Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 264
Filtrer
1.
J Transl Med ; 22(1): 759, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138492

RÉSUMÉ

BACKGROUND: Oral cancer poses a significant health challenge due to limited treatment protocols and therapeutic targets. We aimed to investigate the invasive margins of gingivo-buccal oral squamous cell carcinoma (GB-OSCC) tumors in terms of the localization of genes and cell types within the margins at various distances that could lead to nodal metastasis. METHODS: We collected tumor tissues from 23 resected GB-OSCC samples for gene expression profiling using digital spatial transcriptomics. We monitored differential gene expression at varying distances between the tumor and its microenvironvent (TME), and performed a deconvulation study and immunohistochemistry to identify the cells and genes regulating the TME. RESULTS: We found that the tumor-stromal interface (a distance up to 200 µm between tumor and immune cells) is the most active region for disease progression in GB-OSCC. The most differentially expressed apex genes, such as FN1 and COL5A1, were located at the stromal ends of the margins, and together with enrichment of the extracellular matrix (ECM) and an immune-suppressed microenvironment, were associated with lymph node metastasis. Intermediate fibroblasts, myocytes, and neutrophils were enriched at the tumor ends, while cancer-associated fibroblasts (CAFs) were enriched at the stromal ends. The intermediate fibroblasts transformed into CAFs and relocated to the adjacent stromal ends where they participated in FN1-mediated ECM modulation. CONCLUSION: We have generated a functional organization of the tumor-stromal interface in GB-OSCC and identified spatially located genes that contribute to nodal metastasis and disease progression. Our dataset might now be mined to discover suitable molecular targets in oral cancer.


Sujet(s)
Fibroblastes , Régulation de l'expression des gènes tumoraux , Métastase lymphatique , Tumeurs de la bouche , Cellules myéloïdes , Microenvironnement tumoral , Humains , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/génétique , Métastase lymphatique/anatomopathologie , Fibroblastes/anatomopathologie , Fibroblastes/métabolisme , Cellules myéloïdes/anatomopathologie , Cellules myéloïdes/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/métabolisme , Analyse de profil d'expression de gènes , Femelle , Mâle , Adulte d'âge moyen , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique
2.
J Cell Mol Med ; 28(16): e70025, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39164826

RÉSUMÉ

Metastasis is a crucial stage in tumour progression, and cancer-associated fibroblasts (CAFs) support metastasis through their participation in extracellular matrix (ECM) stiffness. CD248 is a possible biomarker for non-small cell lung cancer (NSCLC)-derived CAFs, but its role in mediating ECM stiffness to promote NSCLC metastasis is unknown. We investigated the significance of CD248+ CAFs in activating the Hippo axis and promoting connective tissue growth factor (CTGF) expression, which affects the stromal collagen I environment and improves ECM stiffness, thereby facilitating NSCLC metastasis. In this study, we found that higher levels of CD248 in CAFs induced the formation of collagen I, which in turn increased extracellular matrix stiffness, thereby enabling NSCLC cell infiltration and migration. Hippo axis activation by CD248+ CAFs induces CTGF expression, which facilitates the formation of the collagen I milieu in the stromal matrix. In a tumour lung metastasis model utilizing fibroblast-specific CD248 gene knockout mice, CD248 gene knockout mice showed a significantly reduced ability to develop tumour lung metastasis compared to that of WT mice. Our findings demonstrate that CD248+ CAFs activate the Hippo pathway, thereby inducing CTGF expression, which in turn facilitates the collagen I milieu of the stromal matrix, which promotes NSCLC metastasis.


Sujet(s)
Fibroblastes associés au cancer , Carcinome pulmonaire non à petites cellules , Facteur de croissance du tissu conjonctif , Matrice extracellulaire , Voie de signalisation Hippo , Tumeurs du poumon , Souris knockout , Protein-Serine-Threonine Kinases , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Animaux , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/secondaire , Matrice extracellulaire/métabolisme , Souris , Facteur de croissance du tissu conjonctif/métabolisme , Facteur de croissance du tissu conjonctif/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Lignée cellulaire tumorale , Antigènes CD/métabolisme , Antigènes CD/génétique , Métastase tumorale , Transduction du signal , Régulation de l'expression des gènes tumoraux , Mouvement cellulaire , Microenvironnement tumoral
3.
Sci Rep ; 14(1): 18030, 2024 08 04.
Article de Anglais | MEDLINE | ID: mdl-39098880

RÉSUMÉ

Pancreatic stellate cells (PSC) are one source of cancer-associated fibroblasts (CAF) and play, therefore, an essential role in pancreatic ductal adenocarcinoma (PDA). Paracrine signalling between PDA cells and CAF has been widely studied, yet external influences on paracrine crosstalk are poorly understood. This study aimed to gain a deeper insight into the communication of PSC and cancer cells under different co-culture conditions via analysis of PSC gene expression profiles. Two contactless co-culture models with tumor cells from the p48-Cre; lox-stop-lox-KrasG12D/+; lox-stop-lox-Trp53R172H/+ mouse model (KPC) and murine PSC separated through a microporous membrane and grown in different compartments (standard co-culture) or on different sides of the same membrane (inverse co-culture), were established. RNA-Sequencing analysis of PSC mRNA was performed 24 h and 72 h after co-culture with KPC cells. For selected genes, results were confirmed by quantitative RT-PCR and immunocytochemistry. Standard co-culture displayed 19 differentially expressed genes (DEG) at 24 h and 52 DEG at 72 h. In inverse co-culture, 800 DEG at 24 h and 2213 DEG at 72 h were enriched. PSC showed great heterogeneity in their gene expression profiles; however, mutually regulated genes of both co-cultures, such as VCAN and CHST11, could be identified. VCAN-protein-protein interaction-network analysis revealed several shared genes between co-culture models, such as SDC4 and FN1. In conclusion, PSC show a varying susceptibility to cancer cell signals depending on the co-culture method, with intensified transcriptome changes with closer proximity.


Sujet(s)
Carcinome du canal pancréatique , Techniques de coculture , Tumeurs du pancréas , Cellules stellaires pancréatiques , Communication paracrine , Transcriptome , Cellules stellaires pancréatiques/métabolisme , Cellules stellaires pancréatiques/anatomopathologie , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Animaux , Souris , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Régulation de l'expression des gènes tumoraux , Lignée cellulaire tumorale , Humains , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Analyse de profil d'expression de gènes
4.
Genome Med ; 16(1): 98, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138551

RÉSUMÉ

BACKGROUND: Cancer-associated fibroblasts (CAFs) are the prominent cell type in the tumor microenvironment (TME), and CAF subsets have been identified in various tumors. However, how CAFs spatially coordinate other cell populations within the liver TME to promote cancer progression remains unclear. METHODS: We combined multi-region proteomics (6 patients, 24 samples), 10X Genomics Visium spatial transcriptomics (11 patients, 25 samples), and multiplexed imaging (92 patients, 264 samples) technologies to decipher the expression heterogeneity, functional diversity, spatial distribution, colocalization, and interaction of fibroblasts. The newly identified CAF subpopulation was validated by cells isolated from 5 liver cancer patients and in vitro functional assays. RESULTS: We identified a liver CAF subpopulation, marked by the expression of COL1A2, COL4A1, COL4A2, CTGF, and FSTL1, and named F5-CAF. F5-CAF is preferentially located within and around tumor nests and colocalizes with cancer cells with higher stemness in hepatocellular carcinoma (HCC). Multiplexed staining of 92 patients and the bulk transcriptome of 371 patients demonstrated that the abundance of F5-CAFs in HCC was associated with a worse prognosis. Further in vitro experiments showed that F5-CAFs isolated from liver cancer patients can promote the proliferation and stemness of HCC cells. CONCLUSIONS: We identified a CAF subpopulation F5-CAF in liver cancer, which is associated with cancer stemness and unfavorable prognosis. Our results provide potential mechanisms by which the CAF subset in the TME promotes the development of liver cancer by supporting the survival of cancer stem cells.


Sujet(s)
Fibroblastes associés au cancer , Carcinome hépatocellulaire , Tumeurs du foie , Cellules souches tumorales , Microenvironnement tumoral , Humains , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Microenvironnement tumoral/génétique , Protéomique/méthodes , Transcriptome , Régulation de l'expression des gènes tumoraux , Génomique/méthodes , Prolifération cellulaire , Analyse de profil d'expression de gènes , Lignée cellulaire tumorale , Pronostic , Multi-omique
5.
J Biochem Mol Toxicol ; 38(9): e23769, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39152098

RÉSUMÉ

Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment (TME) and can induce functional polarization of tumor macrophages. This study aimed to explore the effect of CAFs-derived exosome LINC01833 on the malignant biological behavior of non-small cell lung cancer (NSCLC) cells and its mechanism. Tumor tissues (n = 3) and adjacent noncancerous tissues (n = 3) were collected from patients with NSCLC, and fibroblasts (CAF, NF) were isolated from the two tissues. Expression of LINC01833/miR-335-5p/VAPA in NSCLC clinical tissues and cell lines was detected by RT-qPCR. Exosomes of CAFs and NFs were isolated by ultracentrifugation. Cell proliferation, migration, invasion, and M2 macrophage polarization were detected by MTT, transwell, wound-healing assay, and flow cytometry assay, while western blot was used to verify the expression of M2 macrophage polarization-related proteins. Tumor volume weight and M2 macrophage polarization were detected by tumor xenografts in nude mice. LINC01833 was highly expressed in NSCLC tumor tissues and cells. Knockdown of LINC01833 exosomes could significantly inhibit proliferation, migration, invasion of NSCLC cells, and M2 macrophage polarization of THP-1 cells, while simultaneous knockdown of miR-335-5p on the above basis could reverse the effect of knockdown of LINC01833. In vivo experiments also indicated that knockdown of LINC01833 exosomes suppressed tumor growth and M2 macrophage polarization. CAF-derived LINC01833 exosomes can promote the proliferation, migration and invasion of NSCLC cells and M2 macrophage polarization by inhibiting miR-335-5p and regulating VAPA activity.


Sujet(s)
Fibroblastes associés au cancer , Carcinome pulmonaire non à petites cellules , Exosomes , Tumeurs du poumon , Souris nude , microARN , ARN long non codant , microARN/génétique , microARN/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Humains , Exosomes/métabolisme , Exosomes/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Animaux , ARN long non codant/génétique , ARN long non codant/métabolisme , Souris , Prolifération cellulaire , Mâle , Femelle , Lignée cellulaire tumorale , Mouvement cellulaire , Cellules A549 , Souris de lignée BALB C
6.
Oncol Rep ; 52(4)2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39092576

RÉSUMÉ

Senescent cells are known to secrete proteins, including inflammatory cytokines and damage­associated molecular patterns. This phenomenon is known as the senescence­associated secretory phenotype (SASP). SASP in cancer stromal fibroblasts is involved in cancer growth and progression. Conversely, metformin, an antidiabetic drug, has been reported to inhibit SASP induction by inhibiting the activation of NF­κB, a regulator of SASP. To date, at least to the best of our knowledge, there have been no reports regarding cellular senescence in fibroblasts and tumor progression via the SASP­mediated paracrine pathway. The present study thus aimed to elucidate the induction mechanisms of SASP in radiation­induced fibroblasts and to determine its effects on cancer progression via the paracrine pathway. Furthermore, the present study aimed to determine whether controlling SASP using metformin suppresses cancer progression. A well­differentiated esophageal cancer cell line established by the authors' department and fibroblasts isolated and cultured from the non­cancerous esophageal mucosa of resected esophageal cancer cases were used for the experiments. Fibroblasts were irradiated with 8 Gy radiation, and the changes in the expression of the senescence markers, SA­ß­gal, p21, p16 and NF­κB were evaluated using immunofluorescent staining and western blot analysis in the presence or absence of metformin treatment. The culture supernatants of irradiated fibroblasts treated with metformin and those treated without metformin were collected and added to the cancer cells to evaluate their proliferative, invasive and migratory abilities. Vimentin and E­cadherin expression levels were also evaluated using immunofluorescent staining and western blot analysis. The expression levels of p16, p21 and NF­κB in irradiated fibroblasts were attenuated by treatment with metformin. Supernatants collected from irradiated fibroblasts exhibited the proliferative activity of esophageal cancer cells, and the promotion of migratory and invasion abilities, which may be due to epithelial­mesenchymal transition and changes in cell morphology. These reactions were confirmed to be suppressed by the addition of the supernatant of cultured fibroblasts pre­treated with metformin. On the whole, the present study demonstrates that fibroblasts in the cancer stroma may be involved in tumor progression through cellular senescence.


Sujet(s)
Fibroblastes associés au cancer , Prolifération cellulaire , Vieillissement de la cellule , Tumeurs de l'oesophage , Metformine , Metformine/pharmacologie , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des radiations , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/traitement médicamenteux , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des radiations , Fibroblastes associés au cancer/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Évolution de la maladie , Facteur de transcription NF-kappa B/métabolisme , Lignée cellulaire tumorale , Phénotype sécrétoire associé à la sénescence , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des radiations , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des radiations , Hypoglycémiants/pharmacologie , Fibroblastes/métabolisme , Fibroblastes/effets des radiations , Fibroblastes/effets des médicaments et des substances chimiques
7.
Biol Direct ; 19(1): 68, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160584

RÉSUMÉ

BACKGROUND: Cancer-associated fibroblasts (CAFs) have been reported that can affect cancer cell proliferation, metastasis, ferroptosis, and immune escape. METTL3-mediated N6-methyladenine (m6A) modification is involved in the tumorigenesis of colorectal cancer (CRC). Herein, we investigated whether METTL3-dependent m6A in CAFs-derived exosomes (exo) affected CRC progression. METHODS: qRT-PCR and western blotting analyses detected levels of mRNAs and proteins. Cell proliferation and metastasis were evaluated using MTT, colony formation, transwell, and wound healing assays, respectively. Cell ferroptosis was assessed by detecting cell viability and the levels of Fe+, reactive oxygen species, and glutathione after erastin treatment. Exosomes were isolated from CAFs by ultracentrifugation. The m6A modification profile was determined by methylated RNA immunoprecipitation assay and the interaction between METTL3 and ACSL3 (acyl-CoA synthetase 3) was verified using dual-luciferase reporter assay. Animal models were established for in vivo analysis. RESULTS: CAFs promoted CRC cell proliferation and metastasis, and suppressed cell ferroptosis. METTL3 was enriched in CAFs and was packaged into exosomes. The m6A modification and METTL3 expression were increased in CRC samples. Knockdown of METTL3 in CAFs-exo suppressed CRC cell proliferation and metastasis, and induced cell ferroptosis. Mechanistically, METTL3 induced ACSL3 m6A modification and stabilized its expression. The anticancer effects mediated by METTL3-silenced CAFs-exo could be rescued by ACSL3 overexpression. Moreover, in vivo assay also showed that CAFs-exo with decreased METTL3 could hinder CRC growth and metastasis in mice models. CONCLUSION: CAFs promoted the proliferation and metastasis, and restrained the ferroptosis in CRC by exosomal METTL3-elicited ACSL3 m6A modification.


Sujet(s)
Fibroblastes associés au cancer , Prolifération cellulaire , Coenzyme A ligases , Tumeurs colorectales , Exosomes , Ferroptose , Methyltransferases , Ferroptose/génétique , Methyltransferases/métabolisme , Methyltransferases/génétique , Exosomes/métabolisme , Exosomes/génétique , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/anatomopathologie , Humains , Souris , Fibroblastes associés au cancer/métabolisme , Animaux , Coenzyme A ligases/génétique , Coenzyme A ligases/métabolisme , Métastase tumorale , Lignée cellulaire tumorale , Souris nude , Mâle
8.
Cell Syst ; 15(8): 753-769.e5, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39116880

RÉSUMÉ

This study introduces a new imaging, spatial transcriptomics (ST), and single-cell RNA-sequencing integration pipeline to characterize neoplastic cell state transitions during tumorigenesis. We applied a semi-supervised analysis pipeline to examine premalignant pancreatic intraepithelial neoplasias (PanINs) that can develop into pancreatic ductal adenocarcinoma (PDAC). Their strict diagnosis on formalin-fixed and paraffin-embedded (FFPE) samples limited the single-cell characterization of human PanINs within their microenvironment. We leverage whole transcriptome FFPE ST to enable the study of a rare cohort of matched low-grade (LG) and high-grade (HG) PanIN lesions to track progression and map cellular phenotypes relative to single-cell PDAC datasets. We demonstrate that cancer-associated fibroblasts (CAFs), including antigen-presenting CAFs, are located close to PanINs. We further observed a transition from CAF-related inflammatory signaling to cellular proliferation during PanIN progression. We validate these findings with single-cell high-dimensional imaging proteomics and transcriptomics technologies. Altogether, our semi-supervised learning framework for spatial multi-omics has broad applicability across cancer types to decipher the spatiotemporal dynamics of carcinogenesis.


Sujet(s)
Fibroblastes associés au cancer , Carcinogenèse , Carcinome du canal pancréatique , Tumeurs du pancréas , Humains , Tumeurs du pancréas/génétique , Carcinogenèse/génétique , Fibroblastes associés au cancer/métabolisme , Carcinome du canal pancréatique/génétique , Microenvironnement tumoral/génétique , Analyse sur cellule unique/méthodes , Transcriptome/génétique , Régulation de l'expression des gènes tumoraux/génétique , Épithélioma in situ/génétique , Épithélioma in situ/anatomopathologie
9.
Cell Rep Methods ; 4(8): 100841, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39127046

RÉSUMÉ

Cell-type-specific domains are the anatomical domains in spatially resolved transcriptome (SRT) tissues where particular cell types are enriched coincidentally. It is challenging to use existing computational methods to detect specific domains with low-proportion cell types, which are partly overlapped with or even inside other cell-type-specific domains. Here, we propose De-spot, which synthesizes segmentation and deconvolution as an ensemble to generate cell-type patterns, detect low-proportion cell-type-specific domains, and display these domains intuitively. Experimental evaluation showed that De-spot enabled us to discover the co-localizations between cancer-associated fibroblasts and immune-related cells that indicate potential tumor microenvironment (TME) domains in given slices, which were obscured by previous computational methods. We further elucidated the identified domains and found that Srgn may be a critical TME marker in SRT slices. By deciphering T cell-specific domains in breast cancer tissues, De-spot also revealed that the proportions of exhausted T cells were significantly increased in invasive vs. ductal carcinoma.


Sujet(s)
Tumeurs du sein , Transcriptome , Microenvironnement tumoral , Microenvironnement tumoral/immunologie , Humains , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/immunologie , Femelle , Analyse de profil d'expression de gènes/méthodes , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Souris , Animaux , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie
10.
J Cancer Res Clin Oncol ; 150(8): 388, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39120743

RÉSUMÉ

BACKGROUND: Cancer-associated fibroblasts (CAFs) play a crucial role in the progression of colorectal cancer (CRC). However, the impact of CAF subpopulation trajectory differentiation on CRC remains unclear. METHODS: In this study, we first explored the trajectory differences of CAFs subpopulations using bulk and integrated single-cell sequencing data, and then performed consensus clustering of CRC samples based on the trajectory differential genes of CAFs subpopulations. Subsequently, we analyzed the heterogeneity of CRC subtypes using bioinformatics. Finally, we constructed relevant prognostic signature using machine learning and validated them using spatial transcriptomic data. RESULTS: Based on the differential genes of CAFs subpopulation trajectory differentiation, we identified two CRC subtypes (C1 and C2) in this study. Compared to C1, C2 exhibited worse prognosis, higher immune evasion microenvironment and high CAF characteristics. C1 was primarily associated with metabolism, while C2 was primarily associated with cell metastasis and immune regulation. By combining 101 combinations of 10 machine learning algorithms, we developed a High-CAF risk signatures (HCAFRS) based on the C2 characteristic gene. HCAFRS was an independent prognostic factor for CRC and, when combined with clinical parameters, significantly predicted the overall survival of CRC patients. HCAFRS was closely associated with epithelial-mesenchymal transition, angiogenesis, and hypoxia. Furthermore, the risk score of HCAFRS was mainly derived from CAFs and was validated in the spatial transcriptomic data. CONCLUSION: In conclusion, HCAFRS has the potential to serve as a promising prognostic indicator for CRC, improving the quality of life for CRC patients.


Sujet(s)
Fibroblastes associés au cancer , Tumeurs colorectales , Microenvironnement tumoral , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Humains , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/métabolisme , Pronostic , Microenvironnement tumoral/génétique , Analyse de regroupements , Apprentissage machine , Marqueurs biologiques tumoraux/génétique , Transcriptome , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes/méthodes , Femelle , Mâle
11.
Clin Exp Pharmacol Physiol ; 51(10): e13914, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39139001

RÉSUMÉ

The pleiotropic effect of cancer-associated fibroblasts (CAFs) on tumour progression depends on the environment. circFARP1 is critical for CAFs-induced gemcitabine (GEM) resistance in pancreatic cancer. Its specific role and mechanism in non-small cell lung cancer (NSCLC) have not been reported yet. We prepared a cancer-associated fibroblasts-conditioned medium (CAF-CM) to incubate the A549 cells. Quantitative real-time polymerase chain reaction was used to detect RNA levels. We detected protein expression by immunohistochemistry, immunocytochemistry, western blot and immunofluorescence. We also detected the targeting impact between circFARP1, miR-338-3p and SRY-box transcription factor 4 (SOX4) by using dual-luciferase reporter and RNA pull-down assays. We determined cell proliferation, migration and invasion capabilities through Cell Counting Kit-8 and transwell assays. In addition, we measured tumour volume and weight in vivo by establishing a xenograft tumour model. CircFARP1 levels were remarkably high in the CAFs. The transfection experiments found that circFARP1 downregulation in CAFs caused migration, proliferation and invasion inhibition of CAFs and A549 cells, whereas inhibiting miR-38-3p or overexpressing SOX4 in CAFs could significantly reverse the inhibition. In vivo study in nude mice confirmed that CAFs could promote NSCLC tumour growth and knockdown of circFARP1 could inhibit tumour growth of NSCLC, whereas miR-38-3p downregulation or SOX4 overexpression could significantly reverse the inhibition. circFARP1 promotes NSCLC development by stimulating miR-338-3p/SOX4 signalling axis to regulate CAFs.


Sujet(s)
Fibroblastes associés au cancer , Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , microARN , Invasion tumorale , ARN circulaire , Facteurs de transcription SOX-C , Humains , microARN/génétique , microARN/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Facteurs de transcription SOX-C/génétique , Facteurs de transcription SOX-C/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Animaux , ARN circulaire/génétique , ARN circulaire/métabolisme , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Prolifération cellulaire/génétique , Souris , Cellules A549 , Métastase tumorale , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux , Souris nude , Mâle
12.
Front Immunol ; 15: 1433679, 2024.
Article de Anglais | MEDLINE | ID: mdl-39086477

RÉSUMÉ

Introduction: Fibroblast activation protein (FAP) overexpression on cancer-associated fibroblasts (CAFs) is associated with poor prognosis and worse clinical outcomes. Selective ablation of pro-tumorgenic FAP+ stromal cells with CAR-T cells may be a new therapeutic strategy. However, the clinical use of FAP-CAR T cells is suggested to proceed with caution for occasional poor efficacy and induction of on-target off-tumor toxicity (OTOT), including lethal osteotoxicity and cachexia. Hence, more investigations and preclinical trials are required to optimize the FAP-CAR T cells and to approve their safety and efficacy. Methods: In this study, we designed second-generation CAR T cells targeting FAP with 4-1BB as a co-stimulatory molecule, and tested their cytotoxicity against FAP-positive cells (hFAP-HT1080 cells and a variety of primary CAFs) in vitro and in Cell line-derived xenograft (CDX) and a patient-derived xenograft (PDX) model. Results: Results showed that our FAP-CAR T cells were powerfully potent in killing human and murine FAP-positive tumor cells and CAFs in multiple types of tumors in BALB/c and C57BL/6 mice and in patient-derived xenografts (PDX) model. And they were proved to be biologically safe and exhibit low-level OTOT. Discussion: Taken together, the human/murine cross-reactive FAP-CAR T cells were powerfully potent in killing human and murine FAP positive tumor cells and CAFs. They were biologically safe and exhibit low-level OTOT, warranting further clinical investigation into our FAP-CAR T cells.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Animaux , Femelle , Humains , Souris , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/métabolisme , Lignée cellulaire tumorale , Réactions croisées/immunologie , Endopeptidases , Gelatinases/immunologie , Gelatinases/métabolisme , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Protéines membranaires/immunologie , Protéines membranaires/génétique , Souris de lignée BALB C , Souris de lignée C57BL , Tumeurs/immunologie , Tumeurs/thérapie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Serine endopeptidases/immunologie , Serine endopeptidases/génétique , Serine endopeptidases/métabolisme , Lymphocytes T/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39125655

RÉSUMÉ

Pancreatic cancer is a very aggressive disease with a dismal prognosis. The tumor microenvironment exerts immunosuppressive activities through the secretion of several cytokines, including interleukin (IL)-1. The IL-1/IL-1 receptor (IL-1R) axis is a key regulator in tumor-promoting T helper (Th)2- and Th17-type inflammation. Th2 cells are differentiated by dendritic cells endowed with Th2-polarizing capability by the thymic stromal lymphopoietin (TSLP) that is secreted by IL-1-activated cancer-associated fibroblasts (CAFs). Th17 cells are differentiated in the presence of IL-1 and other IL-1-regulated cytokines. In pancreatic cancer, the use of a recombinant IL-1R antagonist (IL1RA, anakinra, ANK) in in vitro and in vivo models has shown efficacy in targeting the IL-1/IL-1R pathway. In this study, we have developed sphingomyelin nanosystems (SNs) loaded with ANK (ANK-SNs) to compare their ability to inhibit Th2- and Th17-type inflammation with that of the free drug in vitro. We found that ANK-SNs inhibited TSLP and other pro-tumor cytokines released by CAFs at levels similar to ANK. Importantly, inhibition of IL-17 secretion by Th17 cells, but not of interferon-γ, was significantly higher, and at lower concentrations, with ANK-SNs compared to ANK. Collectively, the use of ANK-SNs might be beneficial in reducing the effective dose of the drug and its toxic effects.


Sujet(s)
Antagoniste du récepteur à l'interleukine-1 , Interleukine-1 , Tumeurs du pancréas , Sphingomyéline , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Humains , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Interleukine-1/métabolisme , Sphingomyéline/métabolisme , Cytokines/métabolisme , Lignée cellulaire tumorale , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules Th17/immunologie , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th2/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques
14.
J Gastric Cancer ; 24(3): 300-315, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38960889

RÉSUMÉ

PURPOSE: Gastric cancer (GC) is among the deadliest malignancies and the third leading cause of cancer-related deaths worldwide. Galectin-1 (Gal-1) is a primary protein secreted by cancer-associated fibroblasts (CAFs); however, its role and mechanisms of action of Gal-1 in GC remain unclear. In this study, we stimulated GC cells with exogenous human recombinant galectin-1 protein (rhGal-1) to investigate its effects on the proliferation, migration, and resistance to cisplatin. MATERIALS AND METHODS: We used simulated rhGal-1 protein as a paracrine factor produced by CAFs to induce GC cells and investigated its promotional effects and mechanisms in GC progression and cisplatin resistance. Immunohistochemical (IHC) assay confirmed that Gal-1 expression was associated with clinicopathological parameters and correlated with the expression of neuropilin-1 (NRP-1), c-JUN, and Wee1. RESULTS: Our study reveals Gal-1 expression was significantly associated with poor outcomes. Gal-1 boosts the proliferation and metastasis of GC cells by activating the NRP-1/C-JUN/Wee1 pathway. Gal-1 notably increases GC cell resistance to cisplatin The NRP-1 inhibitor, EG00229, effectively counteracts these effects. CONCLUSIONS: These findings revealed a potential mechanism by which Gal-1 promotes GC growth and contributes to chemoresistance, offering new therapeutic targets for the treatment of GC.


Sujet(s)
Prolifération cellulaire , Cisplatine , Résistance aux médicaments antinéoplasiques , Galectine 1 , Neuropiline 1 , Tumeurs de l'estomac , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Humains , Galectine 1/génétique , Galectine 1/métabolisme , Cisplatine/pharmacologie , Cisplatine/usage thérapeutique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Neuropiline 1/métabolisme , Neuropiline 1/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mâle , Femelle , Évolution de la maladie , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Transduction du signal/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Souris , Animaux , Mouvement cellulaire/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/anatomopathologie
15.
Sci Transl Med ; 16(756): eadi0178, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39018369

RÉSUMÉ

Perineural invasion (PNI) is a biological characteristic commonly observed in pancreatic cancer. Although PNI plays a key role in pancreatic cancer metastasis, recurrence, and poor postoperative survival, its mechanism is largely unclarified. Clinical sample analysis and endoscopic ultrasonographic elasticity scoring indicated that cancer-associated fibroblasts (CAFs) were closely related to the occurrence of PNI. Furthermore, CAF-derived extracellular vesicles (EVs) were involved in PNI in dorsal root ganglion coculture and mouse sciatic nerve models. Next, we demonstrated that CAFs promoted PNI through extracellular vesicle transmission of PNI-associated transcript (PIAT). Mechanistically, PIAT specifically bound to YBX1 and blocked the YBX1-Nedd4l interaction to inhibit YBX1 ubiquitination and degradation. Furthermore, PIAT enhanced the binding of YBX1 and PNI-associated mRNAs in a 5-methylcytosine (m5C)-dependent manner. Mutation of m5C recognition motifs in YBX1 or m5C sites in downstream target genes reversed PIAT-mediated PNI. Consistent with these findings, analyses using a KPC mouse model demonstrated that the PIAT/YBX1 axis enhanced PNI through m5C modification. Clinical data suggested that the PIAT expression in the serum EVs of patients with pancreatic cancer was associated with the degree of neural invasion and prognosis. Our study revealed the important role of the PIAT/YBX1 signaling axis in the tumor microenvironment (TME) in promoting tumor cell PNI and provided a new target for precise interference with CAFs and RNA methylation in the TME to suppress PNI in pancreatic cancer.


Sujet(s)
Fibroblastes associés au cancer , Modèles animaux de maladie humaine , Vésicules extracellulaires , Tumeurs du pancréas , Animaux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Vésicules extracellulaires/métabolisme , Humains , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Souris , Mâle , Lignée cellulaire tumorale , Invasion tumorale , Femelle , Protéine-1 de liaison à la boîte Y
16.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-39000453

RÉSUMÉ

Regulatory T cells (Tregs) possess unique immunosuppressive activity among CD4-positive T cells. Tregs are ubiquitously present in mammals and function to calm excessive immune responses, thereby suppressing allergies or autoimmune diseases. On the other hand, due to their immunosuppressive function, Tregs are thought to promote cancer progression. The tumor microenvironment (TME) is a multicellular system composed of many cell types, including tumor cells, infiltrating immune cells, and cancer-associated fibroblasts (CAFs). Within this environment, Tregs are recruited by chemokines and metabolic factors and impede effective anti-tumor responses. However, in some cases, their presence can also improve patient's survival rates. Their functional consequences may vary across tumor types, locations, and stages. An in-depth understanding of the precise roles and mechanisms of actions of Treg is crucial for developing effective treatments, emphasizing the need for further investigation and validation. This review aims to provide a comprehensive overview of the complex and multifaceted roles of Tregs within the TME, elucidating cellular communications, signaling pathways, and their impacts on tumor progression and highlighting their potential anti-tumor mechanisms through interactions with functional molecules.


Sujet(s)
Évolution de la maladie , Tumeurs , Lymphocytes T régulateurs , Microenvironnement tumoral , Humains , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Microenvironnement tumoral/immunologie , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Animaux , Transduction du signal , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/immunologie , Fibroblastes associés au cancer/anatomopathologie
17.
Mol Genet Genomics ; 299(1): 70, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39017768

RÉSUMÉ

Lung adenocarcinoma (LUAD) is the leading cause of cancer-related death worldwide. Cancer-associated fibroblasts (CAFs) are a special type of fibroblasts, which play an important role in the development and immune escape of tumors. Weighted gene co-expression network analysis (WGCNA) was used to construct the co-expression module. In combination with univariate Cox regression and analysis of least absolute shrinkage operator (LASSO), characteristics associated with CAFs were developed for a prognostic model. The migration and proliferation of lung cancer cells were evaluated in vitro. Finally, the expression levels of proteins were analyzed by Western blot. LASSO Cox regression algorithm was then performed to select hub genes. Finally, a total of 2 Genes (COL5A2, COL6A2) were obtained. We then divided LUAD patients into high- and low-risk groups based on CAFs risk scores. Survival analysis, CAFs score correlation analysis and tumor mutation load analysis showed that COL5A2 and COL6A2 were high-risk genes for LUAD. Human Protein Atlas (HPA), western blot and PCR results showed that COL5A2 and COL6A2 were up-regulated in LUAD tissues. When COL5A2 and COL6A2 were knocked down, the proliferation, invasion and migration of lung cancer cells were significantly decreased. Finally, COL5A2 can affect LUAD progression through the Wnt/ß-Catenin and TGF-ß signaling pathways. Our CAFs risk score model offers a new approach for predicting the prognosis of LUAD patients. Furthermore, the identification of high-risk genes COL5A2 and COL6A2 and drug sensitivity analysis can provide valuable candidate clues for clinical treatment of LUAD.


Sujet(s)
Adénocarcinome pulmonaire , Fibroblastes associés au cancer , Tumeurs du poumon , Humains , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/mortalité , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Pronostic , Régulation de l'expression des gènes tumoraux , Immunothérapie/méthodes , Prolifération cellulaire/génétique , Marqueurs biologiques tumoraux/génétique , Collagène de type V/génétique , Mouvement cellulaire/génétique , Lignée cellulaire tumorale , Mâle , Femelle , Analyse de survie
18.
Sci Adv ; 10(27): eadl1197, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38959305

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) is characterized by increasing fibrosis, which can enhance tumor progression and spread. Here, we undertook an unbiased temporal assessment of the matrisome of the highly metastatic KPC (Pdx1-Cre, LSL-KrasG12D/+, LSL-Trp53R172H/+) and poorly metastatic KPflC (Pdx1-Cre, LSL-KrasG12D/+, Trp53fl/+) genetically engineered mouse models of pancreatic cancer using mass spectrometry proteomics. Our assessment at early-, mid-, and late-stage disease reveals an increased abundance of nidogen-2 (NID2) in the KPC model compared to KPflC, with further validation showing that NID2 is primarily expressed by cancer-associated fibroblasts (CAFs). Using biomechanical assessments, second harmonic generation imaging, and birefringence analysis, we show that NID2 reduction by CRISPR interference (CRISPRi) in CAFs reduces stiffness and matrix remodeling in three-dimensional models, leading to impaired cancer cell invasion. Intravital imaging revealed improved vascular patency in live NID2-depleted tumors, with enhanced response to gemcitabine/Abraxane. In orthotopic models, NID2 CRISPRi tumors had less liver metastasis and increased survival, highlighting NID2 as a potential PDAC cotarget.


Sujet(s)
Carcinome du canal pancréatique , Tumeurs du pancréas , Protéomique , Animaux , Humains , Souris , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/génétique , Molécules d'adhérence cellulaire , Lignée cellulaire tumorale , Désoxycytidine/analogues et dérivés , Désoxycytidine/pharmacologie , Modèles animaux de maladie humaine , Fibrose , , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Protéomique/méthodes
19.
Genes (Basel) ; 15(7)2024 Jul 22.
Article de Anglais | MEDLINE | ID: mdl-39062740

RÉSUMÉ

Heme, an iron-containing tetrapyrrole, is essential in almost all organisms. Heme biosynthesis needs to be precisely regulated particularly given the potential cytotoxicity of protoporphyrin IX, the intermediate preceding heme formation. Here, we report on the porphyrin intermediate accumulation within the tumor microenvironment (TME), which we propose to result from dysregulation of heme biosynthesis concomitant with an enhanced cancer survival dependence on mid-step genes, a process we recently termed "Porphyrin Overdrive". Specifically, porphyrins build up in both lung cancer cells and stromal cells in the TME. Within the TME's stromal cells, evidence supports cancer-associated fibroblasts (CAFs) actively producing porphyrins through an imbalanced pathway. Conversely, normal tissues exhibit no porphyrin accumulation, and CAFs deprived of tumor cease porphyrin overproduction, indicating that both cancer and tumor-stromal porphyrin overproduction is confined to the cancer-specific tissue niche. The clinical relevance of our findings is implied by establishing a correlation between imbalanced porphyrin production and overall poorer survival in more aggressive cancers. These findings illuminate the anomalous porphyrin dynamics specifically within the tumor microenvironment, suggesting a potential target for therapeutic intervention.


Sujet(s)
Porphyrines , Microenvironnement tumoral , Humains , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Lignée cellulaire tumorale , Hème/métabolisme , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Porphyrines/métabolisme , Protoporphyrines/métabolisme , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie
20.
J Cancer Res Clin Oncol ; 150(7): 356, 2024 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-39033089

RÉSUMÉ

PURPOSE: Neoadjuvant chemotherapy serves as an effective strategy for treating osteosarcoma (OS) not only by targeting cancerous cells but also by influencing the tumor's immune and stromal elements. Gaining insights into how chemotherapy reshapes the tumor's local environment is crucial for advancing OS treatment protocols. METHODS: Using single-cell RNA sequencing, this study analyzed tumor samples from patients with advanced osteosarcoma collected both before and after chemotherapy. RESULTS: The results revealed that chemotherapy caused the remaining OS cells to express higher levels of genes associated with stemness. Additionally, this process enhances the presence of cancer-associated fibroblasts, increasing their ability to modify the extracellular matrix (ECM). Chemotherapy also increases the number of endothelial cells, albeit with compromised differentiation capabilities. Importantly, the treatment reduced the immune cell population, including myeloid and T/NK cells, particularly impacting the subpopulations with tumor-fighting capabilities. CONCLUSION: These findings highlight the complex reaction of the tumor environment to chemotherapy, providing valuable insights into how chemotherapy influences OS cells and the tumor microenvironment (TME). This knowledge is essential for understanding OS resistance mechanisms to treatments, potentially guiding the development of novel therapies for managing advanced OS.


Sujet(s)
Tumeurs osseuses , Ostéosarcome , Analyse sur cellule unique , Transcriptome , Microenvironnement tumoral , Ostéosarcome/traitement médicamenteux , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Analyse sur cellule unique/méthodes , Traitement néoadjuvant , Fibroblastes associés au cancer/anatomopathologie , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Femelle , Mâle , Adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE