Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 515
Filtrer
1.
Int Immunopharmacol ; 138: 112608, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-38981221

RÉSUMÉ

BACKGROUND: Abdominal aortic aneurysm (AAA) poses a significant health risk and is influenced by various compositional features. This study aimed to develop an artificial intelligence-driven multiomics predictive model for AAA subtypes to identify heterogeneous immune cell infiltration and predict disease progression. Additionally, we investigated neutrophil heterogeneity in patients with different AAA subtypes to elucidate the relationship between the immune microenvironment and AAA pathogenesis. METHODS: This study enrolled 517 patients with AAA, who were clustered using k-means algorithm to identify AAA subtypes and stratify the risk. We utilized residual convolutional neural network 200 to annotate and extract contrast-enhanced computed tomography angiography images of AAA. A precise predictive model for AAA subtypes was established using clinical, imaging, and immunological data. We performed a comparative analysis of neutrophil levels in the different subgroups and immune cell infiltration analysis to explore the associations between neutrophil levels and AAA. Quantitative polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay were performed to elucidate the interplay between CXCL1, neutrophil activation, and the nuclear factor (NF)-κB pathway in AAA pathogenesis. Furthermore, the effect of CXCL1 silencing with small interfering RNA was investigated. RESULTS: Two distinct AAA subtypes were identified, one clinically more severe and more likely to require surgical intervention. The CNN effectively detected AAA-associated lesion regions on computed tomography angiography, and the predictive model demonstrated excellent ability to discriminate between patients with the two identified AAA subtypes (area under the curve, 0.927). Neutrophil activation, AAA pathology, CXCL1 expression, and the NF-κB pathway were significantly correlated. CXCL1, NF-κB, IL-1ß, and IL-8 were upregulated in AAA. CXCL1 silencing downregulated NF-κB, interleukin-1ß, and interleukin-8. CONCLUSION: The predictive model for AAA subtypes demonstrated accurate and reliable risk stratification and clinical management. CXCL1 overexpression activated neutrophils through the NF-κB pathway, contributing to AAA development. This pathway may, therefore, be a therapeutic target in AAA.


Sujet(s)
Anévrysme de l'aorte abdominale , Intelligence artificielle , Chimiokine CXCL1 , Évolution de la maladie , Granulocytes neutrophiles , Humains , Anévrysme de l'aorte abdominale/immunologie , Mâle , Femelle , Sujet âgé , Granulocytes neutrophiles/immunologie , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Facteur de transcription NF-kappa B/métabolisme , Adulte d'âge moyen , Angiographie par tomodensitométrie , Multi-omique
2.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 78-84, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836678

RÉSUMÉ

Macrophages in the tumor microenvironment can polarize into M1 or M2 forms, with M2 macrophages (M2φ) promoting tumor growth and metastasis in cervical squamous cell carcinoma (CESC). This study explored the effects of M2φ on CESC metabolic reprogramming both in vitro and in vivo. Results showed that M2φ secreted CXCL1, which significantly increased CESC migration and metabolic regulation. Further experiments revealed that CXCL1 upregulated KDM6B to enhance PFKFB2 transcriptional activity, thus regulating CESC glucose metabolism. Transcriptome sequencing screened 5 upregulated genes related to glycolysis, with PFKFB2 showing the most significant increase in cells treated with rCXCL1. Dual-luciferase reporter assay confirmed that rCXCL1 enhances PFKFB2 transcriptional activity. Bioinformatics analysis revealed a high correlation between expressions of KDM6B and PFKFB2 in CESC. Mechanistic experiments demonstrated that KDM6B inhibited H3K27me3 modification to activate PFKFB2 transcriptional expression. In conclusion, M2φ secreted CXCL1 to promote CESC cell migration and invasion, and CXCL1 activated KDM6B expression in CESC cells, inhibiting H3K27 protein methylation modification, and enhanced PFKFB2 transcriptional activity to regulate CESC glucose metabolism. These results provided new insights into the complex interplay between the immune system and cancer metabolism, which may have broader implications for understanding and treating other types of cancer.


Sujet(s)
Carcinome épidermoïde , Mouvement cellulaire , Chimiokine CXCL1 , Régulation de l'expression des gènes tumoraux , Jumonji Domain-Containing Histone Demethylases , Macrophages , Phosphofructokinase-2 , Tumeurs du col de l'utérus , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme , Humains , Femelle , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/génétique , Macrophages/métabolisme , Phosphofructokinase-2/métabolisme , Phosphofructokinase-2/génétique , Mouvement cellulaire/génétique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Animaux , Lignée cellulaire tumorale , Souris , Microenvironnement tumoral/génétique , Glucose/métabolisme , Souris nude , Glycolyse/génétique ,
3.
Int Immunopharmacol ; 136: 112383, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38843642

RÉSUMÉ

The treatment of autoimmune and inflammatory diseases often requires targeting multiple pathogenic pathways. KYS202004A is a novel bispecific fusion protein designed to antagonize TNF-α and IL-17A, pivotal in the pathophysiology of autoimmune and inflammatory diseases. Our initial efforts focused on screening for optimal structure by analyzing expression levels, purity, and binding capabilities. The binding affinity of KYS202004A to TNF-α and IL-17A was evaluated using SPR. In vitro, we assessed the inhibitory capacity of KYS202004A on cytokine-induced CXCL1 expression in HT29 cells. In vivo, its efficacy was tested using a Collagen-Induced Arthritis (CIA) model in transgenic human-IL-17A mice and an imiquimod-induced psoriasis model in cynomolgus monkeys. KYS202004A demonstrated significant inhibition of IL-17A and TNF-α signaling pathways, outperforming the efficacy of monotherapeutic agents ixekizumab and etanercept in reducing CXCL1 expression in vitro and ameliorating disease markers in vivo. In the CIA model, KYS202004A significantly reduced clinical symptoms, joint destruction, and serum IL-6 concentrations. The psoriasis model revealed that KYS202004A, particularly at a 2  mg/kg dose, was as effective as the combination of ixekizumab and etanercept. This discovery represents a significant advancement in treating autoimmune and inflammatory diseases, offering a dual-targeted therapeutic approach with enhanced efficacy over current monotherapies.


Sujet(s)
Arthrite expérimentale , Interleukine-17 , Macaca fascicularis , Psoriasis , Protéines de fusion recombinantes , Facteur de nécrose tumorale alpha , Animaux , Interleukine-17/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Humains , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/induit chimiquement , Protéines de fusion recombinantes/usage thérapeutique , Protéines de fusion recombinantes/pharmacologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/immunologie , Souris , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Cellules HT29 , Maladies auto-immunes/traitement médicamenteux , Maladies auto-immunes/immunologie , Souris transgéniques , Modèles animaux de maladie humaine , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/pharmacologie , Mâle , Évaluation préclinique de médicament , Imiquimod , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Souris de lignée DBA
4.
Cell Death Dis ; 15(6): 447, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918360

RÉSUMÉ

There is a paucity of comprehensive knowledge pertaining to the underlying mechanisms leading to gefitinib resistance in individuals diagnosed NSCLC harboring EGFR-sensitive mutations who inevitably develop resistance to gefitinib treatment within six months to one year. In our preceding investigations, we have noted a marked upregulation of IGFBP2 in the neoplastic tissues of NSCLC, predominantly in the periphery of the tissue, implying its plausible significance in NSCLC. Consequently, in the current research, we delved into the matter and ascertained the molecular mechanisms that underlie the participation of IGFBP2 in the emergence of gefitinib resistance in NSCLC cells. Firstly, the expression of IGFBP2 in the bronchoalveolar lavage fluid and lung cancer tissues of 20 NSCLC patients with gefitinib tolerance was found to be significantly higher than that of non-tolerant patients. Furthermore, in vitro and in vivo experiments demonstrated that IGFBP2 plays a significant role in the acquisition of gefitinib resistance. Mechanistically, IGFBP2 can activate STAT3 to enhance the transcriptional activity of CXCL1, thereby increasing the intracellular expression level of CXCL1, which contributes to the survival of lung cancer cells in the gefitinib environment. Additionally, we identified ITGA5 as a key player in IGFBP2-mediated gefitinib resistance, but it does not function as a membrane receptor in the process of linking IGFBP2 to intracellular signaling transduction. In conclusion, this study demonstrates the promoting role and mechanism of IGFBP2 in acquired gefitinib resistance caused by non-EGFR secondary mutations, suggesting the potential of IGFBP2 as a biomarker for gefitinib resistance and a potential intervention target.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Chimiokine CXCL1 , Résistance aux médicaments antinéoplasiques , Géfitinib , Protéine-2 de liaison aux IGF , Tumeurs du poumon , Facteur de transcription STAT-3 , Animaux , Femelle , Humains , Mâle , Souris , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Géfitinib/pharmacologie , Géfitinib/usage thérapeutique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Protéine-2 de liaison aux IGF/métabolisme , Protéine-2 de liaison aux IGF/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris nude , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription STAT-3/métabolisme
5.
J Clin Invest ; 134(14)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885342

RÉSUMÉ

While inflammation is beneficial for insulin secretion during homeostasis, its transformation adversely affects ß cells and contributes to diabetes. However, the regulation of islet inflammation for maintaining glucose homeostasis remains largely unknown. Here, we identified pericytes as pivotal regulators of islet immune and ß cell function in health. Islets and pancreatic pericytes express various cytokines in healthy humans and mice. To interfere with the pericytic inflammatory response, we selectively inhibited the TLR/MyD88 pathway in these cells in transgenic mice. The loss of MyD88 impaired pericytic cytokine production. Furthermore, MyD88-deficient mice exhibited skewed islet inflammation with fewer cells, an impaired macrophage phenotype, and reduced IL-1ß production. This aberrant pericyte-orchestrated islet inflammation was associated with ß cell dedifferentiation and impaired glucose response. Additionally, we found that Cxcl1, a pericytic MyD88-dependent cytokine, promoted immune IL-1ß production. Treatment with either Cxcl1 or IL-1ß restored the mature ß cell phenotype and glucose response in transgenic mice, suggesting a potential mechanism through which pericytes and immune cells regulate glucose homeostasis. Our study revealed pericyte-orchestrated islet inflammation as a crucial element in glucose regulation, implicating this process as a potential therapeutic target for diabetes.


Sujet(s)
Inflammation , Interleukine-1 bêta , Facteur de différenciation myéloïde-88 , Péricytes , Transduction du signal , Animaux , Facteur de différenciation myéloïde-88/génétique , Facteur de différenciation myéloïde-88/métabolisme , Souris , Péricytes/métabolisme , Péricytes/anatomopathologie , Péricytes/immunologie , Humains , Inflammation/anatomopathologie , Inflammation/métabolisme , Inflammation/génétique , Inflammation/immunologie , Interleukine-1 bêta/métabolisme , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Souris transgéniques , Récepteurs de type Toll/métabolisme , Récepteurs de type Toll/génétique , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Ilots pancréatiques/immunologie , Ilots pancréatiques/métabolisme , Ilots pancréatiques/anatomopathologie , Souris knockout , Cellules à insuline/métabolisme , Cellules à insuline/anatomopathologie , Cellules à insuline/immunologie , Mâle , Glucose/métabolisme
6.
J Pharmacol Sci ; 155(3): 94-100, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38797538

RÉSUMÉ

Interleukin (IL-19) belongs to the IL-10 family of cytokines and plays diverse roles in inflammation, cell development, viral responses, and lipid metabolism. Acute lung injury (ALI) is a severe respiratory condition associated with various diseases, including severe pneumonia, sepsis, and trauma, lacking established treatments. However, the role of IL-19 in acute inflammation of the lungs is unknown. We reported the impact of IL-19 functional deficiency in mice crossed with an ALI model using HCl. Lungs damages, neutrophil infiltration, and pulmonary edema induced by HCl were significantly worse in IL-19 knockout (KO) mice than in wild-type (WT) mice. mRNA expression levels of C-X-C motif chemokine ligand 1 (CXCL1) and IL-6 in the lungs were significantly higher in IL-19 KO mice than in WT mice. Little apoptosis was detected in lung injury in WT mice, whereas apoptosis was observed in exacerbated area of lung injury in IL-19 KO mice. These results are the first to show that IL-19 is involved in acute inflammation of the lungs, suggesting a novel molecular mechanism in acute respiratory failures. If it can be shown that neutrophils have IL-19 receptors and that IL-19 acts directly on them, it would be a novel drug target.


Sujet(s)
Lésion pulmonaire aigüe , Acide chlorhydrique , Interleukines , Souris knockout , Animaux , Lésion pulmonaire aigüe/étiologie , Lésion pulmonaire aigüe/anatomopathologie , Lésion pulmonaire aigüe/génétique , Interleukines/génétique , Interleukines/métabolisme , Souris de lignée C57BL , Interleukine-6/métabolisme , Interleukine-6/génétique , Modèles animaux de maladie humaine , Infiltration par les neutrophiles , Chimiokine CXCL1/génétique , Chimiokine CXCL1/métabolisme , Mâle , Poumon/anatomopathologie , Poumon/métabolisme , Apoptose/génétique , Apoptose/effets des médicaments et des substances chimiques , Souris , Granulocytes neutrophiles , Oedème pulmonaire/étiologie , Expression des gènes
7.
Environ Pollut ; 351: 124081, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38697251

RÉSUMÉ

Microcystin-leucine arginine (MC-LR) is a common cyantotoxin produced by hazardous cyanobacterial blooms, and eutrophication is increasing the contamination level of MC-LR in drinking water supplies and aquatic foods. MC-LR has been linked to colorectal cancer (CRC) progression associated with tumor microenvironment, however, the underlying mechanism is not clearly understood. In present study, by using GEO, KEGG, GESA and ImmPort database, MC-LR related differentially expressed genes (DEGs) and pathway- and gene set-enrichment analysis were performed. Of the three identified DEGs (CXCL1, GUCA2A and GDF15), CXCL1 was shown a positive association with tumor infiltration, and was validated to have a dominantly higher upregulation in MC-LR-treated tumor-associated macrophages (TAMs) rather than in MC-LR-treated CRC cells. Both CRC cell/macrophage co-culture and xenograft mouse models indicated that MC-LR stimulated TAMs to secrete CXCL1 resulting in promoted proliferation, migration, and invasion capability of CRC cells. Furtherly, IP-MS assay found that interaction between TAMs-derived CXCL1 and CRC cell-derived IGHG1 may enhance CRC cell proliferation and migration after MC-LR treatment, and this effect can be attenuated by silencing IGHG1 in CRC cell. In addition, molecular docking analysis, co-immunoprecipitation and immunofluorescence further proved the interactions between CXCL1 and IGHG1. In conclusion, CXCL1 secreted by TAMs can trigger IGHG1 expression in CRC cells, which provides a new clue in elucidating the mechanism of MC-LR-mediated CRC progression.


Sujet(s)
Chimiokine CXCL1 , Tumeurs colorectales , Transduction du signal , Macrophages associés aux tumeurs , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Humains , Animaux , Chimiokine CXCL1/génétique , Chimiokine CXCL1/métabolisme , Souris , Macrophages associés aux tumeurs/métabolisme , Microcystines/toxicité , Toxines de la flore et de la faune marines , Lignée cellulaire tumorale , Évolution de la maladie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Microenvironnement tumoral
8.
Sci Rep ; 14(1): 11062, 2024 05 14.
Article de Anglais | MEDLINE | ID: mdl-38745005

RÉSUMÉ

To evaluate gene expression associated with unfavorable vaginal bleeding in users of the Etonogestrel (ENG) contraceptive implant. Prospective study involving 100 women who intended to use the ENG implant. Exclusion criteria included abnormal uterine bleeding, inability to attend a 1-year follow-up, and implant removal for reasons unrelated to vaginal bleeding or loss of follow-up. We obtained endometrial biopsies before implant placement and assessed the expression of 20 selected genes. Users maintained a uterine bleeding diary for 12 months post-implant placement. For statistical analysis, we categorized women into those with or without favorable vaginal bleeding at 3 and 12 months. Women with lower CXCL1 expression had a 6.8-fold increased risk of unfavorable vaginal bleeding at 3 months (OR 6.8, 95% CI 2.21-20.79, p < 0.001), while those with higher BCL6 and BMP6 expression had 6- and 5.1-fold increased risks, respectively. By the 12-month follow-up, women with lower CXCL1 expression had a 5.37-fold increased risk of unfavorable vaginal bleeding (OR 5.37, 95% CI 1.63-17.73, p = 0.006). Women with CXCL1 expression < 0.0675, BCL6 > 0.65, and BMP6 > 3.4 had a higher likelihood of experiencing unfavorable vaginal bleeding at 3 months, and CXCL1 < 0.158 at 12 months. Users of ENG contraceptive implants with elevated BCL6 and BMP6 expression exhibited a higher risk of breakthrough bleeding at the 3-month follow-up. Conversely, reduced CXCL1 expression was associated with an elevated risk of bleeding at both the 3 and 12-month follow-ups.


Sujet(s)
Contraceptifs féminins , Désogestrel , Hémorragie utérine , Humains , Femelle , Désogestrel/administration et posologie , Désogestrel/effets indésirables , Adulte , Études prospectives , Hémorragie utérine/génétique , Contraceptifs féminins/effets indésirables , Contraceptifs féminins/administration et posologie , Endomètre/métabolisme , Endomètre/effets des médicaments et des substances chimiques , Endomètre/anatomopathologie , Implant pharmaceutique , Chimiokine CXCL1/génétique , Chimiokine CXCL1/métabolisme , Jeune adulte
9.
Nat Commun ; 15(1): 4119, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38750020

RÉSUMÉ

Sepsis results from systemic, dysregulated inflammatory responses to infection, culminating in multiple organ failure. Here, we demonstrate the utility of CD5L for treating experimental sepsis caused by cecal ligation and puncture (CLP). We show that CD5L's important features include its ability to enhance neutrophil recruitment and activation by increasing circulating levels of CXCL1, and to promote neutrophil phagocytosis. CD5L-deficient mice exhibit impaired neutrophil recruitment and compromised bacterial control, rendering them susceptible to attenuated CLP. CD5L-/- peritoneal cells from mice subjected to medium-grade CLP exhibit a heightened pro-inflammatory transcriptional profile, reflecting a loss of control of the immune response to the infection. Intravenous administration of recombinant CD5L (rCD5L) in immunocompetent C57BL/6 wild-type (WT) mice significantly ameliorates measures of disease in the setting of high-grade CLP-induced sepsis. Furthermore, rCD5L lowers endotoxin and damage-associated molecular pattern (DAMP) levels, and protects WT mice from LPS-induced endotoxic shock. These findings warrant the investigation of rCD5L as a possible treatment for sepsis in humans.


Sujet(s)
Souris de lignée C57BL , Souris knockout , Granulocytes neutrophiles , Sepsie , Animaux , Sepsie/immunologie , Sepsie/traitement médicamenteux , Souris , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Phagocytose , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Modèles animaux de maladie humaine , Mâle , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques , Caecum/chirurgie , Protéines recombinantes/usage thérapeutique , Protéines recombinantes/administration et posologie , Humains , Perforines/métabolisme , Ligature , Lipopolysaccharides , Choc septique/immunologie
10.
Cancer Lett ; 592: 216903, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38670307

RÉSUMÉ

High levels of acetyl-CoA are considered a key metabolic feature of metastatic cancers. However, the impacts of acetyl-CoA metabolic accumulation on cancer microenvironment remodeling are poorly understood. In this study, using human hepatocellular carcinoma (HCC) tissues and orthotopic xenograft models, we found a close association between high acetyl-CoA levels in HCCs, increased infiltration of tumor-associated neutrophils (TANs) in the cancer microenvironment and HCC metastasis. Cytokine microarray and enzyme-linked immunosorbent assays (ELISA) revealed the crucial role of the chemokine (C-X-C motif) ligand 1(CXCL1). Mechanistically, acetyl-CoA accumulation induces H3 acetylation-dependent upregulation of CXCL1 gene expression. CXCL1 recruits TANs, leads to neutrophil extracellular traps (NETs) formation and promotes HCC metastasis. Collectively, our work linked the accumulation of acetyl-CoA in HCC cells and TANs infiltration, and revealed that the CXCL1-CXC receptor 2 (CXCR2)-TANs-NETs axis is a potential target for HCCs with high acetyl-CoA levels.


Sujet(s)
Acétyl coenzyme A , Carcinome hépatocellulaire , Chimiokine CXCL1 , Tumeurs du foie , Granulocytes neutrophiles , Microenvironnement tumoral , Animaux , Femelle , Humains , Mâle , Souris , Acétyl coenzyme A/métabolisme , Acétylation , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/génétique , Lignée cellulaire tumorale , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Pièges extracellulaires/métabolisme , Régulation de l'expression des gènes tumoraux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Tumeurs du foie/génétique , Souris nude , Infiltration par les neutrophiles , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/anatomopathologie , Récepteurs à l'interleukine-8B/métabolisme , Récepteurs à l'interleukine-8B/génétique , Adulte , Adulte d'âge moyen , Sujet âgé , Souris de lignée BALB C
11.
Front Immunol ; 15: 1367019, 2024.
Article de Anglais | MEDLINE | ID: mdl-38686389

RÉSUMÉ

Background: Although hyperuricemia is not always associated with acute gouty arthritis, uric acid is a significant risk factor for gout. Therefore, we investigated the specific mechanism of uric acid activity. Methods: Using the gout-associated transcriptome dataset GSE160170, we conducted differential expression analysis to identify differentially expressed genes (DEGs). Moreover, we discovered highly linked gene modules using weighted gene coexpression network analysis (WGCNA) and evaluated their intersection. Subsequently, we screened for relevant biomarkers using the cytoHubba and Mcode algorithms in the STRING database, investigated their connection to immune cells and constructed a competitive endogenous RNA (ceRNA) network to identify upstream miRNAs and lncRNAs. We also collected PBMCs from acute gouty arthritis patients and healthy individuals and constructed a THP-1 cell gout inflammatory model, RT-qPCR and western blotting (WB) were used to detect the expression of C-X-C motif ligand 8 (CXCL8), C-X-C motif ligand 2 (CXCL2), and C-X-C motif ligand 1 (CXCL1). Finally, we predicted relevant drug targets through hub genes, hoping to find better treatments. Results: According to differential expression analysis, there were 76 upregulated and 28 downregulated mRNAs in GSE160170. Additionally, WGCNA showed that the turquoise module was most strongly correlated with primary gout; 86 hub genes were eventually obtained upon intersection. IL1ß, IL6, CXCL8, CXCL1, and CXCL2 are the principal hub genes of the protein-protein interaction (PPI) network. Using RT-qPCR and WB, we found that there were significant differences in the expression levels of CXCL8, CXCL1, and CXCL2 between the gouty group and the healthy group, and we also predicted 10 chemicals related to these proteins. Conclusion: In this study, we screened and validated essential genes using a variety of bioinformatics tools to generate novel ideas for the diagnosis and treatment of gout.


Sujet(s)
Marqueurs biologiques , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Goutte , Humains , Goutte/génétique , Chimiokine CXCL1/génétique , Chimiokine CXCL2/génétique , Chimiokine CXCL2/métabolisme , Biologie informatique/méthodes , Transcriptome , Cellules THP-1 , Interleukine-8/génétique , microARN/génétique , Acide urique , Cartes d'interactions protéiques , Régulation de l'expression des gènes , Bases de données génétiques , Goutte articulaire/génétique
12.
Zhen Ci Yan Jiu ; 49(4): 331-340, 2024 Apr 25.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38649200

RÉSUMÉ

OBJECTIVES: To observe whether acupuncture up-regulates chemokine CXC ligand 1 (CXCL1) in the brain to play an analgesic role through CXCL1/chemokine CXC receptor 2 (CXCR2) signaling in adjuvant induced arthritis (AIA) rats, so as to reveal its neuro-immunological mechanism underlying improvement of AIA. METHODS: BALB/c mice with relatively stable thermal pain reaction were subjected to planta injection of complete Freund adjuvant (CFA) for establishing AIA model, followed by dividing the AIA mice into simple AF750 (fluorochrome) and AF750+CXCL1 groups (n=2 in each group). AF750 labeled CXCL1 recombinant protein was then injected into the mouse's tail vein to induce elevation of CXCL1 level in blood for simulating the effect of acupuncture stimulation which has been demonstrated by our past study. In vivo small animal imaging technology was used to observe the AF750 and AF750+CXCL1-labelled target regions. After thermal pain screening, the Wistar rats with stable pain reaction were subjected to AIA modeling by injecting CFA into the rat's right planta, then were randomized into model and manual acupuncture groups (n=12 in each group). Other 12 rats that received planta injection of saline were used as the control group. Manual acupuncture (uniform reinforcing and reducing manipulations) was applied to bilateral "Zusanli" (ST36) for 4×2 min, with an interval of 5 min between every 2 min, once daily for 7 days. The thermal pain threshold was assessed by detecting the paw withdrawal latency (PWL) using a thermal pain detector. The contents of CXCL1 in the primary somatosensory cortex (S1), medial prefrontal cortex, nucleus accumbens, amygdala, periaqueductal gray and rostroventromedial medulla regions were assayed by using ELISA, and the expression levels of CXCL1, CXCR2 and mu-opioid receptor (MOR) mRNA in the S1 region were detected using real time-quantitative polymerase chain reaction. The immune-fluorescence positive cellular rate of CXCL1 and CXCR2 in S1 region was observed after immunofluorescence stain. The immunofluorescence double-stain of CXCR2 and astrocyte marker glial fibrillary acidic protein (GFAP) or neuron marker NeuN or MOR was used to determine whether there is a co-expression between them. RESULTS: In AIA mice, results of in vivo experiments showed no obvious enrichment signal of AF750 or AF750+CXCL1 in any organ of the body, while in vitro experiments showed that there was a stronger fluorescence signal of CXCL1 recombinant protein in the brain. In rats, compared with the control group, the PWL from day 0 to day 7 was significantly decreased (P<0.01) and the expression of CXCR2 mRNA in the S1 region significantly increased in the model group (P<0.05), while in comparison with the model group, the PWL from day 2 to day 7, CXCL1 content, CXCR2 mRNA expression and CXCR2 content, and MOR mRNA expression in the S1 region were significantly increased in the manual acupuncture group (P<0.05, P<0.01). Immunofluorescence stain showed that CXCR2 co-stained with NeuN and MOR in the S1 region, indicating that CXCR2 exists in neurons and MOR-positive neurons but not in GFAP positive astrocytes. CONCLUSIONS: Acupuncture can increase the content of CXCL1 in S1 region, up-regulate CXCR2 on neurons in the S1 region and improve MOR expression in S1 region of AIA rats, which may contribute to its effect in alleviating inflammatory pain.


Sujet(s)
Thérapie par acupuncture , Arthrite expérimentale , Chimiokine CXCL1 , Récepteurs à l'interleukine-8B , Cortex somatosensoriel , Animaux , Humains , Mâle , Souris , Rats , Points d'acupuncture , Arthrite expérimentale/thérapie , Arthrite expérimentale/métabolisme , Arthrite expérimentale/génétique , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Inflammation/thérapie , Inflammation/métabolisme , Inflammation/génétique , Souris de lignée BALB C , Douleur/métabolisme , Douleur/génétique , Gestion de la douleur , Rat Wistar , Récepteurs à l'interleukine-8B/métabolisme , Récepteurs à l'interleukine-8B/génétique , Transduction du signal , Cortex somatosensoriel/métabolisme
13.
J Exp Clin Cancer Res ; 43(1): 121, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38654356

RÉSUMÉ

BACKGROUND: Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, and chemotherapy still serves as the cornerstone treatment functioning by inducing cytotoxic cell death. Notably, emerging evidence suggests that dying cell-released signals may induce cancer progression and metastasis by modulating the surrounding microenvironment. However, the underlying molecular mechanisms and targeting strategies are yet to be explored. METHODS: Apoptotic TNBC cells induced by paclitaxel or adriamycin treatment were sorted and their released extracellular vesicles (EV-dead) were isolated from the cell supernatants. Chemokine array analysis was conducted to identify the crucial molecules in EV-dead. Zebrafish and mouse xenograft models were used to investigate the effect of EV-dead on TNBC progression in vivo. RESULTS: It was demonstrated that EV-dead were phagocytized by macrophages and induced TNBC metastasis by promoting the infiltration of immunosuppressive PD-L1+ TAMs. Chemokine array identified CXCL1 as a crucial component in EV-dead to activate TAM/PD-L1 signaling. CXCL1 knockdown in EV-dead or macrophage depletion significantly inhibited EV-dead-induced TNBC growth and metastasis. Mechanistic investigations revealed that CXCL1EV-dead enhanced TAM/PD-L1 signaling by transcriptionally activating EED-mediated PD-L1 promoter activity. More importantly, TPCA-1 (2-[(aminocarbonyl) amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide) was screened as a promising inhibitor targeting CXCL1 signals in EVs to enhance paclitaxel chemosensitivity and limit TNBC metastasis without noticeable toxicities. CONCLUSIONS: Our results highlight CXCL1EV-dead as a novel dying cell-released signal and provide TPCA-1 as a targeting candidate to improve TNBC prognosis.


Sujet(s)
Antigène CD274 , Chimiokine CXCL1 , Vésicules extracellulaires , Transduction du signal , Tumeurs du sein triple-négatives , Macrophages associés aux tumeurs , Animaux , Femelle , Humains , Souris , Antigène CD274/métabolisme , Lignée cellulaire tumorale , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Vésicules extracellulaires/métabolisme , Métastase tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/anatomopathologie , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/traitement médicamenteux , Tests d'activité antitumorale sur modèle de xénogreffe , Danio zébré , Macrophages associés aux tumeurs/métabolisme
14.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38673949

RÉSUMÉ

Chemokines play a key role in cancer processes, with CXCL1 being a well-studied example. Due to the lack of a complete summary of CXCL1's role in cancer in the literature, in this study, we examine the significance of CXCL1 in various cancers such as bladder, glioblastoma, hemangioendothelioma, leukemias, Kaposi's sarcoma, lung, osteosarcoma, renal, and skin cancers (malignant melanoma, basal cell carcinoma, and squamous cell carcinoma), along with thyroid cancer. We focus on understanding how CXCL1 is involved in the cancer processes of these specific types of tumors. We look at how CXCL1 affects cancer cells, including their proliferation, migration, EMT, and metastasis. We also explore how CXCL1 influences other cells connected to tumors, like promoting angiogenesis, recruiting neutrophils, and affecting immune cell functions. Additionally, we discuss the clinical aspects by exploring how CXCL1 levels relate to cancer staging, lymph node metastasis, patient outcomes, chemoresistance, and radioresistance.


Sujet(s)
Chimiokine CXCL1 , Tumeurs , Humains , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/génétique , Animaux , Transition épithélio-mésenchymateuse/génétique , Pertinence clinique
15.
Gastroenterology ; 167(2): 281-297, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38492894

RÉSUMÉ

BACKGROUND & AIMS: Because pancreatic cancer responds poorly to chemotherapy and immunotherapy, it is necessary to identify novel targets and compounds to overcome resistance to treatment. METHODS: This study analyzed genomic single nucleotide polymorphism sequencing, single-cell RNA sequencing, and spatial transcriptomics. Ehf-knockout mice, KPC (LSL-KrasG12D/+, LSL-Trp53R172H/+ and Pdx1-Cre) mice, CD45.1+ BALB/C nude mice, and CD34+ humanized mice were also used as subjects. Multiplexed immunohistochemistry and flow cytometry were performed to investigate the proportion of tumor-infiltrated C-X-C motif chemokine receptor 2 (CXCR2)+ neutrophils. In addition, multiplexed cytokines assays and chromatin immunoprecipitation assays were used to examine the mechanism. RESULTS: The TP53 mutation-mediated loss of tumoral EHF increased the recruitment of CXCR2+ neutrophils, modulated their spatial distribution, and further induced chemo- and immunotherapy resistance in clinical cohorts and preclinical syngeneic mice models. Mechanistically, EHF deficiency induced C-X-C motif chemokine ligand 1 (CXCL1) transcription to enhance in vitro and in vivo CXCR2+ neutrophils migration. Moreover, CXCL1 or CXCR2 blockade completely abolished the effect, indicating that EHF regulated CXCR2+ neutrophils migration in a CXCL1-CXCR2-dependent manner. The depletion of CXCR2+ neutrophils also blocked the in vivo effects of EHF deficiency on chemotherapy and immunotherapy resistance. The single-cell RNA-sequencing results of PDAC treated with Nifurtimox highlighted the therapeutic significance of Nifurtimox by elevating the expression of tumoral EHF and decreasing the weightage of CXCL1-CXCR2 pathway within the microenvironment. Importantly, by simultaneously inhibiting the JAK1/STAT1 pathway, it could significantly suppress the recruitment and function of CXCR2+ neutrophils, further sensitizing PDAC to chemotherapy and immunotherapies. CONCLUSIONS: The study demonstrated the role of EHF in the recruitment of CXCR2+ neutrophils and the promising role of Nifurtimox in sensitizing pancreatic cancer to chemotherapy and immunotherapy.


Sujet(s)
Chimiokine CXCL1 , Résistance aux médicaments antinéoplasiques , Infiltration par les neutrophiles , Granulocytes neutrophiles , Tumeurs du pancréas , Récepteurs à l'interleukine-8B , Animaux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Récepteurs à l'interleukine-8B/génétique , Récepteurs à l'interleukine-8B/métabolisme , Récepteurs à l'interleukine-8B/antagonistes et inhibiteurs , Humains , Infiltration par les neutrophiles/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Souris , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Lignée cellulaire tumorale , Souris knockout , Microenvironnement tumoral , Immunothérapie/méthodes , Souris nude , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Souris de lignée BALB C , Antinéoplasiques/pharmacologie , Transduction du signal , Mutation , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/immunologie , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/anatomopathologie
16.
J Leukoc Biol ; 115(6): 1177-1182, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38298146

RÉSUMÉ

CXCL17, a novel member of the CXC chemokine class, has been implicated in several human pathologies, but its role in mediating immune response is not well understood. Characteristic features of immune response include resident macrophages orchestrating successive and structured recruitment of neutrophils and monocytes to the insult site. Here, we show that Cxcl17 knockout (KO) mice, compared with the littermate wild-type control mice, were significantly impaired in peritoneal neutrophil recruitment post-lipopolysaccharide (LPS) challenge. Further, the KO mice show dysregulated Cxcl1, Cxcr2, and interleukin-6 levels, all of which directly impact neutrophil recruitment. Importantly, the KO mice showed no difference in monocyte recruitment post-LPS challenge or in peritoneal macrophage levels in both unchallenged and LPS-challenged mice. We conclude that Cxcl17 is a proinflammatory chemokine and that it plays an important role in the early proinflammatory response by promoting neutrophil recruitment to the insult site.


Sujet(s)
Chimiokines CXC , Lipopolysaccharides , Souris knockout , Granulocytes neutrophiles , Récepteurs à l'interleukine-8B , Animaux , Souris , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Récepteurs à l'interleukine-8B/métabolisme , Récepteurs à l'interleukine-8B/génétique , Chimiokines CXC/métabolisme , Chimiokines CXC/génétique , Lipopolysaccharides/pharmacologie , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Interleukine-6/métabolisme , Interleukine-6/génétique , Infiltration par les neutrophiles , Souris de lignée C57BL , Inflammation/immunologie , Inflammation/anatomopathologie , Inflammation/métabolisme , Macrophages péritonéaux/immunologie , Macrophages péritonéaux/métabolisme , Monocytes/immunologie , Monocytes/métabolisme
17.
J Hepatol ; 81(1): 93-107, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38403027

RÉSUMÉ

BACKGROUND & AIMS: The effectiveness of immune checkpoint inhibitor (ICI) therapy for hepatocellular carcinoma (HCC) is limited by treatment resistance. However, the mechanisms underlying immunotherapy resistance remain elusive. We aimed to identify the role of CT10 regulator of kinase-like (CRKL) in resistance to anti-PD-1 therapy in HCC. METHODS: Gene expression in HCC specimens from 10 patients receiving anti-PD-1 therapy was identified by RNA-sequencing. A total of 404 HCC samples from tissue microarrays were analyzed by immunohistochemistry. Transgenic mice (Alb-Cre/Trp53fl/fl) received hydrodynamic tail vein injections of a CRKL-overexpressing vector. Mass cytometry by time of flight was used to profile the proportion and status of different immune cell lineages in the mouse tumor tissues. RESULTS: CRKL was identified as a candidate anti-PD-1-resistance gene using a pooled genetic screen. CRKL overexpression nullifies anti-PD-1 treatment efficacy by mobilizing tumor-associated neutrophils (TANs), which block the infiltration and function of CD8+ T cells. PD-L1+ TANs were found to be an essential subset of TANs that were regulated by CRKL expression and display an immunosuppressive phenotype. Mechanistically, CRKL inhibits APC (adenomatous polyposis coli)-mediated proteasomal degradation of ß-catenin by competitively decreasing Axin1 binding, and thus promotes VEGFα and CXCL1 expression. Using human HCC samples, we verified the positive correlations of CRKL/ß-catenin/VEGFα and CXCL1. Targeting CRKL using CRISPR-Cas9 gene editing (CRKL knockout) or its downstream regulators effectively restored the efficacy of anti-PD-1 therapy in an orthotopic mouse model and a patient-derived organotypic tumor spheroid model. CONCLUSIONS: Activation of the CRKL/ß-catenin/VEGFα and CXCL1 axis is a critical obstacle to successful anti-PD-1 therapy. Therefore, CRKL inhibitors combined with anti-PD-1 could be useful for the treatment of HCC. IMPACT AND IMPLICATIONS: Here, we found that CRKL was overexpressed in anti-PD-1-resistant hepatocellular carcinoma (HCC) and that CRKL upregulation promotes anti-PD-1 resistance in HCC. We identified that upregulation of the CRKL/ß-catenin/VEGFα and CXCL1 axis contributes to anti-PD-1 tolerance by promoting infiltration of tumor-associated neutrophils. These findings support the strategy of bevacizumab-based immune checkpoint inhibitor combination therapy, and CRKL inhibitors combined with anti-PD-1 therapy may be developed for the treatment of HCC.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Carcinome hépatocellulaire , Résistance aux médicaments antinéoplasiques , Inhibiteurs de points de contrôle immunitaires , Tumeurs du foie , Infiltration par les neutrophiles , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/immunologie , Tumeurs du foie/génétique , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Animaux , Humains , Souris , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteur-1 de mort cellulaire programmée/génétique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Souris transgéniques , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Mâle , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique
18.
Mol Biol Rep ; 51(1): 331, 2024 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-38393465

RÉSUMÉ

BACKGROUND: ER positive breast cancer is currently targeted using various endocrine therapies. Despite the proven therapeutic efficacy, resistance to the drug and reoccurrence of tumor appears to be a complication that many patients deal with. Molecular pathways underlying the development of resistance are being widely studied. METHODS AND RESULTS: In this study, using four established endocrine resistant breast cancer (ERBC) cell lines, we characterized CXCL1 as a secreted factor in crosstalk between ERBC cells and fibroblasts. Protein array revealed upregulation of CXCL1 and we confirmed the CXCL1 expression by real-time qRT-PCR and U-Plex assay. Co-culturing ERBC cells with fibroblasts enhanced the cell growth and migration compared to monoculture. The crosstalk of ERBC cells with fibroblasts significantly activates ERK/MAPK signaling pathway while reparixin, CXCR1/2 receptor inhibitor, attenuates the activity. Reparixin displayed the ERBC cell growth inhibition and the combination treatment with reparixin and CDK4/6 inhibitor (palbociclib and ribociclib) increased these inhibitory effect. CONCLUSIONS: Taken together, our study implicates CXCL1 as a critical role in ERBC growth and metastasis via crosstalk with fibroblast and cotargeting CXCR1/2 and CDK4/6 could potentially overcome endocrine resistant breast cancer.


Sujet(s)
Tumeurs du sein , Chimiokine CXCL1 , Femelle , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Chimiokine CXCL1/génétique , Résistance aux médicaments antinéoplasiques , Fibroblastes/métabolisme , Sulfonamides/pharmacologie
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166988, 2024 02.
Article de Anglais | MEDLINE | ID: mdl-38070583

RÉSUMÉ

Psoriasis is a multifaceted chronic inflammatory skin disease; however, its underlying molecular mechanisms remain unclear. In this study, we explored the role of fucosylation in psoriasis using an imiquimod-induced psoriasis-like mouse model. ABH antigen and fucosyltransferase 1 (Fut1) expression was reduced in the granular layer of lesional skin of patients with psoriasis. In particular, the blood group H antigen type 2 (H2 antigen)-a precursor of blood group A and B antigens-and FUT1 were highly expressed throughout the spinous layer in both patients with psoriasis and the skin of imiquimod-treated mice. Upon the application of imiquimod, Fut1-deficient mice, which lacked the H2 antigen, exhibited higher clinical scores based on erythema, induration, and scaling than those of wild-type mice. Imiquimod-treated Fut1-deficient mice displayed increased skin thickness, trans-epidermal water loss, and Gr-1+ cell infiltration compared with wild-type mice. Notably, the levels of CXCL1 protein and mRNA were significantly higher in Fut1-deficient mice than those in wild-type mice; however, there were no significant differences in other psoriasis-related markers, such as IL-1ß, IL-6, IL-17A, and IL-23. Fut1-deficient primary keratinocytes treated with IL-17A also showed a significant increase in both mRNA and protein levels of CXCL1 compared with IL-17A-treated wild-type primary keratinocytes. Further mechanistic studies revealed that this increased Cxcl1 mRNA in Fut1-deficient keratinocytes was caused by enhanced Cxcl1 mRNA stabilization. In summary, our findings indicated that fucosylation, which is essential for ABH antigen synthesis in humans, plays a protective role in psoriasis-like skin inflammation and is a potential therapeutic target for psoriasis.


Sujet(s)
Antigènes de groupe sanguin , Psoriasis , Humains , Animaux , Souris , Imiquimod/effets indésirables , Interleukine-17/génétique , Interleukine-17/métabolisme , Antigènes H-2/effets indésirables , Psoriasis/induit chimiquement , Psoriasis/génétique , Inflammation/induit chimiquement , ARN messager/génétique , ARN messager/métabolisme , Antigènes de groupe sanguin/effets indésirables , Chimiokine CXCL1/génétique
20.
Exp Biol Med (Maywood) ; 248(23): 2249-2261, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38158808

RÉSUMÉ

Chemokines critically orchestrate the tumorigenesis, metastasis, and stemness features of cancer cells that lead to poor outcomes. High plasma levels of transforming growth factor-ß1 (TGFß1) correlate with poor prognostic features in advanced lung cancer patients, thus suggesting the importance of TGFß1 in the lung tumor microenvironment. However, the role of chemokines in TGFß1-induced tumor stemness features remains unclear. Here, we clarify the previously undocumented role of CXCL1 in TGFß1-induced lung cancer stemness features. CXCL1 and its receptor CXCR2 were significantly upregulated in TGFß1-induced lung cancer stem cells (CSCs). CXCL1 silencing (shCXCL1) suppressed stemness gene expression, tumorsphere formation, colony formation, drug resistance, and in vivo tumorigenicity in TGFß1-induced lung tumorspheres. Immunohistochemistry staining showed that patients with stage II/III lung cancer had higher expression levels of CXCL1. The levels of CXCL1 were positively associated with lymph node metastasis and correlated with the expression of the CSC transcription factor Oct-4. Furthermore, online database analysis revealed that CXCL1 expression was negatively correlated with lung cancer survival in patients. Patients with high TGFß1/CXCL1/CD44 co-expression had a worse survival rate. We suggest that CXCL1 serves as a crucial factor in TGFß1-induced stemness features of lung cancer.


Sujet(s)
Tumeurs du poumon , Humains , Tumeurs du poumon/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Lignée cellulaire tumorale , Chimiokine CXCL1/génétique , Chimiokine CXCL1/métabolisme , Cellules souches tumorales/métabolisme , Microenvironnement tumoral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE