Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 525
Filtrer
1.
Heliyon ; 10(16): e36235, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39247316

RÉSUMÉ

Background: Clear cell renal cell carcinoma (ccRCC) is a highly aggressive cancer associated with higher death rates. However, traditional anti-angiogenic therapies have limited effectiveness due to drug resistance. Vascular mimicry (VM) provides a different way for tumors to develop blood vessels without relying on endothelial cells or angiogenesis. However, the intricate mechanisms and interplay between it and the immune microenvironment in ccRCC remain unclear. Methods: A PubMed and GeneCards literature review was conducted to identify VM-related genes (VMRGs). VMRGs expression profiles were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), developing a novel VM risk score model and nomogram for ccRCC. The EBI ArrayExpress database (the validation set) was obtained to validate the prognostic model. The relationship between VMRGs risk score clinical characteristics and immune infiltration was investigated. Finally, the expression of six model VMRGs was validated using single-cell analysis, GEPIA, Human Protein Atlas (HPA), and quantitative Real-time PCR (qRT-PCR). Results: Cox regression analysis and nomogram identified L1CAM, TEK, CLDN4, EFNA1, SERPINF1, and MALAT1 as independent prognostic risk factors, which could be used to stratify the ccRCC population into two risk groups with distinct immune profiles and responsiveness to immunotherapy. The results of single-cell analysis, GEPIA, HPA, and qRT-PCR validated the model genes' expression. Conclusions: Our novel findings constructed a convenient and reliable 6 gene signatures as potential immunologic and prognostic biomarkers of VM in ccRCC.

2.
Cell Mol Biol Lett ; 29(1): 118, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39237880

RÉSUMÉ

BACKGROUND: Vasculogenic mimicry (VM) is a potential cause of resistance to antiangiogenic therapy and is closely related to the malignant progression of tumors. It has been shown that noncoding RNAs play an important role in the formation of VM in malignant tumors. However, the role of circRNAs in VM of bladder cancer and the regulatory mechanisms are unclear. METHODS: Firstly, hsa_circ_0000520 was identified to have circular character by Sanger sequencing and Rnase R assays. Secondly, the potential clinical value of hsa_circ_0000520 was explored by quantitative real-time polymerase chain reaction (qRT-PCR) and fluorescence in situ hybridization (FISH) of clinical specimens. Thirdly, the role of hsa_circ_0000520 in bladder cancer invasion, migration, and VM formation was examined by in vivo and in vitro experiments. Finally, the regulatory mechanisms of hsa_circ_0000520 in the malignant progression of bladder cancer were elucidated by RNA binding protein immunoprecipitation (RIP), RNA pulldown, co-immunoprecipitation (co-IP), qRT-PCR, Western blot (WB), and fluorescence co-localization. RESULTS: Hsa_circ_0000520 was characterized as a circular RNA and was lowly expressed in bladder cancer compared with the paracancer. Bladder cancer patients with high expression of hsa_circ_0000520 had better survival prognosis. Functionally, hsa_circ_0000520 inhibited bladder cancer invasion, migration, and VM formation. Mechanistically, hsa_circ_0000520 acted as a scaffold to promote binding of UBE2V1/UBC13 to Lin28a, further promoting the ubiquitous degradation of Lin28a, improving PTEN mRNA stability, and inhibiting the phosphorylation of the PI3K/AKT pathway. The formation of hsa_circ_0000520 in bladder cancer was regulated by RNA binding protein QKI. CONCLUSIONS: Hsa_circ_0000520 inhibits metastasis and VM formation in bladder cancer and is a potential target for bladder cancer diagnosis and treatment.


Sujet(s)
Mouvement cellulaire , Phosphohydrolase PTEN , Phosphatidylinositol 3-kinases , ARN circulaire , Protéines de liaison à l'ARN , Transduction du signal , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Humains , ARN circulaire/génétique , ARN circulaire/métabolisme , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Transduction du signal/génétique , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Lignée cellulaire tumorale , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Mouvement cellulaire/génétique , Mâle , Animaux , Régulation de l'expression des gènes tumoraux , Métastase tumorale , Femelle , Néovascularisation pathologique/génétique , Souris nude , Souris , Adulte d'âge moyen , Souris de lignée BALB C
3.
Am J Pathol ; 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39233275

RÉSUMÉ

Traditional anti-angiogenesis drugs are usually unsatisfactory in hepatocellular carcinoma (HCC) treatment. Therefore, it is urgent to find new precise therapeutic targets and further develop more effective drugs for the treatment of HCC. Vasculogenic mimicry (VM) is different from classical endothelium-dependent angiogenesis and associated with poor prognosis in patients with malignant tumor. However, the mechanism underlying VM occurrence is complex and not fully defined. Immunoglobulin-like transcript (ILT) 4, as a negative regulator of immune response, was recently found expressed in many solid tumors. However, whether and how ILT4 regulate VM remains unclear. In this study, we found VM enriched in HCC tissues especially in tissues from patients who experience relapse within 5 years after surgery. Similarly, ILT4 expression level was also higher in HCC tissues from patients who experience relapse within 5 years after surgery. Linear regression analysis revealed a positive correlation between the expression of ILT4 and VM density. Further, Overexpression/knockdown of ILT4 expression upregulated/downregulated VM related marker, three-dimensional tube formation, the migration and invasion of HCC cell lines in vitro. Mechanistic studies showed that ILT4 promoted VM formation via MAPK/ERK signaling. In conclusion, this study provides a rationale and mechanism for ILT4-mediated postoperative relapse via inducing VM in HCC. The related molecular pathways can be used as novel therapeutic targets for the inhibition of HCC angiogenesis and postoperative relapse.

4.
Biochem Pharmacol ; 227: 116458, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39102993

RÉSUMÉ

Vasculogenic mimicry (VM) serves as a vascular-like channel that provides important substances for tumor growth and is a primary factor in glioblastoma (GBM) drug resistance. Human Antigen R (HuR)-an mRNA-binding protein-is highly expressed in GBM, closely related to tumor progression, and deemed a potential drug target. Although some small-molecule compounds have been identified to disrupt HuR binding to target mRNA, they remain in the preclinical research stage, suggesting the need for further validation and development of HuR inhibitors. In our study, we aim to screen for potential HuR inhibitors and investigate their efficacy and molecular mechanisms in GBM. We employed the fluorescence polarization method to identify HuR inhibitors from a natural compound library, confirming the efficacy of juglone in effectively inhibiting the binding of HuR to AREVegf-a. Further validation of the binding of juglone to HuR at the protein level was conducted through electrophoretic mobility shift analysis, surface plasmon resonance, and molecular docking. Furthermore, juglone demonstrated inhibitory effects on glioma growth and VM formation in vitro and in vivo. Moreover, it was observed that juglone reversed epithelial-mesenchymal transition by inhibiting the VEGF-A/VEGFR2/AKT/SNAIL signaling pathway. Finally, we established the capability of juglone to target HuR in U251 cells through HuR knockdown, mRNA stability, and cell thermal shift assays. Therefore, this study identifies juglone as a novel HuR inhibitor, potentially offering promise as a lead compound for anti-VM therapy in GBM by targeting HuR. Abbreviations: AKT, protein kinase B; ARE, adenine-and uridine-rich elements; CETSA, cellular thermal shift assay; DMEM, Dulbecco's modified Eagle's medium; ELISA, enzyme linked immune sorbent assay; EMSA, electrophoretic mobility shift assay; EMT, epithelial mesenchymal transition; FP, fluorescence polarization; GBM, glioblastoma; HTS, high-throughput screening; HuR, human antigen R; IF, Immunofluorescence; PAS, periodic acid-Schiff; PI3K, phosphoinositide-3 kinase; qRT-PCR, quantitative real-time PCR; RRMs, RNA recognition motifs; SPR, surface plasmon resonance. TMZ, temozolomide; VM, vasculogenic mimicry; VEGF-A, Vascular endothelial growth factor-A; VEGFR2, Vascular endothelial growth factor receptor-2.


Sujet(s)
Protéine-1 similaire à ELAV , Naphtoquinones , Facteur de croissance endothéliale vasculaire de type A , Humains , Protéine-1 similaire à ELAV/métabolisme , Protéine-1 similaire à ELAV/génétique , Protéine-1 similaire à ELAV/antagonistes et inhibiteurs , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs , Naphtoquinones/pharmacologie , Animaux , Souris , Lignée cellulaire tumorale , Souris nude , Gliome/métabolisme , Gliome/traitement médicamenteux , Gliome/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Souris de lignée BALB C , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
5.
Exp Ther Med ; 28(4): 373, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39091630

RÉSUMÉ

Frizzled family protein 2 (FZD2) is widely associated with tumor development and metastasis. The present study aimed to gain an insight into the role and regulatory mechanism of FZD2 in glioma. The expression level of FZD2 in normal astrocyte and glioma cells was determined by reverse transcription-quantitative PCR and western blotting, and cell transfection was conducted for FZD2 expression knockdown. Malignant behaviors including cell proliferation, migration and invasion, vasculogenic mimicry (VM) and cell stemness were determined using Cell Counting Kit-8, 5-Ethynyl-2'-deoxyuridine (EdU) staining, colony formation, wound healing, Transwell, 3D culturing and sphere formation assays. The expression levels of proteins related to stemness, epithelial-mesenchymal transition (EMT) and Notch/NF-κB signaling were measured by western blotting. Then, the Notch agonist, Jagged-1 (JAG), was adopted for rescue experiments. The results demonstrated that FZD2 was highly expressed in glioma cells. Interference of FZD2 expression suppressed the proliferation of glioma cells, as evidenced by the reduced cell viability and the number of EdU+ cells and colonies. Meanwhile, the reduced sphere formation ability and decreased protein expression of Nanog, Sox2 and Oct4 following FZD2 knockdown confirmed that FZD2 repressed cell stemness in glioma. Additionally, FZD2 knockdown suppressed the migration, invasion, EMT and VM formation capabilities of glioma cells, and also blocked the Notch/NF-κB signaling pathway. Furthermore, activation of Notch by JAG treatment partially reversed the aforementioned FZD2 knockdown-mediated changes in glioma cell malignant behaviors. In conclusion, FZD2 may contribute to glioma progression through activating the Notch/NF-κB signaling pathway, providing a plausible therapeutic target for the treatment of glioma.

6.
J Gastroenterol ; 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097533

RÉSUMÉ

BACKGROUND: Hypoxic microenvironment is a common feature of most solid tumors including hepatocellular carcinoma (HCC). Vasculogenic mimicry (VM) formation by tumor cells could provide blood supply to tumor cells under hypoxia. NFE2 like basic leucine zipper (bZIP) transcription factor 2 (Nrf2), a regulator of cellular homeostasis, may promote tumor progression in the hypoxic conditions. However, the role and regulatory mechanisms of Nrf2 in HCC are not fully elucidated. METHODS: Nrf2 and assembly factor for spindle microtubules (ASPM) expression modulations were conducted by lentiviral transfections. Western blot, immunofluorescence, ChIP-qPCR, dual-luciferase reporter gene assay, flow cytometry, RNA sequencing, multiple bioinformatics databases analysis, cell function assays in vitro, mouse model in vivo and human HCC tissues were employed to assess the effect of Nrf2/ASPM axis on HCC progression under hypoxia. RESULTS: Nrf2 and ASPM expression facilitated epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs) feature, and VM formation of HCC cells under hypoxia. Furthermore, Nrf2-regulated ASPM expression, via binding directly to the promoter region of ASPM and transcriptionally promoting ASPM expression. ASPM re-expression in Nrf2 knockdown cells or ASPM knockdown in Nrf2 overexpression cells reversed the cellular function caused by Nrf2. Meantime, retinol metabolism pathway was disrupted following abnormal ASPM expression. Nrf2/ASPM axis in murine models accelerated tumor growth and VM, corroborating in vitro findings. All-trans retinoic acid treatment reversed stemness and VM of HCC cells in vitro and in vivo. Clinically, Nrf2 and ASPM expressions were related to poor prognosis of HCC patients. CONCLUSIONS: Nrf2 drives EMT, CSCs characteristics and VM in HCC under hypoxia through the modulation of ASPM. Retinol metabolism pathway was dysregulated in HCC cells with ASPM overexpression. Nrf2/ASPM axis and related pathway provided potential therapeutic target for HCC.

7.
Biomed Environ Sci ; 37(8): 834-849, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39198249

RÉSUMÉ

Objective: Vasculogenic mimicry (VM) is a novel vasculogenic process integral to glioma stem cells (GSCs) in glioblastoma (GBM). However, the relationship between VM and ataxia-telangiectasia mutated (ATM) serine/threonine kinase activation, which confers chemoradiotherapy resistance, remains unclear. Methods: We investigated VM formation and phosphorylated ATM (pATM) levels by CD31/GFAP-periodic acid-Schiff dual staining and immunohistochemical staining in 145 GBM specimens. Glioma stem-like cells (GSLCs) derived from the formatted spheres of U87 and U251 cell lines and their pATM level and VM formation ability were examined using western blot and three-dimensional culture. For the examination of the function of pATM in VM formation by GSLCs, ATM knockdown by shRNAs and deactivated via ATM phosphorylation inhibitor KU55933 were studied. Results: VM and high pATM expression occurred in 38.5% and 41.8% of tumors, respectively, and were significantly associated with reduced progression-free and overall survival. Patients with VM-positive GBMs exhibited higher pATM levels ( r s = 0.425, P = 0.01). The multivariate analysis established VM as an independent negative prognostic factor ( P = 0.002). Furthermore, GSLCs expressed high levels of pATM and formed vascular-like networks in vitro. ATM inactivation or knockdown hindered VM-like network formation concomitant with the downregulation of pVEGFR-2, VE-cadherin, and laminin B2. Conclusion: VM may predict a poor GBM prognosis and is associated with pATM expression. We propose that pATM promotes VM through extracellular matrix modulation and VE-Cadherin / pVEGFR-2 activation, thereby highlighting ATM activation as a potential target for enhancing anti-angiogenesis therapies for GBM.


Sujet(s)
Protéines mutées dans l'ataxie-télangiectasie , Gliome , Cellules souches tumorales , Humains , Protéines mutées dans l'ataxie-télangiectasie/métabolisme , Protéines mutées dans l'ataxie-télangiectasie/génétique , Cellules souches tumorales/métabolisme , Gliome/métabolisme , Gliome/anatomopathologie , Gliome/vascularisation , Lignée cellulaire tumorale , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/vascularisation , Néovascularisation pathologique/métabolisme , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/vascularisation , Adulte , Sujet âgé
8.
Phytomedicine ; 134: 155962, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39214017

RÉSUMÉ

Vasculogenic mimicry (VM) is a novel model for supplying blood to multiple tumors, including gastric cancer (GC), and is a potential target for its treatment. Dihydroartemisinin (DHA) is a potential natural antitumor substance that inhibits the progression of tumors in many ways. The research aimed to evaluate the impact of DHA on VM formation and its mechanisms. The IC50 of DHA, DHA's effect on proliferation, invasion, and migration in GC cells and VM formation in both cell and animal models were determined through wound healing, MTT, EdU, colony formation, and Transwell assays. Genomics was employed to identify genes related to DHA inhibition of VM formation, and to analyze their relationship to VM formation. qRT‒PCR and western blot (WB) analysis were carried out to analyze the changes in protein and mRNA levels after DHA treatment and the changes in VM-associated protein biomarkers after blocking target gene-related pathways. The mechanism by which DHA inhibits VM in GC was elucidated in vivo. DHA reduced the invasion, proliferation, and migration of GC cells and inhibited VM in cells and in vivo. A total of 220 DEGs were identified in the DHA-treated HGC-27 cells. Among the 146 downregulated genes, fibroblast growth Factor 2 (FGF2) was most closely associated with angiogenesis and VM. The level of FGF2 in GC tissues with VM was markedly greater than in VM lacking tissues. Treatment with DHA or FGFR1 blockade suppressed VM formation and reduced VM-related biomarker proteins. DHA suppressed tumor progression and VM formation by reducing FGF2 in xenograft mouse models. Per our knowledge, this is the first study to demonstrate the inhibitory effect of DHA on VM, providing a novel strategy for the treatment of GC.

9.
Bioeng Transl Med ; 9(4): e10648, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39036079

RÉSUMÉ

Vasculogenic mimicry (VM) describes a process by which tumor cells formed a novel microcirculation pattern in an endothelial cell-free manner. Clinically, VM is associated with aggressive phenotype and poor patient survival. However, the current models for investigating VM include 2D monolayer cultures, Matrigel-based cultures, and animal models, each of which has limitations. Matrigel-based models often exhibit batch-to-batch variations, while in vivo tumor models currently produce insufficient amounts of VM. There is currently no suitable tumor model to discover new therapeutic targets against VM. Herein, we establish an extracellular matrix (ECM)-based engineered tumor model in vivo and in vitro. In this study, we demonstrate that matrix proteins enhanced the VM formation in the engineered xenograft model. Furthermore, we also investigated the role of collagen/fibronectin (FN) in melanoma progression and VM formation. Compared with cells cultured on TCPS plates, the B16F10 cells cultured on collagen/FN coated plates showed increased proliferation and stemness, and significantly enhanced invasion and formation of VM networks. Molecular mechanism analysis showed that Integrin/VE-cadherin/EphA2/PI3K/MMP-2 signaling pathways are responsible for VM formation. Our results indicate that collagen/FN matrix plays an important role in VM formation in melanoma, suggesting that ECM protein is a potential therapeutic target for anti-VM therapy for melanoma.

10.
Arch Dermatol Res ; 316(7): 447, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38958761

RÉSUMÉ

Malignant melanoma presents a formidable challenge due to its aggressive metastatic behavior and limited response to current treatments. To address this, our study delves into the impact of anlotinib on angiogenesis and vasculogenic mimicry using malignant melanoma cells and human umbilical vein endothelial cells. Evaluating tubular structure formation, cell proliferation, migration, invasion, and key signaling molecules in angiogenesis, we demonstrated that anlotinib exerts a dose-dependent inhibition on tubular structures and effectively suppresses cell growth and invasion in both cell types. Furthermore, in a mouse xenograft model, anlotinib treatment resulted in reduced tumor growth and vascular density. Notably, the downregulation of VEGFR-2, FGFR-1, PDGFR-ß, and PI3K underscored the multitargeted antitumor activity of anlotinib. Our findings emphasize the therapeutic potential of anlotinib in targeting angiogenesis and vasculogenic mimicry, contributing to the development of novel strategies for combating malignant melanoma.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Cellules endothéliales de la veine ombilicale humaine , Indoles , Mélanome , Néovascularisation pathologique , Quinoléines , Récepteur-2 au facteur croissance endothéliale vasculaire , Tests d'activité antitumorale sur modèle de xénogreffe , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Quinoléines/administration et posologie , Humains , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Animaux , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/anatomopathologie , Indoles/pharmacologie , Indoles/usage thérapeutique , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Mouvement cellulaire/effets des médicaments et des substances chimiques , Récepteur FGFR1/métabolisme , Récepteur FGFR1/antagonistes et inhibiteurs , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/administration et posologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Récepteur au PDGF bêta/métabolisme , Récepteur au PDGF bêta/antagonistes et inhibiteurs , Souris nude ,
11.
Acta Histochem ; 126(5-7): 152174, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38976933

RÉSUMÉ

Choroidal melanoma (CM), a highly metastatic eye tumor, exhibits vasculogenic mimicry (VM) facilitated by hypoxia-induced angiogenesis. This study explored the inhibitory impact of the anti-malarial drug Artesunate (ART) on CM VM through modulation of the HIF-1α/VEGF/PDGF pathway. Immunohistochemistry (IHC) confirmed VM in CM with elevated VEGF and PDGF expression. Hypoxia promoted CM proliferation, upregulating HIF-1α, VEGF and PDGF. VEGF and PDGF enhanced CM migration, invasion and VM, with HIF-1α playing a crucial role. ART mitigated VM formation by suppressing the HIF-1α/VEGF/PDGF pathway, highlighting its potential as an anti-tumor agent in CM.

12.
Front Pharmacol ; 15: 1421470, 2024.
Article de Anglais | MEDLINE | ID: mdl-39050762

RÉSUMÉ

Background: Vasculogenic Mimicry (VM) can reduce the efficacy of anti-angiogenesis and promote distant metastasis in hepatocellular carcinoma (HCC). Our previous studies have found that Celastrus orbiculatus extract (COE) can inhibit the VM formation in HCC by reducing EphA2 expression. However the underlying mechanism related to EphA2 in VM formation is unclear. Purpose: This study aimed to confirm that EphA2 is one of the potential targets of COE, and to explore the effect of EphA2 in VM formation in hypoxia context in HCC. Methods: TCM Systems Pharmacology database and proteomics analysis were used to explore the key targets of COE in HCC treatment. CD31-PAS double staining and VE-CAD staining were used to indicate vasculogenic mimicry. The localization of EphA2 and VE-CAD was examined through fluorescent microscopy. CCK8 assay, cell invasion assay, and tube formation assay were used to indicate the formation of VM under hypoxic conditions. The regulatory relationship of EphA2 upstream and downstream molecules were evaluated through COIP and Western Blot. The nude mouse xenograft tumor models were used to observe the VM formation after knocking down or overexpressing EphA2. Results: EphA2 is identified to the target of COE, and the driving gene of HCC. In HCC surgical specimens, EphA2 expression is closely associated with the VM formation of HCC. COE-regulated EphA2 is involved in hypoxia-induced VM formation in HCC cells in vitro. EphA2 is regulated by HIF directly or indirectly by C-MYC. Overexpression of EphA2 can promote the VM formation of HCC in nude mice, while knocking down EphA2 can inhibit the VM formation. Conclusion: EphA2, as a target of COE, plays a crucial regulatory role in the formation of vasculogenic mimicry in HCC, involving upstream HIF/MYC transcriptional promotion and downstream PI3K/FAK/VE-CAD expression regulation.

13.
Heliyon ; 10(11): e32111, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38947483

RÉSUMÉ

Objective: To deepen the imaging-pathological mechanism of primary central nervous system lymphoma (PCNSL) and provide a theoretical basis for clinical diagnosis and treatment, the functional magnetic resonance imaging (fMRI) characteristics of PCNSL were analyzed, and the relationship between the fMRI characteristics and vasculogenic mimicry (VM) and reticular fiber in PCNSL was discussed. Methods: Ninety-six patients with PCNSL treated in our hospital were divided into three groups according to the pathological examination results, including strong positive group of VM (n = 40), weak positive group of VM (n = 56), strong positive group of reticular fiber (n = 45) and weak positive group of reticular fiber (n = 51). The levels of augmentation index and apparent diffusion coefficient (ADC) were compared among the groups. receiver operator characteristic (ROC) curve analysis was used to analyze the clinical value of ADC value in differential diagnosis of PCNSL. Results: The levels of augmentation index in the strong positive group of VM were significantly higher than that in the weak positive group of VM, and the ADC value in the strong positive group of VM was significantly lower than that in the weak positive group of VM (P < 0.001). The levels of augmentation index in the strong positive group of reticular fiber were significantly higher than that in the weak positive group of reticular fiber, and ADC value in the strong positive group of reticular fiber was significantly lower than that in reticular fiber weak positive group (P < 0.001). Pearson correlation analysis showed that the levels of augmentation index were positively correlated with VM and reticular fiber (r = 0.529, 0.548, P < 0.001) and the ADC value was negatively correlated with VM and reticular fiber (r = -0.485, -0.513, P < 0.001). There was a significant negative correlation between necrotic lesions and VM (r = -0.185, P < 0.05). The area under the curve (AUC) values of average ADC value, minimum ADC value, and maximum ADC value for individual differential diagnosis of PCNSL were 0.920, 0.901, and 0.702, while the AUC of the combined differential diagnosis was 0.985, with a sensitivity of 95.00 % and a specificity of 92.70 %. Conclusion: The levels of augmentation index and the ADC value of PCNSL focus are significantly correlated with VM and reticular fiber, and there is a strong negative correlation between necrotic lesions and VM. MRI imaging technology is of great significance in revealing the biological behavior of PCNSL, which can effectively reveal the relationship between VM and reticular fibers and the MRI characteristics in PCNSL, thereby providing a new imaging basis for the clinical diagnosis and treatment of PCNSL.

14.
Cell Biol Toxicol ; 40(1): 44, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38862832

RÉSUMÉ

BACKGROUND: Vasculogenic mimicry (VM) is an enigmatic physiological feature that influences blood supply within glioblastoma (GBM) tumors for their sustained growth. Previous studies identify NFATC3, FOSL1 and HNRNPA2B1 as significant mediators of VEGFR2, a key player in vasculogenesis, and their molecular relationships may be crucial for VM in GBM. AIMS: The aim of this study was to understand how NFATC3, FOSL1 and HNRNPA2B1 collectively influence VM in GBM. METHODS: We have investigated the underlying gene regulatory mechanisms for VM in GBM cell lines U251 and U373 in vitro and in vivo. In vitro cell-based assays were performed to explore the role of NFATC3, FOSL1 and HNRNPA2B1 in GBM cell proliferation, VM and migration, in the context of RNA interference (RNAi)-mediated knockdown alongside corresponding controls. Western blotting and qRT-PCR assays were used to examine VEGFR2 expression levels. CO-IP was employed to detect protein-protein interactions, ChIP was used to detect DNA-protein complexes, and RIP was used to detect RNA-protein complexes. Histochemical staining was used to detect VM tube formation in vivo. RESULTS: Focusing on NFATC3, FOSL1 and HNRNPA2B1, we found each was significantly upregulated in GBM and positively correlated with VM-like cellular behaviors in U251 and U373 cell lines. Knockdown of NFATC3, FOSL1 or HNRNPA2B1 each resulted in decreased levels of VEGFR2, a key growth factor gene that drives VM, as well as the inhibition of proliferation, cell migration and extracorporeal VM activity. Chromatin immunoprecipitation (ChIP) studies and luciferase reporter gene assays revealed that NFATC3 binds to the promoter region of VEGFR2 to enhance VEGFR2 gene expression. Notably, FOSL1 interacts with NFATC3 as a co-factor to potentiate the DNA-binding capacity of NFATC3, resulting in enhanced VM-like cellular behaviors. Also, level of NFATC3 protein in cells was enhanced through HNRNPA2B1 binding of NFATC3 mRNA. Furthermore, RNAi-mediated silencing of NFATC3, FOSL1 and HNRNPA2B1 in GBM cells reduced their capacity for tumor formation and VM-like behaviors in vivo. CONCLUSION: Taken together, our findings identify NFATC3 as an important mediator of GBM tumor growth through its molecular and epistatic interactions with HNRNPA2B1 and FOSL1 to influence VEGFR2 expression and VM-like cellular behaviors.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Glioblastome , Ribonucléoprotéine nucléaire hétérogène du groupe A-B , Facteurs de transcription NFATC , Néovascularisation pathologique , Protéines proto-oncogènes c-fos , Humains , Protéines proto-oncogènes c-fos/métabolisme , Protéines proto-oncogènes c-fos/génétique , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/génétique , Glioblastome/vascularisation , Lignée cellulaire tumorale , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/métabolisme , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/génétique , Facteurs de transcription NFATC/métabolisme , Facteurs de transcription NFATC/génétique , Animaux , Prolifération cellulaire/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation pathologique/anatomopathologie , Mouvement cellulaire/génétique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Régulation de l'expression des gènes tumoraux , Souris , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/vascularisation , Souris nude
15.
Expert Rev Clin Immunol ; 20(8): 913-926, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38712535

RÉSUMÉ

INTRODUCTION: Vasculogenic mimicry (VM) alludes to the ability of cancer cells to organize on three-dimensional channel-like structures to obtain nutrients and oxygen. This mechanism confers an aggressive phenotype, metastatic potential, and resistance to chemotherapy resulting in a poor prognosis. Recent studies have been focused on the identification of microRNAs (miRNAs) that regulate the VM representing potential therapeutic targets in cancer. AREAS COVERED: An overview of the roles of miRNAs on VM development and their functional relationships with tumor microenvironment. The functions of cancer stem-like cells in VM, and resistance to therapy are also discussed. Moreover, the modulation of VM by natural compounds is explored. The clinical significance of deregulated miRNAs as potential therapeutic targets in tumors showing VM is further highlighted. EXPERT OPINION: The miRNAs are regulators of protein-encoding genes involved in VM; however, their specific expression signatures with clinical value in large cohorts of patients have not been established yet. We considered that genomic profiling of miRNAs could be useful to define some hallmarks of tumors such as stemness, drug resistance, and VM in cancer patients. However, additional studies are needed to establish the relevant role of miRNAs as effective therapeutic targets in tumors that have developed VM.


Sujet(s)
Résistance aux médicaments antinéoplasiques , microARN , Tumeurs , Cellules souches tumorales , Néovascularisation pathologique , Microenvironnement tumoral , Humains , microARN/génétique , Tumeurs/génétique , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/thérapie , Résistance aux médicaments antinéoplasiques/génétique , Néovascularisation pathologique/génétique , Néovascularisation pathologique/traitement médicamenteux , Cellules souches tumorales/anatomopathologie , Animaux , Régulation de l'expression des gènes tumoraux
16.
J Biomed Res ; 38(4): 322-333, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38807414

RÉSUMÉ

Glioblastoma (GBM) is a highly vascularized malignant brain tumor with poor clinical outcomes. Vasculogenic mimicry (VM) formed by aggressive GBM cells is an alternative approach for tumor blood supply and contributes to the failure of anti-angiogenic therapy. To date, there is still a lack of effective drugs that target VM formation in GBM. In the present study, we evaluated the effects of the plant cyclopeptide moroidin on VM formed by GBM cells and investigated its underlying molecular mechanisms. Moroidin significantly suppressed cell migration, tube formation, and the expression levels of α-smooth muscle actin and matrix metalloproteinase-9 in human GBM cell lines at sublethal concentrations. The RNA sequencing data suggested the involvement of the epithelial-mesenchymal transition (EMT) pathway in the mechanism of moroidin. Exposure to moroidin led to a concentration-dependent decrease in the expression levels of the EMT markers N-cadherin and vimentin in GBM cells. Moreover, moroidin significantly reduced the level of phosphorylated extracellular signal-regulated protein kinase (p-ERK) and inhibited the activation of ß-catenin. Finally, we demonstrated that the plant cyclopeptide moroidin inhibited VM formation by GBM cells through inhibiting the ERK/ß-catenin-mediated EMT. Therefore, our study indicates a potential application of moroidin as an anti-VM agent in the treatment of GBM.

17.
Curr Med Chem ; 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38757323

RÉSUMÉ

BACKGROUND: Vasculogenic mimicry, a novel neovascularization pattern of aggressive tumors, is associated with poor clinical outcomes. OBJECTIVE: The aim of this research was to establish a new model, termed VC score, to predict the prognosis, Tumor Microenvironment (TME) components, and immunotherapeutic response in Hepatocellular Carcinoma (HCC). METHODS: The expression data of the public databases were used to develop the prognostic model. Consensus clustering was performed to confirm the molecular subtypes with ideal clustering efficacy. The high- and low-risk groups were stratified utilizing the VC score. Various methodologies, including survival analysis, single-sample Gene Set Enrichment Analysis (ssGSEA), Tumor Immune Dysfunction and Exclusion scores (TIDE), Immunophenoscore (IPS), and nomogram, were utilized for verification of the model performance and to characterize the immune status of HCC tissues. GSEA was performed to mine functional pathway information. RESULTS: The survival and immune characteristics varied between the three molecular subtypes. A five-gene signature (TPX2, CDC20, CFHR4, SPP1, and NQO1) was verified to function as an independent predictive factor for the prognosis of patients with HCC. The high-risk group exhibited lower Overall Survival (OS) rates and higher mortality rates in comparison to the low-risk group. Patients in the low-risk group were predicted to benefit from immune checkpoint inhibitor therapy and exhibit increased sensitivity to immunotherapy. Enrichment analysis revealed that signaling pathways linked to the cell cycle and DNA replication processes exhibited enrichment in the high-risk group. CONCLUSIONS: The VC score holds the potential to establish individualized treatment plans and clinical management strategies for patients with HCC.

18.
Acta Pharm Sin B ; 14(5): 2039-2056, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38799644

RÉSUMÉ

Osteosarcoma, a prevalent primary malignant bone tumor, often presents with lung metastases, severely impacting patient survival rates. Extracellular vesicles, particularly exosomes, play a pivotal role in the formation and progression of osteosarcoma-related pulmonary lesions. However, the communication between primary osteosarcoma and exosome-mediated pulmonary lesions remains obscure, with the potential impact of pulmonary metastatic foci on osteosarcoma progression largely unknown. This study unveils an innovative mechanism by which exosomes originating from osteosarcoma pulmonary metastatic sites transport the miR-194/215 cluster to the primary tumor site. This transportation enhances lung metastatic capability by downregulating myristoylated alanine-rich C-kinase substrate (MARCKS) expression. Addressing this phenomenon, in this study we employ cationic bovine serum albumin (CBSA) to form nanoparticles (CBSA-anta-194/215) via electrostatic interaction with antagomir-miR-194/215. These nanoparticles are loaded into nucleic acid-depleted exosomal membrane vesicles (anta-194/215@Exo) targeting osteosarcoma lung metastatic sites. Intervention with bioengineered exosome mimetics (anta-194/215@Exo) not only impedes osteosarcoma progression but also significantly prolongs the lifespan of tumor-bearing mice. These findings suggest that pulmonary metastatic foci-derived exosomes initiate primary osteosarcoma lung metastasis by transferring the miR-194/215 cluster targeting MARCKS, making the miR-194/215 cluster a promising therapeutic target for inhibiting the progression of patients with osteosarcoma lung metastases.

19.
Anticancer Res ; 44(6): 2377-2392, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821582

RÉSUMÉ

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC) is characterized by early metastasis, clinical resistance and poor prognosis. Recently, we showed that aggressive OSCC cells co-express endothelial cell markers and can form tube-like structures, known as vasculogenic mimicry (VM), a process associated with poor prognosis in head and neck cancers. Given the limited success of current antiangiogenic therapy in treating OSCC, this study sought to explore the efficiency of these drugs in targeting an ex vivo model of VM. MATERIALS AND METHODS: OSCC cell lines from the tongue and floor of the mouth in addition to human endothelial cells were used. The treatments comprised a set of clinically relevant antiangiogenic drugs: sorafenib, sunitinib, and axitinib, which were administered in different doses. Multiple ex vivo approaches including cell tubulogenesis, proliferation, apoptosis, and migration assays were used. RESULTS: Although these drugs inhibited the formation of endothelial cell capillaries, they showed clear differential effects on OSCC cell-derived VM and cell morphology. Sorafenib inhibited the tubulogenesis of aggressive OSCC cells compared with the limited effect of sunitinib and axitinib. Furthermore, our data consistently demonstrated a preferential efficacy of certain drugs over others. Sorafenib and sunitinib exhibited anti-cancer effects on tumor cell proliferation, apoptosis, and cell migration, compared with the limited effect of axitinib. CONCLUSION: The antiangiogenic drugs, except sorafenib, had limited effect on VM formation in vitro and exhibited varying anti-cancer effects on OSCC cells. These data support the notion that VM formation may in part explain the development of drug resistance in OSCC cells.


Sujet(s)
Inhibiteurs de l'angiogenèse , Axitinib , Mouvement cellulaire , Prolifération cellulaire , Tumeurs de la bouche , Néovascularisation pathologique , Sorafénib , Sunitinib , Humains , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/vascularisation , Tumeurs de la bouche/métabolisme , Lignée cellulaire tumorale , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/anatomopathologie , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Sunitinib/pharmacologie , Sunitinib/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Axitinib/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/vascularisation , Carcinome épidermoïde/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/anatomopathologie , Cellules endothéliales/métabolisme , Phénylurées/pharmacologie , Phénylurées/usage thérapeutique , Nicotinamide/analogues et dérivés , Nicotinamide/pharmacologie , Nicotinamide/usage thérapeutique
20.
J Exp Clin Cancer Res ; 43(1): 135, 2024 May 04.
Article de Anglais | MEDLINE | ID: mdl-38702792

RÉSUMÉ

BACKGROUND: Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported. METHODS: Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2. RESULTS: Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS. CONCLUSION: Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.


Sujet(s)
Néovascularisation pathologique , Rhabdomyosarcome , Ribonucléoprotéines , Facteurs de transcription de la famille Snail , Ubiquitin-specific peptidase 7 , Humains , Facteurs de transcription de la famille Snail/métabolisme , Facteurs de transcription de la famille Snail/génétique , Animaux , Souris , Ubiquitin-specific peptidase 7/métabolisme , Ubiquitin-specific peptidase 7/génétique , Rhabdomyosarcome/métabolisme , Rhabdomyosarcome/anatomopathologie , Rhabdomyosarcome/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Femelle , Évolution de la maladie , Prolifération cellulaire , Mâle , Homéostasie , Lignée cellulaire tumorale , Souris nude , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE