Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 205
Filtrer
1.
Nat Commun ; 15(1): 5352, 2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38914547

RÉSUMÉ

Immune checkpoint blockade (ICB) approaches have changed the therapeutic landscape for many tumor types. However, half of cutaneous squamous cell carcinoma (cSCC) patients remain unresponsive or develop resistance. Here, we show that, during cSCC progression in male mice, cancer cells acquire epithelial/mesenchymal plasticity and change their immune checkpoint (IC) ligand profile according to their features, dictating the IC pathways involved in immune evasion. Epithelial cancer cells, through the PD-1/PD-L1 pathway, and mesenchymal cancer cells, through the CTLA-4/CD80 and TIGIT/CD155 pathways, differentially block antitumor immune responses and determine the response to ICB therapies. Accordingly, the anti-PD-L1/TIGIT combination is the most effective strategy for blocking the growth of cSCCs that contain both epithelial and mesenchymal cancer cells. The expression of E-cadherin/Vimentin/CD80/CD155 proteins in cSCC, HNSCC and melanoma patient samples predicts response to anti-PD-1/PD-L1 therapy. Collectively, our findings indicate that the selection of ICB therapies should take into account the epithelial/mesenchymal features of cancer cells.


Sujet(s)
Antigène CD274 , Carcinome épidermoïde , Plasticité cellulaire , Transition épithélio-mésenchymateuse , Inhibiteurs de points de contrôle immunitaires , Immunothérapie , Tumeurs cutanées , Animaux , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/thérapie , Tumeurs cutanées/traitement médicamenteux , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/thérapie , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/traitement médicamenteux , Souris , Humains , Antigène CD274/métabolisme , Antigène CD274/antagonistes et inhibiteurs , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Mâle , Immunothérapie/méthodes , Transition épithélio-mésenchymateuse/immunologie , Plasticité cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Antigène CTLA-4/antagonistes et inhibiteurs , Antigène CTLA-4/métabolisme , Antigène CTLA-4/immunologie , Récepteurs viraux/métabolisme , Récepteurs viraux/génétique , Antigène CD80/métabolisme , Récepteurs immunologiques/métabolisme
3.
Life Sci ; 336: 122294, 2024 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-38007147

RÉSUMÉ

Doublecortin-like kinase 1 (DCLK1), a significant constituent of the protein kinase superfamily and the doublecortin family, has been recognized as a prooncogenic factor that exhibits a strong association with the malignant progression and clinical prognosis of various cancers. DCLK1 serves as a stem cell marker that governs tumorigenesis, tumor cell reprogramming, and epithelial-mesenchymal transition. Multiple studies have indicated the capable of DCLK1 in regulating the DNA damage response and facilitating DNA damage repair. Additionally, DCLK1 is involved in the regulation of the immune microenvironment and the promotion of tumor immune evasion. Recently, DCLK1 has emerged as a promising therapeutic target for a multitude of cancers. Several small-molecule inhibitors of DCLK1 have been identified. Nevertheless, the biological roles of DCLK1 are mainly ambiguous, particularly with the disparities between its α- and ß-form transcripts in the malignant progression of cancers, which impedes the development of more precisely targeted drugs. This article focuses on tumor stem cells, tumor epithelial-mesenchymal transition, the DNA damage response, and the tumor microenvironment to provide a comprehensive overview of the association between DCLK1 and tumor malignant progression, address unsolved questions and current challenges, and project future directions for targeting DCLK1 for the diagnosis and treatment of cancers.


Sujet(s)
Kinases de type doublecortine , Tumeurs , Tumeurs/diagnostic , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Tumeurs/génétique , Kinases de type doublecortine/antagonistes et inhibiteurs , Kinases de type doublecortine/génétique , Kinases de type doublecortine/immunologie , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/immunologie , Cellules souches tumorales , Réparation de l'ADN/génétique , Réparation de l'ADN/immunologie , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Échappement de la tumeur à la surveillance immunitaire/génétique , Inhibiteurs de protéines kinases/usage thérapeutique , Humains , Isoformes de protéines
4.
Toxicol Lett ; 383: 141-151, 2023 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-37394155

RÉSUMÉ

Long-term inhalation of silica nanoparticles (SiNPs) can induce pulmonary fibrosis (PF), nevertheless, the potential mechanisms remain elusive. Herein, we constructed a three-dimensional (3D) co-culture model by using Matrigel to investigate the interaction among different cells and potential regulatory mechanisms after SiNPs exposure. Methodologically, we dynamically observed the changes in cell morphology and migration after exposure to SiNPs by co-culturing mouse monocytic macrophages (RAW264.7), human non-small cell lung cancer cells (A549), and medical research council cell strain-5 (MRC-5) in Matrigel for 24 h. Subsequently, we detected the expression of nuclear factor kappa B (NF-κB), inflammatory factor and epithelial-mesenchymal transition (EMT) markers. The results showed that SiNPs produced toxic effects on cells. In the 3D co-culture state, the cell's movement velocity and displacement increased, and the cell migration ability was enhanced. Meanwhile, the expression of inflammatory factor tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) were upregulated, the epithelial marker E-cadherin (E-cad) was downregulated, the mesenchymal marker N-cadherin (N-cad) and myofibroblast marker alpha-smooth muscle actin (α-SMA) expression were upregulated, while NF-κB expression was also upregulated after SiNPs exposure. We further found that cells were more prone to transdifferentiate into myofibroblasts in the 3D co-culture state. Conversely, utilizing the NF-κB-specific inhibitor BAY 11-7082 effectively downregulated the expression of TNF-α, IL-6, interleukin-1ß (IL-1ß), N-cad, α-SMA, collagen-I (COL I), and fibronectin (FN), the expression of E-cad was upregulated. These findings suggest that NF-κB is involved in regulating SiNPs-induced inflammatory, EMT, and fibrosis in the 3D co-culture state.


Sujet(s)
Transition épithélio-mésenchymateuse , Fibrose , Maladies pulmonaires , Nanoparticules , Silice , Animaux , Humains , Souris , Carcinome pulmonaire non à petites cellules , Techniques de coculture , Transition épithélio-mésenchymateuse/immunologie , Fibrose/étiologie , Fibrose/immunologie , Interleukine-6 , Tumeurs du poumon , Nanoparticules/toxicité , Facteur de transcription NF-kappa B/métabolisme , Silice/toxicité , Facteur de nécrose tumorale alpha/métabolisme , Maladies pulmonaires/étiologie , Maladies pulmonaires/immunologie
5.
Front Immunol ; 13: 941608, 2022.
Article de Anglais | MEDLINE | ID: mdl-35990621

RÉSUMÉ

Chronic rhinosinusitis with nasal polyps (CRSwNP) is caused by prolonged inflammation of the paranasal sinus mucosa. The epithelial to mesenchymal transition (EMT) is involved in the occurrence and development of CRSwNP. The T-cell immunoglobulin domain and the mucin domain 4 (TIM-4) is closely related to chronic inflammation, but its mechanism in CRSwNP is poorly understood. In our study, we found that TIM-4 was increased in the sinonasal mucosa of CRSwNP patients and, especially, in macrophages. TIM-4 was positively correlated with α-SMA but negatively correlated with E-cadherin in CRS. Moreover, we confirmed that TIM-4 was positively correlated with the clinical parameters of the Lund-Mackay and Lund-Kennedy scores. In the NP mouse model, administration of TIM-4 neutralizing antibody significantly reduced the polypoid lesions and inhibited the EMT process. TIM-4 activation by stimulating with tissue extracts of CRSwNP led to a significant increase of TGF-ß1 expression in macrophages in vitro. Furthermore, coculture of macrophages and human nasal epithelial cells (hNECs) results suggested that the overexpression of TIM-4 in macrophages made a contribution to the EMT process in hNECs. Mechanistically, TIM-4 upregulated TGF-ß1 expression in macrophages via the ROS/p38 MAPK/Egr-1 pathway. In conclusion, TIM-4 contributes to the EMT process and aggravates the development of CRSwNP by facilitating the production of TGF-ß1 in macrophages. Inhibition of TIM-4 expression suppresses nasal polyp formation, which might provide a new therapeutic approach for CRSwNP.


Sujet(s)
Transition épithélio-mésenchymateuse , Macrophages , Protéines membranaires , Muqueuse nasale , Polypes du nez , Facteur de croissance transformant bêta-1 , Animaux , Maladie chronique , Cellules épithéliales/immunologie , Transition épithélio-mésenchymateuse/immunologie , Humains , Inflammation/immunologie , Macrophages/immunologie , Protéines membranaires/immunologie , Souris , Muqueuse nasale/immunologie , Polypes du nez/immunologie , Sinus de la face/immunologie , Rhinite/immunologie , Sinusite/immunologie , Facteur de croissance transformant bêta-1/immunologie
6.
Cancer Lett ; 539: 215712, 2022 07 28.
Article de Anglais | MEDLINE | ID: mdl-35490920

RÉSUMÉ

The epithelial-to-mesenchymal transition (EMT) is involved in cancer metastasis; nevertheless, interferon (IFN)-γ induces anticancer activities by causing cell growth suppression, cytotoxicity, and migration inhibition. Regarding the poor response to exogenously administered IFN-γ as anticancer therapy, it was hypothesized that malignant cells may acquire a means of escaping from IFN-γ immunosurveillance, likely through an EMT-related process. A genomic analysis of human lung cancers revealed a negative link between the EMT and IFN-γ signaling, while compared to human lung adenocarcinoma A549 cells, IFN-γ-hyporesponsive AS2 cells exhibited mesenchymal characteristics. Chemically, physically, and genetically engineered EMT attenuated IFN-γ-induced IFN regulatory factor 1 transactivation. Poststimulation of transforming growth factor-ß induced the EMT and also selectively retarded IFN-γ-responsive gene expression as well as IFN-γ-induced signal transducer and activator of transcription 1 activation, major histocompatibility complex I, and CD54 expression, cell migration/invasion inhibition, and direct/indirect cytotoxicity. Without changes in IFN-γ receptors, excessive oxidative activation of Src homology-2 containing phosphatase 2 (SHP2) in cells undergoing the EMT primarily caused cellular hyporesponsiveness to IFN-γ signaling and cytotoxicity, while combining an SHP2 inhibitor or antioxidant sensitized EMT-associated AS2 and mesenchymal A549 cells to IFN-γ-induced priming effects on tumor necrosis factor-related apoptosis-inducing ligand cytotoxicity. In cell line-derived xenograft models, combined treatment with IFN-γ and an SHP2 inhibitor induced enhanced anticancer activities. These results imply that EMT-associated SHP2 activation inhibits IFN-γ signaling, facilitating lung cancer cell escape from IFN-γ immunosurveillance.


Sujet(s)
Interféron gamma , Tumeurs du poumon , Lignée cellulaire tumorale , Mouvement cellulaire/immunologie , Transition épithélio-mésenchymateuse/immunologie , Humains , Surveillance immunologique , Interféron gamma/immunologie , Interféron gamma/pharmacologie , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie
7.
Commun Biol ; 5(1): 41, 2022 01 11.
Article de Anglais | MEDLINE | ID: mdl-35017635

RÉSUMÉ

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive malignancies with a 5-year survival rate of only 9%. Despite the fact that changes in glycosylation patterns during tumour progression have been reported, no systematic approach has been conducted to evaluate its potential for patient stratification. By analysing publicly available transcriptomic data of patient samples and cell lines, we identified here two specific glycan profiles in PDAC that correlated with progression, clinical outcome and epithelial to mesenchymal transition (EMT) status. These different glycan profiles, confirmed by glycomics, can be distinguished by the expression of O-glycan fucosylated structures, present only in epithelial cells and regulated by the expression of GALNT3. Moreover, these fucosylated glycans can serve as ligands for DC-SIGN positive tumour-associated macrophages, modulating their activation and inducing the production of IL-10. Our results show mechanisms by which the glyco-code contributes to the tolerogenic microenvironment in PDAC.


Sujet(s)
Carcinome du canal pancréatique , Glycoprotéines , Tumeurs du pancréas , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/anatomopathologie , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/immunologie , Glycoprotéines/composition chimique , Glycoprotéines/génétique , Glycoprotéines/immunologie , Glycoprotéines/métabolisme , Glycosylation , Humains , Pancréas/métabolisme , Pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Polyosides/composition chimique , Polyosides/génétique , Polyosides/immunologie , Polyosides/métabolisme
8.
Oncogene ; 41(1): 26-36, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34667277

RÉSUMÉ

The EMT (epithelial-to-mesenchymal-transition) subtype of gastric cancer (GC) is associated with poor treatment responses and unfavorable clinical outcomes. Despite the broad physiological roles of the micro-RNA (miR)-200 family, they largely serve to maintain the overall epithelial phenotype. However, during late-stage gastric tumorigenesis, members of the miR-200 family are markedly suppressed, resulting in the transition to the mesenchymal state and the acquisition of invasive properties. As such, the miR-200 family represents a robust molecular marker of EMT, and subsequently, disease severity and prognosis. Most reports have studied the effect of single miR-200 family member knockdown. Here, we employ a multiplex CRISPR/Cas9 system to generate a complete miR-200 family knockout (FKO) to investigate their collective and summative role in regulating key cellular processes during GC pathogenesis. Genetic deletion of all miR-200s in the human GC cell lines induced potent morphological alterations, G1/S cell cycle arrest, increased senescence-associated ß-galactosidase (SA-ß-Gal) activity, and aberrant metabolism, collectively resembling the senescent phenotype. Coupling RNA-seq data with publicly available datasets, we revealed a clear separation of senescent and non-senescent states amongst FKO cells and control cells, respectively. Further analysis identified key senescence-associated secretory phenotype (SASP) components in FKO cells and a positive feedback loop for maintenance of the senescent state controlled by activation of TGF-ß and TNF-α pathways. Finally, we showed that miR-200 FKO associated senescence in cancer epithelial cells significantly recruited stromal cells in the tumor microenvironment. Our work has identified a new role of miR-200 family members which function as an integrated unit serving to link senescence with EMT, two major conserved biological processes.


Sujet(s)
Vieillissement de la cellule/immunologie , Transition épithélio-mésenchymateuse/immunologie , Régulation de l'expression des gènes tumoraux/génétique , microARN/métabolisme , Tumeurs de l'estomac/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Humains , Pronostic , Tumeurs de l'estomac/anatomopathologie , Microenvironnement tumoral
9.
Front Immunol ; 12: 724200, 2021.
Article de Anglais | MEDLINE | ID: mdl-34917071

RÉSUMÉ

The transcription factor Snail1, a key inducer of epithelial-mesenchymal transition (EMT), plays a critical role in tumor metastasis. Its stability is strictly controlled by multiple intracellular signal transduction pathways and the ubiquitin-proteasome system (UPS). Increasing evidence indicates that methylation and acetylation of Snail1 also affects tumor metastasis. More importantly, Snail1 is involved in tumor immunosuppression by inducing chemokines and immunosuppressive cells into the tumor microenvironment (TME). In addition, some immune checkpoints potentiate Snail1 expression, such as programmed death ligand 1 (PD-L1) and T cell immunoglobulin 3 (TIM-3). This mini review highlights the pathways and molecules involved in maintenance of Snail1 level and the significance of Snail1 in tumor immune evasion. Due to the crucial role of EMT in tumor metastasis and tumor immunosuppression, comprehensive understanding of Snail1 function may contribute to the development of novel therapeutics for cancer.


Sujet(s)
Invasion tumorale/anatomopathologie , Facteurs de transcription de la famille Snail/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie , Animaux , Transition épithélio-mésenchymateuse/immunologie , Humains , Microenvironnement tumoral/immunologie
10.
Front Immunol ; 12: 769242, 2021.
Article de Anglais | MEDLINE | ID: mdl-34819935

RÉSUMÉ

Complement Factor H-Related 3 (FHR-3) is a major regulator of the complement system, which is associated with different diseases, such as age-related macular degeneration (AMD). However, the non-canonical local, cellular functions of FHR-3 remained poorly understood. Here, we report that FHR-3 bound to oxidative stress epitopes and competed with FH for interaction. Furthermore, FHR-3 was internalized by viable RPE cells and modulated time-dependently complement component (C3, FB) and receptor (C3aR, CR3) expression of human RPE cells. Independently of any external blood-derived proteins, complement activation products were detected. Anaphylatoxin C3a was visualized in treated cells and showed a translocation from the cytoplasm to the cell membrane after FHR-3 exposure. Subsequently, FHR-3 induced a RPE cell dependent pro-inflammatory microenvironment. Inflammasome NLRP3 activation and pro-inflammatory cytokine secretion of IL-1ß, IL-18, IL-6 and TNF-α were induced after FHR-3-RPE interaction. Our previously published monoclonal anti-FHR-3 antibody, which was chimerized to reduce immunogenicity, RETC-2-ximab, ameliorated the effect of FHR-3 on ARPE-19 cells. Our studies suggest FHR-3 as an exogenous trigger molecule for the RPE cell "complosome" and as a putative target for a therapeutic approach for associated degenerative diseases.


Sujet(s)
Protéines du sang/immunologie , Activation du complément/immunologie , Facteur H du complément/immunologie , Cellules épithéliales/immunologie , Épithélium pigmentaire de la rétine/cytologie , Protéines du sang/génétique , Protéines du sang/métabolisme , Lignée cellulaire , Activation du complément/génétique , Complément C3/génétique , Complément C3/immunologie , Complément C3/métabolisme , Facteur H du complément/génétique , Facteur H du complément/métabolisme , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/immunologie , Expression des gènes/génétique , Expression des gènes/immunologie , Cellules HEK293 , Humains , Inflammasomes/génétique , Inflammasomes/immunologie , Inflammasomes/métabolisme , Antigène macrophage 1/génétique , Antigène macrophage 1/immunologie , Antigène macrophage 1/métabolisme , Dégénérescence maculaire/génétique , Dégénérescence maculaire/immunologie , Dégénérescence maculaire/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Polymorphisme de nucléotide simple/génétique , Polymorphisme de nucléotide simple/immunologie
11.
Aging Cell ; 20(10): e13491, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34605151

RÉSUMÉ

Advanced maternal age (AMA) pregnancies are rapidly increasing and are associated with aberrant trophoblast cell function, poor placentation, and unfavorable pregnancy outcomes, presumably due to premature placental senescence. SIRT1 is an NAD+ -dependent deacetylase with well-known antiaging effects, but its connection with placental senescence is unreported. In this study, human term placentas and first-trimester villi were collected from AMA and normal pregnancies, and a mouse AMA model was established by cross breeding young and aged male and female C57 mice. SIRT1 expression and activity in HTR8/SVneo cells were genetically or pharmacologically manipulated. Trophoblast-specific Sirt1-knockout (KO) mouse placentas were generated by mating Elf5-Cre and Sirt1fl/fl mice. Trophoblast cell mobility was assessed with transwell invasion and wound-healing assays. SIRT1-binding proteins in HTR8/SVneo cells and human placental tissue were identified by mass spectrometry. We identified SIRT1 as the only differentially expressed sirtuin between AMA and normal placentas. It is downregulated in AMA placentas early in the placental life cycle and is barely impacted by paternal age. SIRT1 loss upregulates P53 acetylation and P21 expression and impairs trophoblast invasion and migration. Sirt1-KO mouse placentas exhibit senescence markers and morphological disruption, along with decreased fetal weight. In trophoblasts, SIRT1 interacts with vimentin, regulating its acetylation. In conclusion, SIRT1 promotes trophoblast epithelial-mesenchymal transition (EMT) to enhance invasiveness by modulating vimentin acetylation. AMA placentas are associated with premature senescence during placentation due to SIRT1 loss. Therefore, SIRT1 may be an antiaging therapeutic target for improving placental development and perinatal outcomes in AMA pregnancies.


Sujet(s)
Transition épithélio-mésenchymateuse/immunologie , Sirtuine-1/métabolisme , Trophoblastes/métabolisme , Vimentine/effets indésirables , Acétylation , Sujet âgé , Animaux , Femelle , Humains , Âge maternel , Souris , Grossesse
12.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-34576042

RÉSUMÉ

Epithelial-mesenchymal transition (EMT) and metabolic reprogramming in cancer cells are the key hallmarks of tumor metastasis. Since the relationship between the two has been well studied, researchers have gained increasing interest in the interplay of cancer cell EMT and immune metabolic changes. Whether the mutual influences between them could provide novel explanations for immune surveillance during metastasis is worth understanding. Here, we review the role of immunometabolism in the regulatory loop between tumor-infiltrating immune cells and EMT. We also discuss the challenges and perspectives of targeting immunometabolism in cancer treatment.


Sujet(s)
Transition épithélio-mésenchymateuse/immunologie , Système immunitaire/métabolisme , Tumeurs/métabolisme , Microenvironnement tumoral/immunologie , Transition épithélio-mésenchymateuse/génétique , Humains , Métastase tumorale , Tumeurs/immunologie , Tumeurs/anatomopathologie , Microenvironnement tumoral/génétique
13.
Sci Rep ; 11(1): 18007, 2021 09 09.
Article de Anglais | MEDLINE | ID: mdl-34504204

RÉSUMÉ

Tumor immune microenvironment plays a crucial role in tumor progression. We performed immune profiling to compare immune-related gene expression between ductal carcinoma in situ (DCIS) and invasive carcinoma of the breast using nCounter PanCancer immune Profiling Panel and found that CXCL10 was the most significant gene that had the highest difference in expression between them. Effect of CXCL10 on breast cancer cell proliferation and invasion was examined in vitro, and expression of CXCL10 and its relationship with immune cell infiltration was assessed in breast cancer samples. CXCL10 induced cell proliferation, migration and epithelial-mesenchymal transition in MCF-7 and MDA-MB-231 breast cancer cell lines. We confirmed that CXCL10 mRNA expression was significantly higher in invasive carcinoma than in DCIS, especially in hormone receptor (HR)-negative tumors using a validation set. CXCL10 mRNA expression showed a positive correlation with tumor infiltrating lymphocyte (TIL) density in both DCIS and invasive carcinoma; CXCL10-positive tumors generally showed higher infiltration of CD8+ and FOXP3+TILs as well as PD-L1+ immune cells compared to CXCL10-negative tumors, albeit with different patterns according to HR status. In conclusion, our study showed that CXCL10 promotes tumor cell proliferation, invasion, and immune cell infiltration, implying its contribution in the progression of DCIS to invasive carcinoma of the breast.


Sujet(s)
Tumeurs du sein/génétique , Carcinome canalaire du sein/génétique , Carcinome intracanalaire non infiltrant/génétique , Chimiokine CXCL10/génétique , Transition épithélio-mésenchymateuse/génétique , Lymphocytes TIL/immunologie , Antigène CD274/génétique , Antigène CD274/immunologie , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/anatomopathologie , Carcinome canalaire du sein/immunologie , Carcinome canalaire du sein/anatomopathologie , Carcinome intracanalaire non infiltrant/immunologie , Carcinome intracanalaire non infiltrant/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Chimiokine CXCL10/immunologie , Évolution de la maladie , Transition épithélio-mésenchymateuse/immunologie , Femelle , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/immunologie , Régulation de l'expression des gènes tumoraux , Humains , Lymphocytes TIL/anatomopathologie , Cellules MCF-7 , Invasion tumorale , ARN messager/génétique , ARN messager/immunologie , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie
14.
Respir Res ; 22(1): 216, 2021 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-34344357

RÉSUMÉ

BACKGROUND: Neutrophilic asthmatics (NA) have less response to inhaled corticosteroids. We aimed to find out the predictor of treatment response in NA. METHODS: Asthmatics (n = 115) and healthy controls (n = 28) underwent clinical assessment during 6-month follow-up with standardized therapy. Asthmatics were categorized by sputum differential cell count. The mRNA expressions were measured by RT-qPCR for sputum cytokines (IFN-γ, IL-1ß, IL-27, FOXP3, IL-17A, and IL-5). The protein of IL-1ß in sputum supernatant was detected by ELISA. Reticular basement membranes (RBM) were measured in the biopsy samples. The role and signaling pathways of IL-1ß mediating the epithelial-mesenchymal transition (EMT) process were explored through A549 cells. RESULTS: NA had increased baseline sputum cell IL-1ß expression compared to eosinophilic asthmatics (EA). After follow-up, NA had less improvement in FEV1 compared to EA. For all asthmatics, sputum IL-1ß mRNA was positively correlated with protein expression. Sputum IL-1ß mRNA and protein levels were negatively correlated to FEV1 improvement. After subgrouping, the correlation between IL-1ß mRNA and FEV1 improvement was significant in NA but not in EA. Thickness of RBM in asthmatics was greater than that of healthy controls and positively correlated with neutrophil percentage in bronchoalveolar lavage fluid. In vitro experiments, the process of IL-1ß augmenting TGF-ß1-induced EMT cannot be abrogated by glucocorticoid or montelukast sodium, but can be reversed by MAPK inhibitors. CONCLUSIONS: IL-1ß level in baseline sputum predicts the poor lung function improvement in NA. The potential mechanism may be related to IL-1ß augmenting TGF-ß1-induced steroid-resistant EMT through MAPK signaling pathways. TRIAL REGISTRATION: This study was approved by the Ethics Committee of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (IRB ID: 20150406).


Sujet(s)
Asthme/immunologie , Transition épithélio-mésenchymateuse/immunologie , Interleukine-1 bêta/immunologie , Granulocytes neutrophiles/immunologie , Muqueuse respiratoire/immunologie , Facteur de croissance transformant bêta/administration et posologie , Cellules A549 , Adulte , Asthme/génétique , Asthme/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Femelle , Études de suivi , Humains , Interleukine-1 bêta/génétique , Mâle , Adulte d'âge moyen , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Muqueuse respiratoire/effets des médicaments et des substances chimiques , Muqueuse respiratoire/métabolisme , Jeune adulte
15.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-34299016

RÉSUMÉ

Metaplastic breast carcinoma (MBC) is a heterogeneous group of infrequent triple negative (TN) invasive carcinomas with poor prognosis. MBCs have a different clinical behavior from other types of triple negative breast cancer (TNBC), being more resistant to standard chemotherapy. MBCs are an example of tumors with activation of epithelial-mesenchymal transition (EMT). The mechanisms involved in EMT could be responsible for the increase in the infiltrative and metastatic capacity of MBCs and resistance to treatments. In addition, a relationship between EMT and the immune response has been seen in these tumors. In this sense, MBC differ from other TN tumors showing a lower number of tumor-infiltrating lymphocytes (TILS) and a higher percentage of tumor cells expressing programmed death-ligand 1 (PD-L1). A better understanding of the relationship between the immune system and EMT could provide new therapeutic approaches in MBC.


Sujet(s)
Antigène CD274/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/immunologie , Carcinomes/immunologie , Carcinomes/métabolisme , Transition épithélio-mésenchymateuse/immunologie , Antigène CD274/immunologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/immunologie , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Cadhérines/métabolisme , Carcinomes/traitement médicamenteux , Carcinomes/génétique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Femelle , Humains , Lymphocytes TIL/immunologie , microARN/génétique , microARN/métabolisme , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie
16.
Med Oncol ; 38(9): 101, 2021 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-34302557

RÉSUMÉ

The COVID-19 pandemic has engulfed the entire world and has claimed more than 3 million lives worldwide. This viral disease is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and mainly characterized by fever, dry cough, fatigue, anosmia, anorexia, and dyspnea. The severity of the disease increases with age and presence of comorbidities, including cancer. Multiple clinical studies have shown that the cancer patients are highly susceptible to the severe form of the viral disease. In this review article, we have summarized the available scientific literature regarding the molecular links between COVID-19 and cancer, which make the cancer patients highly susceptible to COVID-19. Few studies have shown that the angiotensin-converting enzyme 2 (ACE2) receptor, transmembrane protease serine 2 (TMPRSS2), and the immune response and inflammation establish the interconnection between the two diseases. Additionally, we have also discussed whether SARS-CoV-2 can contribute to cancer development in COVID-19 patients. A recent study has suggested that SARS-CoV-2 may create a microenvironment that may support cancer cell proliferation and induce the activation of dormant cancer cells (DCCs). In another study, the blood sera of COVID-19 patients were found to activate epithelial-to-mesenchymal transition (EMT) in cancer cells. Overall, this review article will surely help the scientific community to understand why the cancer patients are so much prone to COVID-19 and will also motivate the researchers to find new therapeutic strategies that may save the lives of many COVID-19-infected cancer patients.


Sujet(s)
COVID-19/immunologie , Tumeurs/immunologie , Animaux , Transition épithélio-mésenchymateuse/immunologie , Humains , Immunité/immunologie , Inflammation/immunologie
17.
Int J Med Sci ; 18(12): 2466-2479, 2021.
Article de Anglais | MEDLINE | ID: mdl-34104078

RÉSUMÉ

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with a high mortality rate. Epithelial-to-mesenchymal transition (EMT) confers cancer cells with immune evasive ability by modulating the expression of immune checkpoints in many cancers. Thus, the aim of our study is to examine the interplay between EMT and immune checkpoint molecules in HCC. A reversible EMT model was utilised with transforming growth factor (TGF)-ß1 as an EMT inducer for HCC cell lines Hep3B and PLC/PRF/5. HCC cells were treated with TGF-ß1 for 72 h and the EMT status and immune checkpoint expression were examined. In addition, the migratory ability of HCC cells were examined using wound healing and transwell migration assays in the reversible EMT model. siRNA-mediated knockdown of immune checkpoint molecule, B7-H3, was further utilised to validate the association between TGF-ß1-mediated EMT and immune checkpoint expression in HCC. In addition, a web-based platform, SurvExpress, was utilised to evaluate the association between expression of TGF-ß1 in combination with immune checkpoint molecules and overall survival in HCC patients. We observed induction of EMT upon treatment of HCC cells with TGF-ß1 revealed by reduced expression of epithelial markers along with increased expression of mesenchymal markers. Withdrawal of TGF-ß1 reversed the process of EMT with elevated expression of epithelial markers and reduced expression of mesenchymal markers. TGF-ß1 treatment elevated the migratory potential of HCC cells which was reversed following reversal assay. Notably, during TGF-ß1-induced EMT, there was upregulation of immune checkpoint molecules PD-L1 and B7-H3. However, the reversal of EMT decreased the expression of PD-L1 and B7-H3. In addition, TGF-ß1 driven EMT was reversed following knockdown of B7-H3 in both HCC cells further validating the interplay between TGF-ß1-mediated EMT and immune checkpoint expression in HCC. Furthermore, the coordinate expression of TGF-ß1 with PD-L1 (p=0.01487) and B7-H3 (p=0.009687) was correlated with poor overall survival in 422 HCC patients. Our study has demonstrated a close association between TGF-ß1-mediated EMT and regulation of immune checkpoints in HCC.


Sujet(s)
Antigènes B7/métabolisme , Antigène CD274/métabolisme , Carcinome hépatocellulaire/immunologie , Tumeurs du foie/immunologie , Facteur de croissance transformant bêta-1/métabolisme , Antigènes B7/génétique , Antigène CD274/génétique , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Transition épithélio-mésenchymateuse/immunologie , Régulation de l'expression des gènes tumoraux/immunologie , Techniques de knock-down de gènes , Humains , Tumeurs du foie/génétique , Tumeurs du foie/mortalité , Tumeurs du foie/anatomopathologie , Transduction du signal/immunologie , Régulation positive
18.
JCI Insight ; 6(15)2021 08 09.
Article de Anglais | MEDLINE | ID: mdl-34185707

RÉSUMÉ

We previously reported heightened expression of the human endogenous retroviral protein HERV-K deoxyuridine triphosphate nucleotidohydrolase (dUTPase) in circulating monocytes and pulmonary arterial (PA) adventitial macrophages of patients with PA hypertension (PAH). Furthermore, recombinant HERV-K dUTPase increased IL-6 in PA endothelial cells (PAECs) and caused pulmonary hypertension in rats. Here we show that monocytes overexpressing HERV-K dUTPase, as opposed to GFP, can release HERV-K dUTPase in extracellular vesicles (EVs) that cause pulmonary hypertension in mice in association with endothelial mesenchymal transition (EndMT) related to induction of SNAIL/SLUG and proinflammatory molecules IL-6 as well as VCAM1. In PAECs, HERV-K dUTPase requires TLR4-myeloid differentiation primary response-88 to increase IL-6 and SNAIL/SLUG, and HERV-K dUTPase interaction with melanoma cell adhesion molecule (MCAM) is necessary to upregulate VCAM1. TLR4 engagement induces p-p38 activation of NF-κB in addition to p-pSMAD3 required for SNAIL and pSTAT1 for IL-6. HERV-K dUTPase interaction with MCAM also induces p-p38 activation of NF-κB in addition to pERK1/2-activating transcription factor-2 (ATF2) to increase VCAM1. Thus in PAH, monocytes or macrophages can release HERV-K dUTPase in EVs, and HERV-K dUTPase can engage dual receptors and signaling pathways to subvert PAEC transcriptional machinery to induce EndMT and associated proinflammatory molecules.


Sujet(s)
Rétrovirus endogènes , Transition épithélio-mésenchymateuse/immunologie , Hypertension pulmonaire , Macrophages/immunologie , Monocytes/immunologie , Artère pulmonaire , Pyrophosphatases/métabolisme , Animaux , Antigènes CD146/métabolisme , Rétrovirus endogènes/métabolisme , Rétrovirus endogènes/pathogénicité , Cellules endothéliales/métabolisme , Hypertension pulmonaire/immunologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/virologie , Inflammation/métabolisme , Inflammation/virologie , Souris , Artère pulmonaire/immunologie , Artère pulmonaire/anatomopathologie , Transduction du signal , Facteurs de transcription de la famille Snail/métabolisme
19.
Aging (Albany NY) ; 13(9): 12691-12709, 2021 05 11.
Article de Anglais | MEDLINE | ID: mdl-33973529

RÉSUMÉ

Due to its effectiveness, cancer immunotherapy has attracted widespread attention from clinicians and scientific researchers. Numerous studies have proven that effective stratification of cancer patients would promote the personalized application of immunotherapy. Therefore, we used the transcriptome data of nearly 1,000 patients with non-small cell lung cancer (NSCLC) to construct a new immune subgroup. We found that the new immune subgroup, named cluster 2, was a mixture of lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), and showed poor overall survival, which was further verified in the independent validation set. Immune infiltration correlation analysis showed that the Mast cell type and its status subdivisions had a predictive effect on the prognosis of NSCLC, especially in LUAD. Phenotypic analysis suggested that epithelial-mesenchymal transition (EMT) was positively correlated with immunosuppression, supporting the correlation between tumor phenotype and immune background. Although immune subtypes failed to significantly distinguish the progression-free survival (PFS) of immunotherapy patients, they showed the expected trend; the sample size needs to be further expanded for verification. In addition, some results indicated that the two cancer types, LUAD and LUSC, might require independent analyses.


Sujet(s)
Adénocarcinome pulmonaire/traitement médicamenteux , Antinéoplasiques immunologiques/pharmacologie , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome épidermoïde/traitement médicamenteux , Tumeurs du poumon/traitement médicamenteux , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/immunologie , Adénocarcinome pulmonaire/mortalité , Sujet âgé , Antinéoplasiques immunologiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/mortalité , Carcinome épidermoïde/génétique , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/mortalité , Jeux de données comme sujet , Résistance aux médicaments antinéoplasiques/génétique , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/immunologie , Femelle , Régulation de l'expression des gènes tumoraux/immunologie , Humains , Poumon/immunologie , Poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Tumeurs du poumon/mortalité , Mâle , Mastocytes/immunologie , Adulte d'âge moyen , Pronostic , Survie sans progression , RNA-Seq , Transcriptome/immunologie , Échappement de la tumeur à la surveillance immunitaire/génétique
20.
Int Immunopharmacol ; 97: 107690, 2021 Aug.
Article de Anglais | MEDLINE | ID: mdl-33940323

RÉSUMÉ

It has recently been shown that expression levels of tissue factor (TF) are high in the serum and peripheral blood mononuclear cells of patients with asthma. However, whether TF impacts airway inflammation and remodelling in asthma remains unknown. The aim of this study was to investigate the effect of TF in asthma airway inflammation and remodelling using a house dust mite (HDM)-induced chronic asthma model and human bronchial epithelial (16HBE) cells. A chronic asthma model was constructed in BALB/c mice by the intranasal instillation of HDM. Mice were treated with short hairpin TF (shTF), and airway inflammation and remodelling features of asthma and epithelial-mesenchymal transition (EMT) were assessed. 16HBE cells were induced by transforming growth factor-ß1 (TGF-ß1) and HDM in the presence or absence of shTF; then, EMT markers and invasion and migration ability were determined. TF expression increased in the lung tissue and 16HBE cells when exposed to HDM. TF downregulation in the lung significantly reduced airway hyperresponsiveness, eosinophil inflammation, the EMT process, and levels of interleukin (IL)-4, IL-6, IL-13, and TGF-ß1 in bronchoalveolar lavage fluid of asthmatic mice. Moreover, TF downregulation inhibited migration and incursion and decreased the expression levels of fibronectin 1 and TGF-ß1, but increased the expression of E-cadherin in HDM- and TGF-ß1-stimulated 16HBE cells. These results demonstrated that TF promoted airway pathological features by enhancing the EMT of bronchial epithelial cells both in vitro and in mice with house dust mite-induced asthma.


Sujet(s)
Allergènes/immunologie , Asthme/immunologie , Dermatophagoides pteronyssinus/immunologie , Thromboplastine/métabolisme , Remodelage des voies aériennes/immunologie , Animaux , Asthme/anatomopathologie , Bronches/cytologie , Bronches/immunologie , Bronches/anatomopathologie , Liquide de lavage bronchoalvéolaire , Modèles animaux de maladie humaine , Cellules épithéliales/immunologie , Cellules épithéliales/métabolisme , Transition épithélio-mésenchymateuse/immunologie , Cellules HEK293 , Humains , Souris , Organismes exempts d'organismes pathogènes spécifiques , Thromboplastine/génétique , Régulation positive/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...