Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.680
Filtrer
1.
Gene ; 932: 148880, 2025 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-39181273

RÉSUMÉ

It has been discovered that Trichorhinophalangeal Syndrome-1 (TRPS1), a novel member of the GATA transcription factor family, participates in both normal physiological processes and the development of numerous diseases. Recently, TRPS1 has been identified as a new biomarker to aid in cancer diagnosis and is very common in breast cancer (BC), especially in triple-negative breast cancer (TNBC). In this review, we discussed the structure and function of TRPS1 in various normal cells, focused on its role in tumorigenesis and tumor development, and summarize the research status of TRPS1 in the occurrence and development of BC. We also analyzed the potential use of TRPS1 in guiding clinically personalized precision treatment and the development of targeted drugs.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du sein , Protéines de liaison à l'ADN , Protéines de répression , Facteurs de transcription , Humains , Femelle , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/diagnostic , Tumeurs du sein/métabolisme , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Régulation de l'expression des gènes tumoraux , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Animaux
2.
Cell Physiol Biochem ; 58(5): 510-526, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39305132

RÉSUMÉ

BACKGROUND/AIMS: Osteosarcoma is a prevalent and aggressive primary malignant bone tumor affecting children and adolescents. Despite advancements in sequencing technologies, there remains a lack of reliable prognostic biomarkers and effective targeted therapies for osteosarcoma. This study focuses on identifying key prognostic genes, particularly the role of GNAS, in osteosarcoma progression. METHODS: Bioinformatics analyses were performed on osteosarcoma datasets from the Gene Expression Omnibus (GEO). Differential gene expression analysis, weighted correlation network analysis (WGCNA), and survival analysis identified potential prognostic hub genes. The expression and function of these genes were validated through immunohistochemistry and animal experiments. Specifically, the role of GNAS was investigated through siRNA-mediated knockdown in osteosarcoma cell lines and nude mice models. RESULTS: Five hub genes (PROP1, GNAS, CYP4F2, LHX3, CNGB1) were identified as significantly related to osteosarcoma prognosis. Among these, GNAS was found to be highly expressed in osteosarcoma tissues compared to normal tissues. Immunohistochemical analysis confirmed the elevated expression of GNAS in osteosarcoma samples. GNAS mutation analysis revealed a low mutation rate in osteosarcoma, suggesting its oncogenic role is independent of mutational status. Animal experiments demonstrated that knocking down GNAS significantly inhibited tumor growth and induced apoptosis in osteosarcoma cells. CONCLUSION: GNAS is highly expressed in osteosarcoma and associated with poor prognosis, acting as an oncogene in osteosarcoma progression. Targeting GNAS could be a potential therapeutic strategy for osteosarcoma. Further studies on GNAS-related signaling pathways may provide deeper insights into the molecular mechanisms driving osteosarcoma malignancy.


Sujet(s)
Tumeurs osseuses , Chromogranine , Sous-unités alpha Gs des protéines G , Souris nude , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Humains , Sous-unités alpha Gs des protéines G/génétique , Sous-unités alpha Gs des protéines G/métabolisme , Animaux , Chromogranine/génétique , Chromogranine/métabolisme , Lignée cellulaire tumorale , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Pronostic , Souris , Mâle , Femelle , Mutation , Petit ARN interférent/métabolisme , Prolifération cellulaire , Interférence par ARN , Apoptose , Souris de lignée BALB C , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie
3.
Cancer Lett ; 603: 217200, 2024 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-39222677

RÉSUMÉ

Triple-negative breast cancer (TNBC) is difficult to treat breast cancer subtype due to lack or insignificant expressions of targetable estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2). Therefore, finding a targetable protein or signaling pathway in TNBC would impact patient care. Here, we report that a member of the Mixed Lineage Kinase (MLK) family, MLK3, is an effector of G-protein-coupled protease-activated receptors 1 (PAR1) and targeting MLK3 by a small-molecule inhibitor prevented PAR1-mediated TNBC tumorigenesis. In silico and immunohistochemistry analysis of human breast tumors showed overexpression of PAR1 and MLK3 in TNBC tumors. Treating α-thrombin and PAR1 agonist increased MLK3 and JNK activities and induced cell migration in TNBC cells. The PAR1 positive/high (PAR1+/hi) population of TNBC cells showed aggressive tumor phenotype with increased MLK3 signaling. Moreover, combined inhibition of the PAR1 and MLK3 mitigated the TNBC tumor burden in preclinical TNBC models. Our data suggests that activation of the PAR1-MLK3 axis promotes TNBC tumorigenesis. Therefore, combinatorial therapy targeting MLK3 and PAR1 could effectively reduce TNBC tumor burden.


Sujet(s)
MAP Kinase Kinase Kinases , , Récepteur de type PAR-1 , Tumeurs du sein triple-négatives , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/génétique , Humains , Récepteur de type PAR-1/métabolisme , Récepteur de type PAR-1/génétique , Femelle , Animaux , Lignée cellulaire tumorale , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Mouvement cellulaire , Transduction du signal , Inhibiteurs de protéines kinases/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Carcinogenèse/métabolisme , Carcinogenèse/génétique , Souris , Prolifération cellulaire
4.
Cells ; 13(17)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39273044

RÉSUMÉ

Cancer research has advanced tremendously with the identification of causative genes, proteins, and signaling pathways. Numerous antitumor drugs have been designed and screened for cancer therapeutics; however, designing target-specific drugs for malignant cells with minimal side effects is challenging. Recently, energy-sensing- and homeostasis-associated molecules and signaling pathways playing a role in proliferation, apoptosis, autophagy, and angiogenesis have received increasing attention. Energy-metabolism-based studies have shown the contribution of energetics to cancer development, where tumor cells show increased glycolytic activity and decreased oxidative phosphorylation (the Warburg effect) in order to obtain the required additional energy for rapid division. The role of energy homeostasis in the survival of normal as well as malignant cells is critical; therefore, fuel intake and expenditure must be balanced within acceptable limits. Thus, energy-sensing enzymes detecting the disruption of glycolysis, AMP, ATP, or GTP levels are promising anticancer therapeutic targets. Here, we review the common energy mediators and energy sensors and their metabolic properties, mechanisms, and associated signaling pathways involved in carcinogenesis, and explore the possibility of identifying drugs for inhibiting the energy metabolism of tumor cells. Furthermore, to corroborate our hypothesis, we performed meta-analysis based on transcriptomic profiling to search for energy-associated biomarkers and canonical pathways.


Sujet(s)
Carcinogenèse , Métabolisme énergétique , Tumeurs , Transduction du signal , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/génétique , Métabolisme énergétique/effets des médicaments et des substances chimiques , Carcinogenèse/anatomopathologie , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/métabolisme , Carcinogenèse/génétique , Animaux
5.
Cells ; 13(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39273053

RÉSUMÉ

Lung cancer treatment and detection can be improved by the identification of new biomarkers. Novel approaches in investigating circular RNAs (circRNAs) as biomarkers have yielded promising results. A circRNA molecule circHIPK3 was found to be widely expressed in non-small-cell lung cancer (NSCLC) cells, where it plays a crucial role in lung cancer tumorigenesis. CircHIPK3 promotes lung cancer progression by sponging oncosuppressive miRNAs such as miR-124, miR-381-3p, miR-149, and miR-107, which results in increased cell proliferation, migration, and resistance to therapies. Inhibiting circHIPK3 has been demonstrated to suppress tumour growth and induce apoptosis, which suggests its potential use in the development of new lung cancer treatment strategies targeting circHIPK3-related pathways. As a biomarker, circHIPK3 shows promise for early detection and monitoring of lung cancer. CircHIPK3 increased expression levels in lung cancer cells, and its potential link to metastasis risk highlights its clinical relevance. Given the promising preliminary findings, more clinical trials are needed to validate circHIPK3 efficacy as a biomarker. Moreover, future research should determine if the mechanisms discovered in NSCLC apply to small cell lung cancer (SCLC) to investigate circHIPK3-targeted therapies for SCLC.


Sujet(s)
Marqueurs biologiques tumoraux , Carcinogenèse , Tumeurs du poumon , ARN circulaire , Humains , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , ARN circulaire/génétique , ARN circulaire/métabolisme , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Régulation de l'expression des gènes tumoraux , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , microARN/génétique , microARN/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Animaux
6.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273409

RÉSUMÉ

Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.


Sujet(s)
Tumeurs colorectales , Humains , Tumeurs colorectales/thérapie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/étiologie , Tumeurs colorectales/génétique , Carcinogenèse/génétique , Animaux , Facteurs de risque
7.
Nat Commun ; 15(1): 7856, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39251623

RÉSUMÉ

Despite recent advances in systemic therapy for hepatocellular carcinoma (HCC), the prognosis of hepatitis B virus (HBV)-induced HCC patients remains poor. By screening a sgRNA library targeting human deubiquitinases, we find that ubiquitin-specific peptidase 26 (USP26) deficiency impairs HBV-positive HCC cell proliferation. Genetically engineered murine models with Usp26 knockout confirm that Usp26 drives HCC tumorigenesis. Mechanistically, we find that the HBV-encoded protein HBx binds to the promoter and induces the production of USP26, which is an X-linked gene exclusively expressed in the testis. HBx consequently promotes the association of USP26 with SIRT1 to synergistically stabilize SIRT1 by deubiquitination, which promotes cell proliferation and impedes cell apoptosis to accelerate HCC tumorigenesis. In patients with HBV-positive HCC, USP26 is robustly induced, and its levels correlate with SIRT1 levels and poor prognosis. Collectively, our study highlights a causative link between HBV infection, deubiquitinase induction and development of HCC, identifying a druggable target, USP26.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Épigenèse génétique , Virus de l'hépatite B , Tumeurs du foie , Sirtuine-1 , Transactivateurs , Protéines virales régulatrices ou accessoires , Carcinome hépatocellulaire/virologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Humains , Animaux , Tumeurs du foie/virologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Virus de l'hépatite B/génétique , Souris , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Transactivateurs/métabolisme , Transactivateurs/génétique , Mâle , Prolifération cellulaire/génétique , Protéines virales régulatrices ou accessoires/métabolisme , Carcinogenèse/génétique , Hépatite B/virologie , Hépatite B/complications , Hépatite B/génétique , Hépatite B/métabolisme , Lignée cellulaire tumorale , Souris knockout , Régulation de l'expression des gènes tumoraux , Enzymes de désubiquitinylation/métabolisme , Enzymes de désubiquitinylation/génétique , Apoptose/génétique , Cysteine endopeptidases/métabolisme , Cysteine endopeptidases/génétique , Régions promotrices (génétique)/génétique
8.
J Transl Med ; 22(1): 833, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39256832

RÉSUMÉ

BACKGROUND: Family with sequence similarity 109, member B (FAM109B) is involved in endocytic transport and affects genetic variation in brain methylation. It is one of the important genes related to immune cell-associated diseases. In the tumor immune system, methylation can regulate tumor immunity and influence the maturation and functional response of immune cells. Whether FAM109B is involved in tumor progression and its correlation with the tumor immune microenvironment has not yet been disclosed. METHODS: A comprehensive pan-cancer analysis of FAM109B expression, prognosis, immunity, and TMB was conducted. The expression, clinical features, and prognostic value of FAM109B in low-grade gliomas (LGG) were evaluated using TCGA, CGGA, and Gravendeel databases. The expression of FAM109B was validated by qRT-PCR, immunohistochemistry (IHC), and Western blotting (WB). The relationship between FAM109B and methylation, Copy Number Variation (CNV), prognosis, immune checkpoints (ICs), and common chemotherapy drug sensitivity in LGG was explored through Cox regression, Kaplan-Meier curves, and Spearman correlation analysis. FAM109B levels and their distribution were studied using the TIMER database and single-cell analysis. The potential role of FAM109B in gliomas was further investigated through in vitro and in vivo experiments. RESULTS: FAM109B was significantly elevated in various tumor types and was associated with poor prognosis. Its expression was related to aggressive progression and poor prognosis in low-grade glioma patients, serving as an independent prognostic marker for LGG. Glioma grade was negatively correlated with FAM109B DNA promoter methylation. Immune infiltration and single-cell analysis showed significant expression of FAM109B in tumor-associated macrophages (TAMs). The expression of FAM109B was closely related to gene mutations, immune checkpoints (ICs), and chemotherapy drugs in LGG. In vitro studies showed increased FAM109B expression in LGG, closely related to cell proliferation. In vivo studies showed that mice in the sh-FAM109B group had slower tumor growth, slower weight loss, and longer survival times. CONCLUSIONS: FAM109B, as a novel prognostic biomarker for low-grade gliomas, exhibits specific overexpression in TAMs and may be a potential therapeutic target for LGG patients.


Sujet(s)
Tumeurs du cerveau , Méthylation de l'ADN , Régulation de l'expression des gènes tumoraux , Gliome , Grading des tumeurs , Macrophages associés aux tumeurs , Gliome/génétique , Gliome/anatomopathologie , Gliome/métabolisme , Humains , Macrophages associés aux tumeurs/métabolisme , Macrophages associés aux tumeurs/anatomopathologie , Macrophages associés aux tumeurs/immunologie , Méthylation de l'ADN/génétique , Animaux , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/immunologie , Pronostic , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Variations de nombre de copies de segment d'ADN/génétique , Microenvironnement tumoral , Lignée cellulaire tumorale , Femelle , Mâle , Souris nude , Souris , Estimation de Kaplan-Meier , Bases de données génétiques
9.
Sci Rep ; 14(1): 21570, 2024 09 16.
Article de Anglais | MEDLINE | ID: mdl-39284825

RÉSUMÉ

Heterogeneous nuclear ribonucleoproteins (hnRNPs), a group of proteins that control gene expression, have been implicated in many post-transcriptional processes. SYNCRIP (also known as hnRNP Q), a subtype of hnRNPs, has been reported to be involved in mRNA splicing and translation. In addition, the deregulation of SYNCRIP was found in colorectal cancer (CRC). However, the role of SYNCRIP in regulating CRC growth remains largely unknown. Here, we found that SYNCRIP was highly expressed in colorectal cancer by analyzing TCGA and GEPIA database. Furthermore, we confirmed the expression of SYNCRIP expression in CRC tumor and CRC cell lines. Functionally, SYNCRIP depletion using shRNA in CRC cell lines (SW480 and HCT 116) resulted in increased caspase3/7 activity and decreased cell proliferation, as well as migration. Meanwhile, overexpression of SYNCRIP showed opposite results. Mechanistically, SYNCRIP regulated the expression of DNA methyltransferases (DNMT) 3A, but not DNMT1 or DNMT3B, which affected the expression of tumor suppressor, p16. More importantly, our in vivo experiments showed that SYNCRIP depletion significantly inhibited colorectal tumor growth. Taken all together, our results suggest SYNCRIP as a potent therapeutic target in colorectal cancer.


Sujet(s)
Carcinogenèse , Prolifération cellulaire , Tumeurs colorectales , DNA (cytosine-5-)-methyltransferase , DNA methyltransferase 3A , Régulation de l'expression des gènes tumoraux , Régulation positive , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , DNA (cytosine-5-)-methyltransferase/métabolisme , DNA (cytosine-5-)-methyltransferase/génétique , Prolifération cellulaire/génétique , DNA methyltransferase 3A/métabolisme , Animaux , Carcinogenèse/génétique , Lignée cellulaire tumorale , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Inhibiteur p16 de kinase cycline-dépendante/génétique , Souris , Ribonucléoprotéines nucléaires hétérogènes/métabolisme , Ribonucléoprotéines nucléaires hétérogènes/génétique , Mouvement cellulaire/génétique , Cellules HCT116 , Souris nude
10.
Clin Exp Med ; 24(1): 224, 2024 Sep 19.
Article de Anglais | MEDLINE | ID: mdl-39294405

RÉSUMÉ

As one of the factors regulating tumour angiogenesis, angiopoietin-4 (ANGPT4), which plays an important role in promoting tumour proliferation, survival, expansion and angiogenesis, is highly expressed in some tumours, such as lung adenocarcinoma, glioblastoma and ovarian cancer. This may be related to the fact that ANGPT4 affects the blood vessels and lymphatic system of the tumour. Specifically, ANGPT4 could play an effective role in promoting cancer by affecting the tyrosine kinase receptor TIE2, ERK1/2 and PI3K/AKT signalling pathways. Therefore, ANGPT4 may be an important biomarker for the occurrence and development of cancer and poor prognosis. In addition, the inhibition of ANGPT4 may be a useful cancer treatment. This paper reviews the latest preclinical research on ANGPT4, emphasizes its role in tumourigenesis and broadens our understanding of the carcinogenic function of ANGPT4 and the development of ANGPT4 inhibitors. This is the latest version of the revised version of the previous article.


Sujet(s)
Carcinogenèse , Humains , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Transduction du signal , Néovascularisation pathologique/métabolisme , Animaux , Régulation de l'expression des gènes tumoraux , Protéine-4 similaire à l'angiopoïétine
11.
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
12.
Nat Commun ; 15(1): 8070, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-39277592

RÉSUMÉ

Recent advances in high-resolution mapping of spatial interactions among regulatory elements support the existence of complex topological assemblies of enhancers and promoters known as enhancer-promoter hubs or cliques. Yet, organization principles of these multi-interacting enhancer-promoter hubs and their potential role in regulating gene expression in cancer remain unclear. Here, we systematically identify enhancer-promoter hubs in breast cancer, lymphoma, and leukemia. We find that highly interacting enhancer-promoter hubs form at key oncogenes and lineage-associated transcription factors potentially promoting oncogenesis of these diverse cancer types. Genomic and optical mapping of interactions among enhancer and promoter elements further show that topological alterations in hubs coincide with transcriptional changes underlying acquired resistance to targeted therapy in T cell leukemia and B cell lymphoma. Together, our findings suggest that enhancer-promoter hubs are dynamic and heterogeneous topological assemblies with the potential to control gene expression circuits promoting oncogenesis and drug resistance.


Sujet(s)
Carcinogenèse , Résistance aux médicaments antinéoplasiques , Éléments activateurs (génétique) , Régulation de l'expression des gènes tumoraux , Réseaux de régulation génique , Régions promotrices (génétique) , Humains , Régions promotrices (génétique)/génétique , Éléments activateurs (génétique)/génétique , Résistance aux médicaments antinéoplasiques/génétique , Carcinogenèse/génétique , Femelle , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Oncogènes/génétique , Lignée cellulaire tumorale , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Leucémies/génétique , Leucémies/métabolisme , Lymphomes/génétique , Lymphomes/métabolisme
13.
Sci Adv ; 10(38): eadn2806, 2024 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-39303037

RÉSUMÉ

We previously showed that inhibition of glycolysis in cutaneous squamous cell carcinoma (SCC)-initiating cells had no effect on tumorigenesis, despite the perceived requirement of the Warburg effect, which was thought to drive carcinogenesis. Instead, these SCCs were metabolically flexible and sustained growth through glutaminolysis, another metabolic process frequently implicated to fuel tumorigenesis in various cancers. Here, we focused on glutaminolysis and genetically blocked this process through glutaminase (GLS) deletion in SCC cells of origin. Genetic deletion of GLS had little effect on tumorigenesis due to the up-regulated lactate consumption and utilization for the TCA cycle, providing further evidence of metabolic flexibility. We went on to show that posttranscriptional regulation of nutrient transporters appears to mediate metabolic flexibility in this SCC model. To define the limits of this flexibility, we genetically blocked both glycolysis and glutaminolysis simultaneously and found the abrogation of both of these carbon utilization pathways was enough to prevent both papilloma and frank carcinoma.


Sujet(s)
Carcinome épidermoïde , Glutaminase , Glycolyse , Follicule pileux , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Animaux , Follicule pileux/métabolisme , Glutaminase/métabolisme , Glutaminase/génétique , Souris , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/génétique , Cellules souches/métabolisme , Glutamine/métabolisme , Humains , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/génétique , Carcinogenèse/métabolisme , Carcinogenèse/génétique
14.
Elife ; 132024 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-39320349

RÉSUMÉ

The ß-catenin-dependent canonical Wnt signaling is pivotal in organ development, tissue homeostasis, and cancer. Here, we identified an upstream enhancer of Ctnnb1 - the coding gene for ß-catenin, named ieCtnnb1 (intestinal enhancer of Ctnnb1), which is crucial for intestinal homeostasis. ieCtnnb1 is predominantly active in the base of small intestinal crypts and throughout the epithelia of large intestine. Knockout of ieCtnnb1 led to a reduction in Ctnnb1 transcription, compromising the canonical Wnt signaling in intestinal crypts. Single-cell sequencing revealed that ieCtnnb1 knockout altered epithelial compositions and potentially compromised functions of small intestinal crypts. While deletion of ieCtnnb1 hampered epithelial turnovers in physiologic conditions, it prevented occurrence and progression of Wnt/ß-catenin-driven colorectal cancers. Human ieCTNNB1 drove reporter gene expression in a pattern highly similar to mouse ieCtnnb1. ieCTNNB1 contains a single-nucleotide polymorphism associated with CTNNB1 expression levels in human gastrointestinal epithelia. The enhancer activity of ieCTNNB1 in colorectal cancer tissues was stronger than that in adjacent normal tissues. HNF4α and phosphorylated CREB1 were identified as key trans-factors binding to ieCTNNB1 and regulating CTNNB1 transcription. Together, these findings unveil an enhancer-dependent mechanism controlling the dosage of Wnt signaling and homeostasis in intestinal epithelia.


Sujet(s)
Homéostasie , Muqueuse intestinale , Voie de signalisation Wnt , bêta-Caténine , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Muqueuse intestinale/métabolisme , Animaux , Humains , Souris , Carcinogenèse/génétique , Éléments activateurs (génétique)/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Transcription génétique
15.
Cancer Immunol Immunother ; 73(11): 233, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39271545

RÉSUMÉ

Gastric cancer (GC) is a highly heterogeneous disease with a complex tumor microenvironment (TME) that encompasses multiple cell types including cancer cells, immune cells, stromal cells, and so on. Cancer-associated cells could remodel the TME and influence the progression of GC and therapeutic response. Single-cell RNA sequencing (scRNA-seq), as an emerging technology, has provided unprecedented insights into the complicated biological composition and characteristics of TME at the molecular, cellular, and immunological resolutions, offering a new idea for GC studies. In this review, we discuss the novel findings from scRNA-seq datasets revealing the origin and evolution of GC, and scRNA-seq is a powerful tool for investigating transcriptional dynamics and intratumor heterogeneity (ITH) in GC. Meanwhile, we demonstrate that the vital immune cells within TME, including T cells, B cells, macrophages, and stromal cells, play an important role in the disease progression. Additionally, we also overview that how scRNA-seq facilitates our understanding about the effects on individualized therapy of GC patients. Spatial transcriptomes (ST) have been designed to determine spatial distribution and capture local intercellular communication networks, enabling a further understanding of the relationship between the spatial background of a particular cell and its functions. In summary, scRNA-seq and other single-cell technologies provide a valuable perspective for molecular and pathological disease characteristics and hold promise for advancing basic research and clinical practice in GC.


Sujet(s)
Médecine de précision , Analyse de séquence d'ARN , Analyse sur cellule unique , Tumeurs de l'estomac , Microenvironnement tumoral , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Analyse sur cellule unique/méthodes , Médecine de précision/méthodes , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Analyse de séquence d'ARN/méthodes , Hétérogénéité génétique , Carcinogenèse/génétique , Animaux
16.
Sci Signal ; 17(854): eabq4888, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39288218

RÉSUMÉ

Doublecortin-like kinase 1 (DCLK1) is a proposed driver of gastric cancer (GC) that phosphorylates serine and threonine residues. Here, we showed that the kinase activity of DCLK1 orchestrated cancer cell-intrinsic and-extrinsic processes that led to pro-invasive and pro-metastatic reprogramming of GC cells. Inhibition of the kinase activity of DCLK1 reduced the growth of subcutaneous xenograft tumors formed from MKN1 human gastric carcinoma cells in mice and decreased the abundance of the stromal markers α-Sma, vimentin, and collagen. Similar effects were seen in mice with xenograft tumors formed from MKN1 cells expressing a kinase-inactive DCLK1 mutant (MKN1D511N). MKN1D511N cells also had reduced in vitro migratory potential and stemness compared with control cells. Mice orthotopically grafted with MKN1 cells overexpressing DCLK1 (MKN1DCLK1) showed increased invasiveness and had a greater incidence of lung metastases compared with those grafted with control MKN1 cells. Mechanistically, we showed that the chemokine CXCL12 acted downstream of DCLK1 in cultured MKN1 cells and in mice subcutaneously implanted with gastric tumors formed by MKN1DCLK1 cells. Moreover, inhibition of the kinase activity of DCLK1 or the expression of DCLK1D511N reversed the pro-tumorigenic and pro-metastatic phenotype. Together, this study establishes DCLK1 as a broadly acting and potentially targetable promoter of GC.


Sujet(s)
Évolution de la maladie , Kinases de type doublecortine , Protéines et peptides de signalisation intracellulaire , Phénotype , Protein-Serine-Threonine Kinases , Tumeurs de l'estomac , Kinases de type doublecortine/métabolisme , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Animaux , Humains , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Souris , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Carcinogenèse/génétique , Carcinogenèse/métabolisme
17.
Cell Death Dis ; 15(9): 666, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261464

RÉSUMÉ

Colorectal cancer (CRC) is the second leading cause of cancer-related mortality worldwide. Although CRC patients' survival is improved with surgical resection and immunotherapy, metastasis and recurrence remain major problems leading to poor prognosis. Therefore, exploring pathogenesis and identifying specific biomarkers are crucial for CRC early diagnosis and targeted therapy. CCDC113, a member of CCDC families, has been reported to play roles in ciliary assembly, ciliary activity, PSCI, asthma and early lung cancer diagnosis. However, the functions of CCDC113 in CRC still remain unclear. In this study, we find that CCDC113 is significantly highly expressed in CRC. High expression of CCDC113 is significantly correlated with CRC patients' poor prognosis. CCDC113 is required for CRC tumorigenesis and metastasis. RNA-seq and TCGA database analysis indicate that CCDC113 is positively correlated with TGF-ß signaling pathway. TGF-ß signaling pathway inhibitor galunisertib could reverse the increased proliferation and migration ability of CRC cells caused by CCDC113 overexpression in vitro and in vivo. These results indicate that CCDC113 promotes CRC tumorigenesis and metastasis via TGF-ß signaling pathway. In conclusion, it is the first time to explore the functions and mechanisms of CCDC113 in CRC tumorigenesis and metastasis. And CCDC113 may be a potential biomarker and therapeutic target for CRC intervention.


Sujet(s)
Carcinogenèse , Prolifération cellulaire , Tumeurs colorectales , Transduction du signal , Facteur de croissance transformant bêta , Animaux , Femelle , Humains , Mâle , Souris , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Souris nude , Métastase tumorale , Pronostic , Pyrazoles/pharmacologie , Quinoléines/pharmacologie , Facteur de croissance transformant bêta/métabolisme
18.
Clin Epigenetics ; 16(1): 125, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39261937

RÉSUMÉ

BACKGROUND: Breast tumorigenesis is a complex and multistep process accompanied by both genetic and epigenetic dysregulation. In contrast to the extensive studies on DNA epigenetic modifications 5-hydroxymethylcytosine (5hmC) and 5-methylcytosine (5mC) in malignant breast tumors, their roles in the early phases of breast tumorigenesis remain ambiguous. RESULTS: DNA 5hmC and 5mC exhibited a consistent and significant decrease from usual ductal hyperplasia to atypical ductal hyperplasia and subsequently to ductal carcinoma in situ (DCIS). However, 5hmC showed a modest increase in invasive ductal breast cancer compared to DCIS. Genomic analyses showed that the changes in 5hmC and 5mC levels occurred around the transcription start sites (TSSs), and the modification levels were strongly correlated with gene expression levels. Meanwhile, it was found that differentially hydroxymethylated regions (DhMRs) and differentially methylated regions (DMRs) were overlapped in the early phases and accompanied by the enrichment of active histone marks. In addition, TET2-related DNA demethylation was found to be involved in breast tumorigenesis, and four transcription factor binding sites (TFs: ESR1, FOXA1, GATA3, FOS) were enriched in TET2-related DhMRs/DMRs. Intriguingly, we also identified a certain number of common DhMRs between tumor samples and cell-free DNA (cfDNA). CONCLUSIONS: Our study reveals that dynamic changes in DNA 5hmC and 5mC play a vital role in propelling breast tumorigenesis. Both TFs and active histone marks are involved in TET2-related DNA demethylation. Concurrent changes in 5hmC signals in primary breast tumors and cfDNA may play a promising role in breast cancer screening.


Sujet(s)
5-Méthyl-cytosine , Tumeurs du sein , Protéines de liaison à l'ADN , Dioxygenases , Protéines proto-oncogènes , Humains , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Femelle , Tumeurs du sein/génétique , Protéines de liaison à l'ADN/génétique , Dioxygenases/génétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Carcinogenèse/génétique , Méthylation de l'ADN/génétique , Épigenèse génétique/génétique , Régulation de l'expression des gènes tumoraux , Déméthylation de l'ADN
19.
Theranostics ; 14(13): 4948-4966, 2024.
Article de Anglais | MEDLINE | ID: mdl-39267787

RÉSUMÉ

Rationale: Tumor cells remodel transcriptome to construct an ecosystem with stemness features, which maintains tumor growth and highly malignant characteristics. However, the core regulatory factors involved in this process still need to be further discovered. Methods: Single cell RNA-sequncing (scRNA-seq) and bulk RNA-sequencing profiles derived from fetal liver, normal liver, liver tumors, and their adjacent samples were collected to analyze the ecosystem of liver cancer. Mouse models were established to identify molecular functions of oncofetal-related oncogenes using hydrodynamic tail vein injection. Results: We found that liver cancer rebuilt oncofetal ecosystem to maintain malignant features. Interestingly, we identified a group of RNA-binding proteins (RBPs) that were highly overexpressed with oncofetal features. Among them, TRIM71 was specifically expressed in liver cancers and was associated with poor outcomes. TRIM71 drove the carcinogenesis of hepatocellular carcinoma (HCC), and knockdown of TRIM71 significantly abolished liver cancer cell proliferation. Mechanistically, TRIM71 formed a protein complex with IGF2BP1, bound to and stabilized the mRNA of CEBPA in an m6A-dependent manner, enhance the serine/glycine metabolic pathway, and ultimately promoted liver cancer progression. Furthermore, we identified that all-trans-retinoic acid (ATRA) combined with e1A binding protein p300 (EP300) inhibitor A-485 repressed TRIM71, attenuated glycine/serine metabolism, and inhibited liver cancer cell proliferation with high TRIM71 levels. Conclusions: We demonstrated the oncofetal status in liver cancer and highlighted the crucial role of TRIM71 and provided potential therapeutic strategies and liver cancer-specific biomarker for liver cancer patients.


Sujet(s)
Carcinogenèse , Carcinome hépatocellulaire , Glycine , Tumeurs du foie , Sérine , Animaux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Souris , Humains , Sérine/métabolisme , Carcinogenèse/génétique , Carcinogenèse/métabolisme , Glycine/métabolisme , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Protéines à motif tripartite/métabolisme , Protéines à motif tripartite/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Souris nude
20.
Int J Mol Med ; 54(5)2024 11.
Article de Anglais | MEDLINE | ID: mdl-39219279

RÉSUMÉ

Metastasis is the leading cause of cancer­related death in osteosarcoma (OS). OS stem cells (OSCs) and anoikis resistance are considered to be essential for tumor metastasis formation. However, the underlying mechanisms involved in the maintenance of a stem­cell phenotype and anoikis resistance in OS are mostly unknown. Fos­like antigen 1 (FOSL1) is important in maintaining a stem­like phenotype in various cancers; however, its role in OSCs and anoikis resistance remains unclear. In the present study, the dynamic expression patterns of FOSL1 were investigated during the acquisition of cancer stem­like properties using RNA sequencing, PCR, western blotting and immunofluorescence. Flow cytometry, tumor­sphere formation, clone formation assays, anoikis assays, western blotting and in vivo xenograft and metastasis models were used to further investigate the responses of the stem­cell phenotype and anoikis resistance to FOSL1 overexpression or silencing in OS cell lines. The underlying molecular mechanisms were evaluated, focusing on whether SOX2 is crucially involved in FOSL1­mediated stemness and anoikis in OS. FOSL1 expression was observed to be upregulated in OSCs and promoted tumor­sphere formation, clone formation and tumorigenesis in OS cells. FOSL1 expression correlated positively with the expression of stemness­related factors (SOX2, NANOG, CD117 and Stro1). Moreover, FOSL1 facilitated OS cell anoikis resistance and promoted metastases by regulating the expression of apoptosis related proteins BCL2 and BAX. Mechanistically, FOSL1 upregulated SOX2 expression by interacting with the SOX2 promoter and activating its transcription. The results also showed that SOX2 is critical for FOSL1­mediated stem­like properties and anoikis resistance. The current findings indicated that FOSL1 is an important regulator that promotes a stem cell­like phenotype and anoikis resistance to facilitate tumorigenesis and metastasis in OS by regulating the transcription of SOX2. Thus, FOSL1 might represent an attractive target for therapeutic interventions in OS.


Sujet(s)
Anoïkis , Carcinogenèse , Régulation de l'expression des gènes tumoraux , Cellules souches tumorales , Ostéosarcome , Protéines proto-oncogènes c-fos , Facteurs de transcription SOX-B1 , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Humains , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Facteurs de transcription SOX-B1/métabolisme , Facteurs de transcription SOX-B1/génétique , Anoïkis/génétique , Animaux , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Lignée cellulaire tumorale , Souris , Carcinogenèse/génétique , Carcinogenèse/anatomopathologie , Métastase tumorale , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/secondaire , Souris nude , Mâle , Femelle , Souris de lignée BALB C
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE