical basis for the application of ORI based on the mechanism of ferroptosis, and provides potential natural drug candidates for cancer prevention and treatment.


Sujet(s)
Tumeurs du sein , Diterpènes de type kaurane , Ferroptose , Heme oxygenase-1 , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Ferroptose/effets des médicaments et des substances chimiques , Humains , Facteur-2 apparenté à NF-E2/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Stress oxydatif/effets des médicaments et des substances chimiques , Heme oxygenase-1/métabolisme , Diterpènes de type kaurane/pharmacologie , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Espèces réactives de l'oxygène/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , JNK Mitogen-Activated Protein Kinases/métabolisme , Cellules MCF-7 , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Carbolines
16.
Oncogene ; 43(21): 1608-1619, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38565943

RÉSUMÉ

Cancer cells employ adaptive mechanisms to survive various stressors, including genotoxic drugs. Understanding the factors promoting survival is crucial for developing effective treatments. In this study, we unveil a previously unexplored long non-coding RNA, JUNI (JUN-DT, LINC01135), which is upregulated by genotoxic drugs through the activation of stress-activated MAPKs, JNK, and p38 and consequently exerts positive control over the expression of its adjacent gene product c-Jun, a well-known oncoprotein, which transduces signals to multiple transcriptional outputs. JUNI regulates cellular migration and has a crucial role in conferring cellular resistance to chemotherapeutic drugs or UV radiation. Depletion of JUNI markedly increases the sensitivity of cultured cells and spheroids to chemotherapeutic agents. We identified 57 proteins interacting with JUNI. The activity of one of them the MAPK phosphatase and inhibitor, DUSP14, is counteracted by JUNI, thereby, facilitating efficient JNK phosphorylation and c-Jun induction when cells are exposed to UV radiation. The antagonistic interplay with DUSP14 contributes not only to c-Jun induction but also augments the survival of UV-exposed cells. In summary, we introduce JUNI as a novel stress-inducible regulator of c-Jun, positioning it as a potential target for enhancing the sensitivity of cancer cells to chemotherapy.


Sujet(s)
Mouvement cellulaire , Survie cellulaire , Dual-specificity phosphatases , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Dual-specificity phosphatases/métabolisme , Dual-specificity phosphatases/génétique , Mouvement cellulaire/génétique , Survie cellulaire/effets des radiations , Survie cellulaire/génétique , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-jun/métabolisme , Protéines proto-oncogènes c-jun/génétique , Lignée cellulaire tumorale , Rayons ultraviolets/effets indésirables , Système de signalisation des MAP kinases/génétique , Régulation de l'expression des gènes tumoraux , JNK Mitogen-Activated Protein Kinases/métabolisme
17.
Biosci Biotechnol Biochem ; 88(6): 608-619, 2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38573835

RÉSUMÉ

The huge diversity of secondary bioactive metabolites, such as antibiotic and anticancer compounds produced by Micromonospora sp., makes it an attractive target for study. Here, we explored the anti-proliferative activities of Micromonospora sp. M2 extract (MBE) in relation to its pro-oxidative activities in A549 and MCF7 cell lines. Anti-proliferative effects were assessed by treating cells with MBE. We found that treatment with MBE decreased cell proliferation and increased intracellular reactive oxygen species, and that these observations were facilitated by the suppression of the PI3K-AKT pathway, alterations to the Bcl/Bad ratio, and increased caspase activity. These observations also demonstrated that MBE induced apoptotic cell death in cell lines. In addition, the phosphorylation of P38 and c-Jun N-terminal kinase (JNK) were upregulated following MBE treatment in both cell lines. Collectively, these results indicate that MBE acts as an anticancer agent via oxidative stress and JNK/mitogen-activated protein kinase pathway activation, enhancing apoptotic cell death in cell lines.


Sujet(s)
Apoptose , Prolifération cellulaire , Micromonospora , Espèces réactives de l'oxygène , Humains , Cellules A549 , Cellules MCF-7 , Apoptose/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , JNK Mitogen-Activated Protein Kinases/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique
18.
Biomolecules ; 14(4)2024 Mar 25.
Article de Anglais | MEDLINE | ID: mdl-38672411

RÉSUMÉ

Intervertebral disc degeneration (IDD) is a major cause of lower back pain. The pathophysiological development of IDD is closely related to the stimulation of various stressors, including proinflammatory cytokines, abnormal mechanical stress, oxidative stress, metabolic abnormalities, and DNA damage, among others. These factors prevent normal intervertebral disc (IVD) development, reduce the number of IVD cells, and induce senescence and apoptosis. Stress-activated protein kinases (SAPKs), particularly, c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK), control cell signaling in response to cellular stress. Previous studies have shown that these proteins are highly expressed in degenerated IVD tissues and are involved in complex biological signal-regulated processes. Therefore, we summarize the research reports on IDD related to JNK and p38 MAPK. Their structure, function, and signal regulation mechanisms are comprehensively and systematically described and potential therapeutic targets are proposed. This work could provide a reference for future research and help improve molecular therapeutic strategies for IDD.


Sujet(s)
Dégénérescence de disque intervertébral , JNK Mitogen-Activated Protein Kinases , p38 Mitogen-Activated Protein Kinases , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Humains , p38 Mitogen-Activated Protein Kinases/métabolisme , JNK Mitogen-Activated Protein Kinases/métabolisme , Animaux , Système de signalisation des MAP kinases , Transduction du signal , Stress oxydatif , Disque intervertébral/métabolisme , Disque intervertébral/anatomopathologie , Disque intervertébral/enzymologie
19.
Sci Rep ; 14(1): 7277, 2024 03 27.
Article de Anglais | MEDLINE | ID: mdl-38538669

RÉSUMÉ

MicroRNAs (miRNAs) are vital regulators of tumor pathogenesis, including that of retinoblastoma (Rb). This study investigated the functions and mechanisms of action of miR-889-3p in Rb. BMPR2 and miR-889-3p levels were assessed by quantitative reverse transcription PCR (qRT-PCR) or western blotting. Through several cell function tests, the effects of miR-889-3p and BMPR2 on cell proliferation, migration, and JNK/MAPK/ERK signaling were evaluated. The interaction between miR-889-3p and BMPR2 was investigated using a luciferase reporter assay. In vivo tumor development was investigated using a xenograft test. The association between miR-889-3p and BMPR2 expression was identified using Pearson's correlation analysis. miR-889-3p was increased in Rb cells, and miR-889-3p knockdown inhibited Rb cell proliferation, migration, and phosphorylation of c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), and ERK1/2 in vitro, as well as tumor growth in vivo. Further, they were inversely associated in Rb tissues and miR-889-3p may directly attached to the 3'-UTR of BMPR2 mRNA. Finally, the inhibition of BMPR2 inverted the negative effects of the miR-889-3p inhibitor on migration, proliferation, and activation of JNK, p38 MAPK, and ERK1/2 in Rb cells. Our results indicate that miR-889-3p, which targets BMPR2 and promotes Rb growth by controlling the JNK/MAPK/ERK pathway, is an oncogene in Rb. These results suggested that the miR-889-3p/BMPR2 axis may be a new therapeutic target for Rb.


Sujet(s)
microARN , Tumeurs de la rétine , Rétinoblastome , Humains , Rétinoblastome/anatomopathologie , JNK Mitogen-Activated Protein Kinases/métabolisme , microARN/métabolisme , Transduction du signal , Prolifération cellulaire/génétique , Tumeurs de la rétine/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Apoptose/génétique , Récepteurs de la protéine morphogénique osseuse de type II/génétique , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme
20.
Int J Biol Macromol ; 264(Pt 1): 130542, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38432272

RÉSUMÉ

Pathological cardiac hypertrophy (CH) is driven by maladaptive changes in myocardial cells in response to pressure overload or other stimuli. CH has been identified as a significant risk factor for the development of various cardiovascular diseases, ultimately resulting in heart failure. Melanoma differentiation-associated protein 5 (MDA5), encoded by interferon-induced with helicase C domain 1 (IFIH1), is a cytoplasmic sensor that primarily functions as a detector of double-stranded ribonucleic acid (dsRNA) viruses in innate immune responses; however, its role in CH pathogenesis remains unclear. Thus, the aim of this study was to examine the relationship between MDA5 and CH using cellular and animal models generated by stimulating neonatal rat cardiomyocytes with phenylephrine and by performing transverse aortic constriction on mice, respectively. MDA5 expression was upregulated in all models. MDA5 deficiency exacerbated myocardial pachynsis, fibrosis, and inflammation in vivo, whereas its overexpression hindered CH development in vitro. In terms of the underlying molecular mechanism, MDA5 inhibited CH development by promoting apoptosis signal-regulating kinase 1 (ASK1) phosphorylation, thereby suppressing c-Jun N-terminal kinase/p38 signaling pathway activation. Rescue experiments using an ASK1 activation inhibitor confirmed that ASK1 phosphorylation was essential for MDA5-mediated cell death. Thus, MDA5 protects against CH and is a potential therapeutic target.


Sujet(s)
Apoptose , MAP Kinase Kinase Kinase 5 , Souris , Rats , Animaux , Hélicase IFIH1 inductrice de l'interféron/génétique , Hélicase IFIH1 inductrice de l'interféron/métabolisme , MAP Kinase Kinase Kinase 5/métabolisme , Apoptose/physiologie , Cardiomégalie/métabolisme , Transduction du signal , JNK Mitogen-Activated Protein Kinases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE