Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.514
Filtrer
1.
Clin Transl Med ; 14(10): e70036, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39350478

RÉSUMÉ

Solid tumours exhibit a well-defined architecture, comprising a differentiated core and a dynamic border that interfaces with the surrounding tissue. This border, characterised by distinct cellular morphology and molecular composition, serves as a critical determinant of the tumour's invasive behaviour. Notably, the invasive border of the primary tumour represents the principal site for intravasation of metastatic cells. These cells, known as circulating tumour cells (CTCs), function as 'seeds' for distant dissemination and display remarkable heterogeneity. Advancements in spatial sequencing technology are progressively unveiling the spatial biological features of tumours. However, systematic investigations specifically targeting the characteristics of the tumour border remain scarce. In this comprehensive review, we illuminate key biological insights along the tumour body-border-haematogenous metastasis axis over the past five years. We delineate the distinctive landscape of tumour invasion boundaries and delve into the intricate heterogeneity and phenotype of CTCs, which orchestrate haematogenous metastasis. These insights have the potential to explain the basis of tumour invasion and distant metastasis, offering new perspectives for the development of more complex and precise clinical interventions and treatments.


Sujet(s)
Invasion tumorale , Métastase tumorale , Cellules tumorales circulantes , Analyse sur cellule unique , Humains , Métastase tumorale/génétique , Analyse sur cellule unique/méthodes , Invasion tumorale/génétique , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Tumeurs/génétique
2.
Arch Esp Urol ; 77(8): 865-874, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39385481

RÉSUMÉ

BACKGROUND: Ferroptosis is an iron-dependent cell death mode. Ferroptosis resistance is related to prostate cancer (PCa) invasion; However, there is vague understanding with regard to the underlying mechanism. This study was undertaken to clarify the role and mechanism of upstream stimulatory factor 1 (USF1) in ferroptosis resistance in invasive PCa. METHODS: USF1 was silenced in the human PCa cell lines C4-2B and PC-3. After these cells were treated with a ferroptosis inhibitor, cell viability and invasion and the expression of glutathione peroxidase 4 (GPX4) were evaluated. Chromatin immunoprecipitation and Dual-luciferase reporter assay suggested an interaction between USF1 and brain-expressed X-linked protein 1 (BEX1). Consequently, BEX1 was overexpressed in USF1-silenced C4-2B and PC-3 cells and its effects on cell viability and invasion and GPX4 expression were examined. RESULTS: USF1 silencing mitigated PCa cell viability and invasion. Treatment with a ferroptosis inhibitor counteracted the inhibitory roles of USF1 silencing in cell invasion and GPX4 expression. Additionally, USF1 silencing decreased BEX1 expression and USF1 was found to bind to the BEX1 promoter. BEX1 overexpression reversed the influences of USF1 silencing on the viability, BEX1 protein expression, invasive ability and ferroptosis of PCa cells. CONCLUSIONS: USF1 activates the transcription of BEX1, preventing PCa cell ferroptosis and promoting cell invasion. Therefore, USF1 silencing may inhibit the progression of PCa by reducing ferroptosis resistance.


Sujet(s)
Ferroptose , Extinction de l'expression des gènes , Tumeurs de la prostate , Facteurs de transcription USF , Humains , Ferroptose/génétique , Mâle , Facteurs de transcription USF/génétique , Facteurs de transcription USF/métabolisme , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Lignée cellulaire tumorale , Cellules cancéreuses en culture , Invasion tumorale/génétique
3.
Cancer Control ; 31: 10732748241281716, 2024.
Article de Anglais | MEDLINE | ID: mdl-39236066

RÉSUMÉ

INTRODUCTION: The role of SMU1 in DNA replication and RNA splicing is well-established, yet its specific function and dysregulated mechanisms in gastric cancer (GC) remain inadequately elucidated. This study seeks to investigate the potential oncogenic and progression-promoting effects of SMU1 in GC, with the ultimate goal of informing novel approaches for treatment and diagnosis. METHODS: The study investigated the expression levels of SMU1 in GC and adjacent normal tissues by analyzing data from the TCGA (27 tissue pairs) and GEO (47 tissue pairs) databases. Immunohistochemistry was used to examine 277 tumor tissue and adjacent non-tumor tissue spots from GC tissue chips, along with relevant follow-up information. The study further assessed the proliferation, invasion, and migration capabilities of cells by manipulating SMU1 expression levels and conducting various assays, including CCK-8, EdU incorporation, colony formation, transwells, flow cytometry, and subcutaneous tumorigenesis assays. RESULTS: Our study revealed a significant upregulation of SMU1 mRNA and protein levels in GC tissues compared to adjacent tissues. Univariate and multivariate Cox analysis demonstrated that elevated levels of SMU1 were independent prognostic factors for GC prognosis (P = 0.036). Additionally, median survival analysis indicated a significant association between high SMU1 expression and poor prognosis in GC patients (P = 0.0002). In experiments conducted both in vivo and in vitro, it was determined that elevated levels of SMU1 can enhance the proliferation, invasion, and migration of GC cells, whereas suppression of SMU1 can impede the progression of GC by modulating the G1/S checkpoint of the cell cycle. CONCLUSIONS: Our research introduces the novel idea that SMU1 could serve as a prognostic marker for GC progression, influencing cell proliferation through cell cycle activation. These results offer valuable insights into the understanding, diagnosis, and management of gastric carcinoma.


Sujet(s)
Cycle cellulaire , Mouvement cellulaire , Prolifération cellulaire , Invasion tumorale , Tumeurs de l'estomac , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Cycle cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Souris nude , Invasion tumorale/génétique , Pronostic , Facteurs d'épissage des ARN/génétique , Facteurs d'épissage des ARN/métabolisme , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/métabolisme
4.
Eur Thyroid J ; 13(5)2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39312732

RÉSUMÉ

Objective: This study examined the effect of sirtuin 4 (SIRT4), a NAD+-dependent deacetylase, on the proliferation and progression of papillary thyroid carcinoma (PTC). Methods: Data from The Cancer Genome Atlas (TCGA) were analyzed to identify SIRT4 expression in thyroid cancer. Subsequently, the correlation between SIRT4 expression and clinical characteristics was examined in 205 PTC tissue samples. In vitro assays using three human thyroid cancer cell lines (B-CPAP, TPC-1, and SNU-790) were conducted to assess the effects of regulated SIRT4 expression on cell growth, apoptosis, invasion, and migration. Furthermore, in vivo experiments were performed in a xenograft mouse model. Results: Gene Expression Omnibus (GEO) and TCGA data indicated that SIRT4 expression is lower in thyroid cancer and SIRT4 downregulation is associated with poor overall survival. In PTC tissues, positive SIRT4 expression was associated with decreased extracapsular extension. In in vitro experiments using three human thyroid cancer cell lines, overexpression of SIRT4 decreased cell survival, clonogenic potential, and invasion and migratory capabilities, as well as inducing apoptosis and increasing reactive oxygen species levels. SIRT4 overexpression upregulated E-cadherin and downregulated N-cadherin, suggesting its potential involvement in the regulation of epithelial-mesenchymal transition. These findings were confirmed in vivo using a xenograft mouse model. Conclusion: This study provides novel insight into the potential contribution of SIRT4 to the regulation of the pathological progression of PTC. The data suggest that SIRT4 plays a tumor-suppressive role in PTC by inhibiting growth, survival, and invasive potential. Future research should investigate the molecular mechanisms underlying these effects of SIRT4.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Invasion tumorale , Sirtuines , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Humains , Sirtuines/génétique , Sirtuines/métabolisme , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/métabolisme , Cancer papillaire de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/métabolisme , Animaux , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Invasion tumorale/génétique , Souris , Mâle , Femelle , Apoptose , Cadhérines/métabolisme , Cadhérines/génétique , Souris nude , Adulte d'âge moyen , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Régulation négative , Protéines mitochondriales
5.
Biochem Biophys Res Commun ; 733: 150676, 2024 Nov 12.
Article de Anglais | MEDLINE | ID: mdl-39303527

RÉSUMÉ

BACKGROUND: Non-Small Cell Lung Cancer (NSCLC) presents as a highly metastatic disease with Kras and P53 as prevalent oncogenic driver mutations. Endocytosis, through its role in receptor recycling and enrichment, is important for cancer cell proliferation and metastasis. Huntingtin Interacting Protein 1 (HIP1) is a clathrin mediated endocytic adapter protein found overexpressed in different cancers. However, conflicting roles both as a tumour promoter and suppressor are reported. HIP1 expression is found repressed at advanced stages and some HIP1-ALK fusions are reported in NSCLC patients. However, the molecular mechanisms and implications of HIP1 depletion are not completely understood. METHODS: HIP1 depletion was performed using siRNA transient transfection and validated using immunoblotting for each experiment. Gene expression dataset from TCGA, GTEX and GEO databases was analysed to explore HIP1 expression in Lung cancer patients. Kaplan-Meier Plotter database was used to analyse the survival correlation between HIP1 mRNA expression in lung cancer patients. HIP1 depleted A549 cells were analysed for deregulated global proteome using label-free LC-MS and this data is available via ProteomeXchange with identifier PXD054307. Various functional assays such as matrigel based invasion, trans-well migration, soft agar colony and angiogenesis tube formation were performed after HIP1 depletion. NRF2 inhibitor was used after HIP1 knockdown to assess its effect on invasion and soft agar colony formation. RESULTS: In silico analysis of HIP1 transcript expression reveals that it is reduced in high-grade and metastatic lung cancer patients correlating with poor survival. Global proteome profiling reveals that HIP1 depleted A549 cells are enriched in pathways associated with metabolism, proliferation and survival. Molecular and functional analysis indicate higher invasive ability of HIP1 depleted cells. The secretome from HIP1 depleted cells also increases the angiogenic potential of HUVEC cells. NRF2 inhibition significantly reverses invasion of HIP1 depleted NSCLC cells with different driver mutations. CONCLUSION: Our study shows that HIP1 depletion leads to activation of various molecular pathways responsible for cell proliferation and survival. Additionally, enhancement of invasion and anchorage-independent growth in HIP1 depleted subsets of NSCLC cells is via upregulation of NRF2 and can be reversed by its inhibitor.


Sujet(s)
Tumeurs du poumon , Facteur-2 apparenté à NF-E2 , Invasion tumorale , Humains , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Facteur-2 apparenté à NF-E2/antagonistes et inhibiteurs , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Invasion tumorale/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Mouvement cellulaire/génétique , Cellules A549 , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire/génétique , Métastase tumorale , Lignée cellulaire tumorale , Protéines de liaison à l'ADN
6.
Arch Dermatol Res ; 316(9): 634, 2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-39305302

RÉSUMÉ

Ferroptosis, a key factor in tumor progression, is poorly understood at the molecular level. This study investigates how ELK4 and CHMP6 regulate skin cutaneous melanoma (SKCM) cell proliferation and ferroptosis. Analysis of TCGA data reveals high expression of ELK4 and CHMP6 in SKCM. Overexpression of ELK4 or CHMP6 enhances cell proliferation, invasion, and migration while reducing ROS and Fe2 + levels. It also increases GPX4 and xCT expression and decreases ACSL4 levels in SKCM cells. The opposite effects are observed with ELK4 or CHMP6 knockdown. ELK4 binds to the CHMP6 promoter, promoting CHMP6 transcription. Knockdown of CHMP6 reverses the oncogenic effects of ELK4 overexpression. In conclusion, ELK4 enhances proliferation, invasion, and migration while inhibiting ferroptosis in SKCM cells by upregulating CHMP6 transcription. This study sheds light on the intricate mechanisms involved in SKCM progression and identifies potential therapeutic targets in melanoma treatment.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Ferroptose , Régulation de l'expression des gènes tumoraux , Mélanome , Tumeurs cutanées , Humains , Système y+ de transport d'acides aminés/métabolisme , Système y+ de transport d'acides aminés/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Coenzyme A ligases/métabolisme , Coenzyme A ligases/génétique , Ferroptose/génétique , Mélanome/anatomopathologie , Mélanome/génétique , Mélanome/métabolisme , Melanoma, Cutaneous Malignant , Invasion tumorale/génétique , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/génétique , Tumeurs cutanées/métabolisme
7.
Aging (Albany NY) ; 16(17): 12239-12251, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39248711

RÉSUMÉ

The relationship between metastasis-associated protein 2 (MTA2) overexpression and tumor growth and metastasis has been extensively studied in a variety of tumor cells but not in human osteosarcoma cells. This study aims to elucidate the clinical significance, underlying molecular mechanisms, and biological functions of MTA2 in human osteosarcoma in vitro and in vivo. Our results show that MTA2 was elevated in osteosarcoma cell lines and osteosarcoma tissues and was associated with tumor stage and overall survival of osteosarcoma patients. Knockdown of MTA2 inhibited osteosarcoma cell migration and invasion by reducing the expression of urokinase-type plasminogen activator (uPA). Bioinformatic analysis demonstrated that high levels of uPA in human osteosarcoma tissues correlated positively with MTA2 expression. Furthermore, treatment with recombinant human uPA (Rh-uPA) caused significant restoration of OS cell migration and invasion in MTA2 knockdown osteosarcoma cells. We found that ERK1/2 depletion increased the expression of uPA, facilitating osteosarcoma cell migration and invasion. Finally, MTA2 depletion significantly reduced tumor metastasis and the formation of lung nodules in vivo. Overall, our study suggests that MTA2 knockdown suppresses osteosarcoma cell metastasis by decreasing uPA expression via ERK signaling. This finding provides new insight into potential treatment strategies against osteosarcoma metastasis by targeting MTA2.


Sujet(s)
Tumeurs osseuses , Mouvement cellulaire , Techniques de knock-down de gènes , Histone deacetylases , Ostéosarcome , Protéines de répression , Activateur du plasminogène de type urokinase , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Humains , Activateur du plasminogène de type urokinase/génétique , Activateur du plasminogène de type urokinase/métabolisme , Lignée cellulaire tumorale , Protéines de répression/génétique , Protéines de répression/métabolisme , Mouvement cellulaire/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Animaux , Histone deacetylases/métabolisme , Histone deacetylases/génétique , Mâle , Femelle , Souris , Régulation de l'expression des gènes tumoraux , Invasion tumorale/génétique , Métastase tumorale , Souris nude , Système de signalisation des MAP kinases/génétique
8.
Pathol Res Pract ; 262: 155563, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39217772

RÉSUMÉ

BACKGROUND: The actin-binding protein anillin (ANLN) functions as an oncogene in various cancers but has not been fully studied in oral squamous cell carcinoma (OSCC). This study aimed to investigate the expression of ANLN in OSCC tissues and cell lines, to better understand its role in mediating proliferative, angiogenic, invasive, and metastatic capabilities in this type of cancer. METHODS: ANLN mRNA and protein levels were assessed using qPCR and western immunoblotting. The expression intensity of ANLN was evaluated using immunohistochemical (IHC) staining. Biological functional assays were employed to characterize the behavior of OSCC cells influenced by ANLN. Additionally, comprehensive bioinformatics analysis, including GO analysis and KEGG enrichment analysis, was performed on differentially expressed genes in ANLN-mediated pathways. RESULTS: OSCC tumors and cell lines exhibited higher ANLN expression. Silencing of ANLN significantly suppressed OSCC cell proliferation, as evidenced by a significant reduction in the Ki-67 index both in vitro and in vivo. The migration and invasive ability of OSCC cells were markedly diminished, coinciding with a decrease in epithelial-mesenchymal transition activity. ANLN was also found to promote angiogenic activity in OSCC cells, partly through synergistic effects mediated by vascular endothelial growth factor A (VEGFA). Downregulation of ANLN expression led to decreased VEGFA levels, resulting in reduced angiogenesis characterized by fewer vascular branches. CONCLUSIONS: Our findings highlight the promising role of ANLN as a biomarker for both diagnostic and prognostic in OSCC. Targeting ANLN with inhibitory strategies could impede the oncogenesis processes at the core of OSCC development, presenting significant opportunities for advancing therapeutic interventions.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Protéines des microfilaments , Tumeurs de la bouche , Invasion tumorale , Néovascularisation pathologique , Humains , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/génétique , Tumeurs de la bouche/métabolisme , Prolifération cellulaire/génétique , Néovascularisation pathologique/génétique , Néovascularisation pathologique/anatomopathologie , Néovascularisation pathologique/métabolisme , Mouvement cellulaire/génétique , Invasion tumorale/génétique , Protéines des microfilaments/métabolisme , Protéines des microfilaments/génétique , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Carcinome épidermoïde/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Animaux , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/métabolisme , Extinction de l'expression des gènes , Souris , Transition épithélio-mésenchymateuse/génétique , Femelle , Mâle , Angiogenesis
9.
Int J Med Sci ; 21(11): 2170-2188, 2024.
Article de Anglais | MEDLINE | ID: mdl-39239548

RÉSUMÉ

Purpose: Matrix metalloproteinase-11 (MMP11), which belongs to the stromelysin subgroup, has been reported to play a role in the progression of colorectal cancer (CRC). However, the significance of MMP11 in the tumor microenvironment, immune/stromal cells, and its mechanism in CRC remain unclear. Methods: The impact of MMP11 knockdown using specific short hairpin RNAs (shRNAs) on the metastasis and invasion of colorectal cancer RKO and SW480 cells was investigated using western blot, quantitative real-time polymerase chain reaction (qRT-PCR), transwell assays, and immunohistochemistry. Results: MMP11 mRNA expression was significantly higher in CRC cells than in normal cells, and its expression was stimulated in CCD-18Co fibroblasts. Additionally, MMP11 expression was found to be higher in individuals aged ≤ 65 years, the T4/T3 group, and Stage III/IV patients. Overall survival (OS) and disease-free survival rates were significantly different between the high and low MMP11 groups. Furthermore, the receiver operating characteristic (ROC) curves for MMP11 at 1-, 3-, and 5-years were 0.450, 0.552, and 0.560, respectively. Moreover, MMP11 promoted the migration and invasion of CRC cells by elevating the expression of Slug protein. Most importantly, MMP11 was positively associated with M0-macrophages and negatively associated with M1-macrophages, NK cells activated, NK cells resting, T cells CD4 memory activated, and T cells follicular helper, indicating the remarkable interactions of MMP11 with tumor immunology. Conclusions: MMP11 plays an important role in colorectal cancer development, and its mechanism in CRC needs to be further explored in the future.


Sujet(s)
Mouvement cellulaire , Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , Matrix metalloproteinase 11 , Invasion tumorale , Facteurs de transcription de la famille Snail , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/mortalité , Facteurs de transcription de la famille Snail/métabolisme , Facteurs de transcription de la famille Snail/génétique , Matrix metalloproteinase 11/génétique , Matrix metalloproteinase 11/métabolisme , Invasion tumorale/génétique , Mouvement cellulaire/génétique , Mâle , Lignée cellulaire tumorale , Femelle , Adulte d'âge moyen , Sujet âgé , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Survie sans rechute
10.
Tissue Cell ; 90: 102510, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39126833

RÉSUMÉ

The epithelial-mesenchymal transition (EMT) plays a crucial role in lung cancer metastasis, rendering it a promising therapeutic target. Research has shown that non-small cell lung cancer (NSCLC) with p53 mutations exhibits an increased tendency for cancer metastasis. However, the exact contribution of the p53-R273H mutation to tumor metastasis remains uncertain in the current literature. Our study established the H1299-p53-R273H cell model successfully by transfecting the p53-R273H plasmid into H1299 cells. We observed that p53-R273H promotes cell proliferation, migration, invasion, and EMT through CCK-8, wound healing, transwell, western blot and immunofluorescence assays. Notably, the expression of EGR1 was increased in H1299-p53-R273H cells. Knocking out EGR1 in these cells hindered the progression of EMT. ChIP-PCR experiments revealed that p53-R273H binds to the EGR1 promoter sequence, thereby regulating its expression. These findings suggest that p53-R273H triggers EMT by activating EGR1, thereby offering a potential therapeutic approach for lung cancer treatment.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Facteur de transcription EGR-1 , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Tumeurs du poumon , Protéine p53 suppresseur de tumeur , Transition épithélio-mésenchymateuse/génétique , Facteur de transcription EGR-1/métabolisme , Facteur de transcription EGR-1/génétique , Humains , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Lignée cellulaire tumorale , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Mutation , Invasion tumorale/génétique
11.
Eur Thyroid J ; 13(5)2024 10 01.
Article de Anglais | MEDLINE | ID: mdl-39121020

RÉSUMÉ

Objective: This study examined the effect of sirtuin 4 (SIRT4), a NAD+-dependent deacetylase, on the proliferation and progression of papillary thyroid carcinoma (PTC). Methods: Data from The Cancer Genome Atlas (TCGA) were analyzed to identify SIRT4 expression in thyroid cancer. Subsequently, the correlation between SIRT4 expression and clinical characteristics was examined in 205 PTC tissue samples. In vitro assays using three human thyroid cancer cell lines (B-CPAP, TPC-1, and SNU-790) were conducted to assess the effects of regulated SIRT4 expression on cell growth, apoptosis, invasion, and migration. Furthermore, in vivo experiments were performed in a xenograft mouse model. Results: Gene Expression Omnibus (GEO) and TCGA data indicated that SIRT4 expression is lower in thyroid cancer and SIRT4 downregulation is associated with poor overall survival. In PTC tissues, positive SIRT4 expression was associated with decreased extracapsular extension. In in vitro experiments using three human thyroid cancer cell lines, overexpression of SIRT4 decreased cell survival, clonogenic potential, and invasion and migratory capabilities, as well as inducing apoptosis and increasing reactive oxygen species levels. SIRT4 overexpression upregulated E-cadherin and downregulated N-cadherin, suggesting its potential involvement in the regulation of epithelial-mesenchymal transition. These findings were confirmed in vivo using a xenograft mouse model. Conclusion: This study provides novel insight into the potential contribution of SIRT4 to the regulation of the pathological progression of PTC. The data suggest that SIRT4 plays a tumor-suppressive role in PTC by inhibiting growth, survival, and invasive potential. Future research should investigate the molecular mechanisms underlying these effects of SIRT4.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Invasion tumorale , Sirtuines , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Humains , Sirtuines/génétique , Sirtuines/métabolisme , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/métabolisme , Cancer papillaire de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/métabolisme , Animaux , Prolifération cellulaire/génétique , Lignée cellulaire tumorale , Invasion tumorale/génétique , Souris , Mâle , Femelle , Apoptose , Cadhérines/métabolisme , Cadhérines/génétique , Souris nude , Adulte d'âge moyen , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Régulation négative , Protéines mitochondriales
12.
JAMA Dermatol ; 160(9): 964-971, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39141363

RÉSUMÉ

Importance: It is unknown whether germline genetic factors influence in situ melanoma risk differently than invasive melanoma risk. Objective: To determine whether differences in risk of in situ melanoma and invasive melanoma are heritable. Design, Setting, and Participants: Three genome-wide association study meta-analyses were conducted of in situ melanoma vs controls, invasive melanoma vs controls, and in situ vs invasive melanoma (case-case) using 4 population-based genetic cohorts: the UK Biobank, the FinnGen cohort, the QSkin Sun and Health Study, and the Queensland Study of Melanoma: Environmental and Genetic Associations (Q-MEGA). Melanoma status was determined using International Statistical Classification of Diseases and Related Health Problems codes from cancer registry data. Data were collected from 1987 to 2022, and data were analyzed from September 2022 to June 2023. Exposure: In situ and invasive cutaneous melanoma. Main Outcomes and Measures: To test whether in situ and invasive melanoma have independent heritable components, genetic effect estimates were calculated for single-nucleotide variants (SNV; formerly single-nucleotide polymorphisms) throughout the genome for each melanoma. Then, SNV-based heritability was estimated, the genetic correlation between melanoma subtypes was assessed, and polygenic risk scores (PRS) were generated for in situ vs invasive status in Q-MEGA participants. Results: A total of 6 genome-wide significant loci associated with in situ melanoma and 18 loci with invasive melanoma were identified. A strong genetic correlation (genetic r = 0.96; 95% CI, 0.76-1.15) was observed between the 2 classifications. Notably, loci near IRF4, KLF4, and HULC had significantly larger effects for in situ melanoma compared with invasive melanoma, while MC1R had a significantly larger effect on invasive melanoma compared with in situ melanoma. Heritability estimates were consistent for both, with in situ melanoma heritability of 6.7% (95% CI, 4.1-9.3) and invasive melanoma heritability of 4.9% (95% CI, 2.8-7.2). Finally, a PRS, derived from comparing invasive melanoma with in situ melanoma genetic risk, was on average significantly higher in participants with invasive melanoma (odds ratio per 1-SD increase in PRS, 1.43; 95% CI, 1.16-1.77). Conclusions and Relevance: There is much shared genetic architecture between in situ melanoma and invasive melanoma. Despite indistinguishable heritability estimates between the melanoma classifications, PRS suggest germline genetics may influence whether a person gets in situ melanoma or invasive melanoma. PRS could potentially help stratify populations based on invasive melanoma risk, informing future screening programs without exacerbating the current burden of melanoma overdiagnosis.


Sujet(s)
Prédisposition génétique à une maladie , Étude d'association pangénomique , Mélanome , Invasion tumorale , Tumeurs cutanées , Humains , Mutation germinale , Mélanome/génétique , Mélanome/anatomopathologie , Melanoma, Cutaneous Malignant , Invasion tumorale/génétique , Invasion tumorale/anatomopathologie , Polymorphisme de nucléotide simple , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie
13.
Hum Cell ; 37(6): 1734-1741, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39174825

RÉSUMÉ

Synovial sarcoma (SS) is identified as a sarcoma with monomorphic blue spindle cells that display variable epithelial differentiation and is characterized by the SS18::SSX fusion gene. SS accounts for approximately 5-10% of all soft tissue sarcomas, making it a relatively common type within this group of tumors. Since SS is generally sensitive to chemotherapy, the standard treatment for SS includes extensive surgical resection, complemented by neoadjuvant chemotherapy with several approved anticancer drugs. However, in advanced and metastatic cases, the efficacy of these drugs is limited, resulting in poor prognoses. This underscores the need for innovative therapeutic strategies. Patient-derived cancer cell lines are essential tools for basic and preclinical research, yet only four SS cell lines are publicly available. To facilitate the studies of SS, we have developed a novel SS cell line, named NCC-SS6-C1, derived from surgically excised tumor tissue of an SS patient. NCC-SS6-C1 cells preserve the SS18::SSX1 fusion gene, consistent with the genetic characteristics of the original tumor. The cells exhibit continuous proliferation, invasiveness, and the ability to form spheroids. Additionally, we confirmed that this cell line was useful for evaluating the efficacy of anticancer drugs. Our results suggest that NCC-SS6-C1 is a useful tool for basic and pre-clinical studies of SS.


Sujet(s)
Sarcome synovial , Humains , Sarcome synovial/génétique , Sarcome synovial/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Protéines de fusion oncogènes/génétique , Antinéoplasiques/pharmacologie , Invasion tumorale/génétique , Protéines proto-oncogènes , Protéines de répression
14.
Hum Cell ; 37(6): 1719-1733, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39181971

RÉSUMÉ

Uveal melanoma is a malignant tumor originating from melanocytes in the eye's uvea, often detected during routine ophthalmic examinations due to its typically asymptomatic nature. Despite effective local treatments, up to 50% of patients develop hematogenous metastases, highlighting the need for better prognostic markers and therapeutic targets. In this study, we developed an innovative Metastasis-Related Gene Signature (MERGS) score to classify patients from various cohorts. By establishing this scoring method, we discovered underlying mechanisms responsible for significant differences between samples with high and low MERGS scores. We identified a set of ten genes to construct MERGS, which showed a high predictive accuracy for patient survival. Further, Monoglyceride Lipase (MGLL) emerged as the most important gene in distinguishing uveal melanoma metastasis. Functional studies demonstrated that knocking down MGLL significantly inhibited proliferation, invasion, and migration of uveal melanoma cells in vitro and in vivo, while overexpression of MGLL enhanced these malignant behaviors. Additionally, MGLL modulated free fatty acid (FFA) levels within these cells. Our findings reveal MGLL as a crucial player in uveal melanoma progression and propose it as a novel therapeutic target, potentially leading to improved management and outcomes for patients with this disease.


Sujet(s)
Prolifération cellulaire , Mélanome , Acylglycerol lipase , Métastase tumorale , Tumeurs de l'uvée , Tumeurs de l'uvée/génétique , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/métabolisme , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/métabolisme , Humains , Prolifération cellulaire/génétique , Acylglycerol lipase/génétique , Acylglycerol lipase/métabolisme , Métastase tumorale/génétique , Acide gras libre/métabolisme , Lignée cellulaire tumorale , Expression des gènes/génétique , Mouvement cellulaire/génétique , Invasion tumorale/génétique
15.
J Neurooncol ; 170(1): 161-171, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39117967

RÉSUMÉ

PURPOSE: This study investigated the effect of an isocitrate dehydrogenase 1 (IDH1) mutation (mutIDH1) on the invasion and angiogenesis of human glioma cells. METHODS: Doxycycline was used to induce the expression of mutIDH1 in glioma cells. Transwell and wound healing assays were conducted to assess glioma cell migration and invasion. Western blotting and cell immunofluorescence were used to measure the expression levels of various proteins. The influence of bone morphogenetic protein 2 (BMP2) on invasion, angiogenesis-related factors, BMP2-related receptor expression, and changes in Smad signaling pathway-related proteins were evaluated after treatment with BMP2. Differential gene expression and reference transcription analysis were performed. RESULTS: Successful infection with recombinant lentivirus expressing mutIDH1 was demonstrated. The IDH1 mutation promoted glioma cell migration and invasion while positively regulating the expression of vascularization-related factors and BMP2-related receptors. BMP2 exhibited a positive regulatory effect on the migration, invasion, and angiogenesis of mutIDH1-glioma cells, possibly mediated by BMP2-induced alterations in Smad signaling pathway-related factors.After BMP2 treatment, the differential genes of MutIDH1-glioma cells are closely related to the regulation of cell migration and cell adhesion, especially the regulation of Smad-related proteins. KEGG analysis confirmed that it was related to BMP signaling pathway and TGF-ß signaling pathway and cell adhesion. Enrichment analysis of gene ontology and genome encyclopedia further confirmed the correlation of these pathways. CONCLUSION: Mutation of isocitrate dehydrogenase 1 promotes the migration, invasion, and angiogenesis of glioma cells, through its effects on the BMP2-driven Smad signaling pathway. In addition, BMP2 altered the transcriptional patterns of mutIDH1 glioma cells, enriching different gene loci in pathways associated with invasion, migration, and angiogenesis.


Sujet(s)
Protéine morphogénétique osseuse de type 2 , Tumeurs du cerveau , Mouvement cellulaire , Gliome , Isocitrate dehydrogenases , Mutation , Invasion tumorale , Néovascularisation pathologique , Isocitrate dehydrogenases/génétique , Isocitrate dehydrogenases/métabolisme , Humains , Gliome/génétique , Gliome/métabolisme , Gliome/anatomopathologie , Protéine morphogénétique osseuse de type 2/métabolisme , Protéine morphogénétique osseuse de type 2/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Invasion tumorale/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/métabolisme , Transduction du signal , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéines Smad/métabolisme , Protéines Smad/génétique , Angiogenesis
16.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 85-91, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097891

RÉSUMÉ

Prostate cancer (PCa) belongs to a prevailing neoplasm globally. Circular RNAs (circRNAs) are critical regulators in various tumors, but the role of circRNAs in PCa is obscure. In this research, a circRNA derived from the TADA2A gene (hsa_circ_0006220) was high-expressed in PCa tissues along with cell lines. Elevated Circ-0006220 expression was also related to PCa poor prognosis. Besides, circ-0006220 accelerated PCa cells malignant behaviors in vitro; it also promoted PCa tumor growth together with metastasis in vivo. Moreover, circ-0006220 competed with the Cell Division Cycle Associated 7 (CDCA7) for binding to miR-520f-3p. Circ-0006220 sponged miR-520f-3p to regulate CDCA7 expression, thereby promoting PCa cell proliferation, migration, invasion, along with metastasis. All above data suggested that circ-0006220 may be a worthy target for PCa therapeutics.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , microARN , Tumeurs de la prostate , ARN circulaire , Humains , Mâle , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , microARN/génétique , microARN/métabolisme , Invasion tumorale/génétique , Métastase tumorale , Pronostic , Tumeurs de la prostate/génétique , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , ARN circulaire/génétique , ARN circulaire/métabolisme
17.
Pol J Pathol ; 75(2): 157-165, 2024.
Article de Anglais | MEDLINE | ID: mdl-39166524

RÉSUMÉ

Long non-coding RNA LINC00908 is a functional biomarker in regulating tumour progression. Its dysregulation in gastric cancer implies its potential functional role. Few studies have noted the functional role of LINC00908 in gastric cancer. The potential of LINC00908 to serve as a biomarker in gastric cancer was evaluated. A total of 113 paired gastric cancer tissues and normal tissues were collected from patients with gastric cancer. LINC00908 levels were evaluated by polymerase chain reaction, and its significance in disease progression and patients' prognosis was assessed. In vitro, the function of LINC00908 in tumour-related cellular processes was evaluated with CCK8 and Transwell assay. Significant downregulation of LINC00908 was observed in gastric cancer and was negatively associated with disease development and overall survival of patients. LINC00908 showed significant inhibitory effects on the proliferation, migration, and invasion of gastric cancer cells. Additionally, miR-627-3p was sponged by LINC00908 and therefore mediated the function of LINC00908 in gastric cancer cells. LINC00908 functioned as a prognostic biomarker and tumour suppressor of gastric cancer, providing a therapeutic target for gastric cancer.


Sujet(s)
Marqueurs biologiques tumoraux , Prolifération cellulaire , Régulation négative , Régulation de l'expression des gènes tumoraux , ARN long non codant , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , ARN long non codant/génétique , Femelle , Mâle , Pronostic , Adulte d'âge moyen , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire/génétique , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Sujet âgé , microARN/génétique , microARN/métabolisme , Invasion tumorale/génétique
18.
BMC Gastroenterol ; 24(1): 285, 2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39179979

RÉSUMÉ

Microsatellite instability (MSI) has been widely acknowledged as an important factor regulating tumor intrinsic biological behavior and affecting the survival of gastric cancer patients. Here, we firstly identified the RARB as a gene associated with MSI gastric cancer. RARB was downregulated in human gastric cancer tissues compared to paired paracancerous tissues, Knockdown of RARB accelerated the proliferation, invasion and migration of cancer cells in vitro. Mechanismly, RARB knockdown promoted epithelial-mesenchymal transition (EMT) process of gastric cancer. However, RARBLow patients exhibited better survival compared to RARBHigh patients. Further study revealed that RARB expression was inversely correlated with MSI status and immune infiltrates in vivo. Thus, RARB may be a potential target for the treatment of gastric cancer.


Sujet(s)
Prolifération cellulaire , Évolution de la maladie , Transition épithélio-mésenchymateuse , Instabilité des microsatellites , Récepteurs à l'acide rétinoïque , Tumeurs de l'estomac , Femelle , Humains , Mâle , Adulte d'âge moyen , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation négative , Transition épithélio-mésenchymateuse/génétique , Techniques de knock-down de gènes , Invasion tumorale/génétique , Pronostic , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/mortalité , Récepteurs à l'acide rétinoïque/génétique , Récepteurs à l'acide rétinoïque/métabolisme
19.
Gen Physiol Biophys ; 43(5): 445-455, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39140681

RÉSUMÉ

This study aims to investigate the impacts of SLC12A8 on the invasion, migration, and epithelial-mesenchymal transition (EMT) of non-small cell lung cancer (NSCLC) cells. GEPIA database was employed to examine SLC12A8 expression pattern in lung cancer cells. Subsequently, qRT-PCR and Western blot analyses were conducted to assess SLC12A8 expression in NSCLC tissues and cell lines. The overall prognosis of NSCLC patients was evaluated using Kaplan-Meier plot and univariate and multivariate COX regression curves. The knockdown of SLC12A8 was established using lentivirus-mediated shRNA in A549 and H1299 cells. Cell proliferation, invasion, migration, and apoptosis were evaluated using CCK-8 assay, transwell, and flow cytometry techniques, respectively. Western blot analysis was performed to measure the expression levels of EMT-related proteins (E-cadherin and vimentin). The expression level of SLC12A8 was found to be significantly higher in both NSCLC cell lines and tissues. High SLC12A8 expression was correlated with a poor prognosis in NSCLC patients. Knocking down SLC12A8 led to a significant decrease in proliferation, migration, and invasion abilities, while promoting apoptosis in NSCLC cells. Additionally, SLC12A8 knockdown resulted in decreased levels of N-cadherin and vimentin, along with increased E-cadherin expression. The results indicate that reducing SLC12A8 expression may suppress the malignant phenotype of NSCLC cells, as well as the EMT. SLC12A8 may serve as a target for the clinical management of NSCLC progression.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Mouvement cellulaire , Tumeurs du poumon , Invasion tumorale , Humains , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Mouvement cellulaire/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Invasion tumorale/génétique , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/génétique , Prolifération cellulaire/génétique , Apoptose/génétique , Cellules A549
20.
Pediatr Surg Int ; 40(1): 221, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-39133317

RÉSUMÉ

PURPOSE: The aim of this study was to detect candidate oncogenes of rhabdoid tumor of the kidney (RTK) and evaluate their roles in RTK in vitro. METHODS: An integrated analysis of messenger RNA (mRNA) and microRNA (miRNA) sequencing was performed to determine the expression profile of exosome-derived miRNAs and mRNAs in human RTK-derived cell lines and a human embryonic renal cell line. A Gene Ontology enrichment analysis was performed to analyze the functional characteristics of differentially expressed mRNAs in RTK cells. Matrigel invasion and wound-healing assays were performed to evaluate the cell invasion and migration abilities. RESULTS: Forty mRNAs were highly expressed in RTK cells targeted by exosomal miRNAs, the expression of which was lower in RTK cells than in the controls. These mRNAs were primarily related to cell adhesion. Of these mRNAs, we selected neuropilin 1 (NRP1) as a candidate oncogene because its upregulated expression is associated with a poor prognosis of several types of tumors. RTK cells in which NRP1 had been knocked down exhibited decreased invasive and migratory abilities. CONCLUSION: Our study indicates that NRP1 acts as an oncogene by promoting the invasion and migration of RTK cells and that it could serve as a therapeutic target.


Sujet(s)
Mouvement cellulaire , Tumeurs du rein , Invasion tumorale , Neuropiline 1 , Tumeur rhabdoïde , Humains , Neuropiline 1/génétique , Neuropiline 1/métabolisme , Mouvement cellulaire/génétique , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Invasion tumorale/génétique , Tumeur rhabdoïde/génétique , Tumeur rhabdoïde/anatomopathologie , Lignée cellulaire tumorale , ARN messager/génétique , ARN messager/métabolisme , Régulation de l'expression des gènes tumoraux , microARN/génétique , Techniques de knock-down de gènes/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE