Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 9.844
Filtrer
1.
Int J Nanomedicine ; 19: 7165-7183, 2024.
Article de Anglais | MEDLINE | ID: mdl-39050873

RÉSUMÉ

Background: Exosomal microRNAs (miRNAs) in the tumor microenvironment play crucial roles in tumorigenesis and tumor progression by participating in intercellular cross-talk. However, the functions of exosomal miRNAs and the mechanisms by which they regulate esophageal squamous cell carcinoma (ESCC) progression are unclear. Methods: RNA sequencing and GEO analysis were conducted to identify candidate exosomal miRNAs involved in ESCC development. Receiver operating characteristic curve analysis was performed to assess the diagnostic value of plasma exosomal miR-493-5p. EdU, tube formation and Transwell assays were used to investigate the effects of exosomal miR-493-5p on human umbilical vein endothelial cells (HUVECs). A subcutaneous xenograft model was used to evaluate the antitumor effects of miR-493-5p and decitabine (a DNA methyltransferase inhibitor). The relationship between miR-493-5p and SP1/SP3 was revealed via a dual-luciferase reporter assay. A series of rescue assays were subsequently performed to investigate whether SP1/SP3 participate in exosomal miR-493-5p-mediated ESCC angiogenesis. Results: We found that miR-493-5p expression was notably reduced in the plasma exosomes of ESCC patients, which showed the high potential value in early ESCC diagnosis. Additionally, miR-493-5p, as a candidate tumor suppressor, inhibited the proliferation, migration and tube formation of HUVECs by suppressing the expression of VEGFA and exerted its angiostatic effect via exosomes. Moreover, we found that SP1/SP3 are direct targets of miR-493-5p and that re-expression of SP1/SP3 could reverse the inhibitory effects of miR-493-5p. Further investigation revealed that miR-493-5p expression could be regulated by DNA methyltransferase 3A (DNMT3A) and DNMT3B, and either miR-493-5p overexpression or restoration of miR-493-5p expression with decitabine increased the antitumor effects of bevacizumab. Conclusion: Exosomal miR-493-5p is a highly valuable ESCC diagnosis marker and inhibits ESCC-associated angiogenesis. miR-493-5p can be silenced via DNA methylation, and restoration of miR-493-5p expression with decitabine increases the antitumor effects of bevacizumab, suggesting its potential as a therapeutic target for ESCC treatment.


Sujet(s)
Méthylation de l'ADN , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Exosomes , Cellules endothéliales de la veine ombilicale humaine , microARN , Néovascularisation pathologique , Facteur de croissance endothéliale vasculaire de type A , Humains , Exosomes/métabolisme , Exosomes/génétique , microARN/génétique , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/anatomopathologie , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Animaux , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Lignée cellulaire tumorale , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/métabolisme , Décitabine/pharmacologie , Souris , Souris nude , Facteur de transcription Sp1/métabolisme , Facteur de transcription Sp1/génétique , Régulation de l'expression des gènes tumoraux , Mâle , Souris de lignée BALB C , Femelle ,
2.
Curr Protein Pept Sci ; 25(7): 567-576, 2024.
Article de Anglais | MEDLINE | ID: mdl-39044556

RÉSUMÉ

BACKGROUND: Vascular Endothelial Growth Factor Receptors (VEGFR1 and VEGFR2) are tyrosine kinase receptors expressed on endothelial cells and tumor vessels and play an important role in angiogenesis. In this study, three repeats of VEGFR1 and VEGFR2 binding peptide (VGB3) were genetically fused to the truncated diphtheria toxin (TDT), and its in vitro activity was evaluated. METHODS: The recombinant construct (TDT-triVGB3) was expressed in bacteria cells and purified with nickel affinity chromatography. The binding capacity and affinity of TDT-triVGB3 were evaluated using the enzyme-linked immunosorbent assay. The inhibitory activity of TDT-triVGB3 on viability, migration, and tube formation of human endothelial cells was evaluated using MTT, migration, and tube formation assays. RESULTS: TDT-triVGB3 selectively detected VEGFR1 and VEGFR2 with high affinity in an enzyme- linked immunosorbent assay and significantly inhibited viability, migration, and tube formation of human endothelial cells. CONCLUSION: The developed TDT-triVGB3 is potentially a novel agent for targeting VEGFR1/ VEGFR2 over-expressing cancer cells.


Sujet(s)
Inhibiteurs de l'angiogenèse , Mouvement cellulaire , Toxine diphtérique , Cellules endothéliales de la veine ombilicale humaine , Récepteur-1 au facteur croissance endothéliale vasculaire , Récepteur-2 au facteur croissance endothéliale vasculaire , Humains , Récepteur-1 au facteur croissance endothéliale vasculaire/génétique , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Toxine diphtérique/génétique , Toxine diphtérique/pharmacologie , Toxine diphtérique/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/génétique , Inhibiteurs de l'angiogenèse/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/pharmacologie , Protéines de fusion recombinantes/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Expression des gènes , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques
3.
Cells ; 13(14)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39056780

RÉSUMÉ

Background: Angiogenesis is essential for various physiological and pathological processes, such as embryonic development and cancer cell proliferation, migration, and invasion. Long noncoding RNAs (lncRNAs) play pivotal roles in normal homeostasis and disease processes by regulating gene expression through various mechanisms, including competing endogenous RNAs (ceRNAs) of target microRNAs (miRNAs). The lncRNA MYU is known to promote prostate cancer proliferation via the miR-184/c-Myc regulatory axis and to be upregulated in vascular endothelial cells under hypoxic conditions, which often occurs in solid tumors. In the present study, we investigated whether MYU might affect cancer growth by regulating angiogenesis in vascular endothelial cells under hypoxia. Methods: The expression of MYU-regulated miR-23a-3p and interleukin-8 (IL-8) in HUVEC cell lines was examined using qRT-PCR. The CCK-8 assay, EdU assay, wound-healing assay, and tube-formation assay were used to assess the effects of MYU on cell proliferation, migration, and tube formation of HUVEC cells in vitro. The dual-luciferase reporter assay was performed to examine the effects of miR-23a-3p on MYU and IL-8 expression. Results: We found that the overexpression of MYU and knockdown of miR-23a-3p in human umbilical vein endothelial cells (HUVECs) under hypoxia promoted cell proliferation, migration, and tube formation. Mechanistically, MYU was shown to bind competitively to miR-23a-3p, thereby preventing miR-23a-3p binding to the 3' untranslated region of IL-8 mRNA. In turn, increased production of pro-angiogenic IL-8 promoted HUVEC proliferation, migration, and tube formation under hypoxia. Conclusion: This study identified a new role for lncRNA MYU as a ceRNA for miR-23a-3p and uncovered a novel MYU-miR-23a-3p-IL-8 regulatory axis for angiogenesis. MYU and/or miR-23a-3p may thus represent new targets for the treatment of hypoxia-related diseases by promoting angiogenesis.


Sujet(s)
Hypoxie cellulaire , Mouvement cellulaire , Prolifération cellulaire , Cellules endothéliales de la veine ombilicale humaine , Interleukine-8 , microARN , ARN long non codant , Humains , microARN/génétique , microARN/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Prolifération cellulaire/génétique , Hypoxie cellulaire/génétique , Mouvement cellulaire/génétique , Interleukine-8/métabolisme , Interleukine-8/génétique , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Cellules endothéliales/métabolisme ,
4.
Aging (Albany NY) ; 16(13): 10799-10812, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38967628

RÉSUMÉ

OBJECTIVE: This investigation seeks to elucidate the role of the Granulocyte Colony-Stimulating Factor (G-CSF) in the progression of hepatocellular carcinoma (HCC), as well as the impact of the substance on related signaling pathways within the disease matrix. METHODS: Nude mouse tumor-bearing assay was used to detect tumor progression. Levels of Mannose/CD68 and CD34/Mannose within these samples and the concentrations of Mannose and inducible Nitric Oxide Synthase (iNOS) in macrophages were quantified using immunofluorescence techniques. The angiogenic capability was assessed via tube formation assays, and protein expressions of G-CSF, Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor-beta (TGF-ß), Matrix Metalloproteinases 2 and 9 (MMP2/9), SH2-containing protein tyrosine phosphatase-2 (SHP-2), phosphorylated PI3K/total PI3K (P-PI3K/t-PI3K), phosphorylated AKT/total AKT (P-AKT/t-AKT), and phosphorylated mTOR/total mTOR (P-mTOR/t-mTOR) were measured through Western Blot analysis in both tumor tissues and macrophages. RESULTS: Administration of G-CSF resulted in a marked augmentation of tumor volume. Macrophage Mannose expression was significantly elevated upon G-CSF treatment, while iNOS levels were conspicuously diminished. G-CSF substantially enhanced the secretion of VEGF, TGF-ß, and MMPs in tumor tissues. Macrophage parameters, following incubation in G-CSF pre-treated conditioned medium, indicated enhanced tube-forming capabilities relative to the control, an effect mitigated by the introduction of specific inhibitors. Furthermore, the G-CSF group exhibited a notable reduction in SHP-2 expression, alongside a substantial elevation in the phosphorylation levels of the PI3K/AKT/mTOR pathway proteins across all tumor-bearing paradigms. CONCLUSION: G-CSF ostensibly facilitates the advancement of hepatocellular carcinoma by activating the PI3K/AKT/mTOR signaling cascade within Tumor-Associated Macrophages (TAM).


Sujet(s)
Carcinome hépatocellulaire , Facteur de stimulation des colonies de granulocytes , Tumeurs du foie , Souris nude , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Animaux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Facteur de stimulation des colonies de granulocytes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Souris , Humains , Macrophages associés aux tumeurs/métabolisme , Néovascularisation pathologique/métabolisme , Lignée cellulaire tumorale , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Mâle
5.
Mol Biol Rep ; 51(1): 821, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39023636

RÉSUMÉ

BACKGROUND: Our previous study has demonstrated that Nischarin (NISCH) exerts its antitumor effects in breast cancer (BC) by suppressing cell migration and invasion. This study aims to explore the underlying mechanism through which NISCH functions in BC. METHODS AND RESULTS: The relevance between EGF Like Repeats and Discoidin Domains 3 (EDIL3) mRNA expression and the overall survival of tumor patients was depicted by the Kaplan-Meier curve. The findings revealed that overexpressed NISCH attenuated cell motility and colony-forming capacities of Hs578T cells, yet silenced NISCH in MDA-MB-231 cells led to contrasting results. Western blot (WB) analysis indicated that overexpression of NISCH significantly down-regulated the Vimentin and Slug expression, and inactivated the FAK/ERK signaling pathway. RNA sequencing (RNA-seq) was performed in NISCH-overexpressed Hs578T cells and the control cells to analyze differentially expressed genes (DeGs), and the results showed a significant down-regulation of EDIL3 mRNA level upon overexpression of NISCH. Subsequent functional analyses demonstrated that overexpression of EDIL3 attenuated the inhibitory effect of NISCH on cell migration, invasion, colony formation, and tube formation. CONCLUSION: In summary, our finding preliminarily revealed that NISCH inhibits the epithelial-mesenchymal transition (EMT) process and angiogenesis in BC cells by down-regulating EDIL3 to inactivate the FAK/ERK signaling pathway, thereby suppressing the progression of BC. Our results hold promise for contributing to the deep understanding of BC pathogenesis and identifying new therapeutic strategies for clinical application.


Sujet(s)
Tumeurs du sein , Mouvement cellulaire , Transition épithélio-mésenchymateuse , Régulation de l'expression des gènes tumoraux , Système de signalisation des MAP kinases , Néovascularisation pathologique , Humains , Transition épithélio-mésenchymateuse/génétique , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Système de signalisation des MAP kinases/génétique , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Focal adhesion kinase 1/métabolisme , Focal adhesion kinase 1/génétique , Prolifération cellulaire/génétique , Vimentine/métabolisme , Vimentine/génétique , Transduction du signal , Protéines de la matrice extracellulaire/métabolisme , Protéines de la matrice extracellulaire/génétique , Facteurs de transcription de la famille Snail/métabolisme , Facteurs de transcription de la famille Snail/génétique , , Protéines de liaison au calcium , Molécules d'adhérence cellulaire
6.
Int J Mol Sci ; 25(13)2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-39000375

RÉSUMÉ

Angiogenesis is critical for rheumatoid arthritis (RA) progression. The effects of tofacitinib, a JAK-STAT inhibitor used for RA treatment, on angiogenesis in RA are unclear. We, therefore, evaluated the levels of angiogenic factors in two systems of a human co-culture of fibroblast (HT1080) and monocytic (U937) cell lines treated with tofacitinib and in serum samples from RA patients before and after six months of tofacitinib treatment. Tofacitinib reduced CD147 levels, matrix metalloproteinase-9 (MMP-9) activity, and angiogenic potential but increased endostatin levels and secreted proteasome 20S activity. In vitro, tofacitinib did not change CD147 mRNA but increased miR-146a-5p expression and reduced STAT3 phosphorylation. We recently showed that CD147 regulates the ability of MMP-9 and secreted proteasome 20S to cleave collagen XVIIIA into endostatin. We show here that tofacitinib-enhanced endostatin levels are mediated by CD147, as CD147-siRNA or an anti-CD147 antibody blocked proteasome 20S activity. The correlation between CD147 and different disease severity scores supported this role. Lastly, tofacitinib reduced endostatin' s degradation by inhibiting cathepsin S activity and recombinant cathepsin S reversed this in both systems. Thus, tofacitinib inhibits angiogenesis by reducing pro-angiogenic factors and enhancing the anti-angiogenic factor endostatin in a dual effect mediated partly through CD147 and partly through cathepsin S.


Sujet(s)
Polyarthrite rhumatoïde , Antigènes CD147 , Cathepsines , Endostatines , Pipéridines , Pyrimidines , Humains , Antigènes CD147/métabolisme , Antigènes CD147/génétique , Pipéridines/pharmacologie , Endostatines/métabolisme , Endostatines/pharmacologie , Pyrimidines/pharmacologie , Cathepsines/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Matrix metalloproteinase 9/métabolisme , Matrix metalloproteinase 9/génétique , Facteur de transcription STAT-3/métabolisme , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Inhibiteurs de l'angiogenèse/pharmacologie , Femelle , Adulte d'âge moyen , Mâle , Pyrroles/pharmacologie , Lignée cellulaire
7.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000466

RÉSUMÉ

It is acknowledged that conventional renal cell carcinoma (cRCC), which makes up 85% of renal malignancies, is a highly vascular tumor. Humanized monoclonal antibodies were developed to inhibit tumor neo-angiogenesis, which is driven by VEGFA/KDR signaling. The results largely met our expectations, and in several cases, adverse events occurred. Our study aimed to analyze the expression of VEGFA and its receptor KDR by immunohistochemistry in tissue multi-array containing 811 cRCC and find a correlation between VEGFA/KDR signaling and new vessel formation. None of the 811 cRCC displayed VEGFA-positive immunostaining. However, each glomerulus in normal kidney showed VEGFA-positive endothelial cells. KDR expression in endothelial meshwork was found in only 9% of cRCC, whereas 2% of the cRCC displayed positive KDR reaction in the cytoplasm of tumor cells. Our results disclose the involvement of VEGFA/KDR signaling in the neo-vascularization of cRCC and explain the frequent resistance to drugs targeting the VEGFA/KDR signaling and the high frequency of adverse events.


Sujet(s)
Néphrocarcinome , Tumeurs du rein , Transduction du signal , Facteur de croissance endothéliale vasculaire de type A , Récepteur-2 au facteur croissance endothéliale vasculaire , Néphrocarcinome/métabolisme , Néphrocarcinome/traitement médicamenteux , Néphrocarcinome/anatomopathologie , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Tumeurs du rein/métabolisme , Tumeurs du rein/traitement médicamenteux , Tumeurs du rein/anatomopathologie , Femelle , Mâle , Adulte d'âge moyen , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Sujet âgé , Thérapie moléculaire ciblée , Adulte
8.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-39000507

RÉSUMÉ

Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide, with 20% of patients presenting with metastatic disease at diagnosis. TGF-ß signaling plays a crucial role in various cellular processes, including growth, differentiation, apoptosis, epithelial-mesenchymal transition (EMT), regulation of the extracellular matrix, angiogenesis, and immune responses. TGF-ß signals through SMAD proteins, which are intracellular molecules that transmit TGF-ß signals from the cell membrane to the nucleus. Alterations in the TGF-ß pathway and mutations in SMAD proteins are common in metastatic CRC (mCRC), making them critical factors in CRC tumorigenesis. This review first analyzes normal TGF-ß signaling and then investigates its role in CRC pathogenesis, highlighting the mechanisms through which TGF-ß influences metastasis development. TGF-ß promotes neoangiogenesis via VEGF overexpression, pericyte differentiation, and other mechanisms. Additionally, TGF-ß affects various elements of the tumor microenvironment, including T cells, fibroblasts, and macrophages, promoting immunosuppression and metastasis. Given its strategic role in multiple processes, we explored different strategies to target TGF-ß in mCRC patients, aiming to identify new therapeutic options.


Sujet(s)
Tumeurs colorectales , Transduction du signal , Facteur de croissance transformant bêta , Microenvironnement tumoral , Humains , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Facteur de croissance transformant bêta/métabolisme , Transition épithélio-mésenchymateuse , Animaux , Néovascularisation pathologique/métabolisme
9.
Int J Biol Sci ; 20(9): 3442-3460, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993554

RÉSUMÉ

In this study, we explored the oncogenic mechanism of cleavage and polyadenylation-specific factor 6 (CPSF6) in hepatocellular carcinoma (HCC). CPSF6 was overexpressed in HCC tissues with poor survival rates compared to normal tissues. Hence, CPSF6 depletion suppressed cell viability and colony formation, induced apoptosis via PARP cleavage, and increased the sub-G1 population of Hep3B and Huh7 cells. In addition, CPSF6 enhanced the stability of c-Myc via their binding through nuclear co-localization by binding to c-Myc at the site of 258-360. Furthermore, c-Myc degradation by CPSF6 depletion was disturbed by FBW7 depletion or treatment with the proteasomal inhibitor MG132. Additionally, CPSF6 depletion suppressed the Warburg effect by inhibiting glucose, HK2, PKM2, LDH, and lactate; showed a synergistic effect with Sorafenib in Hep3B cells; and inhibited angiogenesis by tube formation and CAM assays, along with decreased expression and production of vascular endothelial growth factor (VEGF). Notably, CPSF6 depletion attenuated PD-L1 expression and increased Granzyme B levels, along with an increase in the percentage of CD4/CD8 cells in the splenocytes of BALB/c nude mice bearing Hep3B cells. Consistently, immunohistochemistry showed that CPSF6 depletion reduced the growth of Hep3B cells in BALB/c mice in orthotopic and xenograft tumor models by inhibiting tumor microenvironment-associated proteins. Overall, these findings suggest that CPSF6 enhances the Warburg effect for immune escape and angiogenesis, leading to cancer progression via c-Myc, mediated by the HK, PD-L1, and VEGF networks, with synergistic potential with sorafenib as a molecular target for liver cancer therapy.


Sujet(s)
Carcinome hépatocellulaire , Tumeurs du foie , Protéines proto-oncogènes c-myc , Transduction du signal , Humains , Tumeurs du foie/métabolisme , Tumeurs du foie/traitement médicamenteux , Lignée cellulaire tumorale , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Protéines proto-oncogènes c-myc/métabolisme , Animaux , Néovascularisation pathologique/métabolisme , Souris , Sorafénib/usage thérapeutique , Sorafénib/pharmacologie , Effet Warburg en oncologie , Souris nude , Souris de lignée BALB C , Apoptose ,
10.
Cells ; 13(13)2024 Jun 23.
Article de Anglais | MEDLINE | ID: mdl-38994941

RÉSUMÉ

Tumors are a heterogeneous group of cell masses originating in various organs or tissues. The cellular composition of the tumor cell mass interacts in an intricate manner, influenced by humoral, genetic, molecular, and tumor microenvironment cues that dictate tumor growth or suppression. As a result, tumors undergo a period of a dormant state before their clinically discernible stage, which surpasses the clinical dormancy threshold. Moreover, as a genetically imprinted strategy, early-seeder cells, a distinct population of tumor cells, break off to dock nearby or extravasate into blood vessels to secondary tissues, where they form disseminated solitary dormant tumor cells with reversible capacity. Among the various mechanisms underlying the dormant tumor mass and dormant tumor cell formation, heat shock proteins (HSPs) might play one of the most important roles in how the dormancy program plays out. It is known that numerous aberrant cellular processes, such as malignant transformation, cancer cell stemness, tumor invasion, metastasis, angiogenesis, and signaling pathway maintenance, are influenced by the HSPs. An accumulating body of knowledge suggests that HSPs may be involved in the angiogenic switch, immune editing, and extracellular matrix (ECM) remodeling cascades, crucial genetically imprinted strategies important to the tumor dormancy initiation and dormancy maintenance program. In this review, we highlight the biological events that orchestrate the dormancy state and the body of work that has been conducted on the dynamics of HSPs in a tumor mass, as well as tumor cell dormancy and reactivation. Additionally, we propose a conceptual framework that could possibly underlie dormant tumor reactivation in metastatic relapse.


Sujet(s)
Protéines du choc thermique , Tumeurs , Humains , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Protéines du choc thermique/métabolisme , Animaux , Microenvironnement tumoral , Néovascularisation pathologique/métabolisme , Transduction du signal
11.
Molecules ; 29(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38999055

RÉSUMÉ

Lignans, a class of secondary metabolites found in plants, along with their derivatives, exhibit diverse pharmacological activities, including antioxidant, antimicrobial, anti-inflammatory, and antiangiogenic ones. Angiogenesis, the formation of new blood vessels from pre-existing ones, is a crucial process for cancer growth and development. Several studies have elucidated the synergistic relationship between angiogenesis and inflammation in various inflammatory diseases, highlighting a correlation between inflammation and vascular endothelial growth factor (VEGF)-induced angiogenesis. Thus, the identification of novel molecules capable of modulating VEGF effects presents promising prospects for developing therapies aimed at stabilizing, reversing, or even arresting disease progression. Lignans often suffer from low aqueous solubility and, for their use, encapsulation in a delivery system is needed. In this research, a bioinspired benzoxantene has been encapsulated in solid lipid nanoparticles that have been characterized for their pharmacotechnical properties and their thermotropic behavior. The effects of these encapsulated nanoparticles on angiogenic parameters and inflammation in VEGF-induced angiogenesis were evaluated using human brain microvascular endothelial cells (HBMECs) as a human blood-brain barrier model.


Sujet(s)
Barrière hémato-encéphalique , Inflammation , Nanoparticules , Facteur de croissance endothéliale vasculaire de type A , Humains , Nanoparticules/composition chimique , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Lipides/composition chimique , Néovascularisation physiologique/effets des médicaments et des substances chimiques , , Liposomes
12.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38999976

RÉSUMÉ

Tumor angiogenesis, the formation of new blood vessels to support tumor growth and metastasis, is a complex process regulated by a multitude of signaling pathways. Dysregulation of signaling pathways involving protein kinases has been extensively studied, but the role of protein phosphatases in angiogenesis within the tumor microenvironment remains less explored. However, among angiogenic pathways, protein phosphatases play critical roles in modulating signaling cascades. This review provides a comprehensive overview of the involvement of protein phosphatases in tumor angiogenesis, highlighting their diverse functions and mechanisms of action. Protein phosphatases are key regulators of cellular signaling pathways by catalyzing the dephosphorylation of proteins, thereby modulating their activity and function. This review aims to assess the activity of the protein tyrosine phosphatases and serine/threonine phosphatases. These phosphatases exert their effects on angiogenic signaling pathways through various mechanisms, including direct dephosphorylation of angiogenic receptors and downstream signaling molecules. Moreover, protein phosphatases also crosstalk with other signaling pathways involved in angiogenesis, further emphasizing their significance in regulating tumor vascularization, including endothelial cell survival, sprouting, and vessel maturation. In conclusion, this review underscores the pivotal role of protein phosphatases in tumor angiogenesis and accentuate their potential as therapeutic targets for anti-angiogenic therapy in cancer.


Sujet(s)
Tumeurs , Néovascularisation pathologique , Phosphoprotein Phosphatases , Transduction du signal , Humains , Néovascularisation pathologique/métabolisme , Tumeurs/vascularisation , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Animaux , Phosphoprotein Phosphatases/métabolisme , Protein Phosphatase 2/métabolisme , Microenvironnement tumoral , Phosphorylation ,
13.
J Exp Clin Cancer Res ; 43(1): 185, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965575

RÉSUMÉ

BACKGROUND: Metastasis is the leading cause of mortality in patients with colorectal cancer (CRC) and angiogenesis is a crucial factor in tumor invasion and metastasis. Long noncoding RNAs (lncRNAs) play regulatory functions in various biological processes in tumor cells, however, the roles of lncRNAs in CRC-associated angiogenesis remain to be elucidated in CRC, as do the underlying mechanisms. METHODS: We used bioinformatics to screen differentially expressed lncRNAs from TCGA database. LOC101928222 expression was assessed by qRT-PCR. The impact of LOC101928222 in CRC tumor development was assessed both in vitro and in vivo. The regulatory mechanisms of LOC101928222 in CRC were investigated by cellular fractionation, RNA-sequencing, mass spectrometric, RNA pull-down, RNA immunoprecipitation, RNA stability, and gene-specific m6A assays. RESULTS: LOC101928222 expression was upregulated in CRC and was correlated with a worse outcome. Moreover, LOC101928222 was shown to promote migration, invasion, and angiogenesis in CRC. Mechanistically, LOC101928222 synergized with IGF2BP1 to stabilize HMGCS2 mRNA through an m6A-dependent pathway, leading to increased cholesterol synthesis and, ultimately, the promotion of CRC development. CONCLUSIONS: In summary, these findings demonstrate a novel, LOC101928222-based mechanism involved in the regulation of cholesterol synthesis and the metastatic potential of CRC. The LOC101928222-HMGCS2-cholesterol synthesis pathway may be an effective target for diagnosing and managing CRC metastasis.


Sujet(s)
Cholestérol , Tumeurs colorectales , Néovascularisation pathologique , ARN long non codant , ARN messager , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Souris , Cholestérol/métabolisme , Animaux , ARN messager/génétique , ARN messager/métabolisme , Hydroxymethylglutaryl-coA synthase/génétique , Hydroxymethylglutaryl-coA synthase/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Mâle , Femelle ,
14.
Biochem Pharmacol ; 226: 116414, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38972427

RÉSUMÉ

Lung adenocarcinoma (LUAD) is the most common histologic subtype of lung cancer. Angiogenesis plays a pivotal role in LUAD progression via supplying oxygen and nutrients for cancer cells. Non-coding miR-1293, a significantly up-regulated miRNA in LUAD tissues, can be potentially used as a novel biomarker for predicting the prognosis of LUAD patients. However, little information is available about the function of miR-1293 in LUAD progression especially cancer-induced angiogenesis. Herein, we found that miR-1293 knockdown could obviously attenuate LUAD-induced angiogenesis in vitro and down-regulate two most important pro-angiogenic cytokines VEGF-A and bFGF expression and secretion. Indeed, miR-1293 abrogation inactivated the angiogenesis-promoting ERK1/2 signaling characterized by decreased ERK1/2 phosphorylation and translocation from nucleus to cytoplasm. Next we found that miR-1293 knockdown reactivated the endogenous ERK1/2 pathway inhibitor Spry4 expression and Spry4 perturbance with specific siRNA transfection abolished the inhibition of ERK1/2 pathway and LUAD-induced angiogenesis by miR-1293 knockdown. Finally, with in vivo assay, we found obvious Spry4 up-regulation and VEGF-A, bFGF, ERK1/2 phosphorylation, micro-vessel density marker CD31 expression down-regulation in vivo, respectively. Collectively, these results indicated that miR-1293 knockdown could significantly attenuate LUAD angiogenesis via Spry4-mediated ERK1/2 signaling inhibition, which might be helpful for uncovering more functions of miR-1293 in LUAD and providing experimental basis for possible LUAD therapeutic strategy targeting miR-1293.


Sujet(s)
Adénocarcinome pulmonaire , Techniques de knock-down de gènes , Tumeurs du poumon , Système de signalisation des MAP kinases , microARN , Néovascularisation pathologique , Régulation positive , Humains , microARN/génétique , microARN/métabolisme , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Animaux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/métabolisme , Système de signalisation des MAP kinases/physiologie , Système de signalisation des MAP kinases/génétique , Souris , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Souris nude , Mâle , Souris de lignée BALB C , Cellules A549 , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Femelle ,
15.
Tissue Cell ; 89: 102466, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38986346

RÉSUMÉ

The gut microbiota is responsible for several metabolic functions, producing various metabolites with numerous roles for the host. The gut microbiota plays a key role in constructing the microvascular network in the intestinal villus, depending on the Paneth cells, strategically positioned to coordinate the development of both the microbiota and the microvasculature. The gut microbiota secretes several molecules and chemokines involved in the induction of the secretion of pro-angiogenic factors.


Sujet(s)
Microbiome gastro-intestinal , Néovascularisation physiologique , Microbiome gastro-intestinal/physiologie , Humains , Animaux , Intestins/microbiologie , Intestins/vascularisation , Muqueuse intestinale/microbiologie , Muqueuse intestinale/métabolisme , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/microbiologie , Néovascularisation pathologique/anatomopathologie , Cellules de Paneth/métabolisme ,
16.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062818

RÉSUMÉ

Exosomal microRNAs (miRNAs) from cancer cells play a key role in mediating the oral squamous cell carcinoma (OSCC) microenvironment. The objective of this study was to investigate how the long non-coding RNA (lncRNA) MEG3 affects OSCC angiogenesis through exosomal miR-421. Global miRNA microarray analysis and quantitative real-time PCR (qRT-PCR) were performed to determine the level of miRNAs in OSCC cell-derived exosomes. Cell migration, invasion, tube formation, immunohistochemistry, and hemoglobin concentrations were used to study the effects of exosomal miR-421 in angiogenesis. Western blotting was used to determine the expression level of HS2ST1 and VEGFR2-related downstream proteins. MiRNA array and qRT-PCR identified the upregulation of miR-421 in OSCC cell-derived exosomes. Furthermore, exosomal miR-421 can be taken up by human umbilical vein endothelial cells (HUVECs) and then target HS2ST1 through VEGF-mediated ERK and AKT phosphorylation, thereby promoting HUVEC migration, invasion, and tube formation. Additionally, forced expression of the lncRNA MEG3 in OSCC cells reduced exosomal miR-421 levels and then increased HS2ST1 expression, thereby reducing the VEGF/VEGFR2 pathway in HUVECs. Our results demonstrate a novel mechanism by which lncRNA MEG3 can act as a tumor suppressor and regulate endothelial angiogenesis through the exosomal miR-421/HS2ST1 axis, which provides a potential therapeutic strategy for OSCC angiogenesis.


Sujet(s)
Carcinome épidermoïde , Mouvement cellulaire , Exosomes , Régulation de l'expression des gènes tumoraux , Cellules endothéliales de la veine ombilicale humaine , microARN , Tumeurs de la bouche , Néovascularisation pathologique , ARN long non codant , Humains , microARN/génétique , microARN/métabolisme , Exosomes/métabolisme , Exosomes/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/génétique , Tumeurs de la bouche/anatomopathologie , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/génétique , Carcinome épidermoïde/anatomopathologie , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique ,
17.
Int J Mol Sci ; 25(14)2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39063232

RÉSUMÉ

Glioma cells overexpress different peptide receptors that are useful for research, diagnosis, management, and treatment of the disease. Oncogenic peptides favor the proliferation, migration, and invasion of glioma cells, as well as angiogenesis, whereas anticancer peptides exert antiproliferative, antimigration, and anti-angiogenic effects against gliomas. Other peptides exert a dual effect on gliomas, that is, both proliferative and antiproliferative actions. Peptidergic systems are therapeutic targets, as peptide receptor antagonists/peptides or peptide receptor agonists can be administered to treat gliomas. Other anticancer strategies exerting beneficial effects against gliomas are discussed herein, and future research lines to be developed for gliomas are also suggested. Despite the large amount of data supporting the involvement of peptides in glioma progression, no anticancer drugs targeting peptidergic systems are currently available in clinical practice to treat gliomas.


Sujet(s)
Antinéoplasiques , Gliome , Peptides , Humains , Gliome/traitement médicamenteux , Gliome/métabolisme , Gliome/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Peptides/pharmacologie , Peptides/usage thérapeutique , Animaux , Récepteurs peptidiques/métabolisme , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme
18.
Oncotarget ; 15: 424-438, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953895

RÉSUMÉ

Single-agent TAS102 (trifluridine/tipiracil) and regorafenib are FDA-approved treatments for metastatic colorectal cancer (mCRC). We previously reported that regorafenib combined with a fluoropyrimidine can delay disease progression in clinical case reports of multidrug-resistant mCRC patients. We hypothesized that the combination of TAS102 and regorafenib may be active in CRC and other gastrointestinal (GI) cancers and may in the future provide a treatment option for patients with advanced GI cancer. We investigated the therapeutic effect of TAS102 in combination with regorafenib in preclinical studies employing cell culture, colonosphere assays that enrich for cancer stem cells, and in vivo. TAS102 in combination with regorafenib has synergistic activity against multiple GI cancers in vitro including colorectal and gastric cancer, but not liver cancer cells. TAS102 inhibits colonosphere formation and this effect is potentiated by regorafenib. In vivo anti-tumor effects of TAS102 plus regorafenib appear to be due to anti-proliferative effects, necrosis and angiogenesis inhibition. Growth inhibition by TAS102 plus regorafenib occurs in xenografted tumors regardless of p53, KRAS or BRAF mutations, although more potent tumor suppression was observed with wild-type p53. Regorafenib significantly inhibits TAS102-induced angiogenesis and microvessel density in xenografted tumors, as well inhibits TAS102-induced ERK1/2 activation regardless of RAS or BRAF status in vivo. TAS102 plus regorafenib is a synergistic drug combination in preclinical models of GI cancer, with regorafenib suppressing TAS102-induced increase in microvessel density and p-ERK as contributing mechanisms. The TAS102 plus regorafenib drug combination may be further tested in gastric and other GI cancers.


Sujet(s)
Association médicamenteuse , Synergie des médicaments , Tumeurs gastro-intestinales , Mutation , Cellules souches tumorales , Néovascularisation pathologique , Phénylurées , Protéines proto-oncogènes B-raf , Protéines proto-oncogènes p21(ras) , Pyridines , Pyrrolidines , Facteur de transcription STAT-3 , Thymine , Trifluorothymidine , Uracile , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Trifluorothymidine/pharmacologie , Phénylurées/pharmacologie , Animaux , Pyridines/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes p21(ras)/génétique , Protéines proto-oncogènes p21(ras)/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/génétique , Néovascularisation pathologique/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Tumeurs gastro-intestinales/traitement médicamenteux , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/métabolisme , Uracile/pharmacologie , Uracile/analogues et dérivés , Souris , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Thymine/pharmacologie , Lignée cellulaire tumorale , Pyrrolidines/pharmacologie , Pyrrolidines/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques ,
19.
Theranostics ; 14(9): 3509-3525, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948065

RÉSUMÉ

Rationale: Current treatments for ocular angiogenesis primarily focus on blocking the activity of vascular endothelial growth factor (VEGF), but unfavorable side effects and unsatisfactory efficacy remain issues. The identification of novel targets for anti-angiogenic treatment is still needed. Methods: We investigated the role of tsRNA-1599 in ocular angiogenesis using endothelial cells, a streptozotocin (STZ)-induced diabetic model, a laser-induced choroidal neovascularization model, and an oxygen-induced retinopathy model. CCK-8 assays, EdU assays, transwell assays, and matrigel assays were performed to assess the role of tsRNA-1599 in endothelial cells. Retinal digestion assays, Isolectin B4 (IB4) staining, and choroidal sprouting assays were conducted to evaluate the role of tsRNA-1599 in ocular angiogenesis. Transcriptomic analysis, metabolic analysis, RNA pull-down assays, and mass spectrometry were utilized to elucidate the mechanism underlying angiogenic effects mediated by tsRNA-1599. Results: tsRNA-1599 expression was up-regulated in experimental ocular angiogenesis models and endothelial cells in response to angiogenic stress. Silencing of tsRNA-1599 suppressed angiogenic effects in endothelial cells in vitro and inhibited pathological ocular angiogenesis in vivo. Mechanistically, tsRNA-1599 exhibited little effect on VEGF signaling but could cause reduced glycolysis and NAD+/NADH production in endothelial cells by regulating the expression of HK2 gene through interacting with YBX1, thus affecting endothelial effects. Conclusions: Targeting glycolytic reprogramming of endothelial cells by a tRNA-derived small RNA represents an exploitable therapeutic approach for ocular neovascular diseases.


Sujet(s)
Néovascularisation choroïdienne , Cellules endothéliales , Glycolyse , Animaux , Glycolyse/effets des médicaments et des substances chimiques , Souris , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Néovascularisation choroïdienne/traitement médicamenteux , Néovascularisation choroïdienne/métabolisme , Humains , Protéine-1 de liaison à la boîte Y/métabolisme , Protéine-1 de liaison à la boîte Y/génétique , Inhibiteurs de l'angiogenèse/pharmacologie , Hexokinase/métabolisme , Hexokinase/génétique , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Souris de lignée C57BL , Mâle , Modèles animaux de maladie humaine , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Rétinopathie diabétique/traitement médicamenteux , Rétinopathie diabétique/métabolisme , Rétinopathie diabétique/génétique , Cellules endothéliales de la veine ombilicale humaine , Petit ARN non traduit/génétique , Petit ARN non traduit/métabolisme
20.
Gen Physiol Biophys ; 43(4): 301-312, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38953570

RÉSUMÉ

Vascular endothelial growth factor A (VEGFA) is an important regulator for non-small cell lung cancer (NSCLC). Our study aimed to reveal its upstream pathway to provide new ideas for developing the therapeutic targets of NSCLC. The mRNA and protein levels of VEGFA, ubiquitin-specific peptidase 35 (USP35), and FUS were determined by quantitative real-time PCR and Western blot. Cell proliferation, apoptosis, invasion and angiogenesis were detected using CCK8 assay, EdU assay, flow cytometry, transwell assay and tube formation assay. The interaction between USP35 and VEGFA was assessed by Co-IP assay and ubiquitination assay. Animal experiments were performed to assess USP35 and VEGFA roles in vivo. VEGFA had elevated expression in NSCLC tissues and cells. Interferences of VEGFA inhibited NSCLC cell proliferation, invasion, angiogenesis, and increased apoptosis. USP35 could stabilize VEGFA protein level by deubiquitination, and USP35 knockdown suppressed NSCLC cell growth, invasion and angiogenesis via reducing VEGFA expression. FUS interacted with USP35 to promote its mRNA stability, thereby positively regulating VEGFA expression. Also, USP35 silencing could reduce NSCLC tumorigenesis by downregulating VEGFA. FUS-stabilized USP35 facilitated NSCLC cell growth, invasion and angiogenesis through deubiquitinating VEGFA, providing a novel idea for NSCLC treatment.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Tumeurs du poumon , Invasion tumorale , Néovascularisation pathologique , Protéine FUS de liaison à l'ARN , Ubiquitination , Facteur de croissance endothéliale vasculaire de type A , Humains , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Protéine FUS de liaison à l'ARN/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Prolifération cellulaire/génétique , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/génétique , Invasion tumorale/génétique , Lignée cellulaire tumorale , Souris , Animaux , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Souris nude ,
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE