Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.107
Filtrer
1.
Int J Med Sci ; 21(11): 2040-2051, 2024.
Article de Anglais | MEDLINE | ID: mdl-39239540

RÉSUMÉ

Myofibrillar myopathy (MFM) is a group of hereditary myopathies that mainly involves striated muscles. This study aimed to use tandem mass tag (TMT)-based proteomics to investigate the underlying pathomechanisms of two of the most common MFM subtypes, desminopathy and titinopathy. Muscles from 7 patients with desminopathy, 5 with titinopathy and 5 control individuals were included. Samples were labelled with TMT and then underwent high-resolution liquid chromatography-mass spectrometry analysis. Compared with control samples, there were 436 differentially abundant proteins (DAPs) in the desminopathy group and 269 in the titinopathy group. When comparing the desminopathy with the titinopathy group, there were 113 DAPs. In desminopathy, mitochondrial ATP production, muscle contraction, and cytoskeleton organization were significantly suppressed. Activated cellular components and pathways were mostly related to extracellular matrix (ECM). In titinopathy, mitochondrial-related pathways and the cellular component ECM were downregulated, while gluconeogenesis was activated. Direct comparison between desminopathy and titinopathy revealed hub genes that were all involved in glycolytic process. The disparity in glycolysis in the two MFM subtypes is likely due to fiber type switching. This study has revealed disorganization of cytoskeleton and mitochondrial dysfunction as the common pathophysiological processes in MFM, and glycolysis and ECM as the differential pathomechanism between desminopathy and titinopathy. This offers a future direction for targeted therapy for MFM.


Sujet(s)
Connectine , Humains , Mâle , Femelle , Adulte , Adulte d'âge moyen , Connectine/génétique , Connectine/métabolisme , Protéomique/méthodes , Myopathies congénitales structurales/génétique , Myopathies congénitales structurales/anatomopathologie , Myopathies congénitales structurales/métabolisme , Muscles squelettiques/anatomopathologie , Muscles squelettiques/métabolisme , Desmine/génétique , Desmine/métabolisme , Glycolyse/génétique , Mitochondries/métabolisme , Mitochondries/génétique , Mitochondries/anatomopathologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/anatomopathologie , Dystrophies musculaires , Cardiomyopathies
2.
Cell Mol Life Sci ; 81(1): 391, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39254854

RÉSUMÉ

Human spermatogonial stem cells (SSCs) have significant applications in reproductive medicine and regenerative medicine because of their great plasticity. Nevertheless, it remains unknown about the functions and mechanisms of long non-coding RNA (LncRNA) in regulating the fate determinations of human SSCs. Here we have demonstrated that LncRNA ACVR2B-as1 (activin A receptor type 2B antisense RNA 1) controls the self-renewal and apoptosis of human SSCs by interaction with ALDOA via glycolysis activity. LncRNA ACVR2B-as1 is highly expressed in human SSCs. LncRNA ACVR2B-as1 silencing suppresses the proliferation and DNA synthesis and enhances the apoptosis of human SSCs. Mechanistically, our ChIRP-MS and RIP assays revealed that ACVR2B-as1 interacted with ALDOA in human SSCs. High expression of ACVR2B-as1 enhanced the proliferation, DNA synthesis, and glycolysis of human SSCs but inhibited their apoptosis through up-regulation of ALDOA. Importantly, overexpression of ALDOA counteracted the effect of ACVR2B-as1 knockdown on the aforementioned biological processes. Collectively, these results indicate that ACVR2B-as1 interacts with ALDOA to control the self-renewal and apoptosis of human SSCs by enhancing glycolysis activity. This study is of great significance because it sheds a novel insight into molecular mechanisms underlying the fate decisions of human SSCs and it may offer innovative approaches to address the etiology of male infertility.


Sujet(s)
Apoptose , Prolifération cellulaire , Glycolyse , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Apoptose/génétique , Glycolyse/génétique , Mâle , Prolifération cellulaire/génétique , Récepteur activine, type 2/métabolisme , Récepteur activine, type 2/génétique , Spermatogonies/métabolisme , Spermatogonies/cytologie , Cellules souches germinales adultes/métabolisme , Auto-renouvellement cellulaire/génétique , Cellules cultivées
3.
J Cell Mol Med ; 28(17): e70053, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39224032

RÉSUMÉ

MitoAMPK was proved to inhibit the Warburg effect, but the specific mechanisms on non-small-cell lung cancer remain unclear. Here, we selected SIRT6 and MZF1 to clarify the mechanism. By western blotting, quantitative polymerase chain reaction, the CCK-8 assay, and immunohistochemistry assays, we found SIRT6 expression was lower in NSCLC tissues and cell lines than normal tissues and cells. Moreover, SIRT6 could inhibit the Warburg effect by regulating glycolysis-related genes of SLC2A2, SLC2A4 and PKM2. Finally, we demonstrated the interaction between SIRT6 and MZF1 using ChIP-qPCR. In conclusion, mitoAMPK inhibits the Warburg effect by regulating the expression of the MZF1-SIRT6 complex.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Régulation de l'expression des gènes tumoraux , Facteurs de transcription Krüppel-like , Tumeurs du poumon , Sirtuines , Effet Warburg en oncologie , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Sirtuines/métabolisme , Sirtuines/génétique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Lignée cellulaire tumorale , Glycolyse/génétique , Femelle , Mâle
4.
Cell Death Dis ; 15(9): 645, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-39227375

RÉSUMÉ

lncRNA can regulate tumorigenesis development and distant metastasis of colorectal cancer (CRC). However, the detailed molecular mechanisms are still largely unknown. Using RNA-sequencing data, RT-qPCR, and FISH assay, we found that HIF1A-AS2 was upregulated in CRC tissues and associated with poor prognosis. Functional experiments were performed to determine the roles of HIF1A-AS2 in tumor progression and we found that HIF1A-AS2 can promote the proliferation, metastasis, and aerobic glycolysis of CRC cells. Mechanistically, HIF1A-AS2 can promote FOXC1 expression by sponging miR-141-3p. SP1 can transcriptionally activate HIF1A-AS2. Further, HIF1A-AS2 can be packaged into exosomes and promote the malignant phenotype of recipient tumor cells. Taken together, we discovered that SP1-induced HIF1A-AS2 can promote the metabolic reprogramming and progression of CRC via miR-141-3p/FOXC1 axis. HIF1A-AS2 is a promising diagnostic marker and treatment target in CRC.


Sujet(s)
Tumeurs colorectales , Évolution de la maladie , Facteurs de transcription Forkhead , Régulation de l'expression des gènes tumoraux , microARN , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , microARN/métabolisme , microARN/génétique , Animaux , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Lignée cellulaire tumorale , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris , Souris nude , Prolifération cellulaire/génétique , Facteur de transcription Sp1/métabolisme , Facteur de transcription Sp1/génétique , Glycolyse/génétique , Souris de lignée BALB C , Mâle , Femelle , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Mouvement cellulaire/génétique ,
5.
Sci Rep ; 14(1): 20794, 2024 09 05.
Article de Anglais | MEDLINE | ID: mdl-39242716

RÉSUMÉ

Cytokine-induced apoptosis inhibitor 1 (CIAPIN1) is a protein that regulates apoptosis and programmed cell death. This research aims to evaluate its potential role in inhibiting breast cancer cell proliferation, migration, and glycolysis and uncover its underlying molecular mechanism. We collected breast cancer tissue samples from eight patients between January 2019 and June 2023 in our Hospital to analyse CIAPIN1 expression. We transfected human breast cancer cell lines (MCF7, MDA-MB-231, MDA-MB-453, and MDA-MB-468) with siRNA of CIAPIN1. Finally, we determined protein expression using RT-qPCR and Western blotting. CIAPIN1 expression was elevated in both breast cancer tissue and serum. Overexpression of CIAPIN1 detected in the breast cancer cell lines MCF7 and MDA-MB-468. In addition, CIAPIN1 overexpression increased cell proliferation and migration rate. CIAPIN1 downregulation suppressed cell proliferation while elevated cellular apoptosis, reactive oxygen species (ROS) production and oxidative stress in breast cancer cells. Moreover, CIAPIN1 inhibition remarkably suppressed pyruvate, lactate and adenosine triphosphate (ATP) production and reduced the pyruvate kinase M2 (PKM2) protein expression and phosphorylation of signal transducer and activator of transcription 3 (STAT3) in breast cancer cells. Downregulation of CIAPIN1 suppresses cell proliferation, migration and glycolysis capacity in breast cancer cells by inhibiting the STAT3/PKM2 pathway.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Prolifération cellulaire , Régulation négative , Glycolyse , Protéines et peptides de signalisation intracellulaire , Facteur de transcription STAT-3 , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Glycolyse/génétique , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Mouvement cellulaire/génétique , Femelle , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Transduction du signal , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Apoptose/génétique , Cellules MCF-7 , Espèces réactives de l'oxygène/métabolisme
6.
J Ovarian Res ; 17(1): 185, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39272131

RÉSUMÉ

BACKGROUND: In-depth understanding of dynamic expression profiles of human granulosa cells (GCs) during follicular development will contribute to the diagnostic and targeted interventions for female infertility. However, genome-scale analysis of long non-coding ribonucleic acid (lncRNA) in GCs across diverse developmental stages is challenging. Meanwhile, further research is needed to determine how aberrant lncRNA expression participates in ovarian diseases. METHODS: Granulosa cell-related lncRNAs data spanning five follicular development stages were retrieved and filtered from the NCBI dataset (GSE107746). Stage-specific lncRNA expression patterns and mRNA-lncRNA co-expression networks were identified with bioinformatic approaches. Subsequently, the expression pattern of SNHG18 was detected in GCs during ovarian aging. And SNHG18 siRNA or overexpression plasmids were transfected to SVOG cells in examining the regulatory roles of SNHG18 in GC proliferation and apoptosis. Moreover, whether PKCɛ/SNHG18 signaling take part in GC glycolysis via ENO1 were verified in SVOG cells. RESULTS: We demonstrated that GC-related lncRNAs were specifically expressed across different developmental stages, and coordinated crucial biological functions like mitotic cell cycle and metabolic processes in the folliculogenesis. Thereafter, we noticed a strong correlation of PRKCE and SNHG18 expression in our analysis. With downregulated SNHG18 of GCs identified in the context of ovarian aging, SNHG18 knockdown could further induce cell apoptosis, retard cell proliferation and exacerbate DNA damage in SVOG cell. Moreover, downregulated PKCɛ/SNHG18 pathway interrupted the SVOG cell glycolysis by lowering the ENO1 expression. CONCLUSIONS: Altogether, our results revealed that folliculogenesis-related lncRNA SNHG18 participated in the pathogenesis of ovarian aging, which may provide novel biomarkers for ovarian function and new insights for the infertility treatment.


Sujet(s)
Apoptose , Glycolyse , Cellules de la granulosa , ARN long non codant , Femelle , Humains , Vieillissement/génétique , Vieillissement/métabolisme , Apoptose/génétique , Glycolyse/génétique , Cellules de la granulosa/métabolisme , Ovaire/métabolisme , Ovaire/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme
7.
Cell Biol Toxicol ; 40(1): 78, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39289194

RÉSUMÉ

The N7-methylguanosine (m7G) modification and circular RNAs (circRNAs) have been shown to play important roles in the development of lung cancer. However, the m7G modification of circRNAs has not been fully elucidated. This study revealed the presence of the m7G modification in circFAM126A. We propose the novel hypothesis that the methyltransferase TRMT10C mediates the m7G modification of circFAM126A and that the stability of m7G-modified circFAM126A is reduced. circFAM126A is downregulated in lung cancer and significantly inhibits lung cancer growth both in vitro and in vivo. The expression of circFAM126A correlates with the stage of lung cancer and with the tumour diameter, and circFAM126A can be used as a potential molecular target for lung cancer. The molecular mechanism by which circFAM126A increases HSP90 ubiquitination and suppresses AKT1 expression to regulate cellular glycolysis, ultimately inhibiting the progression of lung cancer, is elucidated. This study not only broadens the knowledge regarding the expression and regulatory mode of circRNAs but also provides new insights into the molecular mechanisms that regulate tumour cell metabolism and affect tumour cell fate from an epigenetic perspective. These findings will facilitate the development of new strategies for lung cancer prevention and treatment.


Sujet(s)
Prolifération cellulaire , Glycolyse , Tumeurs du poumon , Methyltransferases , ARN circulaire , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Glycolyse/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Animaux , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Souris nude , Souris , Protéines proto-oncogènes c-akt/métabolisme , Protéines du choc thermique HSP90/métabolisme , Protéines du choc thermique HSP90/génétique , Cellules A549 , Guanosine/analogues et dérivés , Guanosine/métabolisme , Mâle , Femelle , Souris de lignée BALB C , Ubiquitination
8.
Biol Direct ; 19(1): 83, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39294751

RÉSUMÉ

BACKGROUND: Breast cancer (BC) is a great clinical challenge because of its aggressiveness and poor prognosis. Zinc Finger Protein 64 (ZFP64), as a transcriptional factor, is responsible for the development and progression of cancers. This study aims to investigate whether ZFP64 regulates stem cell-like properties and tumorigenesis in BC by the glycolytic pathway. RESULTS: It was demonstrated that ZFP64 was overexpressed in BC specimens compared to adjacent normal tissues, and patients with high ZFP64 expression had shorter overall survival and disease-free survival. The analysis of the association of ZFP64 expression with clinicopathological characteristics showed that high ZFP64 expression is closely associated with N stage, TNM stage, and progesterone receptor status. Knockdown of ZFP64 suppressed the viability and colony formation capacity of BC cells by CCK8 and colony formation assays. The subcutaneous xenograft models revealed that ZFP64 knockdown reduced the volume of formatted tumors, and decreased Ki67 expression in tumors. The opposite effects on cell proliferation and tumorigenesis were demonstrated by ZFP64 overexpression. Furthermore, we suggested that the stem cell-like properties of BC cells were inhibited by ZFP64 depletion, as evidenced by the decreased size and number of formatted mammospheres, the downregulated expressions of OCT4, Nanog, and SOX2 proteins, as well as the reduced proportion of CD44+/CD24- subpopulations. Mechanistically, glycolysis was revealed to mediate the effect of ZFP64 using mRNA-seq analysis. Results showed that ZFP64 knockdown blocked the glycolytic process, as indicated by decreasing glycolytic metabolites, inhibiting glucose consumption, and reducing lactate and ATP production. As a transcription factor, we identified that ZFP64 was directly bound to the promoters of glycolysis-related genes (ALDOC, ENO2, HK2, and SPAG4), and induced the transcription of these genes by ChIP and dual-luciferase reporter assays. Blocking the glycolytic pathway by the inhibition of glycolytic enzymes ENO2/HK2 suppressed the high proliferation and stem cell-like properties of BC cells induced by ZFP64 overexpression. CONCLUSIONS: These data support that ZFP64 promotes stem cell-like properties and tumorigenesis of BC by activating glycolysis in a transcriptional mechanism.


Sujet(s)
Tumeurs du sein , Carcinogenèse , Glycolyse , Cellules souches tumorales , Humains , Glycolyse/génétique , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Cellules souches tumorales/métabolisme , Carcinogenèse/génétique , Animaux , Souris , Lignée cellulaire tumorale , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Prolifération cellulaire , Adulte d'âge moyen
9.
Biotechnol J ; 19(9): e2400163, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39295558

RÉSUMÉ

The 3D multicellular tumor spheroid (MTS) model exhibits enhanced fidelity in replicating the tumor microenvironment and demonstrates exceptional resistance to clinical drugs compared to the 2D monolayer model. In this study, we used multiomics (transcriptome, proteomics, and metabolomics) tools to explore the molecular mechanisms and metabolic differences of the two culture models. Analysis of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathways revealed that the differentially expressed genes between the two culture models were mainly enriched in cellular components and biological processes associated with extracellular matrix, extracellular structural organization, and mitochondrial function. An integrated analysis of three omics data revealed 11 possible drug resistance targets. Among these targets, seven genes, AKR1B1, ALDOC, GFPT2, GYS1, LAMB2, PFKFB4, and SLC2A1, exhibited significant upregulation. Conversely, four genes, COA7, DLD, IFNGR1, and QRSL1, were significantly downregulated. Clinical prognostic analysis using the TCGA survival database indicated that high-expression groups of SLC2A1, ALDOC, and PFKFB4 exhibited a significant negative correlation with patient survival. We further validated their involvement in chemotherapy drug resistance, indicating their potential significance in improving prognosis and chemotherapy outcomes. These results provide valuable insights into potential therapeutic targets that can potentially enhance treatment efficacy and patient outcomes.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Transporteur de glucose de type 1 , Glycolyse , Phosphofructokinase-2 , Sphéroïdes de cellules , Humains , Résistance aux médicaments antinéoplasiques/génétique , Phosphofructokinase-2/génétique , Phosphofructokinase-2/métabolisme , Sphéroïdes de cellules/métabolisme , Sphéroïdes de cellules/anatomopathologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Glycolyse/génétique , Glycolyse/effets des médicaments et des substances chimiques , Cellules HeLa , Transporteur de glucose de type 1/génétique , Transporteur de glucose de type 1/métabolisme , Régulation de l'expression des gènes tumoraux , Antinéoplasiques/pharmacologie
10.
Exp Cell Res ; 442(1): 114228, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39197578

RÉSUMÉ

Anterior gradient-2 (AGR2) is highly expressed in several tumors and plays an important role in tumor development. However, the biological function of AGR2 in teratomas has not yet been thoroughly studied. In this study, AGR2 was found to be upregulated in teratoma tissues and in human testicular teratoma cell lines by Western blotting and qRT-PCR assays. A DNA Methylation-Specific PCR assay demonstrated that AGR2 upregulation resulted from hypomethylated AGR2 in teratoma cells. NCC-IT and NT2-D1 cells were transfected with pcDNA-AGR2 or sh-AGR2 to obtain AGR2-overexpressed or -silenced cells, and cell proliferation, invasion and glycolysis were determined using CCK-8, 5-ethynyl-2'-deoxyuridine (EdU), Transwell assays, and commercial kits. The results revealed that overexpression of AGR2 promoted teratoma cell proliferation and invasion and elevated glycolysis levels evidencing by the increase in lactate secretion, glucose consumption, ATP levels and the expression of glycolysis-related proteins, while knockdown of AGR2 showed the opposite results. The interactions between AGR2 and annexin A2 (AnXA2), as well as between AnXA2 and epidermal growth factor receptor (EGFR) were verified by co-immunoprecipitation assay. Mechanistic studies revealed that AGR2 interacts with AnXA2 and increases the level of AnXA2 to recruit more AnXA2 to EGFR, there by promoting EGFR expression. A series of rescue experiments showed that knockdown of AnXA2 or EGFR weakened the promotional effects of AGR2 overexpression on the proliferation, invasion, and glycolysis of teratoma cells. Finally, tumorigenicity assays were performed using NT2-D1 cells stably transfected with either LV-NC-shRNA or LV-shAGR2. The results showed that AGR2 knockdown significantly inhibited teratoma tumor growth in vivo. In conclusion, our data suggested that AGR2 facilitates glycolysis in teratomas through promoting EGFR expression by interacting with AnXA2, thereby promoting teratoma cells proliferation and invasion.


Sujet(s)
Annexine A2 , Prolifération cellulaire , Récepteurs ErbB , Glycolyse , Mucoprotéines , Protéines oncogènes , Tumeurs du testicule , Humains , Mucoprotéines/génétique , Mucoprotéines/métabolisme , Glycolyse/génétique , Protéines oncogènes/métabolisme , Protéines oncogènes/génétique , Animaux , Prolifération cellulaire/génétique , Mâle , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Souris , Annexine A2/métabolisme , Annexine A2/génétique , Tumeurs du testicule/anatomopathologie , Tumeurs du testicule/génétique , Tumeurs du testicule/métabolisme , Lignée cellulaire tumorale , Souris nude , Régulation de l'expression des gènes tumoraux , Transduction du signal , Protéines/métabolisme , Protéines/génétique , Mouvement cellulaire/génétique , Souris de lignée BALB C , Invasion tumorale
11.
Signal Transduct Target Ther ; 9(1): 216, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39143065

RÉSUMÉ

Third-generation EGFR tyrosine kinase inhibitors (TKIs), exemplified by osimertinib, have demonstrated promising clinical efficacy in the treatment of non-small cell lung cancer (NSCLC). Our previous work has identified ASK120067 as a novel third-generation EGFR TKI with remarkable antitumor effects that has undergone New Drug Application (NDA) submission in China. Despite substantial progress, acquired resistance to EGFR-TKIs remains a significant challenge, impeding the long-term effectiveness of therapeutic approaches. In this study, we conducted a comprehensive investigation utilizing high-throughput proteomics analysis on established TKI-resistant tumor models, and found a notable upregulation of branched-chain amino acid transaminase 1 (BCAT1) expression in both osimertinib- and ASK120067-resistant tumors compared with the parental TKI-sensitive NSCLC tumors. Genetic depletion or pharmacological inhibition of BCAT1 impaired the growth of resistant cells and partially re-sensitized tumor cells to EGFR TKIs. Mechanistically, upregulated BCAT1 in resistant cells reprogrammed branched-chain amino acid (BCAA) metabolism and promoted alpha ketoglutarate (α-KG)-dependent demethylation of lysine 27 on histone H3 (H3K27) and subsequent transcriptional derepression of glycolysis-related genes, thereby enhancing glycolysis and promoting tumor progression. Moreover, we identified WQQ-345 as a novel BCAT1 inhibitor exhibiting antitumor activity both in vitro and in vivo against TKI-resistant lung cancer with high BCAT1 expression. In summary, our study highlighted the crucial role of BCAT1 in mediating resistance to third-generation EGFR-TKIs through epigenetic activation of glycolysis in NSCLC, thereby supporting BCAT1 as a promising therapeutic target for the treatment of TKI-resistant NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Résistance aux médicaments antinéoplasiques , Épigenèse génétique , Récepteurs ErbB , Glycolyse , Tumeurs du poumon , Inhibiteurs de protéines kinases , Transaminases , Humains , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Transaminases/génétique , Transaminases/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Glycolyse/effets des médicaments et des substances chimiques , Glycolyse/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Épigenèse génétique/effets des médicaments et des substances chimiques , Épigenèse génétique/génétique , Souris , Tumeurs du poumon/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Acrylamides/pharmacologie , Animaux , Dérivés de l'aniline/pharmacologie , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Indoles , Pyrimidines
12.
Front Biosci (Landmark Ed) ; 29(8): 308, 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39206892

RÉSUMÉ

BACKGROUND: Aerobic glycolysis and the cell cycle are well-established tumor hallmarks. Understanding their relationship could help to unravel the pathogenic mechanisms of breast cancer (BC) and suggest potential new strategies for treatment. METHODS: Glycolysis-related genes (GRGs) were downloaded from the Reactome database and screened using univariate Cox analysis. The consensus clustering method was employed to identify a glycolytic activity signature (GAS) using the Gene Expression Omnibus (GEO) dataset. A nomogram risk prediction model was constructed using coefficients from univariate Cox analysis. Immune cell infiltration was evaluated using single-sample gene set enrichment analysis (ssGSEA) and the ESTIMATE algorithm. Gene co-expression modules were created using weighted correlation network analysis (WGCNA) to identify hub genes. Gene expression in three BC cell lines was quantified using Quantitative Reverse Transcriptase Polymera (qRT-PCR). Single-cell RNA sequencing (scRNA-seq) data was used to examine the relationship between GAS and hub genes. The sensitivity of different groups to cell cycle-related clinical drugs was also examined. RESULTS: BC with high GAS (HGAS) showed high tumor grade and recurrence rate. HGAS was a prognostic indicator of worse overall survival (OS) in BC patients. HGAS BC showed more abundant immune cells and significantly higher expression of immunomodulators compared to BC with low GAS (LGAS). HGAS BC also showed enhanced cell cycle pathway, with high mRNA and protein expression levels of Cyclin B2 (CCNB2), a key component of the cell cycle pathway. Importantly, scRNA-seq analysis revealed that elevated CCNB2 expression was positively correlated with HGAS in triple-negative BC (TNBC). This was validated in clinical samples from TNBC patients. High expression of CCNB2 was found in three BC cell lines, and was also an indicator of poor prognosis. HGAS BC showed high sensitivity to several cell cycle-related clinical drugs, with 9 of these also showing activity in BC with high CCNB2 expression. CONCLUSIONS: HGAS was associated with enhanced cell cycle pathway and immune activity in BC. These results suggest that CCNB2 is a potential key therapeutic target in BC patients.


Sujet(s)
Cycline B2 , Régulation de l'expression des gènes tumoraux , Glycolyse , Tumeurs du sein triple-négatives , Humains , Glycolyse/génétique , Femelle , Cycline B2/génétique , Cycline B2/métabolisme , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Lignée cellulaire tumorale , Pronostic , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Cycle cellulaire/génétique , Analyse de profil d'expression de gènes/méthodes , Nomogrammes
13.
Int J Mol Sci ; 25(16)2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39201646

RÉSUMÉ

Accumulating evidence has indicated that stemness-related genes are associated with the aggressiveness of triple-negative breast cancer (TNBC). Because no universal markers for breast CSCs are available, we applied the density gradient centrifugation method to enrich breast CSCs. We demonstrated that the density centrifugation method allows for the isolation of cancer stem cells (CSCs) from adherent and non-adherent MCF7 (Luminal A), MDA-MB-231 (TNBC) and MDA-MB-468 (TNBC) breast cancer cells. The current study shows that the CSCs' enriched fraction from Luminal A and TNBC cells have an increased capacity to grow anchorage-independently. CSCs from adherent TNBC are mainly characterized by metabolic plasticity, whereas CSCs from Luminal A have an increased mitochondrial capacity. Moreover, we found that non-adherent growth CSCs isolated from large mammospheres have a higher ability to grow anchorage-independently compared to CSCs isolated from small mammospheres. In CSCs, a metabolic shift towards glycolysis was observed due to the hypoxic environment of the large mammosphere. Using a bioinformatic analysis, we indicate that hypoxia HYOU1 gene overexpression is associated with the aggressiveness, metastasis and poor prognosis of TNBC. An in vitro study demonstrated that HYOU1 overexpression increases breast cancer cells' stemness and hyperactivates their metabolic activity. In conclusion, we show that density gradient centrifugation is a non-marker-based approach to isolate metabolically flexible (normoxia) CSCs and glycolytic (hypoxic) CSCs from aggressive TNBC.


Sujet(s)
Centrifugation en gradient de densité , Cellules souches tumorales , Tumeurs du sein triple-négatives , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Humains , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Centrifugation en gradient de densité/méthodes , Femelle , Lignée cellulaire tumorale , Séparation cellulaire/méthodes , Hypoxie cellulaire , Cellules MCF-7 , Glycolyse/génétique
14.
Genes (Basel) ; 15(8)2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39202395

RÉSUMÉ

The determination of sex in mammals is established and controlled by various complex mechanisms. In contrast, sex control in poultry remains an unresolved issue. In this study, RNA-sequencing was conducted for male gonads and ovarian tissues in chicken embryos of up to 18.5 days to identify metabolic factors influencing male and female sex differentiation, as well as gonadal development. Our results reveal that PKM2, a critical glycolysis-related protein, plays a significant role in chicken sex differentiation via PPARG, a crucial hormone gene. We propose that our discoveries bolster the notion that glycolysis and oxidative phosphorylation function as antecedent contributors to sexual phenotypic development and preservation.


Sujet(s)
Poulets , Métabolisme énergétique , Différenciation sexuelle , Transcriptome , Animaux , Différenciation sexuelle/génétique , Mâle , Métabolisme énergétique/génétique , Femelle , Poulets/génétique , Poulets/croissance et développement , Transcriptome/génétique , Embryon de poulet , Régulation de l'expression des gènes au cours du développement , Glycolyse/génétique , Phosphorylation oxydative , , Gonades/métabolisme , Gonades/croissance et développement
15.
Nat Commun ; 15(1): 6755, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39117659

RÉSUMÉ

Histone lysine methyltransferase 2D (KMT2D) is the most frequently mutated epigenetic modifier in head and neck squamous cell carcinoma (HNSCC). However, the role of KMT2D in HNSCC tumorigenesis and whether its mutations confer any therapeutic vulnerabilities remain unknown. Here we show that KMT2D deficiency promotes HNSCC growth through increasing glycolysis. Additionally, KMT2D loss decreases the expression of Fanconi Anemia (FA)/BRCA pathway genes under glycolytic inhibition. Mechanistically, glycolytic inhibition facilitates the occupancy of KMT2D to the promoter/enhancer regions of FA genes. KMT2D loss reprograms the epigenomic landscapes of FA genes by transiting their promoter/enhancer states from active to inactive under glycolytic inhibition. Therefore, combining the glycolysis inhibitor 2-DG with DNA crosslinking agents or poly (ADP-ribose) polymerase (PARP) inhibitors preferentially inhibits tumor growth of KMT2D-deficient mouse HNSCC and patient-derived xenografts (PDXs) harboring KMT2D-inactivating mutations. These findings provide an epigenomic basis for developing targeted therapies for HNSCC patients with KMT2D-inactivating mutations.


Sujet(s)
Glycolyse , Carcinome épidermoïde de la tête et du cou , Animaux , Humains , Souris , Glycolyse/génétique , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/métabolisme , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Protéine BRCA1/déficit , Protéine BRCA2/génétique , Protéine BRCA2/métabolisme , Protéine BRCA2/déficit , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/métabolisme , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/anatomopathologie , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Régulation de l'expression des gènes tumoraux , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Femelle , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Transduction du signal , Régions promotrices (génétique)/génétique , Protéine de la leucémie myéloïde-lymphoïde
16.
Aging (Albany NY) ; 16(16): 11955-11969, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39167430

RÉSUMÉ

BACKGROUND: Clear cell renal carcinoma is a common urological malignancy with poor prognosis and treatment outcomes. lncRNAs are important in metabolic reprogramming and the tumor immune microenvironment, but their role in clear cell renal carcinoma is unclear. METHODS: Renal clear cell carcinoma sample data from The Cancer Genome Atlas was used to establish a new risk profile by glycolysis-associated lncRNAs via machine learning. Risk profile-associated column-line plots were constructed to provide a quantitative tool for clinical practice. Patients with renal clear cell carcinoma were divided into high- and low-risk groups. Clinical features, tumor immune microenvironments, and immunotherapy responses were systematically analyzed. We experimentally confirmed the role of LINC01138 and LINC01605 in renal clear cell carcinoma. RESULTS: The risk profile, consisting of LUCAT1, LINC01138, LINC01605, and HOTAIR, reliably predicted survival in patients with renal clear cell carcinoma and was validated in multiple external datasets. The high-risk group presented higher levels of immune cell infiltration and better immunotherapy responses than the low-risk group. LINC01138 and LINC01605 knockdown inhibited the proliferation of renal clear cell carcinoma. CONCLUSIONS: The identified risk profiles can accurately assess the prognosis of patients with clear cell renal carcinoma and identify patient populations that would benefit from immunotherapy, providing valuable insights and therapeutic targets for the clinical management of clear cell renal carcinoma.


Sujet(s)
Néphrocarcinome , Glycolyse , Immunothérapie , Tumeurs du rein , ARN long non codant , Microenvironnement tumoral , ARN long non codant/génétique , ARN long non codant/métabolisme , Néphrocarcinome/génétique , Néphrocarcinome/immunologie , Néphrocarcinome/thérapie , Néphrocarcinome/anatomopathologie , Humains , Tumeurs du rein/immunologie , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/thérapie , Glycolyse/génétique , Immunothérapie/méthodes , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Pronostic , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Multi-omique
17.
Nat Commun ; 15(1): 6915, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39134530

RÉSUMÉ

Protein post-translational modifications (PTMs) are crucial for cancer cells to adapt to hypoxia; however, the functional significance of lysine crotonylation (Kcr) in hypoxia remains unclear. Herein we report a quantitative proteomics analysis of global crotonylome under normoxia and hypoxia, and demonstrate 128 Kcr site alterations across 101 proteins in MDA-MB231 cells. Specifically, we observe a significant decrease in K131cr, K156cr and K220cr of phosphoglycerate kinase 1 (PGK1) upon hypoxia. Enoyl-CoA hydratase 1 (ECHS1) is upregulated and interacts with PGK1, leading to the downregulation of PGK1 Kcr under hypoxia. Abolishment of PGK1 Kcr promotes glycolysis and suppresses mitochondrial pyruvate metabolism by activating pyruvate dehydrogenase kinase 1 (PDHK1). A low PGK1 K131cr level is correlated with malignancy and poor prognosis of breast cancer. Our findings show that PGK1 Kcr is a signal in coordinating glycolysis and the tricarboxylic acid (TCA) cycle and may serve as a diagnostic indicator for breast cancer.


Sujet(s)
Tumeurs du sein , Cycle citrique , Glycolyse , Phosphoglycerate kinase , Phosphoglycerate kinase/métabolisme , Phosphoglycerate kinase/génétique , Humains , Glycolyse/génétique , Lignée cellulaire tumorale , Femelle , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Lysine/métabolisme , Maturation post-traductionnelle des protéines , Animaux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Régulation négative , Souris , Protéomique/méthodes , Souris nude , Régulation de l'expression des gènes tumoraux , Mitochondries/métabolisme , Hypoxie cellulaire , Pyruvate dehydrogenase acetyl-transferring kinase/métabolisme , Pyruvate dehydrogenase acetyl-transferring kinase/génétique
18.
Cell Mol Biol Lett ; 29(1): 106, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095708

RÉSUMÉ

BACKGROUND: The RNA N6-methyladenosine (m6A) modification has become an essential hotspot in epigenetic modulation. Serine-arginine protein kinase 1 (SRPK1) is associated with the pathogenesis of various cancers. However, the m6A modification of SRPK1 and its association with the mechanism of in lung adenocarcinoma (LUAD) remains unclear. METHODS: Western blotting and polymerase chain reaction (PCR) analyses were carried out to identify gene and protein expression. m6A epitranscriptomic microarray was utilized to the assess m6A profile. Loss and gain-of-function assays were carried out elucidate the impact of METTL3 and SRPK1 on LUAD glycolysis and tumorigenesis. RNA immunoprecipitation (RIP), m6A RNA immunoprecipitation (MeRIP), and RNA stability tests were employed to elucidate the SRPK1's METTL3-mediated m6A modification mechanism in LUAD. Metabolic quantification and co-immunoprecipitation assays were applied to investigate the molecular mechanism by which SRPK1 mediates LUAD metabolism. RESULTS: The epitranscriptomic microarray assay revealed that SRPK1 could be hypermethylated and upregulated in LUAD. The main transmethylase METTL3 was upregulated and induced the aberrant high m6A levels of SRPK1. Mechanistically, SRPK1's m6A sites were directly methylated by METTL3, which also stabilized SRPK1 in an IGF2BP2-dependent manner. Methylated SRPK1 subsequently promoted LUAD progression through enhancing glycolysis. Further metabolic quantification, co-immunoprecipitation and western blot assays revealed that SRPK1 interacts with hnRNPA1, an important modulator of PKM splicing, and thus facilitates glycolysis by upregulating PKM2 in LUAD. Nevertheless, METTL3 inhibitor STM2457 can reverse the above effects in vitro and in vivo by suppressing SRPK1 and glycolysis in LUAD. CONCLUSION: It was revealed that in LUAD, aberrantly expressed METTL3 upregulated SRPK1 levels via an m6A-IGF2BP2-dependent mechanism. METTL3-induced SRPK1 fostered LUAD cell proliferation by enhancing glycolysis, and the small-molecule inhibitor STM2457 of METTL3 could be an alternative novel therapeutic strategy for individuals with LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Adénosine , Glycolyse , Tumeurs du poumon , Methyltransferases , Protein-Serine-Threonine Kinases , Humains , Adénosine/analogues et dérivés , Adénosine/métabolisme , Glycolyse/génétique , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/métabolisme , Adénocarcinome pulmonaire/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Animaux , Régulation de l'expression des gènes tumoraux , Souris , Lignée cellulaire tumorale , Souris nude , Épissage des ARN/génétique , , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Prolifération cellulaire/génétique
19.
BMC Cancer ; 24(1): 979, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118022

RÉSUMÉ

BACKGROUND: Gastric cancer (GC) is a major contributor to cancer-related mortality. Glycolysis plays a pivotal role in tumor microenvironment (TME) reprogramming. In this research, the functions of glycolysis-associated genes (GRGs) were evaluated to predict the outcome and reveal the characteristics of the immune microenvironment in individuals with stomach cancer. METHODS: The Cancer Genome Atlas (TCGA)-stomach adenocarcinoma (STAD) cohort provided gene expression and clinical data for gastric cancer (GC) patients, which were further authenticated using datasets sourced from the Gene Expression Omnibus (GEO). By referencing the Molecular Signatures Database (MSigDB), a total of 326 GRGs were pinpointed. The various subtypes of GC were outlined through consensus clustering, derived from the expression patterns of these GRGs. Utilizing multivariate Cox regression analysis, a multigene risk score model was formulated. Both the CIBERSORT and ESTIMATE algorithms played a pivotal role in assessing the immune microenvironment. To delve into the biological functions of the key genes, wound healing, transwell invasion, and MTT assays were conducted. RESULTS: Based on the expression patterns of GRGs, patients were categorized into two distinct groups: the metabolic subtype, designated as cluster A, and the immune subtype, labeled as cluster B. Patients belonging to cluster B exhibited a poorer prognosis. A prognostic risk score model, formulated upon the expression levels of six key GRGs - ME1, PLOD2, NUP50, CXCR4, SLC35A3, and SRD35A3 - emerged as a viable tool for predicting patient outcomes. The downregulation of CXCR4 notably diminished the glycolytic capacity of gastric cancer (GC) cells, alongside their migratory, invasive, and proliferative capabilities. Intriguingly, despite the adverse prognostic implications associated with both the immune subtype (cluster B) and the high-risk cohort, these groups exhibited a favorable immune microenvironment coupled with elevated expression of immune checkpoint genes. Our investigations revealed a positive correlation between high CXCR4 expression and low ME1 expression with the infiltration of CD8+ T cells, as well as an enhanced responsiveness to treatment with an anti-PD-1 immune checkpoint inhibitor. CONCLUSIONS: In this study, we discovered that the expression profiles of GRGs hold the potential to forecast the prognosis of gastric cancer (GC) patients, thereby possibly aiding in clinical treatment decision-making.


Sujet(s)
Glycolyse , Tumeurs de l'estomac , Microenvironnement tumoral , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/mortalité , Humains , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Pronostic , Glycolyse/génétique , Régulation de l'expression des gènes tumoraux , Mâle , Marqueurs biologiques tumoraux/génétique , Femelle , Analyse de profil d'expression de gènes , Adulte d'âge moyen , Adénocarcinome/génétique , Adénocarcinome/immunologie , Adénocarcinome/anatomopathologie , Lignée cellulaire tumorale
20.
Int J Med Sci ; 21(10): 1976-1989, 2024.
Article de Anglais | MEDLINE | ID: mdl-39113895

RÉSUMÉ

Aortic aneurysm and dissection (AD) represent a critical cardiovascular emergency with an alarmingly high mortality rate. Recent research has spotlighted the overexpression of genes associated with the m6A modification in AD patients, linking them to the presence of inflammatory M1-type macrophages. Moreover, glycolysis is widely recognized as a key feature of inflammatory M1-type macrophages, but biomarkers linking glycolysis and macrophage function to promote disease progression in AD have not been reported. We conducted an analysis of aortic immune cell infiltration, macrophages, and m6A-related biomarkers in AD patients using bioinformatics techniques. Subsequently, we employed a combination of RT-PCR, WB, and immunofluorescence assays to elucidate the alterations in the expression of M1- and M2-type macrophages, as well as markers of glycolysis, following the overexpression of key biomarkers. These findings were further validated in vivo through the creation of a rat model of AD with knockdown of the aforementioned key biomarkers. The findings revealed that the m6A-modified related gene RBM15 exhibited heightened expression in AD samples and was correlated with macrophage polarization. Upon overexpression of RBM15 in macrophages, there was an observed increase in the expression of M1-type macrophage markers CXCL9 and CXCL10, alongside a decrease in the expression of M2-type macrophage markers CCL13 and MRC1. Furthermore, there was an elevation in the expression of glycolytic enzymes GLUT1 and Hexokinase, as well as HIF1α, GAPDH, and PFKFB3 after RBM15 overexpression. Moreover, in vivo knockdown of RBM15 led to an amelioration of aortic aneurysm in the rat AD model. This knockdown also resulted in a reduction of the M1-type macrophage marker iNOS, while significantly increasing the expression of the M2-type macrophage marker CD206. In conclusion, our findings demonstrate that RBM15 upregulates glycolysis in macrophages, thus contributing to the progression of AD through the promotion of M1-type macrophage polarization. Conversely, downregulation of RBM15 suppresses M1-type macrophage polarization, thereby decelerating the advancement of AD. These results unveil potential novel targets for the treatment of AD.


Sujet(s)
Anévrysme de l'aorte , , Évolution de la maladie , Glycolyse , Macrophages , Protéines de liaison à l'ARN , Glycolyse/génétique , Humains , Animaux , Macrophages/métabolisme , Macrophages/immunologie , Rats , /anatomopathologie , /génétique , /métabolisme , Anévrysme de l'aorte/métabolisme , Anévrysme de l'aorte/génétique , Anévrysme de l'aorte/anatomopathologie , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Mâle , Modèles animaux de maladie humaine , Chimiokine CXCL10/métabolisme , Chimiokine CXCL10/génétique , Marqueurs biologiques/métabolisme , Chimiokine CXCL9/métabolisme , Chimiokine CXCL9/génétique , Femelle , Adénosine/analogues et dérivés
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE