Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 95.251
Filtrer
1.
BMC Urol ; 24(1): 144, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997703

RÉSUMÉ

BACKGROUND: Prostate cancer, characterized by its insidious onset and short overall survival, and has seen a rise in incidence over recent decades. This study aims to investigate the expression and molecular mechanism of lncRNA PTCSC3 (PTCSC3) in prostate cancer in order to develop new prognostic and therapeutic biomarkers. METHODS: The level of PTCSC3 in serum and cell samples of prostate cancer was quantitatively measured using RT-qPCR assays. The correlation between the variation in PTCSC3 levels and clinical indicators of patients was evaluated. The survival status of the prostate cancer patients included in the study was evaluated using Kaplan-Meier curve and multivariable Cox analysis. The impact of PTCSC3 overexpression on cell growth and activity was revealed by CCK-8 and Transwell assays. The targeting relationship between PTCSC3 and miR-182-5p was determined by bioinformatics prediction and luciferase activity. RESULTS: PTCSC3 was found to be downregulated in prostate cancer, and its low levels were associated with short overall survival in patients. It influenced the progression of prostate cancer by targeting miR-182-5p. Increasing PTCSC3 levels suppressed the proliferation, migration and invasion levels of cells, and miR-182-5p mimic counteracted PTCSC3's effects on cells. CONCLUSIONS: As a potential prognostic biological factor for prostate cancer, PTCSC3 may regulate the progression of prostate cancer by sponging miR-182-5p and affect the prognosis of patients.


Sujet(s)
microARN , Tumeurs de la prostate , ARN long non codant , Mâle , Humains , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , microARN/génétique , microARN/sang , Pronostic , ARN long non codant/génétique , ARN long non codant/sang , Adulte d'âge moyen , Sujet âgé , Taux de survie , Régulation négative
2.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000128

RÉSUMÉ

Epigenetic changes are common in cancer and include aberrant DNA methylation and histone modifications, including both acetylation or methylation. DNA methylation in the promoter regions and histone deacetylation are usually accompanied by gene silencing, and may lead to the suppression of tumor suppressors in cancer cells. An interaction between epigenetic pathways has been reported that could be exploited to more efficiently target aggressive cancer cells, particularly those against which current treatments usually fail, such as pancreatic cancer. In this study, we explored the possibility to combine the DNA demethylating agent 5-AZA with HDAC inhibitor SAHA to treat pancreatic cancer cell lines, focusing on the acetylation of mutp53 and the consequences on its stability, as well as on the interaction of this protein with c-myc and BRCA-1, key molecules in cancer survival. The results obtained suggest that SAHA/5-AZA combination was more effective than single treatments to promote the degradation of mutp53, to upregulate p21 and downregulate c-Myc and BRCA-1, thus increasing DNA damage and cytotoxicity in pancreatic cancer cells.


Sujet(s)
Protéine BRCA1 , Inhibiteur p21 de kinase cycline-dépendante , Régulation de l'expression des gènes tumoraux , Tumeurs du pancréas , Protéines proto-oncogènes c-myc , Protéine p53 suppresseur de tumeur , Vorinostat , Humains , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Acétylation/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Vorinostat/pharmacologie , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Azacitidine/pharmacologie , Régulation négative/effets des médicaments et des substances chimiques , Protéolyse/effets des médicaments et des substances chimiques , Régulation positive/effets des médicaments et des substances chimiques , Inhibiteurs de désacétylase d'histone/pharmacologie
3.
Oncol Res ; 32(7): 1173-1184, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948026

RÉSUMÉ

Background: Inhibitor of NF-κB kinase-interacting protein (IKIP) is known to promote proliferation of glioblastoma (GBM) cells, but how it affects migration and invasion by those cells is unclear. Methods: We compared levels of IKIP between glioma tissues and normal brain tissue in clinical samples and public databases. We examined the effects of IKIP overexpression and knockdown on the migration and invasion of GBM using transwell and wound healing assays, and we compared the transcriptomes under these different conditions to identify the molecular mechanisms involved. Results: Based on data from our clinical samples and from public databases, IKIP was overexpressed in GBM tumors, and its expression level correlated inversely with survival. IKIP overexpression in GBM cells inhibited migration and invasion in transwell and wound healing assays, whereas IKIP knockdown exerted the opposite effects. IKIP overexpression in GBM cells that were injected into mouse brain promoted tumor growth but inhibited tumor invasion of surrounding tissue. The effects of IKIP were associated with downregulation of THBS1 mRNA and concomitant inhibition of THBS1/FAK signaling. Conclusions: IKIP inhibits THBS1/FAK signaling to suppress migration and invasion of GBM cells.


Sujet(s)
Tumeurs du cerveau , Mouvement cellulaire , Focal adhesion kinase 1 , Glioblastome , Invasion tumorale , Transduction du signal , Thrombospondine-1 , Humains , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Glioblastome/génétique , Animaux , Souris , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Thrombospondine-1/métabolisme , Thrombospondine-1/génétique , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Régulation négative , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire
4.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38969364

RÉSUMÉ

The transmembrane E3 ligases RNF43 and ZNRF3 perform key tumour suppressor roles by inducing endocytosis of members of the Frizzled (FZD) family, the primary receptors for WNT. Loss-of-function mutations in RNF43 and ZNRF3 mediate FZD stabilisation and a WNT-hypersensitive growth state in various cancer types. Strikingly, RNF43 and ZNRF3 mutations are differentially distributed across cancer types, raising questions about their functional redundancy. Here, we compare the efficacy of RNF43 and ZNRF3 of targeting different FZDs for endocytosis. We find that RNF43 preferentially down-regulates FZD1/FZD5/FZD7, whereas ZNRF3 displays a preference towards FZD6. We show that the RNF43 transmembrane domain (TMD) is a key molecular determinant for inducing FZD5 endocytosis. Furthermore, a TMD swap between RNF43 and ZNRF3 re-directs their preference for FZD5 down-regulation. We conclude that RNF43 and ZNRF3 preferentially down-regulate specific FZDs, in part by a TMD-dependent mechanism. In accordance, tissue-specific expression patterns of FZD homologues correlate with the incidence of RNF43 or ZNRF3 cancer mutations in those tissues. Consequently, our data point to druggable vulnerabilities of specific FZD receptors in RNF43- or ZNRF3-mutant human cancers.


Sujet(s)
Endocytose , Récepteurs Frizzled , Ubiquitin-protein ligases , Récepteurs Frizzled/métabolisme , Récepteurs Frizzled/génétique , Humains , Endocytose/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Cellules HEK293 , Mutation , Voie de signalisation Wnt/génétique , Régulation négative/génétique
5.
Life Sci Alliance ; 7(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38977311

RÉSUMÉ

The ubiquitin-like modifier FAT10 is upregulated under pro-inflammatory conditions, targets its substrates for proteasomal degradation and functions as a negative regulator of the type-I IFN response. Influenza A virus infection upregulates the production of type-I IFN and the expression of the E3 ligase TRIM21, which regulates type-I IFN production in a positive feedback manner. In this study, we show that FAT10 becomes covalently conjugated to TRIM21 and that this targets TRIM21 for proteasomal degradation. We further show that the coiled-coil and PRYSPRY domains of TRIM21 and the C-terminal diglycine motif of FAT10 are important for the TRIM21-FAT10 interaction. Moreover, upon influenza A virus infection and in the presence of FAT10 the total ubiquitination of TRIM21 is reduced and our data reveal that the FAT10-mediated degradation of TRIM21 diminishes IFNß production. Overall, this study provides strong evidence that FAT10 down-regulates the antiviral type-I IFN production by modulating additional molecules of the RIG-I signaling pathway besides the already published OTUB1. In addition, we elucidate a novel mechanism of FAT10-mediated proteasomal degradation of TRIM21 that regulates its stability.


Sujet(s)
Interféron de type I , Proteasome endopeptidase complex , Ribonucléoprotéines , Ubiquitination , Ubiquitines , Humains , Ribonucléoprotéines/métabolisme , Ribonucléoprotéines/génétique , Interféron de type I/métabolisme , Ubiquitines/métabolisme , Ubiquitines/génétique , Proteasome endopeptidase complex/métabolisme , Régulation négative , Cellules HEK293 , Transduction du signal , Virus de la grippe A/physiologie , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protéolyse , Animaux
6.
PLoS One ; 19(7): e0306429, 2024.
Article de Anglais | MEDLINE | ID: mdl-38980867

RÉSUMÉ

Brucella abortus (Ba) is a pathogen that survives inside macrophages. Despite being its preferential niche, Ba infects other cells, as shown by the multiple signs and symptoms humans present. This pathogen can evade our immune system. Ba displays a mechanism of down-modulating MHC-I on monocytes/macrophages in the presence of IFN-γ (when Th1 response is triggered) without altering the total expression of MHC-I. The retained MHC-I proteins are located within the Golgi Apparatus (GA). The RNA of Ba is one of the PAMPs that trigger this phenomenon. However, we acknowledged whether this event could be triggered in other cells relevant during Ba infection. Here, we demonstrate that Ba RNA reduced the surface expression of MHC-I induced by IFN-γ in the human bronchial epithelium (Calu-6), the human alveolar epithelium (A-549) and the endothelial microvasculature (HMEC) cell lines. In Calu-6 and HMEC cells, Ba RNA induces the retention of MHC-I in the GA. This phenomenon was not observed in A-549 cells. We then evaluated the effect of Ba RNA on the secretion of IL-8, IL-6 and MCP-1, key cytokines in Ba infection. Contrary to our expectations, HMEC, Calu-6 and A-549 cells treated with Ba RNA had higher IL-8 and IL-6 levels compared to untreated cells. In addition, we showed that Ba RNA down-modulates the MHC-I surface expression induced by IFN-γ on human monocytes/macrophages via the pathway of the Epidermal Growth Factor Receptor (EGFR). So, cells were stimulated with an EGFR ligand-blocking antibody (Cetuximab) and Ba RNA. Neutralization of the EGFR to some extent reversed the down-modulation of MHC-I mediated by Ba RNA in HMEC and A-549 cells. In conclusion, this is the first study exploring a central immune evasion strategy, such as the downregulation of MHC-I surface expression, beyond monocytes and could shed light on how it persists effectively within the host, enduring unseen and escaping CD8+ T cell surveillance.


Sujet(s)
Brucella abortus , Cellules endothéliales , Cellules épithéliales , Antigènes d'histocompatibilité de classe I , Interféron gamma , Humains , Interféron gamma/métabolisme , Interféron gamma/pharmacologie , Cellules endothéliales/métabolisme , Cellules endothéliales/microbiologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/immunologie , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Cellules épithéliales/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/génétique , ARN bactérien/génétique , Lignée cellulaire , Régulation négative/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Brucellose/immunologie , Brucellose/métabolisme , Brucellose/microbiologie , Brucellose/génétique , Appareil de Golgi/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Monocytes/métabolisme , Monocytes/immunologie , Monocytes/effets des médicaments et des substances chimiques
7.
Theranostics ; 14(10): 3945-3962, 2024.
Article de Anglais | MEDLINE | ID: mdl-38994035

RÉSUMÉ

Rationale: NLRP3 inflammasome is critical in the development and progression of many metabolic diseases driven by chronic inflammation, but its effect on the pathology of postmenopausal osteoporosis (PMOP) remains poorly understood. Methods: We here firstly examined the levels of NLRP3 inflammasome in PMOP patients by ELISA. Then we investigated the possible mechanisms underlying the effect of NLRP3 inflammasome on PMOP by RNA sequencing of osteoblasts treated with NLRP3 siRNA and qPCR. Lastly, we accessed the effect of decreased NLRP3 levels on ovariectomized (OVX) rats. To specifically deliver NLRP3 siRNA to osteoblasts, we constructed NLRP3 siRNA wrapping osteoblast-specific aptamer (CH6)-functionalized lipid nanoparticles (termed as CH6-LNPs-siNLRP3). Results: We found that the levels of NLRP3 inflammasome were significantly increased in patients with PMOP, and were negatively correlated with estradiol levels. NLRP3 knock-down influenced signal pathways including immune system process, interferon signal pathway. Notably, of the top ten up-regulated genes in NLRP3-reduced osteoblasts, nine genes (except Mx2) were enriched in immune system process, and five genes were related to interferon signal pathway. The in vitro results showed that CH6-LNPs-siNLRP3 was relatively uniform with a dimeter of 96.64 ± 16.83 nm and zeta potential of 38.37 ± 1.86 mV. CH6-LNPs-siNLRP3 did not show obvious cytotoxicity and selectively delivered siRNA to bone tissue. Moreover, CH6-LNPs-siNLRP3 stimulated osteoblast differentiation by activating ALP and enhancing osteoblast matrix mineralization. When administrated to OVX rats, CH6-LNPs-siNLRP3 promoted bone formation and bone mass, improved bone microarchitecture and mechanical properties by decreasing the levels of NLRP3, IL-1ß and IL-18 and increasing the levels of OCN and Runx2. Conclusion: NLRP3 inflammasome may be a new biomarker for PMOP diagnosis and plays a key role in the pathology of PMOP. CH6-LNPs-siNLRP3 has potential application for the treatment of PMOP.


Sujet(s)
Inflammasomes , Liposomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Nanoparticules , Ostéoblastes , Ostéoporose post-ménopausique , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Femelle , Humains , Rats , Inflammasomes/métabolisme , Nanoparticules/composition chimique , Ostéoporose post-ménopausique/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Petit ARN interférent/administration et posologie , Aptamères nucléotidiques/pharmacologie , Aptamères nucléotidiques/administration et posologie , Modèles animaux de maladie humaine , Adulte d'âge moyen , Ovariectomie
8.
Int J Mol Med ; 54(3)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38994756

RÉSUMÉ

Drug resistance is a key factor underlying the failure of tumor chemotherapy. It enhances the stem­like cell properties of cancer cells, tumor metastasis and relapse. Luteolin is a natural flavonoid with strong anti­tumor effects. However, the mechanism(s) by which luteolin protects against paclitaxel (PTX)­resistant cancer cell remains to be elucidated. The inhibitory effect of luteolin on the proliferation of EC1/PTX and EC1 cells was detected by cell counting kit­8 assay. Colony formation and flow cytometry assays were used to assess clonogenic capacity, cell cycle and apoptosis. Wound healing and Transwell invasion tests were used to investigate the effects of luteolin on the migration and invasion of EC1/PTX cells. Western blotting was used to detect the protein levels of EMT­related proteins and stem cell markers after sphere formation. Parental cells and drug­resistant cells were screened by high­throughput sequencing to detect the differential expression of RNA and differential genes. ELISA and western blotting were used to verify the screened PI3K/Akt signaling pathway, key proteins of which were explored by molecular docking. Hematoxylin and eosin staining and TUNEL staining were used to observe tumor xenografts on morphology and apoptosis in nude mice. The present study found that luteolin inhibited tumor resistance (inhibited proliferation, induced cell cycle arrest and apoptosis and hindered migration invasion, EMT and stem cell spherification) in vitro in PTX­resistant esophageal squamous cell carcinoma (ESCC) cells. In addition, luteolin enhanced drug sensitivity and promoted the apoptosis of drug­resistant ESCC cells in combination with PTX. Mechanistically, luteolin may inhibit the PI3K/AKT signaling pathway by binding to the active sites of focal adhesion kinase (FAK), Src and AKT. Notably, luteolin lowered the tumorigenic potential of PTX­resistant ESCC cells but did not show significant toxicity in vivo. Luteolin enhanced drug chemosensitivity by downregulating the FAK/PI3K/AKT pathway in PTX­resistant ESCC and could be a promising agent for the treatment of PTX­resistant ESCC cancers.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Lutéoline , Paclitaxel , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Lutéoline/pharmacologie , Paclitaxel/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Humains , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Animaux , Lignée cellulaire tumorale , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Carcinome épidermoïde de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/anatomopathologie , Souris nude , Mouvement cellulaire/effets des médicaments et des substances chimiques , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Tests d'activité antitumorale sur modèle de xénogreffe , Antinéoplasiques d'origine végétale/pharmacologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Femelle , Mâle
9.
PeerJ ; 12: e17657, 2024.
Article de Anglais | MEDLINE | ID: mdl-39011384

RÉSUMÉ

Background: Our previous studies have successfully reported the reprogramming of fibroblasts into induced mammary epithelial cells (iMECs). However, the regulatory relationships and functional roles of MicroRNAs (miRNAs) in the progression of fibroblasts achieving the cell fate of iMECs are insufficiently understood. Methods: First, we performed pre-and post-induction miRNAs sequencing analysis by using high-throughput sequencing. Following that, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment studies were used to determine the primary roles of the significantly distinct miRNAs and targeted genes. Finally, the effect of miR-222-3p on iMECs fate reprogramming in vitro by transfecting. Results: As a result goat ear fibroblasts (GEFs) reprogramming into iMECs activates a regulatory program, involving 79 differentially expressed miRNAs. Besides, the programming process involved changes in multiple signaling pathways such as adherens junction, TGF-ß signaling pathway, GnRH secretion and the prolactin signaling pathway, etc. Furthermore, it was discovered that the expression of miR-222-3p downregulation by miR-222-3p inhibitor significantly increase the reprogramming efficiency and promoted lipid accumulation of iMECs.


Sujet(s)
Reprogrammation cellulaire , Cellules épithéliales , Fibroblastes , Capra , microARN , microARN/génétique , microARN/métabolisme , Animaux , Fibroblastes/métabolisme , Cellules épithéliales/métabolisme , Femelle , Reprogrammation cellulaire/génétique , Glandes mammaires animales/cytologie , Glandes mammaires animales/métabolisme , Transduction du signal , Cellules cultivées , Régulation négative
10.
Nat Commun ; 15(1): 5887, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003305

RÉSUMÉ

Memory engrams are a subset of learning activated neurons critical for memory recall, consolidation, extinction and separation. While the transcriptional profile of engrams after learning suggests profound neural changes underlying plasticity and memory formation, little is known about how memory engrams are selected and allocated. As epigenetic factors suppress memory formation, we developed a CRISPR screening in the hippocampus to search for factors controlling engram formation. We identified histone lysine-specific demethylase 4a (Kdm4a) as a negative regulator for engram formation. Kdm4a is downregulated after neural activation and controls the volume of mossy fiber boutons. Mechanistically, Kdm4a anchors to the exonic region of Trpm7 gene loci, causing the stalling of nascent RNAs and allowing burst transcription of Trpm7 upon the dismissal of Kdm4a. Furthermore, the YTH domain containing protein 2 (Ythdc2) recruits Kdm4a to the Trpm7 gene and stabilizes nascent RNAs. Reducing the expression of Kdm4a in the hippocampus via genetic manipulation or artificial neural activation facilitated the ability of pattern separation in rodents. Our work indicates that Kdm4a is a negative regulator of engram formation and suggests a priming state to generate a separate memory.


Sujet(s)
Hippocampe , Mémoire , Canaux cationiques TRPM , Animaux , Hippocampe/métabolisme , Souris , Mémoire/physiologie , Canaux cationiques TRPM/métabolisme , Canaux cationiques TRPM/génétique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Humains , Régulation négative/génétique , Neurones/métabolisme , Mâle , Souris de lignée C57BL , Rats , Systèmes CRISPR-Cas , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Plasticité neuronale/génétique , Cellules HEK293 , Histone Demethylases
11.
Mol Biol Rep ; 51(1): 789, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990383

RÉSUMÉ

BACKGROUND: Syringin, a phenylpropanoid glycoside, has exhibited numerous biological properties including inhibitory activities against various immune and inflammatory disorders. In this study, syringin isolated from Tinospora crispa was evaluated for its ability to down-regulate activated nuclear factor-kappa B (NF-κB), phosphoinositide-3-kinase-Akt (PI3K-Akt) and mitogen-activated protein kinases (MAPKs) signal transducing networks in U937 macrophages activated by lipopolysaccharide. METHODS: The attenuating effects of syringin on the productions of prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), interleukin-1ß (IL-1ß), and tumor necrosis factor-α (TNF-α), and the expressions of signaling molecules of the signaling pathways were investigated by using ELISA, Western blot, and qRT-PCR. RESULTS: Syringin downregulated the NF-κB, MAPKs, and PI3K-Akt signal networks by significantly reducing PGE2 production in the macrophages via suppression of COX-2 gene and protein expression levels. It also reduced TNF-α and IL-1ß secretion and their mRNA expression, suppressed phosphorylation of NF-κB (p65), IKKα/ß, and IκBα, and restored ability of IκBα to degrade. Syringin dose-dependently attenuated Akt, p38 MAPKs, JNK, and ERK phosphorylation. Also, the expression of corresponding upstream signaling molecules toll-like receptor 4 (TLR4) and myeloid differentiation primary response gene 88 (MyD88) were down-regulated in response to syringin treatment. CONCLUSION: The suppressive effect of syringin on the inflammatory signaling molecules in MyD88-dependent pathways suggested it's potential as a drug candidate for development into an agent for treatment of various immune-mediated inflammatory disorders.


Sujet(s)
Glucosides , Lipopolysaccharides , Macrophages , Facteur de différenciation myéloïde-88 , Facteur de transcription NF-kappa B , Phénylpropionates , Transduction du signal , Tinospora , Humains , Facteur de différenciation myéloïde-88/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Lipopolysaccharides/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Tinospora/composition chimique , Glucosides/pharmacologie , Phénylpropionates/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Cellules U937 , Dinoprostone/métabolisme , Interleukine-1 bêta/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Cyclooxygenase 2/métabolisme , Cyclooxygenase 2/génétique , Médiateurs de l'inflammation/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Récepteur de type Toll-4/métabolisme
12.
Arch Dermatol Res ; 316(7): 455, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967656

RÉSUMÉ

Tirbanibulin 1% ointment is a synthetic antiproliferative agent approved in 2021 by the European Union for treating actinic keratoses (AK). Topical tirbanibulin has clinically resolved HPV-57 ( +) squamous cell carcinoma (SCC), HPV-16 ( +) vulvar high-grade squamous intraepithelial lesion, epidermodysplasia verruciformis, and condyloma. We examined how tirbanibulin might affect HPV oncoprotein expression and affect other cellular pathways involved in cell proliferation and transformation. We treated the HeLa cell line, containing integrated HPV-18, with increasing doses of tirbanibulin to determine the effects on cell proliferation. Immunoblotting was performed with antibodies against the Src canonical pathway, HPV 18 E6 and E7 transcription regulation, apoptosis, and invasion and metastasis pathways. Cell proliferation assays with tirbanibulin determined the half-maximal inhibitory concentration (IC50) of HeLa cells to be 31.49 nmol/L. Increasing concentrations of tirbanibulin downregulates the protein expression of Src (p < 0.001), phospho-Src (p < 0.001), Ras (p < 0.01), c-Raf (p < 0.001), ERK1 (p < 0.001), phospho-ERK1 (p < 0.001), phospho-ERK2 (p < 0.01), phospho-Mnk1 (p < 0.001), eIF4E (p < 0.01), phospho-eIF4E (p < 0.001), E6 (p < 0.01), E7 (p < 0.01), Rb (p < 0.01), phospho-Rb (p < 0.001), MDM2 (p < 0.01), E2F1 (p < 0.001), phospho-FAK (p < 0.001), phospho-p130 Cas (p < 0.001), Mcl-1 (p < 0.01), and Bcl-2 (p < 0.001), but upregulates cPARP (p < 0.001), and cPARP/fPARP (p < 0.001). These results demonstrate that tirbanibulin may impact expression of HPV oncoproteins via the Src- MEK- pathway. Tirbanibulin significantly downregulates oncogenic proteins related to cell cycle regulation and cell proliferation while upregulating apoptosis pathways.


Tirbanibulin is Promising Novel Therapy for Human Papillomavirus (HPV)-associated Diseases.Tirbanibulin 1% ointment is an approved synthetic topical ointment for treating actinic keratoses (AK), a precancer of skin cancer. Topical tirbanibulin has previously been reported to clinically resolve human papillomavirus (HPV)-( +) diseases.In this study, we examine how tirbanibulin may affect the HPV and pathways associated with cancer.We treated the HeLa cell line to determine the effects on HPV cell proliferation. Increasing the concentration of tirbanibulin statistically significantly affected numerous cellular pathways often associated with cancer.These results demonstrate that tirbanibulin may impact expression of HPV oncoproteins and thereby kill cancer cells.


Sujet(s)
Prolifération cellulaire , Régulation négative , Papillomavirus humain de type 18 , Protéines des oncogènes viraux , Humains , Cellules HeLa , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines des oncogènes viraux/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Infections à papillomavirus/virologie , Infections à papillomavirus/traitement médicamenteux , Protéines E7 de papillomavirus/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , src-Family kinases/métabolisme , src-Family kinases/antagonistes et inhibiteurs , Femelle , Virus des Papillomavirus humains , Protéines de liaison à l'ADN
13.
Sci Rep ; 14(1): 15175, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956251

RÉSUMÉ

In the current study, we aimed to investigate whether disulfiram (DSF) exerts a neuroprotective role in cerebral ischemiareperfusion (CI-RI) injury by modulating ferredoxin 1 (FDX1) to regulate copper ion (Cu) levels and inhibiting inflammatory responses. To simulate CI-RI, a transient middle cerebral artery occlusion (tMCAO) model in C57/BL6 mice was employed. Mice were administered with or without DSF before and after tMCAO. Changes in infarct volume after tMCAO were observed using TTC staining. Nissl staining and hematoxylin-eosin (he) staining were used to observe the morphological changes of nerve cells at the microscopic level. The inhibitory effect of DSF on initial inflammation was verified by TUNEL assay, apoptosis-related protein detection and iron concentration detection. FDX1 is the main regulatory protein of copper death, and the occurrence of copper death will lead to the increase of HSP70 stress and inflammatory response. Cuproptosis-related proteins and downstream inflammatory factors were detected by western blotting, immunofluorescence staining, and immunohistochemistry. The content of copper ions was detected using a specific kit, while electron microscopy was employed to examine mitochondrial changes. We found that DSF reduced the cerebral infarction volume, regulated the expression of cuproptosis-related proteins, and modulated copper content through down regulation of FDX1 expression. Moreover, DSF inhibited the HSP70/TLR-4/NLRP3 signaling pathway. Collectively, DSF could regulate Cu homeostasis by inhibiting FDX1, acting on the HSP70/TLR4/NLRP3 pathway to alleviate CI/RI. Accordingly, DSF could mitigate inflammatory responses and safeguard mitochondrial integrity, yielding novel therapeutic targets and mechanisms for the clinical management of ischemia-reperfusion injury.


Sujet(s)
Cuivre , Disulfirame , Homéostasie , Inflammation , Souris de lignée C57BL , Lésion d'ischémie-reperfusion , Animaux , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Disulfirame/pharmacologie , Souris , Cuivre/métabolisme , Homéostasie/effets des médicaments et des substances chimiques , Mâle , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Régulation négative/effets des médicaments et des substances chimiques , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Modèles animaux de maladie humaine , Ferrosulfoprotéines/métabolisme , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/traitement médicamenteux , Encéphalopathie ischémique/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Récepteur de type Toll-4/métabolisme
14.
Sci Rep ; 14(1): 15089, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956394

RÉSUMÉ

Morgana is a ubiquitous HSP90 co-chaperone protein coded by the CHORDC1 gene. Morgana heterozygous mice develop with age a myeloid malignancy resembling human atypical myeloid leukemia (aCML), now renamed MDS/MPN with neutrophilia. Patients affected by this pathology exhibit low Morgana levels in the bone marrow (BM), suggesting that Morgana downregulation plays a causative role in the human malignancy. A decrease in Morgana expression levels is also evident in the BM of a subgroup of Philadelphia-positive (Ph+) chronic myeloid leukemia (CML) patients showing resistance or an incomplete response to imatinib. Despite the relevance of these data, the mechanism through which Morgana expression is downregulated in patients' bone marrow remains unclear. In this study, we investigated the possibility that Morgana expression is regulated by miRNAs and we demonstrated that Morgana is under the control of four miRNAs (miR-15a/b and miR-26a/b) and that miR-15a may account for Morgana downregulation in CML patients.


Sujet(s)
Protéines du choc thermique HSP90 , Leucémie myéloïde chronique BCR-ABL positive , microARN , microARN/génétique , microARN/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Humains , Protéines du choc thermique HSP90/métabolisme , Protéines du choc thermique HSP90/génétique , Animaux , Souris , Régulation de l'expression des gènes dans la leucémie , Régulation négative , Moelle osseuse/métabolisme , Moelle osseuse/anatomopathologie , Chaperons moléculaires/métabolisme , Chaperons moléculaires/génétique
16.
JCI Insight ; 9(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38973609

RÉSUMÉ

Lipoprotein lipase (LPL) hydrolyzes circulating triglycerides (TGs), releasing fatty acids (FA) and promoting lipid storage in white adipose tissue (WAT). However, the mechanisms regulating adipose LPL and its relationship with the development of hypertriglyceridemia are largely unknown. WAT from obese humans exhibited high PAR2 expression, which was inversely correlated with the LPL gene. Decreased LPL expression was also inversely correlated with elevated plasma TG levels, suggesting that adipose PAR2 might regulate hypertriglyceridemia by downregulating LPL. In mice, aging and high palmitic acid diet (PD) increased PAR2 expression in WAT, which was associated with a high level of macrophage migration inhibitory factor (MIF). MIF downregulated LPL expression and activity in adipocytes by binding with CXCR2/4 receptors and inhibiting Akt phosphorylation. In a MIF overexpression model, high-circulating MIF levels suppressed adipose LPL, and this suppression was associated with increased plasma TGs but not FA. Following PD feeding, adipose LPL expression and activity were significantly reduced, and this reduction was reversed in Par2-/- mice. Recombinant MIF infusion restored high plasma MIF levels in Par2-/- mice, and the levels decreased LPL and attenuated adipocyte lipid storage, leading to hypertriglyceridemia. These data collectively suggest that downregulation of adipose LPL by PAR2/MIF may contribute to the development of hypertriglyceridemia.


Sujet(s)
Régulation négative , Hypertriglycéridémie , Lipoprotein lipase , Récepteur de type PAR-2 , Animaux , Lipoprotein lipase/métabolisme , Lipoprotein lipase/génétique , Hypertriglycéridémie/métabolisme , Hypertriglycéridémie/génétique , Souris , Humains , Récepteur de type PAR-2/métabolisme , Récepteur de type PAR-2/génétique , Mâle , Souris knockout , Triglycéride/métabolisme , Triglycéride/sang , Tissu adipeux blanc/métabolisme , Facteurs inhibiteurs de la migration des macrophages/métabolisme , Facteurs inhibiteurs de la migration des macrophages/génétique , Adipocytes/métabolisme , Obésité/métabolisme , Obésité/génétique , Acide palmitique/métabolisme , Femelle , Souris de lignée C57BL , Adulte d'âge moyen
17.
Immun Inflamm Dis ; 12(7): e1301, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967361

RÉSUMÉ

OBJECTIVE: Acute pancreatitis (AP) stands as a frequent cause for clinical emergency hospital admissions. The X-box binding protein 1 (XBP1) was found to be implicated in pancreatic acinar cell apoptosis. The objective is to unveil the potential mechanisms governed by XBP1 and SIRT6 in the context of AP. METHODS: Caerulein-treated human pancreatic duct epithelial (HPDE) cells to establish an in vitro research model. The levels and regulatory role of SIRT6 in the treated cells were evaluated, including its effects on inflammatory responses, oxidative stress, apoptosis, and endoplasmic reticulum stress. The relationship between XBP1 and SIRT6 was explored by luciferase and ChIP experiments. Furthermore, the effect of XBP1 overexpression on the regulatory function of SIRT6 on cells was evaluated. RESULTS: Caerulein promoted the decrease of SIRT6 and the increase of XBP1 in HPDE cells. Overexpression of SIRT6 slowed down the secretion of inflammatory factors, oxidative stress, apoptosis level, and endoplasmic reticulum stress in HPDE cells. However, XBP1 negatively regulated SIRT6, and XBP1 overexpression partially reversed the regulation of SIRT6 on the above aspects. CONCLUSION: Our study illuminates the role of XBP1 in downregulating SIRT6 in HPDE cells, thereby promoting cellular injury. Inhibiting XBP1 or augmenting SIRT6 levels holds promise in preserving cell function and represents a potential therapeutic avenue in the management of AP.


Sujet(s)
Apoptose , Régulation négative , Cellules épithéliales , Conduits pancréatiques , Pancréatite , Sirtuines , Protéine-1 liant la boite X , Humains , Sirtuines/métabolisme , Sirtuines/génétique , Cellules épithéliales/métabolisme , Protéine-1 liant la boite X/métabolisme , Protéine-1 liant la boite X/génétique , Pancréatite/métabolisme , Pancréatite/anatomopathologie , Conduits pancréatiques/métabolisme , Conduits pancréatiques/anatomopathologie , Stress du réticulum endoplasmique , Stress oxydatif , Lignée cellulaire , Céruléine/toxicité
18.
Front Immunol ; 15: 1427200, 2024.
Article de Anglais | MEDLINE | ID: mdl-38989284

RÉSUMÉ

Introduction: Glioma, a prevalent and deadly brain tumor, is marked by significant cellular heterogeneity and metabolic alterations. However, the comprehensive cell-of-origin and metabolic landscape in high-grade (Glioblastoma Multiforme, WHO grade IV) and low-grade (Oligoastrocytoma, WHO grade II) gliomas remains elusive. Methods: In this study, we undertook single-cell transcriptome sequencing of these glioma grades to elucidate their cellular and metabolic distinctions. Following the identification of cell types, we compared metabolic pathway activities and gene expressions between high-grade and low-grade gliomas. Results: Notably, astrocytes and oligodendrocyte progenitor cells (OPCs) exhibited the most substantial differences in both metabolic pathways and gene expression, indicative of their distinct origins. The comprehensive analysis identified the most altered metabolic pathways (MCPs) and genes across all cell types, which were further validated against TCGA and CGGA datasets for clinical relevance. Discussion: Crucially, the metabolic enzyme phosphodiesterase 8B (PDE8B) was found to be exclusively expressed and progressively downregulated in astrocytes and OPCs in higher-grade gliomas. This decreased expression identifies PDE8B as a metabolism-related oncogene in IDH-mutant glioma, marking its dual role as both a protective marker for glioma grading and prognosis and as a facilitator in glioma progression.


Sujet(s)
Tumeurs du cerveau , Analyse de profil d'expression de gènes , Gliome , Isocitrate dehydrogenases , Mutation , Analyse sur cellule unique , Humains , Isocitrate dehydrogenases/génétique , Gliome/génétique , Gliome/anatomopathologie , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Régulation de l'expression des gènes tumoraux , Transcriptome , Astrocytes/métabolisme , Oncogènes , Régulation négative , Précurseurs des oligodendrocytes/métabolisme , Grading des tumeurs , Marqueurs biologiques tumoraux/génétique
19.
Clinics (Sao Paulo) ; 79: 100428, 2024.
Article de Anglais | MEDLINE | ID: mdl-38972248

RÉSUMÉ

The use of stem cells capable of multilineage differentiation in treating Pelvic Floor Dysfunction (PFD) holds great promise since they are susceptible to entering connective tissue of various cell types and repairing damaged tissues. This research investigated the effect of microRNA-181a-5p (miR-181a-5p) on Bone Marrow Mesenchymal Stem Cells (BMSCs) in rats with PFD. BMSCs were transfected and analyzed for their fibroblast differentiation ability. miR-181a-5p, MFN1, and fibroblast-related genes were quantitatively analyzed. Whether MFN1 is a target gene of miR-181a-5p was predicted and confirmed. The efficacy of BMSCs in vivo rats with PFD was evaluated by measuring Leak Point Pressure (LPP), Conscious Cystometry (CMG), hematoxylin and eosin staining, and Masson staining. The present results discovered that miR-181a-5p was up-regulated and MFN1 was down-regulated during the differentiation of BMSCs into fibroblasts. Fibroblast differentiation of BMSCs was promoted after miR-181a-5p was induced or MFN1 was suppressed, but it was suppressed after miR-181a-5p was silenced. miR-181a-5p improved LPP and conscious CMG outcomes in PDF rats by targeting MFN1 expression, thereby accelerating fibroblast differentiation of BMSCs. In brief, miR-181a-5p induces fibroblast differentiation of BMSCs in PDF rats by MFN1, potentially targeting PDF therapeutics.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Cellules souches mésenchymateuses , microARN , Animaux , Cellules souches mésenchymateuses/métabolisme , microARN/génétique , microARN/métabolisme , Femelle , Rat Sprague-Dawley , Troubles du plancher pelvien/génétique , Troubles du plancher pelvien/thérapie , Rats , Régulation positive , Modèles animaux de maladie humaine , Régulation négative , Cellules cultivées
20.
Methods Mol Biol ; 2816: 175-191, 2024.
Article de Anglais | MEDLINE | ID: mdl-38977599

RÉSUMÉ

The trabecular meshwork (TM) from primary open-angle glaucoma (POAG) cases has been found to contain decreased levels of intracellular plasmalogens. Plasmalogens are a subset of lipids involved in diverse cellular processes such as intracellular signaling, membrane asymmetry, and protein regulation. Proper plasmalogen biosynthesis is regulated by rate-limiting enzyme fatty acyl-CoA reductase (Far1). ATPase phospholipid transporting 8B2 (ATP8B2) is a type IV P-type ATPase responsible for the asymmetric distribution of plasmalogens between the intracellular and extracellular leaflets of the plasma membranes. Here we describe the methodology for extraction and culturing of TM cells from corneal tissue and subsequent downregulation of ATP8B2 using siRNA transfection. Further quantification and downstream effects of ATP8B2 gene knockdown will be analyzed utilizing immunoblotting techniques.


Sujet(s)
Glaucome à angle ouvert , Acétalphosphatides , Réseau trabéculaire de la sclère , Réseau trabéculaire de la sclère/métabolisme , Réseau trabéculaire de la sclère/cytologie , Humains , Acétalphosphatides/métabolisme , Glaucome à angle ouvert/métabolisme , Glaucome à angle ouvert/génétique , Glaucome à angle ouvert/anatomopathologie , Adenosine triphosphatases/métabolisme , Adenosine triphosphatases/génétique , Petit ARN interférent/génétique , Régulation négative , Cellules cultivées , Techniques de knock-down de gènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...