Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.493
Filtrer
1.
Exp Cell Res ; 440(1): 114116, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38830568

RÉSUMÉ

During the progression of diabetic kidney disease, proximal tubular epithelial cells respond to high glucose to induce hypertrophy and matrix expansion leading to renal fibrosis. Recently, a non-canonical PTEN has been shown to be translated from an upstream initiation codon CUG (leucine) to produce a longer protein called PTEN-Long (PTEN-L). Interestingly, the extended sequence present in PTEN-L contains cell secretion/penetration signal. Role of this non-canonical PTEN-L in diabetic renal tubular injury is not known. We show that high glucose decreases expression of PTEN-L. As a mechanism of its function, we find that reduced PTEN-L activates Akt-2, which phosphorylates and inactivate tuberin and PRAS40, resulting in activation of mTORC1 in tubular cells. Antibacterial agent acriflavine and antiviral agent ATA regulate translation from CUG codon. Acriflavine and ATA, respectively, decreased and increased expression of PTEN-L to altering Akt-2 and mTORC1 activation in the absence of change in expression of canonical PTEN. Consequently, acriflavine and ATA modulated high glucose-induced tubular cell hypertrophy and lamininγ1 expression. Importantly, expression of PTEN-L inhibited high glucose-stimulated Akt/mTORC1 activity to abrogate these processes. Since PTEN-L contains secretion/penetration signals, addition of conditioned medium containing PTEN-L blocked Akt-2/mTORC1 activity. Notably, in renal cortex of diabetic mice, we found reduced PTEN-L concomitant with Akt-2/mTORC1 activation, leading to renal hypertrophy and lamininγ1 expression. These results present first evidence for involvement of PTEN-L in diabetic kidney disease.


Sujet(s)
Néphropathies diabétiques , Glucose , Tubules contournés proximaux , Complexe-1 cible mécanistique de la rapamycine , Phosphohydrolase PTEN , Néphropathies diabétiques/métabolisme , Néphropathies diabétiques/anatomopathologie , Néphropathies diabétiques/génétique , Animaux , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Glucose/métabolisme , Glucose/pharmacologie , Tubules contournés proximaux/métabolisme , Tubules contournés proximaux/anatomopathologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Régulation négative/effets des médicaments et des substances chimiques , Souris , Humains , Mâle , Souris de lignée C57BL , Transduction du signal
2.
Exp Cell Res ; 440(1): 114125, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38880324

RÉSUMÉ

Bladder cancer(BC) is one of the most prevalent cancers in the urinary tract, with high recurrence and fatality rates. Research indicates that go-ichi-ni-san complex subunit 1 (GINS1) crucially influences cancer progression by regulating DNA replication through cell cycle modulation. Thus, suppressing the active proliferation of cells in tumor tissues may require silencing GINS1. However, the consequences of GINS1 in bladder cancer aren't to be determined. In this paper, we examine the role and mechanism of GINS1 in the development of bladder cancer. GINS1 expression levels and prognostic relevance in bladder cancer were validated using Western blotting, immunohistochemistry, and Kaplan-Meier survival analysis. The influence of GINS1 on bladder cancer was investigated using a variety of approaches, including cell transfection, cell counts, transwell migrations, colony formation, and flow cytometry. Immunohistochemistry studies demonstrate that GINS1 expression is increased in bladder cancer tissues. GINS1 silencing resulted in an arrest of the cell cycle at the phase of G0/G1, which inhibited BC cell growth both in vitro and in vivo. GINS1 knockdown also hindered the AKT/mTOR pathway. Furthermore, increased GINS1 expression affects the cell cycle and stimulates the AKT/mTOR pathway, allowing BC to develop more quickly. Consequently, GINS1 occurs as a latent therapeutic target, particularly for individuals with BC.


Sujet(s)
Prolifération cellulaire , Protéines chromosomiques nonhistones , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/métabolisme , Humains , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Protéines chromosomiques nonhistones/métabolisme , Protéines chromosomiques nonhistones/génétique , Prolifération cellulaire/génétique , Animaux , Lignée cellulaire tumorale , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Régulation de l'expression des gènes tumoraux , Souris , Évolution de la maladie , Souris nude , Mâle , Femelle , Pronostic , Souris de lignée BALB C , Protéines de liaison à l'ADN
3.
Int J Mol Sci ; 25(11)2024 May 27.
Article de Anglais | MEDLINE | ID: mdl-38891994

RÉSUMÉ

The PI3K/AKT pathway plays a pivotal role in cellular processes, and its dysregulation is implicated in various cancers, including colorectal cancer. The present study correlates the expression levels of critical genes (PIK3CA, PTEN, AKT1, FOXO1, and FRAP) in 60 tumor tissues with clinicopathological and demographic characteristics. The results indicate age-related variation in FOXO1 gene expression, with higher levels observed in patients aged 68 and above. In addition, tumors originating from the rectum exhibit higher FOXO1 expression compared to colon tumors, suggesting region-specific differences in expression. The results also identify the potential correlation between PTEN, PIK3CA gene expression, and parameters such as tumor grade and neuroinvasion. The bioinformatic comparative analysis found that PTEN and FOXO1 expressions were downregulated in colorectal cancer tissue compared to normal colon tissue. Relapse-free survival analysis based on gene expression identified significant correlations, highlighting PTEN and FRAP as potential indicators of favorable outcomes. Our findings provide a deeper understanding of the role of the PI3K/AKT pathway in colorectal cancer and the importance of understanding the molecular basis of colorectal cancer development and progression.


Sujet(s)
Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , Phosphohydrolase PTEN , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Transduction du signal/génétique , Sujet âgé , Mâle , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Femelle , Adulte d'âge moyen , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases de classe I/génétique , Phosphatidylinositol 3-kinases de classe I/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Protéine O1 à motif en tête de fourche/métabolisme , Adulte , Sujet âgé de 80 ans ou plus
4.
Poult Sci ; 103(7): 103866, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38833957

RÉSUMÉ

Avian pathogenic Escherichia coli (APEC) is the causative agent of chicken colibacillosis. Paeoniflorin, a natural ingredient extracted from Paeonia lactiflora, has a variety of pharmacological effects including anti-inflammatory and immunomodulatory. However, its effects and mechanism in APEC-induced acute lung injury (ALI) in chicken is not clear. The aim of this study was to investigate the protective effect of paeoniflorin on APEC-induced ALI and its possible mechanism. Paeoniflorin (25, 50, and 100 mg/kg) was administered by gavage for 5 d starting at 9 d of age and the chicken were infected with APEC by intraperitoneal injection at 12 d of age. The tissues were collected after APEC infection for 36 h for analysis. The results showed that paeoniflorin significantly alleviated the symptoms, increased the survival rate and body weight gain of APEC-infected chicken, and improved the histopathological damages, and reduced APEC loads in lung tissues. In addition, paeoniflorin restored the gene expression of ZO-1, Occludin and Claudin-3 during APEC infection. Moreover, paeoniflorin pretreatment significantly affected the endocannabinoid system (ECs) by increasing DAGL, decreasing MAGL, increasing secretion of 2-AG. Then, paeoniflorin significantly decreased the secretion of IL-1ß, IL-6 and TNF-α in lung tissues, and decreased the mRNA expression of CXCL8, CXCL12, CCL1, CCL5, and CCL17. In addition, paeoniflorin significantly reduced the phosphorylation levels of PI3K, AKT, P65, and IκB. In summary, we found that paeoniflorin inhibited APEC-induced ALI, and its mechanism may be through affecting ECs and inhibiting the activation of PI3K/AKT and NF-κB signaling pathways, which provides a new idea for the prevention and treatment of chicken colibacillosis.


Sujet(s)
Lésion pulmonaire aigüe , Poulets , Infections à Escherichia coli , Glucosides , Monoterpènes , Facteur de transcription NF-kappa B , Phosphatidylinositol 3-kinases , Maladies de la volaille , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Lésion pulmonaire aigüe/prévention et contrôle , Lésion pulmonaire aigüe/étiologie , Lésion pulmonaire aigüe/médecine vétérinaire , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/traitement médicamenteux , Glucosides/pharmacologie , Glucosides/administration et posologie , Monoterpènes/pharmacologie , Monoterpènes/administration et posologie , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/génétique , Infections à Escherichia coli/médecine vétérinaire , Infections à Escherichia coli/traitement médicamenteux , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Protéines aviaires/métabolisme , Protéines aviaires/génétique , Relation dose-effet des médicaments , Escherichia coli/effets des médicaments et des substances chimiques
5.
PLoS Genet ; 20(6): e1011326, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38857279

RÉSUMÉ

The development of ectodermal organs begins with the formation of a stratified epithelial placode that progressively invaginates into the underlying mesenchyme as the organ takes its shape. Signaling by secreted molecules is critical for epithelial morphogenesis, but how that information leads to cell rearrangement and tissue shape changes remains an open question. Using the mouse dentition as a model, we first establish that non-muscle myosin II is essential for dental epithelial invagination and show that it functions by promoting cell-cell adhesion and persistent convergent cell movements in the suprabasal layer. Shh signaling controls these processes by inducing myosin II activation via AKT. Pharmacological induction of AKT and myosin II can also rescue defects caused by the inhibition of Shh. Together, our results support a model in which the Shh signal is transmitted through myosin II to power effective cellular rearrangement for proper dental epithelial invagination.


Sujet(s)
Adhérence cellulaire , Mouvement cellulaire , Protéines Hedgehog , Myosine de type II , Transduction du signal , Animaux , Souris , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Adhérence cellulaire/génétique , Myosine de type II/métabolisme , Myosine de type II/génétique , Mouvement cellulaire/génétique , Épithélium/métabolisme , Morphogenèse/génétique , Dent/métabolisme , Dent/croissance et développement , Cellules épithéliales/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Régulation de l'expression des gènes au cours du développement
6.
Sci Rep ; 14(1): 14145, 2024 06 19.
Article de Anglais | MEDLINE | ID: mdl-38898042

RÉSUMÉ

This study probes the utility of biomarkers for microsatellite instability (MSI) detection and elucidates the molecular dynamics propelling colorectal cancer (CRC) progression. We synthesized a primer panel targeting 725 MSI loci, informed by The Cancer Genome Atlas (TCGA) and ancillary databases, to construct an amplicon library for next-generation sequencing (NGS). K-means clustering facilitated the distillation of 8 prime MSI loci, including activin A receptor type 2A (ACVR2A). Subsequently, we explored ACVR2A's influence on CRC advancement through in vivo tumor experiments and hematoxylin-eosin (HE) staining. Transwell assays gauged ACVR2A's role in CRC cell migration and invasion, while colony formation assays appraised cell proliferation. Western blotting illuminated the impact of ACVR2A suppression on CRC's PI3K/AKT/mTOR pathway protein expressions under hypoxia. Additionally, ACVR2A's influence on CRC-induced angiogenesis was quantified via angiogenesis assays. K-means clustering of NGS data pinpointed 32 MSI loci specific to tumor and DNA mismatch repair deficiency (dMMR) tissues. ACVR2A emerged as a pivotal biomarker, discerning MSI-H tissues with 90.97% sensitivity. A curated 8-loci set demonstrated 100% sensitivity and specificity for MSI-H detection in CRC. In vitro analyses corroborated ACVR2A's critical role, revealing its suppression of CRC proliferation, migration, and invasion. Moreover, ACVR2A inhibition under CRC-induced hypoxia markedly escalated MMP3, CyclinA, CyclinD1, and HIF1α protein expressions, alongside angiogenesis, by triggering the PI3K/AKT/mTOR cascade. The 8-loci ensemble stands as the optimal marker for MSI-H identification in CRC. ACVR2A, a central element within this group, deters CRC progression, while its suppression amplifies PI3K/AKT/mTOR signaling and angiogenesis under hypoxic stress.


Sujet(s)
Récepteur activine, type 2 , Mouvement cellulaire , Tumeurs colorectales , Évolution de la maladie , Instabilité des microsatellites , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Humains , Récepteur activine, type 2/génétique , Récepteur activine, type 2/métabolisme , Animaux , Mouvement cellulaire/génétique , Souris , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Transduction du signal , Mâle , Séquençage nucléotidique à haut débit , Femelle , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique
7.
Biol Direct ; 19(1): 42, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831379

RÉSUMÉ

Triple-negative breast cancer (TNBC) is more aggressive and has a higher metastasis rate compared with other subtypes of breast cancer. Due to the lack of drug-targetable receptors, chemotherapy is now the only available systemic treatment for TNBC. However, some patients might still develop drug resistance and have poor prognosis. Therefore, novel molecular biomarkers and new treatment targets are urgently needed for patients with TNBC. To provide molecular insights into TNBC progression, we investigated the function and the underlying mechanism of Defective in cullin neddylation 1 domain containing 5 (DCUN1D5) in the regulation of TNBC. By TCGA dataset and surgical specimens with immunohistochemical (IHC) staining method, DCUN1D5 was identified to be significantly upregulated in TNBC tumor tissues and negatively associated with prognosis. A series of in vitro and in vivo experiments were performed to confirm the oncogenic role of DCUN1D5 in TNBC. Overexpression of FN1 or PI3K/AKT activator IGF-1 could restore the proliferative and invasive ability induced by DCUN1D5 knockdown and DCUN1D5 could act as a novel transcriptional target of transcription factor Yin Yang 1 (YY1). In conclusion, YY1-enhanced DCUN1D5 expression could promote TNBC progression by FN1/PI3K/AKT pathway and DCUN1D5 might be a potential prognostic biomarker and therapeutic target for TNBC treatment.


Sujet(s)
Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Tumeurs du sein triple-négatives , Facteur de transcription YY1 , Animaux , Femelle , Humains , Souris , Lignée cellulaire tumorale , Évolution de la maladie , Fibronectines , Régulation de l'expression des gènes tumoraux , Souris nude , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Transduction du signal , Activation de la transcription , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Facteur de transcription YY1/métabolisme , Facteur de transcription YY1/génétique
8.
J Cell Mol Med ; 28(11): e18450, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38842133

RÉSUMÉ

Dynactin subunit 2 (DCTN2) has been reported to play a role in progression of several tumours; however, the involvement of DCTN2 in potential mechanism or the tumour immune microenvironment among various cancers still remains largely unknown. Therefore, the objective of this study was to comprehensively investigate the expression status and potential function of DCTN2 in various malignancies through different database, such as The Cancer Genome Atlas, the Genotype-Tissue Expression and Gene Expression Omnimus databases. We discovered that DCTN2 expression was high in many type of tumours tissues compared to adjacent non-tumour ones. High DCTN2 signified poor prognosis for patients with tumours. Additionally, Gene Set Enrichment Analysis (GSEA) analysis revealed that DCTN2 was positively correlated with oncogenic pathways, including cell cycle, tumour metastasis-related pathway, while it was negatively with anti-tumour immune signalling pathway, such as INF-γ response. More importantly, we elucidated the functional impact of DCTN2 on hepatocellular carcinoma (HCC) progression and its underlying mechanisms. DCTN2 expression was much higher in HCC tissues than in adjacent non-tumour tissues. Silencing DCTN2 dramatically suppressed the proliferative and metastasis capacities of tumour cell in vitro. Mechanistically, DCTN2 exerted tumour-promoting effects by modulating the AKT signalling pathway. DCTN2 knockdown in HCC cells inhibited AKT phosphorylation and its downstream targets as well. Rescue experiments revealed that the anti-tumour effects of DCTN2 knockdown were partially reversed upon AKT pathway activation. Overall, DCTN2 may be a potent biomarker signifying tumour prognosis and a promising therapeutic target for tumour treatment, particularly in HCC.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du foie , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Complexe dynactine/métabolisme , Complexe dynactine/génétique , Tumeurs du foie/génétique , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Pronostic , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Microenvironnement tumoral/génétique
9.
J Agric Food Chem ; 72(25): 14349-14363, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38869217

RÉSUMÉ

Deoxynivalenol (DON) is a common agricultural mycotoxin that is chemically stable and not easily removed from cereal foods. When organisms consume food made from contaminated crops, it can be hazardous to their health. Numerous studies in recent years have found that hesperidin (HDN) has hepatoprotective effects on a wide range of toxins. However, few scholars have explored the potential of HDN in attenuating DON-induced liver injury. In this study, we established a low-dose DON exposure model and intervened with three doses of HDN, acting on male C57 BL/6 mice and AML12 cells, which served as in vivo and in vitro models, respectively, to investigate the protective mechanism of HDN against DON exposure-induced liver injury. The results suggested that DON disrupted hepatic autophagic fluxes, thereby impairing liver structure and function, and HDN significantly attenuated these changes. Further studies revealed that HDN alleviated DON-induced excessive autophagy through the mTOR pathway and DON-induced lysosomal dysfunction through the AKT/GSK3ß/TFEB pathway. Overall, our study suggested that HDN could ameliorate DON-induced autophagy flux disorders via the mTOR pathway and the AKT/GSK3ß/TFEB pathway, thereby reducing liver injury.


Sujet(s)
Autophagie , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines , Glycogen synthase kinase 3 beta , Hespéridine , Foie , Souris de lignée C57BL , Protéines proto-oncogènes c-akt , Sérine-thréonine kinases TOR , Trichothécènes , Animaux , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Trichothécènes/toxicité , Mâle , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , Souris , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/métabolisme , Facteurs de transcription à motifs basiques hélice-boucle-hélice et à glissière à leucines/génétique , Hespéridine/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Humains , Lésions hépatiques dues aux substances/métabolisme , Lésions hépatiques dues aux substances/traitement médicamenteux , Lésions hépatiques dues aux substances/prévention et contrôle , Lignée cellulaire
10.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892296

RÉSUMÉ

As we move into the era of precision medicine, the growing relevance of genetic alterations to prostate cancer (PCa) development and treatment demonstrates the importance of characterizing preclinical models at the genomic level. Our study investigated the genomic characterization of eight PCa cell lines to understand which models are clinically relevant. We designed a custom AmpliSeq DNA gene panel that encompassed key molecular pathways targeting AR signaling, apoptosis, DNA damage repair, and PI3K/AKT/PTEN, in addition to tumor suppressor genes. We examined the relationship between cell line genomic alterations and therapeutic response. In addition, using DepMap's Celligner tool, we identified which preclinical models are most representative of specific prostate cancer patient populations on cBioPortal. These data will help investigators understand the genetic differences in preclinical models of PCa and determine which ones are relevant for use in their translational research.


Sujet(s)
Génomique , Tumeurs de la prostate , Humains , Mâle , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Lignée cellulaire tumorale , Génomique/méthodes , Transduction du signal , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Réparation de l'ADN
11.
J Cardiothorac Surg ; 19(1): 322, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844975

RÉSUMÉ

AIM: The most common type of cancer that leads to death worldwide is lung cancer. Despite significant surgery and chemotherapy improvements, lung cancer patient's survival rate is still poor. The RNA polymerase I subunit D (POLR1D) gene can induce various cancers. A current study reported that POLR1D plays a vital role in cancer prognosis. However, its biological function in the development of lung cancer remains unclear. METHODS: Reverse transcription PCR (RT-PCR) measured the relative POLR1D protein expression level in lung cancer cell lines. Lung cancer cell proliferation, migration, and invasion were analyzed by performing cell counting kit-8 (CCK-8), and transwell. The phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/AKT) signaling pathway-related protein expressions were examined by Western blotting assay. RESULTS: POLR1D protein expression was elevated in lung cancer. Lung cancer cell loss-of-function tests showed that POLR1D silencing could attenuate cell viability both in SK-MES-1 and in H2170 cells. Furthermore, silencing POLR1D inhibited SK-MES-1 and H2170 cells proliferation, migration, and invasion. Moreover, SK-MES-1 and H2170 cells' migration and invasion capacity were potentially suppressed by the knockdown of POLR1D. The progression of multiple cancers has been implicated in the PI3K/AKT pathway. Here, we observed that POLR1D silencing suppressed lung cancer progression by inhibition of the PI3K-Akt pathway. CONCLUSIONS: The study speculated that POLR1D might provide a new potential therapeutic possibility for treating lung cancer patients via targeting PI3K/AKT.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Tumeurs du poumon , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , RNA polymerase I/génétique , RNA polymerase I/métabolisme
12.
Int J Nanomedicine ; 19: 6085-6098, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911502

RÉSUMÉ

Introduction: Endometriosis (EM) is an estrogen-dependent benign gynecologic disease affecting approximately 10% of reproductive-age women with a high recurrence rate, but lacks reliable biomarkers. No previous studies have investigated the possible use of extracellular vesicle (EV)-associated micro RNAs (miRNAs) from menstrual blood (MB) as candidate diagnostic or prognostic markers of EM. Methods: Specimens were obtained from endometriosis and non-endometriosis patients at the International Peace Maternity and Child Health Hospital in Shanghai. Microarray was used to screen differentially expressed miRNAs among peritoneal fluid (PF), fallopian tube fluid (FF), and MB. Dual-luciferase reporter gene assay was carried out to verify the relationship between miR-4443 and ACSS2. Cell proliferation and Transwell invasion assays were performed in vitro after intervention on miR-4443 and ACSS2 in hEM15A human endometrial stromal cells and primary human endometrial stromal cells (hESCs). Spearman correlation analysis, receiver operating characteristic (ROC) curve analysis, and survival analysis were applied to clinical data, including severity of symptoms and relapse of EM among EM patients. Results: EV-associated miR-4443 was abundant in MB of endometriosis patients. ACSS2 knockdown and miR-4443 overexpression promoted cell proliferation and migration via the PI3K/AKT pathway. miR-4443 levels in MB-EVs were positively correlated with the degree of dyspareunia (r=0.64; P<0.0001) and dysmenorrhea (r=0.42; P<0.01) in the endometriosis group. ROC curve analyses showed an area under the curve (AUC) of 0.741 (95% CI 0.624-0.858; P<0.05) for miR-4443 and an AUC of 0.929 (95% CI 0.880-0.978; P<0.05) for the combination of miR-4443 and dysmenorrhea. Conclusion: MB-derived EV-associated miR-4443 might participate in endometriosis development, thus providing a new candidate biomarker for the noninvasive prediction of endometriosis recurrence.


Sujet(s)
Prolifération cellulaire , Endométriose , Vésicules extracellulaires , microARN , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Humains , Endométriose/métabolisme , Endométriose/génétique , Femelle , microARN/génétique , microARN/métabolisme , Vésicules extracellulaires/génétique , Vésicules extracellulaires/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Adulte , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Évolution de la maladie , Mouvement cellulaire , Transduction du signal , Lignée cellulaire , Endomètre/métabolisme , Endomètre/anatomopathologie
13.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38928092

RÉSUMÉ

Lung adenocarcinoma (LUAD) is the most widespread cancer in the world, and its development is associated with complex biological mechanisms that are poorly understood. Here, we revealed a marked upregulation in the mRNA level of C1orf131 in LUAD samples compared to non-tumor tissue samples in The Cancer Genome Atlas (TCGA). Depletion of C1orf131 suppressed cell proliferation and growth, whereas it stimulated apoptosis in LUAD cells. Mechanistic investigations revealed that C1orf131 knockdown induced cell cycle dysregulation via the AKT and p53/p21 signalling pathways. Additionally, C1orf131 knockdown blocked cell migration through the modulation of epithelial-mesenchymal transition (EMT) in lung adenocarcinoma. Notably, we identified the C1orf131 protein nucleolar localization sequence, which included amino acid residues 137-142 (KKRKLT) and 240-245 (KKKRKG). Collectively, C1orf131 has potential as a novel therapeutic marker for patients in the future, as it plays a vital role in the progression of lung adenocarcinoma.


Sujet(s)
Adénocarcinome pulmonaire , Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , Protéines proto-oncogènes c-akt , Transduction du signal , Humains , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/génétique , Apoptose/génétique , Évolution de la maladie , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Cellules A549
14.
Curr Med Sci ; 44(3): 589-602, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38748370

RÉSUMÉ

OBJECTIVE: Renal fibrosis is the ultimate pathway of various forms of acute and chronic kidney damage. Notably, the knockout of transient receptor potential channel 6 (TRPC6) has shown promise in alleviating renal fibrosis. However, the regulatory impact of TRPC6 on renal fibrosis remains unclear. METHODS: In vivo, TRPC6 knockout (TRPC6-/-) mice and age-matched 129 SvEv (WT) mice underwent unilateral renal ischemia-reperfusion (uIR) injury surgery on the left renal pedicle or sham operation. Kidneys and serum were collected on days 7, 14, 21, and 28 after euthanasia. In vitro, primary tubular epithelial cells (PTECs) were isolated from TRPC6-/- and WT mice, followed by treatment with transforming growth factor ß1 (TGFß1) for 72 h. The anti-fibrotic effect of TRPC6-/- and the underlying mechanisms were assessed through hematoxylin-eosin staining, Masson staining, immunostaining, qRT-PCR, and Western blotting. RESULTS: Increased TRPC6 expression was observed in uIR mice and PTECs treated with TGFß1. TRPC6-/- alleviated renal fibrosis by reducing the expression of fibrotic markers (Col-1, α-SMA, and vimentin), as well as decreasing the apoptosis and inflammation of PTECs during fibrotic progression both in vivo and in vitro. Additionally, we found that the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/glycogen synthase kinase 3 beta (GSK3ß) signaling pathway, a pivotal player in renal fibrosis, was down-regulated following TRPC6 deletion. CONCLUSION: These results suggest that the ablation of TRPC6 may mitigate renal fibrosis by inhibiting the apoptosis and inflammation of PTECs through down-regulation of the PI3K/AKT/GSK3ß pathway. Targeting TRPC6 could be a novel therapeutic strategy for preventing chronic kidney disease.


Sujet(s)
Fibrose , Glycogen synthase kinase 3 beta , Souris knockout , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Membre-6 de la sous-famille C de canaux cationiques à potentiel de récepteur transitoire , Animaux , Membre-6 de la sous-famille C de canaux cationiques à potentiel de récepteur transitoire/génétique , Membre-6 de la sous-famille C de canaux cationiques à potentiel de récepteur transitoire/métabolisme , Glycogen synthase kinase 3 beta/métabolisme , Glycogen synthase kinase 3 beta/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Souris , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Lésion d'ischémie-reperfusion/génétique , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/anatomopathologie , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Mâle , Rein/anatomopathologie , Rein/métabolisme , Maladies du rein/métabolisme , Maladies du rein/génétique , Maladies du rein/anatomopathologie , Maladies du rein/étiologie , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Apoptose
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167232, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38759814

RÉSUMÉ

Focal malformations of cortical development (FMCDs) are brain disorders mainly caused by hyperactive mTOR signaling due to both inactivating and activating mutations of genes in the PI3K-AKT-mTOR pathway. Among them, mosaic and somatic activating mutations of the mTOR pathway activators are more frequently linked to severe form of FMCDs. A human stem cell-based FMCDs model to study these activating mutations is still lacking. Herein, we genetically engineer human embryonic stem cell lines carrying these activating mutations to generate cortical organoids. Mosaic and somatic expression of AKT3 activating mutations in cortical organoids mimicking the disease presentation with overproliferation and the formation of dysmorphic neurons. In parallel comparison of various AKT3 activating mutations reveals that stronger mutation is associated with more severe neuronal migratory and overgrowth defects. Together, we have established a feasible human stem cell-based model for FMCDs that could help to better understand pathogenic mechanism and develop novel therapeutic strategy.


Sujet(s)
Malformations corticales , Organoïdes , Protéines proto-oncogènes c-akt , Humains , Organoïdes/métabolisme , Organoïdes/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Malformations corticales/génétique , Malformations corticales/anatomopathologie , Malformations corticales/métabolisme , Cellules souches embryonnaires humaines/métabolisme , Transduction du signal/génétique , Cortex cérébral/anatomopathologie , Cortex cérébral/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Mutation , Neurones/métabolisme , Neurones/anatomopathologie , Lignée cellulaire
16.
Thorac Cancer ; 15(18): 1397-1409, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38717936

RÉSUMÉ

BACKGROUND: Alterations in epigenetic factors are recognized as key contributors to the emergence of human cancer. The active and reversible alteration of N6-methyladenosine (m6A) RNA is crucial for controlling gene activity and determining cellular destiny. Even with these insights, the triggering of KIAA1429 (also called VIRMA) and its role in lung adenocarcinoma (LUAD) is mostly unclear. As a result, the objective of this study was to elucidate how KIAA1429 contributes to cancer development in LUAD. METHODS: This study utilized multiple methods for investigation, encompassing the in vitro functional examination of KIAA1429 in lung adenocarcinoma cells, transcriptome sequencing, methylation RNA immunoprecipitation sequencing (MeRIP-seq), as well as RNA stability tests to ascertain the half-life and stability of the target genes. RESULTS: The results indicated that modifying the expression of KIAA1429 regulated the proliferation and metastasis of LUAD. By employing transcriptome sequencing alongside MeRIP-seq analysis, the research pinpointed genes affected by m6A alterations triggered by KIAA1429. In a more detailed manner, it was discovered that KIAA1429 plays a regulatory role in the expression of ARHGAP30. Suppressing KIAA1429 results in reduced m6A levels in the mRNA of the target gene ARHGAP30, boosting its stability and expression, thus inhibiting tumor proliferation and metastasis. CONCLUSION: This study revealed the activation mechanism and pivotal function of KIAA1429 in LUAD tumor development, paving the way for molecular-based interventions for LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Prolifération cellulaire , Protéines d'activation de la GTPase , Tumeurs du poumon , Protéines proto-oncogènes c-akt , Humains , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Protéines d'activation de la GTPase/métabolisme , Protéines d'activation de la GTPase/génétique , Souris , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Animaux , Régulation de l'expression des gènes tumoraux , Métastase tumorale , Hyaluronoglucosaminidase/génétique , Hyaluronoglucosaminidase/métabolisme , Transduction du signal , Protéines de liaison à l'ARN
17.
Biomed Environ Sci ; 37(4): 354-366, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38727158

RÉSUMÉ

Objective: This study investigated the impact of occupational mercury (Hg) exposure on human gene transcription and expression, and its potential biological mechanisms. Methods: Differentially expressed genes related to Hg exposure were identified and validated using gene expression microarray analysis and extended validation. Hg-exposed cell models and PTEN low-expression models were established in vitro using 293T cells. PTEN gene expression was assessed using qRT-PCR, and Western blotting was used to measure PTEN, AKT, and PI3K protein levels. IL-6 expression was determined by ELISA. Results: Combined findings from gene expression microarray analysis, bioinformatics, and population expansion validation indicated significant downregulation of the PTEN gene in the high-concentration Hg exposure group. In the Hg-exposed cell model (25 and 10 µmol/L), a significant decrease in PTEN expression was observed, accompanied by a significant increase in PI3K, AKT, and IL-6 expression. Similarly, a low-expression cell model demonstrated that PTEN gene knockdown led to a significant decrease in PTEN protein expression and a substantial increase in PI3K, AKT, and IL-6 levels. Conclusion: This is the first study to report that Hg exposure downregulates the PTEN gene, activates the PI3K/AKT regulatory pathway, and increases the expression of inflammatory factors, ultimately resulting in kidney inflammation.


Sujet(s)
Inflammation , Mercure , Phosphohydrolase PTEN , Humains , Régulation négative , Cellules HEK293 , Inflammation/induit chimiquement , Inflammation/métabolisme , Interleukine-6/génétique , Interleukine-6/métabolisme , Interleukine-6/sang , Mercure/toxicité , Exposition professionnelle/effets indésirables , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Phosphohydrolase PTEN/génétique , Phosphohydrolase PTEN/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
18.
Biochem Pharmacol ; 225: 116256, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38729448

RÉSUMÉ

Endocrine treatment, particularly tamoxifen, has shown significant improvement in the prognosis of patients with estrogen receptor-positive (ER-positive) breast cancer. However, the clinical utility of this treatment is often hindered by the development of endocrine resistance. Therefore, a comprehensive understanding of the underlying mechanisms driving ER-positive breast cancer carcinogenesis and endocrine resistance is crucial to overcome this clinical challenge. In this study, we investigated the expression of MICAL-L2 in ER-positive breast cancer and its impact on patient prognosis. We observed a significant upregulation of MICAL-L2 expression in ER-positive breast cancer, which correlated with a poorer prognosis in these patients. Furthermore, we found that estrogen-ERß signaling promoted the expression of MICAL-L2. Functionally, our study demonstrated that MICAL-L2 not only played an oncogenic role in ER-positive breast cancer tumorigenesis but also influenced the sensitivity of ER-positive breast cancer cells to tamoxifen. Mechanistically, as an estrogen-responsive gene, MICAL-L2 facilitated the activation of the AKT/mTOR signaling pathway in ER-positive breast cancer cells. Collectively, our findings suggest that MICAL-L2 could serve as a potential prognostic marker for ER-positive breast cancer and represent a promising molecular target for improving endocrine treatment and developing therapeutic approaches for this subtype of breast cancer.


Sujet(s)
Antinéoplasiques hormonaux , Tumeurs du sein , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Tamoxifène , Tamoxifène/pharmacologie , Tamoxifène/usage thérapeutique , Humains , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Femelle , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Antinéoplasiques hormonaux/pharmacologie , Antinéoplasiques hormonaux/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/physiologie , Animaux , Oestrogènes/pharmacologie , Oestrogènes/métabolisme , Souris nude , Souris , Récepteurs des oestrogènes/métabolisme , Récepteurs des oestrogènes/génétique , Cellules MCF-7 , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/physiologie , Évolution de la maladie , Lignée cellulaire tumorale , Souris de lignée BALB C
19.
PLoS One ; 19(5): e0289187, 2024.
Article de Anglais | MEDLINE | ID: mdl-38809881

RÉSUMÉ

Recently, a low-level somatic mutation in the NRAS gene (c.182 A > G, Q61R) was identified in various specimens from patients with kaposiform lymphangiomatosis. However, it is unknown how these low-frequency mutated cells can affect the characterization and surrounding environment of their lesions. To understand the pathogenesis and association of these gene abnormalities, we established NRASQ61R mutated lymphatic endothelial cells transfected with lentivirus vector and undertook morphological and functional characterization, protein expression profiling, and metabolome analysis. NRASQ61R human dermal lymphatic endothelial cells showed poor tube formation, a low proliferation rate, and high migration ability, with an increase in the ratio of mutated cells. An analysis of signaling pathways showed inactivation of the PIK3/AKT/mTOR pathway and hyperactivation of the RAS/MAPK/ERK pathway, which was improved by MAPK kinase (MEK) inhibitor treatment. This study shows the theoretical circumstances induced in vitro by NRASQ61R-mutated cells in the affected lesions of kaposiform lymphangiomatosis patients.


Sujet(s)
Cellules endothéliales , dGTPases , Protéines membranaires , Mutation , Humains , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , dGTPases/génétique , dGTPases/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Prolifération cellulaire/génétique , Transduction du signal/génétique , Mouvement cellulaire/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique
20.
Cancer Genet ; 284-285: 48-57, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38729078

RÉSUMÉ

Although lncRNAs are recognized to contribute to the development of oral squamous-cell carcinoma (OSCC), their exact function in invasion and cell migration is not clear. In this research, we explored the molecular and cellular mechanisms of FOXD2-AS1 in OSCC. Prognostic and bioinformatics analyses were used to test for the differential expression of FOXD2-AS1-PLOD1. Following FOXD2-AS1 suppression or overexpression, changes in cell viability were measured using the CCK-8 test; changes in cell migration and invasion abilities were measured using the migration and the Transwell assay. The expression of associated genes and proteins was found using Western blot and RT-qPCR. Analysis of luciferase reporter genes was done to look for regulatory connections between various molecules. The FOXD2-AS1-PLOD1 pair, which was highly expressed in OSCC, was analyzed and experimentally verified to be closely related to the prognosis of OSCC, and a nomogram model and correction curve were constructed. The inhibition of FOXD2-AS1 resulted in the reduction of cell activity, migration, invasion ability and changes in genes related to invasion and migration. In vivo validation showed that inhibition of FOXD2-AS1 expression slowed tumor growth, and related proteins changed accordingly. The experiments verified that FOXD2-AS1 negatively regulated miR-185-5 p and that miR-185-5 p negatively regulated PLOD1. In addition, it was found that the expression of PLOD1, p-Akt and p-mTOR proteins in OSCC cells was reduced by the inhibition of FOXD2-AS1, and FOXD2-AS1 and PLOD1 were closely related to the Akt/mTOR pathway. Increased expression of FOXD2-AS1 promotes OSCC growth, invasion and migration, which is important in part by targeting miR-185-5 p/PLOD1/Akt/mTOR pathway activity.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , microARN , Tumeurs de la bouche , Invasion tumorale , Protéines proto-oncogènes c-akt , ARN long non codant , Sérine-thréonine kinases TOR , Humains , microARN/génétique , ARN long non codant/génétique , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Mouvement cellulaire/génétique , Tumeurs de la bouche/génétique , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/métabolisme , Prolifération cellulaire/génétique , Souris , Animaux , Procollagen-lysine, 2-oxoglutarate 5-dioxygenase/génétique , Procollagen-lysine, 2-oxoglutarate 5-dioxygenase/métabolisme , Lignée cellulaire tumorale , Transduction du signal/génétique , Régulation de l'expression des gènes tumoraux , Femelle , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/métabolisme , Mâle , Pronostic , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Souris nude
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...