Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 974
Filtrer
1.
J Extracell Vesicles ; 13(7): e12493, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39051750

RÉSUMÉ

Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and chemotherapy is the cornerstone treatment for TNBC. Regrettably, emerging findings suggest that chemotherapy facilitates pro-metastatic changes in the tumour microenvironment. Extracellular vesicles (EVs) have been highly implicated in cancer drug resistance and metastasis. However, the effects of the EVs released from dying cancer cells on TNBC prognosis and corresponding therapeutic strategies have been poorly investigated. This study demonstrated that paclitaxel chemotherapy elicited CXCL1-enriched EVs from apoptotic TNBC cells (EV-Apo). EV-Apo promoted the chemoresistance and invasion of co-cultured TNBC cells by polarizing M2 macrophages through activating PD-L1 signalling. However, baohuoside I (BHS) remarkably sensitized the co-cultured TNBC cells to paclitaxel chemotherapy via modulating EV-Apo signalling. Mechanistically, BHS remarkably decreased C-X-C motif chemokine ligand 1 (CXCL1) cargo within EV-Apo and therefore attenuated macrophage M2 polarization by suppressing PD-L1 activation. Additionally, BHS decreased EV-Apo release by diminishing the biogenesis of intraluminal vesicles (ILVs) within multivesicular bodies (MVBs) of TNBC cells. Furthermore, BHS bound to the LEU104 residue of flotillin 2 (FLOT2) and interrupted its interaction with RAS oncogene family member 31 (RAB31), leading to the blockage of RAB31-FLOT2 complex-driven ILV biogenesis. Importantly, BHS remarkably chemosensitised paclitaxel to inhibit TNBC metastasis in vivo by suppressing EV-ApoCXCL1-induced PD-L1 activation and M2 polarization of tumour-associated macrophages (TAMs). This pioneering study sheds light on EV-ApoCXCL1 as a novel therapeutic target to chemosensitise TNBC, and presents BHS as a promising chemotherapy adjuvant to improve TNBC chemosensitivity and prognosis by disturbing EV-ApoCXCL1 biogenesis.


Sujet(s)
Apoptose , Chimiokine CXCL1 , Vésicules extracellulaires , Paclitaxel , Tumeurs du sein triple-négatives , Humains , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Femelle , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/effets des médicaments et des substances chimiques , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques , Chimiokine CXCL1/métabolisme , Lignée cellulaire tumorale , Animaux , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques
2.
BMC Cancer ; 24(1): 799, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965454

RÉSUMÉ

BACKGROUND: Craniopharyngioma (CP) is a rare malformational tumor characterized by high rates of recurrence and morbid obesity. However, the role of inflammatory mediators in obesity and the prognosis of patients with CP remains unknown. Therefore, the present study aimed to analyze associations of inflammatory mediators with weight-related outcomes and the prognosis of patients with CP. METHODS: A total of 130 consecutive patients with CP were included in this study. The expression levels of seven inflammatory mediators and the plasma leptin concentration were investigated. Clinical parameters, weight changes, new-onset obesity, and progression-free survival (PFS) were recorded. The relationships between inflammatory mediators, clinicopathologic parameters, weight-related outcomes, and PFS were explored. RESULTS: Compared with those in normal pituitary tissue, the expressions of inflammatory mediators in tumor tissue were higher. Higher expression levels of CXCL1 and CXCL8 were identified as independent risk factors for significant weight gain, and CXCL1 and TNF were identified as independent risk factors for new-onset postoperative obesity. Poor PFS was associated with higher expression levels of CXCL1, CXCL8, IL1A, IL6, and TNF. CONCLUSION: The present study revealed that inflammatory mediators are associated with morbid obesity in patients with CP. Inflammatory mediators may be the critical bridge between elevated leptin and weight-related outcomes. Additionally, PFS was associated with the expression of inflammatory mediators. Further research is needed to elucidate the underlying mechanisms of inflammatory mediators and their potential as targets for novel therapies for CP.


Sujet(s)
Craniopharyngiome , Médiateurs de l'inflammation , Leptine , Tumeurs de l'hypophyse , Survie sans progression , Humains , Craniopharyngiome/métabolisme , Craniopharyngiome/anatomopathologie , Craniopharyngiome/mortalité , Craniopharyngiome/complications , Femelle , Mâle , Adulte , Tumeurs de l'hypophyse/mortalité , Tumeurs de l'hypophyse/métabolisme , Tumeurs de l'hypophyse/anatomopathologie , Tumeurs de l'hypophyse/sang , Adulte d'âge moyen , Médiateurs de l'inflammation/métabolisme , Leptine/sang , Leptine/métabolisme , Pronostic , Obésité/complications , Obésité/métabolisme , Obésité morbide/complications , Obésité morbide/métabolisme , Obésité morbide/mortalité , Jeune adulte , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/sang , Âge de début , Facteurs de risque , Pertinence clinique , Interleukine-8
3.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000295

RÉSUMÉ

Olprinone (OLP) is a selective inhibitor of phosphodiesterase III and is used clinically in patients with heart failure and those undergoing cardiac surgery; however, little is known about the effects of OLP on hepatoprotection. The purpose of this study aimed to determine whether OLP has protective effects in in vivo and in vitro rat models of endotoxin-induced liver injury after hepatectomy and to clarify the mechanisms of action of OLP. In the in vivo model, rats underwent 70% partial hepatectomy and lipopolysaccharide treatment (PH/LPS). OLP administration increased survival by 85.7% and decreased tumor necrosis factor-α, C-X-C motif chemokine ligand 1, and inducible nitric oxide synthase (iNOS) mRNA expression in the livers of rats treated with PH/LPS. OLP also suppressed nuclear translocation and/or DNA binding ability of nuclear factor kappa B (NF-κB). Pathological liver damage induced by PH/LPS was alleviated and neutrophil infiltration was reduced by OLP. Primary cultured rat hepatocytes treated with the pro-inflammatory cytokine interleukin-1ß (IL-1ß) were used as a model of in vitro liver injury. Co-treatment with OLP inhibited dose-dependently IL-1ß-stimulated iNOS induction and NF-κB activation. Our results demonstrate that OLP may partially inhibit the induction of several inflammatory mediators through the suppression of NF-κB and thus prevent liver injury induced by endotoxin after liver resection.


Sujet(s)
Modèles animaux de maladie humaine , Hépatectomie , Hépatocytes , Imidazoles , Facteur de transcription NF-kappa B , Nitric oxide synthase type II , Pyridones , Animaux , Hépatectomie/effets indésirables , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Rats , Mâle , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Facteur de transcription NF-kappa B/métabolisme , Imidazoles/pharmacologie , Nitric oxide synthase type II/métabolisme , Inhibiteurs de la phosphodiestérase-3/pharmacologie , Inhibiteurs de la phosphodiestérase-3/usage thérapeutique , Interleukine-1 bêta/métabolisme , Lipopolysaccharides/effets indésirables , Lipopolysaccharides/toxicité , Sepsie/traitement médicamenteux , Rat Sprague-Dawley , Cellules cultivées , Facteur de nécrose tumorale alpha/métabolisme , Chimiokine CXCL1/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme
4.
Nat Commun ; 15(1): 5817, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38987270

RÉSUMÉ

Respiratory infections caused by the human fungal pathogen Aspergillus fumigatus are a major cause of mortality for immunocompromised patients. Exposure to these pathogens occurs through inhalation, although the role of the respiratory epithelium in disease pathogenesis has not been fully defined. Employing a primary human airway epithelial model, we demonstrate that fungal melanins potently block the post-translational secretion of the chemokines CXCL1 and CXCL8 independent of transcription or the requirement of melanin to be phagocytosed, leading to a significant reduction in neutrophil recruitment to the apical airway both in vitro and in vivo. Aspergillus-derived melanin, a major constituent of the fungal cell wall, dampened airway epithelial chemokine secretion in response to fungi, bacteria, and exogenous cytokines. Furthermore, melanin muted pathogen-mediated calcium fluxing and hindered actin filamentation. Taken together, our results reveal a critical role for melanin interaction with airway epithelium in shaping the host response to fungal and bacterial pathogens.


Sujet(s)
Aspergillus fumigatus , Calcium , Chimiokine CXCL1 , Interleukine-8 , Mélanines , Mélanines/métabolisme , Humains , Interleukine-8/métabolisme , Calcium/métabolisme , Chimiokine CXCL1/métabolisme , Animaux , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/microbiologie , Souris , Cellules épithéliales/métabolisme , Cellules épithéliales/microbiologie , Chimiokines/métabolisme , Souris de lignée C57BL
5.
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
6.
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 ,
7.
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
8.
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
9.
J Reprod Immunol ; 164: 104278, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38901109

RÉSUMÉ

The objective of this study was to investigate the immune mechanisms involved in preterm labor (PTL), preterm prelabor rupture of the membranes (PPROM), and normal pregnancies. The second objective was to explore immune profiles in PTL for association with early ( < 34 gestational weeks (gw)) or instant ( < 48 h) delivery. This prospective observational multi-center study included women with singleton pregnancies with PTL (n = 80) or PPROM (n = 40) before 34 gw, women with normal pregnancies scheduled for antenatal visits (n = 44), and women with normal pregnancies in active labor at term (n = 40). Plasma samples obtained at admission were analyzed for cytokine and chemokine quantification using a multiplex bead assay in order to compare the immune profiles between PTL, PPROM, and normal pregnancies. In PTL, CXCL1 and CCL17 were significantly higher compared to gestational age-matched women at antenatal visits, whereas for PPROM, CXCL1 and IL-6 were increased. Women in term labor had a more pronounced inflammatory pattern with higher levels of CXCL1, CXCL8, and IL-6 compared with PTL (p = 0.007, 0.003, and 0.013, respectively), as well as higher levels of CCL17, CXCL1 and IL-6 (all p < 0.001) compared with the women at antenatal visits. In PTL, CXCL8 was higher in women with delivery before 34 gw, whereas CXCL8, GM-CSF, and IL-6 were significantly higher in women with delivery within 48 h. To conclude, PTL and PPROM were associated with a complex pattern of inflammation, both involving Th17 (CXCL1) responses. Although further studies are needed, CXCL8, GM-CSF, and IL-6 may be potential candidates for predicting preterm birth in PTL.


Sujet(s)
Rupture prématurée des membranes foetales , Travail obstétrical prématuré , Humains , Femelle , Grossesse , Rupture prématurée des membranes foetales/sang , Rupture prématurée des membranes foetales/immunologie , Adulte , Travail obstétrical prématuré/immunologie , Travail obstétrical prématuré/sang , Travail obstétrical prématuré/diagnostic , Études prospectives , Cytokines/sang , Chimiokines/sang , Interleukine-6/sang , Âge gestationnel , Chimiokine CXCL1/sang , Chimiokine CXCL1/métabolisme , Chimiokine CCL17
10.
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
11.
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
12.
J Immunother Cancer ; 12(5)2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724465

RÉSUMÉ

BACKGROUND: CD276 (B7-H3), a pivotal immune checkpoint, facilitates tumorigenicity, invasiveness, and metastasis by escaping immune surveillance in a variety of tumors; however, the underlying mechanisms facilitating immune escape in esophageal squamous cell carcinoma (ESCC) remain enigmatic. METHODS: We investigated the expression of CD276 in ESCC tissues from patients by using immunohistochemistry (IHC) assays. In vivo, we established a 4-nitroquinoline 1-oxide (4NQO)-induced CD276 knockout (CD276wKO) and K14cre; CD276 conditional knockout (CD276cKO) mouse model of ESCC to study the functional role of CD276 in ESCC. Furthermore, we used the 4NQO-induced mouse model to evaluate the effects of anti-CXCL1 antibodies, anti-Ly6G antibodies, anti-NK1.1 antibodies, and GSK484 inhibitors on tumor growth. Moreover, IHC, flow cytometry, and immunofluorescence techniques were employed to measure immune cell proportions in ESCC. In addition, we conducted single-cell RNA sequencing analysis to examine the alterations in tumor microenvironment following CD276 depletion. RESULTS: In this study, we elucidate that CD276 is markedly upregulated in ESCC, correlating with poor prognosis. In vivo, our results indicate that depletion of CD276 inhibits tumorigenesis and progression of ESCC. Furthermore, conditional knockout of CD276 in epithelial cells engenders a significant downregulation of CXCL1, consequently reducing the formation of neutrophil extracellular trap networks (NETs) via the CXCL1-CXCR2 signaling axis, while simultaneously augmenting natural killer (NK) cells. In addition, overexpression of CD276 promotes tumorigenesis via increasing NETs' formation and reducing NK cells in vivo. CONCLUSIONS: This study successfully elucidates the functional role of CD276 in ESCC. Our comprehensive analysis uncovers the significant role of CD276 in modulating immune surveillance mechanisms in ESCC, thereby suggesting that targeting CD276 might serve as a potential therapeutic approach for ESCC treatment.


Sujet(s)
Antigènes B7 , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Pièges extracellulaires , Animaux , Femelle , Humains , Mâle , Souris , Antigènes B7/métabolisme , Chimiokine CXCL1/métabolisme , Tumeurs de l'oesophage/génétique , Tumeurs de l'oesophage/immunologie , Tumeurs de l'oesophage/métabolisme , Tumeurs de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/immunologie , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/métabolisme , Pièges extracellulaires/métabolisme , Souris knockout , Récepteurs à l'interleukine-8B/métabolisme , Échappement de la tumeur à la surveillance immunitaire , Microenvironnement tumoral
13.
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
14.
Toxicology ; 506: 153845, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38801935

RÉSUMÉ

We investigated the intratracheal instillation of Polyacrylic acid (PAA) in rats to determine if it would cause pulmonary disorders, and to see what factors would be associated with the pathological changes. Male F344 rats were intratracheally instilled with low (0.2 mg/rat) and high (1.0 mg/rat) doses of PAA. They were sacrificed at 3 days, 1 week, 1 month, 3 months, and 6 months after PAA exposure to examine inflammatory and fibrotic changes in the lungs. There was a persistent increase in the neutrophil count, lactate dehydrogenase (LDH) levels, cytokine-induced neutrophil chemoattractant (CINC) values in bronchoalveolar lavage fluid (BALF), and heme oxygenase-1 (HO-1) in lung tissue. Transforming growth factor-beta 1 (TGF-ß1), a fibrotic factor, showed a sustained increase in the BALF until 6 months after intratracheal instillation, and connective tissue growth factor (CTGF) in lung tissue was elevated at 3 days after exposure. Histopathological findings in the lung tissue showed persistent (more than one month) inflammation, fibrotic changes, and epithelial-mesenchymal transition (EMT) changes. There was also a strong correlation between TGF-ß1 in the BALF and, especially, in the fibrosis score of histopathological specimens. Intratracheal instillation of PAA induced persistent neutrophilic inflammation, fibrosis, and EMT in the rats' lungs, and TGF-ß1 and CTGF appeared to be associated with the persistent fibrosis.


Sujet(s)
Résines acryliques , Liquide de lavage bronchoalvéolaire , Facteur de croissance du tissu conjonctif , Fibrose pulmonaire , Rats de lignée F344 , Facteur de croissance transformant bêta-1 , Animaux , Mâle , Facteur de croissance transformant bêta-1/métabolisme , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Résines acryliques/toxicité , Résines acryliques/administration et posologie , Facteur de croissance du tissu conjonctif/métabolisme , Liquide de lavage bronchoalvéolaire/composition chimique , Liquide de lavage bronchoalvéolaire/cytologie , Rats , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , L-Lactate dehydrogenase/métabolisme , Heme oxygenase-1/métabolisme , Chimiokine CXCL1/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Heme oxygenase (decyclizing)
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE