Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.373
Filtrer
1.
Sci Rep ; 14(1): 19811, 2024 08 27.
Article de Anglais | MEDLINE | ID: mdl-39191856

RÉSUMÉ

The tumor microenvironment (TME) plays an important role in the occurrence and progression of Acute Myeloid Leukemia (AML). Single-cell sequencing has enabled researchers to explore the correlation between TME subgroups and tumor prognosis, distinguish the existence of drug-resistant subgroups of tumor cells, and unravel the complexity of the AML cellular heterogeneity. We used bone marrow immune cell enrichment analysis from public databases to screen prognostic genes, construct prognostic models, and validate their prognostic significance on independent external datasets and patient samples. A total of 18,251 single cells were obtained to establish prognostic scoring models for 10 key genes including CCL5, ETLS2, and IL2RA.The AML cases were divided into two groups: high-risk and low-risk. The low-risk group exhibited a higher survival rate than the high-risk group. The areas under curves (AUC) of 1-, 3- and 5-year survival curves in the TCGA and GEO training sets were greater than 0.8 and 0.6, respectively, indicating effective prediction. The model's prognostic efficacy was confirmed across multiple validation sets. It demonstrated increased expression of ETS2, CCL5, and IL2RA in AML samples compared to controls, which was associated with decreased overall survival (OS). This prognostic scoring model based on tumor immune infiltration provides a reference for developing novel treatment strategies for recurrent/refractory AML.


Sujet(s)
Leucémie aigüe myéloïde , Analyse sur cellule unique , Transcriptome , Microenvironnement tumoral , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/mortalité , Leucémie aigüe myéloïde/anatomopathologie , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Pronostic , Analyse sur cellule unique/méthodes , Femelle , Mâle , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme , Marqueurs biologiques tumoraux/génétique , Adulte d'âge moyen , Protéine proto-oncogène c-ets-2
2.
Sci Immunol ; 9(98): eadk2612, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093956

RÉSUMÉ

Aberrant activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway causes autoimmunity in humans and mice; however, the exact mechanism by which the cGAS-STING pathway initiates adaptive immunity and tissue pathology is still not fully understood. Here, we used a cGAS knockin (KI) mouse model that develops systemic autoimmunity. In the lungs of cGAS-KI mice, blood vessels were enclosed by organized lymphoid tissues that resemble tertiary lymphoid structures (TLSs). Cell-intrinsic cGAS induction promoted up-regulation of CCR5 in CD8+ T cells and led to CCL5 production in vascular endothelial cells. Peripheral CD8+ T cells were recruited to the lungs and produced CXCL13 and interferon-γ. The latter triggered endothelial cell death, potentiated CCL5 production, and was essential for TLS establishment. Blocking CCL5 or CCR5, or depleting CD8+ T cells, impaired TLS formation. cGAS-mediated TLS formation also enhanced humoral and antitumor responses. These data demonstrate that cGAS signaling drives a specialized lymphoid structure that underlies autoimmune tissue pathology.


Sujet(s)
Lymphocytes T CD8+ , Cellules endothéliales , Nucleotidyltransferases , Structures lymphoïdes tertiaires , Animaux , Nucleotidyltransferases/immunologie , Nucleotidyltransferases/génétique , Nucleotidyltransferases/métabolisme , Souris , Cellules endothéliales/immunologie , Structures lymphoïdes tertiaires/immunologie , Lymphocytes T CD8+/immunologie , Chimiokine CCL5/immunologie , Chimiokine CCL5/génétique , Souris de lignée C57BL , Souris transgéniques , Transduction du signal/immunologie , Récepteurs CCR5/immunologie , Récepteurs CCR5/génétique , Récepteurs CCR5/métabolisme , Auto-immunité/immunologie
3.
Elife ; 132024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38949655

RÉSUMÉ

Secreted chemokines form concentration gradients in target tissues to control migratory directions and patterns of immune cells in response to inflammatory stimulation; however, how the gradients are formed is much debated. Heparan sulfate (HS) binds to chemokines and modulates their activities. In this study, we investigated the roles of HS in the gradient formation and chemoattractant activity of CCL5 that is known to bind to HS. CCL5 and heparin underwent liquid-liquid phase separation and formed gradient, which was confirmed using CCL5 immobilized on heparin-beads. The biological implication of HS in CCL5 gradient formation was established in CHO-K1 (wild-type) and CHO-677 (lacking HS) cells by Transwell assay. The effect of HS on CCL5 chemoattractant activity was further proved by Transwell assay of human peripheral blood cells. Finally, peritoneal injection of the chemokines into mice showed reduced recruitment of inflammatory cells either by mutant CCL5 (lacking heparin-binding sequence) or by addition of heparin to wild-type CCL5. Our experimental data propose that co-phase separation of CCL5 with HS establishes a specific chemokine concentration gradient to trigger directional cell migration. The results warrant further investigation on other heparin-binding chemokines and allows for a more elaborate insight into disease process and new treatment strategies.


Sujet(s)
Chimiokine CCL5 , Chimiotaxie , Cricetulus , Héparitine sulfate , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Animaux , Héparitine sulfate/métabolisme , Humains , Cellules CHO , Souris , Héparine/métabolisme , Héparine/pharmacologie ,
4.
Int J Mol Sci ; 25(12)2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38928033

RÉSUMÉ

Bladder cancer (BC) is the 12th most commonly diagnosed cancer worldwide. Although there are several well-established molecular and immunological classifications, prognostic and predictive markers for tumor cells and immune cells are still needed. Using a tissue microarray, we analyzed the expression of the chemokine CC motif ligand 5 (CCL5) by immunohistochemistry (IHC) in 175 muscle-invasive BC samples. The application of a single cutoff for the staining status of tumor cells (TCs; positive vs. negative) and immune cells (ICs; positive vs. negative) revealed 75 patients (42.9%) and 123 patients (70.3%) with CCL5-positive TCs or ICs, respectively. IHC results were associated with prognostic and predictive data. Multivariate Cox regression analysis revealed that positive CCL5 staining in TCs was associated with significantly shorter disease-specific survival (DSS; RR = 1.51; p = 0.047), but CCL5-negative ICs were associated with significantly shorter overall survival (OS; RR = 1.66; p = 0.005), DSS (RR = 2.02; p = 0.001) and recurrence-free survival (RFS; RR = 1.94; p = 0.002). Adjuvant chemotherapy was favorable for patients with CCL5-negative ICs for OS (RR = 0.30; p = 0.006), DSS (RR = 0.36; p = 0.022) and RFS (RR = 0.41; p = 0.046) but not for patients with CCL5-positive ICs, except in the subgroup of N1 + N2 patients, where it was associated with better OS. We suggest that CCL5 expression can be a prognostic and predictive marker for muscle-invasive bladder cancer patients.


Sujet(s)
Marqueurs biologiques tumoraux , Chimiokine CCL5 , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/mortalité , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Mâle , Femelle , Sujet âgé , Pronostic , Adulte d'âge moyen , Marqueurs biologiques tumoraux/métabolisme , Invasion tumorale , Sujet âgé de 80 ans ou plus , Adulte , Immunohistochimie
5.
Adv Sci (Weinh) ; 11(29): e2400611, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38873823

RÉSUMÉ

Immunosuppression is a major hallmark of tumor progression in non-small cell lung cancer (NSCLC). Cluster of differentiation 147 (CD147), an important pro-tumorigenic factor, is closely linked to NSCLC immunosuppression. However, the role of CD147 di-methylation in the immunosuppressive tumor microenvironment (TME) remains unclear. Here, di-methylation of CD147 at Lys148 (CD147-K148me2) is identified as a common post-translational modification (PTM) in NSCLC that is significantly associated with unsatisfying survival outcomes among NSCLC sufferers, especially those in the advanced stages of the disease. The methyltransferase NSD2 catalyzes CD147 to generate CD147-K148me2. Further analysis demonstrates that CD147-K148me2 reestablishes the immunosuppressive TME and promotes NSCLC progression. Mechanistically, this modification promotes the interaction between cyclophilin A (CyPA) and CD147, and in turn, increases CCL5 gene transcription by activating p38-ZBTB32 signaling, leading to increased NSCLC cell-derived CCL5 secretion. Subsequently, CD147-K148me2-mediated CCL5 upregulation facilitates M2-like tumor-associated macrophage (TAM) infiltration in NSCLC tissues via CCL5/CCR5 axis-dependent intercellular crosstalk between tumor cells and macrophages, which is inhibited by blocking CD147-K148me2 with the targeted antibody 12C8. Overall, this study reveals the role of CD147-K148me2-driven intercellular crosstalk in the development of NSCLC immunosuppression, and provides a potential interventional strategy for PTM-targeted NSCLC therapy.


Sujet(s)
Antigènes CD147 , Carcinome pulmonaire non à petites cellules , Chimiokine CCL5 , Tumeurs du poumon , Récepteurs CCR5 , Microenvironnement tumoral , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Humains , Tumeurs du poumon/métabolisme , Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Antigènes CD147/métabolisme , Antigènes CD147/génétique , Souris , Animaux , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Microenvironnement tumoral/immunologie , Macrophages/métabolisme , Macrophages/immunologie , Lignée cellulaire tumorale , Immunosuppression thérapeutique , Modèles animaux de maladie humaine , Transduction du signal
6.
J Cell Mol Med ; 28(12): e18489, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38899522

RÉSUMÉ

This study explores the impact of senescence on autocrine C-C motif chemokine ligand 5 (CCL5) in human endothelial progenitor cell (EPCs), addressing the poorly understood decline in number and function of EPCs during ageing. We examined the effects of replication-induced senescence on CCL5/CCL5 receptor (CCR5) signalling and angiogenic activity of EPCs in vitro and in vivo. We also explored microRNAs controlling CCL5 secretion in senescent EPCs, its impact on EPC angiogenic activity, and validated our findings in humans. CCL5 secretion and CCR5 levels in senescent EPCs were reduced, leading to attenuated angiogenic activity. CCL5 enhanced EPC proliferation via the CCR5/AKT/P70S6K axis and increased vascular endothelial growth factor (VEGF) secretion. Up-regulation of miR-409 in senescent EPCs resulted in decreased CCL5 secretion, inhibiting the angiogenic activity, though these negative effects were counteracted by the addition of CCL5 and VEGF. In a mouse hind limb ischemia model, CCL5 improved the angiogenic activity of senescent EPCs. Analysis involving 62 healthy donors revealed a negative association between CCL5 levels, age and Framingham Risk Score. These findings propose CCL5 as a potential biomarker for detection of EPC senescence and cardiovascular risk assessment, suggesting its therapeutic potential for age-related cardiovascular disorders.


Sujet(s)
Vieillissement de la cellule , Chimiokine CCL5 , Progéniteurs endothéliaux , microARN , Néovascularisation physiologique , Animaux , Humains , Mâle , Souris , , Prolifération cellulaire , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Régulation négative/génétique , Progéniteurs endothéliaux/métabolisme , Progéniteurs endothéliaux/cytologie , Ischémie/métabolisme , Ischémie/anatomopathologie , Ischémie/génétique , microARN/génétique , microARN/métabolisme , Néovascularisation physiologique/génétique , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Transduction du signal , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique
7.
J Immunother Cancer ; 12(5)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38719543

RÉSUMÉ

The CCR/L5 axis is known for its role in immune regulation in a variety of settings and has been shown to have dichotomous functions in cancer, influencing both tumor progression and immune responses. Battaglin et al investigated its role using genomic and transcriptomic data from several datasets of patients with advanced colorectal cancer (CRC), including patients treated on CALGB/SWOG 80405, a trial of chemotherapy plus cetuximab versus bevacizumab, as well as a larger population of patients whose CRCs underwent commercially available Caris NGS and CODEai assays. These authors showed that CCR/L5 expression was both prognostic and predictive. They reported that low expression of the CCR/L5 axis was correlated with improved survival broadly, with particular benefit in patients treated with chemotherapy plus cetuximab. They demonstrated that high expression of CCR/L5 was associated with infiltration by negatively prognostic Tregs, M1 and M2 macrophages, myeloid-derived suppressor cells, and cancer-associated fibroblasts. They also showed that increased expression was correlated a wide variety of immune suppressive proteins, including PD-1, PD-L1, PD-L2, CTLA4, CD80, CD86, TIM3, IDO1, LAG3, and IFN-γ. This suggests mechanisms by which CRC resists anti-cancer immune responses. This study enhances our understanding of the role of the CCR/L5 axis in advanced CRC.


Sujet(s)
Chimiokine CCL5 , Tumeurs colorectales , Récepteurs CCR5 , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Récepteurs CCR5/métabolisme , Récepteurs CCR5/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Métastase tumorale
8.
Oncogene ; 43(28): 2215-2227, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38802647

RÉSUMÉ

Approximately 40% of patients with lung adenocarcinoma (LUAD) often develop bone metastases during the course of their disease. However, scarcely any in vivo model of LUAD bone metastasis has been established, leading to a poor understanding of the mechanisms underlying LUAD bone metastasis. Here, we established a multiorgan metastasis model via the left ventricular injection of luciferase-labeled LUAD cells into nude mice and then screened out lung metastasis (LuM) and bone metastasis (BoM) cell subpopulations. BoM cells exhibited greater stemness and epithelial-mesenchymal transition (EMT) plasticity than LuM cells and initially colonized the bone and subsequently disseminated to distant organs after being reinjected into mice. Moreover, a CD74-ROS1 fusion mutation (C6; R34) was detected in BoM cells but not in LuM cells. Mechanistically, BoM cells bearing the CD74-ROS1 fusion highly secrete the C-C motif chemokine ligand 5 (CCL5) protein by activating STAT3 signaling, recruiting macrophages in tumor microenvironment and strongly inducing M2 polarization of macrophages. BoM cell-activated macrophages produce a high level of TGF-ß1, thereby facilitating EMT and invasion of LUAD cells via TGF-ß/SMAD2/3 signaling. Targeting the CD74-ROS1/CCL5 axis with Crizotinib (a ROS1 inhibitor) and Maraviroc (a CCL5 receptor inhibitor) in vivo strongly impeded bone metastasis and secondary metastasis of BoM cells. Our findings reveal the critical role of the CD74-ROS1/STAT3/CCL5 axis in the interaction between LUAD bone metastasis cells and macrophages for controlling LUAD cell dissemination, highlighting the significance of the bone microenvironment in LUAD bone metastasis and multiorgan secondary metastasis, and suggesting that targeting CD74-ROS1 and CCL5 is a promising therapeutic strategy for LUAD bone metastasis.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs osseuses , Transition épithélio-mésenchymateuse , Tumeurs du poumon , Macrophages , Protein-tyrosine kinases , Protéines proto-oncogènes , Animaux , Humains , Souris , Tumeurs osseuses/secondaire , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Tumeurs du poumon/secondaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/secondaire , Adénocarcinome pulmonaire/métabolisme , Transition épithélio-mésenchymateuse/génétique , Protein-tyrosine kinases/métabolisme , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Antigènes de différenciation des lymphocytes B/métabolisme , Antigènes de différenciation des lymphocytes B/génétique , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Antigènes d'histocompatibilité de classe II/génétique , Antigènes d'histocompatibilité de classe II/métabolisme , Souris nude , Lignée cellulaire tumorale , Microenvironnement tumoral , Protéines de fusion oncogènes/génétique , Protéines de fusion oncogènes/métabolisme , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Transduction du signal
9.
J Transl Med ; 22(1): 502, 2024 May 26.
Article de Anglais | MEDLINE | ID: mdl-38797830

RÉSUMÉ

BACKGROUND: Inflammation and dysregulated immunity play vital roles in idiopathic pulmonary arterial hypertension (IPAH), while the mechanisms that initiate and promote these processes are unclear. METHODS: Transcriptomic data of lung tissues from IPAH patients and controls were obtained from the Gene Expression Omnibus database. Weighted gene co-expression network analysis (WGCNA), differential expression analysis, protein-protein interaction (PPI) and functional enrichment analysis were combined with a hemodynamically-related histopathological score to identify inflammation-associated hub genes in IPAH. The monocrotaline-induced rat model of pulmonary hypertension was utilized to confirm the expression pattern of these hub genes. Single-cell RNA-sequencing (scRNA-seq) data were used to identify the hub gene-expressing cell types and their intercellular interactions. RESULTS: Through an extensive bioinformatics analysis, CXCL9, CCL5, GZMA and GZMK were identified as hub genes that distinguished IPAH patients from controls. Among these genes, pulmonary expression levels of Cxcl9, Ccl5 and Gzma were elevated in monocrotaline-exposed rats. Further investigation revealed that only CCL5 and GZMA were highly expressed in T and NK cells, where CCL5 mediated T and NK cell interaction with endothelial cells, smooth muscle cells, and fibroblasts through multiple receptors. CONCLUSIONS: Our study identified a new inflammatory pathway in IPAH, where T and NK cells drove heightened inflammation predominantly via the upregulation of CCL5, providing groundwork for the development of targeted therapeutics.


Sujet(s)
Chimiokine CCL5 , Hypertension artérielle pulmonaire primitive familiale , Cellules tueuses naturelles , RNA-Seq , Analyse sur cellule unique , Lymphocytes T , Animaux , Humains , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/immunologie , Hypertension artérielle pulmonaire primitive familiale/génétique , Hypertension artérielle pulmonaire primitive familiale/anatomopathologie , Hypertension artérielle pulmonaire primitive familiale/métabolisme , Lymphocytes T/métabolisme , Lymphocytes T/immunologie , Mâle , Communication cellulaire/génétique , Rat Sprague-Dawley , Poumon/anatomopathologie , Rats , Réseaux de régulation génique , Monocrotaline , Cartes d'interactions protéiques/génétique , Biologie informatique
10.
J Immunother ; 47(6): 195-204, 2024.
Article de Anglais | MEDLINE | ID: mdl-38654631

RÉSUMÉ

SUMMARY: Immunocytokines are a promising immunotherapeutic approach in cancer therapy. Anti-VEGFR2-interferon α (IFNα) suppressed colorectal cancer (CRC) growth and enhanced CD8 + T-cell infiltration in the tumor microenvironment, exhibiting great clinical translational potential. However, the mechanism of how the anti-VEGFR2-IFNα recruits T cells has not been elucidated. Here, we demonstrated that anti-VEGFR2-IFNα suppressed CRC metastasis and enhanced CD8 + T-cell infiltration. RNA sequencing revealed a transcriptional activation of CCL5 in metastatic CRC cells, which was correlated with T-cell infiltration. IFNα but not anti-VEGFR2 could further upregulate CCL5 in tumors. In immunocompetent mice, both IFNα and anti-VEGFR2-IFNα increased the subset of tumor-infiltrating CD8 + T cells through upregulation of CCL5. Knocking down CCL5 in tumor cells attenuated the infiltration of CD8 + T cells and dampened the antitumor efficacy of anti-VEGFR2-IFNα treatment. We, therefore, propose upregulation of CCL5 is a key to enhance infiltration of CD8 + T cells in metastatic CRC with IFNα and IFNα-based immunocytokine treatments. These findings may help the development of IFNα related immune cytokines for the treatment of less infiltrated tumors.


Sujet(s)
Lymphocytes T CD8+ , Chimiokine CCL5 , Tumeurs colorectales , Interféron alpha , Lymphocytes TIL , Récepteur-2 au facteur croissance endothéliale vasculaire , Animaux , Femelle , Humains , Souris , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Interféron alpha/métabolisme , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Microenvironnement tumoral/immunologie , Régulation positive , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique
11.
Am J Physiol Cell Physiol ; 326(5): C1320-C1333, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38497114

RÉSUMÉ

Intramuscular fat (IMF) refers to the lipid stored in skeletal muscle tissue. The number and size of intramuscular adipocytes are the primary factors that regulate IMF content. Intramuscular adipocytes can be derived from either in situ or ectopic migration. In this study, it was discovered that the regulation of IMF levels is achieved through the chemokine (C-C motif) ligand 5 (CCL5)/chemokine (C-C motif) receptor 5 (CCR5) pathway by modulating adipocyte migration. In coculture experiments, C2C12 myotubes were more effective in promoting the migration of 3T3-L1 preadipocytes than C2C12 myoblasts, along with increasing CCL5. Correspondingly, overexpressing the CCR5, one of the receptors of CCL5, in 3T3-L1 preadipocytes facilitated their migration. Conversely, the application of the CCL5/CCR5 inhibitor, MARAVIROC (MVC), reduced this migration. In vivo, transplanted experiments of subcutaneous adipose tissue (SCAT) from transgenic mice expressing green fluorescent protein (GFP) provided evidence that injecting recombinant CCL5 (rCCL5) into skeletal muscle promotes the migration of subcutaneous adipocytes to the skeletal muscle. The level of CCL5 in skeletal muscle increased with obesity. Blocking the CCL5/CCR5 axis by MVC inhibited IMF deposition, whereas elevated skeletal muscle CCL5 promoted IMF deposition in obese mice. These results establish a link between the IMF and the CCL5/CCR5 pathway, which could have a potential application for modulating IMF through adipocyte migration.NEW & NOTEWORTHY C2C12 myotubes attract 3T3-L1 preadipocyte migration regulated by the chemokine (C-C motif) ligand 5 (CCL5)/ chemokine (C-C motif) receptor 5 (CCR5) axis. High levels of skeletal muscle-specific CCL5 promote the migration of subcutaneous adipocytes to skeletal muscle and induce the intramuscular fat (IMF) content.


Sujet(s)
Adipocytes , Chimiokine CCL5 , , Obésité , Animaux , Souris , Chimiokine CCL5/génétique , Chimiokine CCL5/pharmacologie , Ligands , Souris obèse , Muscles squelettiques/métabolisme , Récepteurs CCR/métabolisme , Adipocytes/métabolisme , Obésité/génétique , Obésité/métabolisme , Obésité/anatomopathologie
12.
Medicine (Baltimore) ; 103(8): e36897, 2024 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-38394497

RÉSUMÉ

BACKGROUND: Atopic dermatitis (AD) is a common and recurrent inflammatory disease with strong genetic susceptibility. The abnormal production of chemokines plays an important role in the occurrence and development of AD. METHODS: A comprehensive online literature search was performed in databases of China National Knowledge Infrastructure, Wanfang, VIP China Science and Technology Journal Database, China Biomedical Literature Database, PubMed, Embase and Cochrane Library to retrieve relevant articles published from January 2000 to October 2022. The odds ratio (OR) with its 95% confidence interval (CI) was employed to calculate this relationship. RESULTS: A total of 7 studies were finally screened out, including 1316 AD patients and 1099 controls. There were 3 studies for CC chemokine ligand 5 (CCL5) polymorphisms, 2 for CCL11 polymorphisms, and 2 for CCL17 polymorphisms, respectively. The meta-analysis revealed a significant association between the CCL5 - 403G/A polymorphism and AD under the allelic model (A vs G: OR = 1.25, 95% CI = 1.02-1.52, P = .03), heterozygous model (AG vs GG: OR = 1.40, 95% CI = 1.08-1.80, P = .01) and dominant model (AA + AG vs GG: OR = 1.38, 95% CI = 1.08-1.76, P = .01) in a fixed-effect model. The allelic model (G vs C: OR = 1.46, 95% CI = 1.07-1.98, P < .01) and dominant model (GG + GC vs CC: OR = 1.74, 95% CI = 1.23-2.47, P < .001) of the CCL5 - 28C/G polymorphism were also associated with an increased risk of AD. However, this significant association was not found in other alleles and genotypes (P > .05). CONCLUSION: Our results show that the A allele, AG and AA + AG genotypes of the CCL5 - 403G/A polymorphism, the G allele and GG + GC genotype of the CCL5 - 28C/G polymorphism are risk factors for AD. Future studies with large population are still needed to further explore those correlations.


Sujet(s)
Chimiokine CCL11 , Chimiokine CCL17 , Chimiokine CCL5 , Eczéma atopique , Humains , Chimiokine CCL11/génétique , Chimiokine CCL17/génétique , Chimiokine CCL5/génétique , Eczéma atopique/génétique , Prédisposition génétique à une maladie , Génotype , Ligands , Polymorphisme de nucléotide simple , Facteurs de risque
13.
Clin Cancer Res ; 30(9): 1934-1944, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38372707

RÉSUMÉ

PURPOSE: Neoadjuvant anti-PD1 (aPD1) therapies are being explored in surgically resectable head and neck squamous cell carcinoma (HNSCC). Encouraging responses have been observed, but further insights into the mechanisms underlying resistance and approaches to improve responses are needed. EXPERIMENTAL DESIGN: We integrated data from syngeneic mouse oral carcinoma (MOC) models and neoadjuvant pembrolizumab HNSCC patient tumor RNA-sequencing data to explore the mechanism of aPD1 resistance. Tumors and tumor-draining lymph nodes (DLN) from MOC models were analyzed for antigen-specific priming. CCL5 expression was enforced in an aPD1-resistant model. RESULTS: An aPD1-resistant mouse model showed poor priming in the tumor DLN due to type 1 conventional dendritic cell (cDC1) dysfunction, which correlated with exhausted and poorly responsive antigen-specific T cells. Tumor microenvironment analysis also showed decreased cDC1 in aPD1-resistant tumors compared with sensitive tumors. Following neoadjuvant aPD1 therapy, pathologic responses in patients also positively correlated with baseline transcriptomic cDC1 signatures. In an aPD1-resistant model, intratumoral cDC1 vaccine was sufficient to restore aPD1 response by enhancing T-cell infiltration and increasing antigen-specific responses with improved tumor control. Mechanistically, CCL5 expression significantly correlated with neoadjuvant aPD1 response and enforced expression of CCL5 in an aPD1-resistant model, enhanced cDC1 tumor infiltration, restored antigen-specific responses, and recovered sensitivity to aPD1 treatment. CONCLUSIONS: These data highlight the contribution of tumor-infiltrating cDC1 in HNSCC aPD1 response and approaches to enhance cDC1 infiltration and function that may circumvent aPD1 resistance in patients with HNSCC.


Sujet(s)
Cellules dendritiques , Résistance aux médicaments antinéoplasiques , Tumeurs de la tête et du cou , Carcinome épidermoïde de la tête et du cou , Microenvironnement tumoral , Animaux , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris , Humains , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/génétique , Tumeurs de la tête et du cou/anatomopathologie , Tumeurs de la tête et du cou/traitement médicamenteux , Tumeurs de la tête et du cou/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/génétique , Carcinome épidermoïde de la tête et du cou/anatomopathologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Carcinome épidermoïde de la tête et du cou/métabolisme , Microenvironnement tumoral/immunologie , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Modèles animaux de maladie humaine , Traitement néoadjuvant/méthodes , Femelle , Lignée cellulaire tumorale
14.
Sci China Life Sci ; 67(6): 1226-1241, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38300441

RÉSUMÉ

Ovarian cancer is the most lethal and aggressive gynecological cancer with a high recurrence rate and is often diagnosed late. In ovarian cancer, multiple metabolic enzymes of lipid metabolism are abnormally expressed, resulting in metabolism disorder. As a characteristic pathway in polyunsaturated fatty acid (PUFA) metabolism, arachidonic acid (AA) metabolism is disturbed in ovarian cancer. Therefore, we established a 10-gene signature model to evaluate the prognostic risk of PUFA-related genes. This 10-gene signature has strong robustness and can play a stable predictive role in datasets of various platforms (TCGA, ICGC, and GSE17260). The high association between the risk subgroups and clinical characteristics indicated a good performance of the model. Our data further indicated that the high expression of LTA4H was positively correlated with poor prognosis in ovarian cancer. Deficiency of LTA4H enhanced sensitivity to Cisplatin and modified the characteristics of immune cell infiltration in ovarian cancer. Additionally, our results indicate that CCL5 was involved in the aberrant metabolism of the AA/LTA4H axis, which contributes to the reduction of tumor-infiltrating CD8+ T cells and immune escape in ovarian cancer. These findings provide new insights into the prognosis and potential target of LTA4H/CCL5 in treating ovarian cancer.


Sujet(s)
Chimiokine CCL5 , Cisplatine , Epoxide hydrolase , Tumeurs de l'ovaire , Microenvironnement tumoral , Femelle , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Humains , Chimiokine CCL5/métabolisme , Chimiokine CCL5/génétique , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Cisplatine/usage thérapeutique , Cisplatine/pharmacologie , Epoxide hydrolase/métabolisme , Epoxide hydrolase/génétique , Lignée cellulaire tumorale , Pronostic , Régulation de l'expression des gènes tumoraux , Acide arachidonique/métabolisme , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Animaux , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Lymphocytes TIL/effets des médicaments et des substances chimiques , Souris
15.
J Immunother Cancer ; 12(1)2024 01 11.
Article de Anglais | MEDLINE | ID: mdl-38212126

RÉSUMÉ

BACKGROUND: The C-C motif chemokine receptor 5 (CCR5)/C-C motif chemokine ligand 5 (CCL5) axis plays a major role in colorectal cancer (CRC). We aimed to characterize the molecular features associated with CCR5/CCL5 expression in CRC and to determine whether CCR5/CCL5 levels could impact treatment outcomes. METHODS: 7604 CRCs tested with NextGen Sequencing on DNA and RNA were analyzed. Molecular features were evaluated according to CCR5 and CCL5 tumor gene expression quartiles. The impact on treatment outcomes was assessed in two cohorts, including 6341 real-world patients and 429 patients from the Cancer and Leukemia Group B (CALGB)/SWOG 80405 trial. RESULTS: CCR5/CCL5 expression was higher in right-sided versus left-sided tumors, and positively associated with consensus molecular subtypes 1 and 4. Higher CCR5/CCL5 expression was associated with higher tumor mutational burden, deficiency in mismatch repair and programmed cell death ligand 1 (PD-L1) levels. Additionally, high CCR5/CCL5 were associated with higher immune cell infiltration in the tumor microenvironment (TME) of MMR proficient tumors. Ingenuity pathway analysis revealed upregulation of the programmed cell death protein 1 (PD-1)/PD-L1 cancer immunotherapy pathway, phosphatase and tensin homolog (PTEN) and peroxisome proliferator-activated receptors (PPAR) signaling, and cytotoxic T-lymphocyte antigen 4 (CTLA-4) signaling in cytotoxic T lymphocytes, whereas several inflammation-related pathways were downregulated. Low CCR5/CCL5 expression was associated with increased benefit from cetuximab-FOLFOX treatment in the CALGB/SWOG 80405 trial, where significant treatment interaction was observed with biologic agents and chemotherapy backbone. CONCLUSIONS: Our data show a strong association between CCR5/CCL5 gene expression and distinct molecular features, gene expression profiles, TME cell infiltration, and treatment benefit in CRC. Targeting the CCR5/CCL5 axis may have clinical applications in selected CRC subgroups and may play a key role in developing and deploying strategies to modulate the immune TME for CRC treatment.


Sujet(s)
Tumeurs colorectales , Récepteurs aux chimiokines , Humains , Antigène CD274/génétique , Ligands , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme , Chimiokines/génétique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Tumeurs colorectales/anatomopathologie , Expression des gènes , Microenvironnement tumoral , Récepteurs CCR5/génétique , Récepteurs CCR5/métabolisme
16.
Hypertension ; 81(4): 776-786, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38240165

RÉSUMÉ

BACKGROUND: Aldosterone has been described to initiate cardiovascular diseases by triggering exacerbated sterile vascular inflammation. The functions of CCL5 (C-C motif chemokine ligand 5) and its receptor CCR5 (C-C motif chemokine receptor 5) are well known in infectious diseases, their contributions to aldosterone-induced vascular injury and hypertension remain unknown. METHODS: We analyzed the vascular profile, blood pressure, and renal damage in wild-type (CCR5+/+) and CCR5 knockout (CCR5-/-) mice treated with aldosterone (600 µg/kg per day for 14 days) while receiving 1% saline to drink. Vascular function was analyzed in aorta and mesenteric arteries, blood pressure was measured by telemetry and renal injury and inflammation were analyzed via histology and flow cytometry. Endothelial cells were used to study the molecular signaling whereby CCL5 induces endothelial dysfunction. RESULTS: Aldosterone treatment resulted in exaggerated CCL5 circulating levels and vascular CCR5 expression in CCR5+/+ mice accompanied by endothelial dysfunction, hypertension, and renal inflammation and damage. CCR5-/- mice were protected from these aldosterone-induced effects. Mechanistically, we demonstrated that CCL5 increased NOX1 (NADPH oxidase 1) expression, reactive oxygen species formation, NFκB (nuclear factor kappa B) activation, and inflammation and reduced NO production in isolated endothelial cells. These effects were abolished by antagonizing CCR5 with Maraviroc. Finally, aorta incubated with CCL5 displayed severe endothelial dysfunction, which is prevented by blocking NOX1, NFκB, or CCR5. CONCLUSIONS: Our data demonstrate that CCL5/CCR5, through activation of NFκB and NOX1, is critically involved in aldosterone-induced vascular and renal damage and hypertension placing CCL5 and CCR5 as potential therapeutic targets for conditions characterized by aldosterone excess.


Sujet(s)
Aldostérone , Chimiokine CCL5 , Hypertension artérielle , Récepteurs CCR5 , Animaux , Souris , Aldostérone/pharmacologie , Cellules endothéliales/métabolisme , Hypertension artérielle/induit chimiquement , Hypertension artérielle/métabolisme , Inflammation , Récepteurs CCR5/génétique , Récepteurs CCR5/métabolisme , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme
17.
Cancer Res ; 84(2): 276-290, 2024 01 16.
Article de Anglais | MEDLINE | ID: mdl-37890164

RÉSUMÉ

Heat shock factor 1 (HSF1) is a stress-responsive transcription factor that promotes cancer cell malignancy. To provide a better understanding of the biological processes regulated by HSF1, here we developed an HSF1 activity signature (HAS) and found that it was negatively associated with antitumor immune cells in breast tumors. Knockdown of HSF1 decreased breast tumor size and caused an influx of several antitumor immune cells, most notably CD8+ T cells. Depletion of CD8+ T cells rescued the reduction in growth of HSF1-deficient tumors, suggesting HSF1 prevents CD8+ T-cell influx to avoid immune-mediated tumor killing. HSF1 suppressed expression of CCL5, a chemokine for CD8+ T cells, and upregulation of CCL5 upon HSF1 loss significantly contributed to the recruitment of CD8+ T cells. These findings indicate that HSF1 suppresses antitumor immune activity by reducing CCL5 to limit CD8+ T-cell homing to breast tumors and prevent immune-mediated destruction, which has implications for the lack of success of immune modulatory therapies in breast cancer. SIGNIFICANCE: The stress-responsive transcription factor HSF1 reduces CD8+ T-cell infiltration in breast tumors to prevent immune-mediated killing, indicating that cellular stress responses affect tumor-immune interactions and that targeting HSF1 could improve immunotherapies.


Sujet(s)
Tumeurs du sein , Protéines de liaison à l'ADN , Humains , Femelle , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Tumeurs du sein/anatomopathologie , Facteurs de transcription de choc thermique/génétique , Lignée cellulaire tumorale , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Lymphocytes T CD8+/métabolisme , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme
18.
Curr Cancer Drug Targets ; 24(3): 308-318, 2024.
Article de Anglais | MEDLINE | ID: mdl-37581517

RÉSUMÉ

BACKGROUND: Osteosarcoma (OS) is the most common primary malignant tumor of bone tissue, which has an insidious onset and is difficult to detect early, and few early diagnostic markers with high specificity and sensitivity. Therefore, this study aims to identify potential biomarkers that can help diagnose OS in its early stages and improve the prognosis of patients. METHODS: The data sets of GSE12789, GSE28424, GSE33382 and GSE36001 were combined and normalized to identify Differentially Expressed Genes (DEGs). The data were analyzed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG) and Disease Ontology (DO). The hub gene was selected based on the common DEG that was obtained by applying two regression methods: the Least Absolute Shrinkage and Selection Operator (LASSO) and Support vVector Machine (SVM). Then the diagnostic value of the hub gene was evaluated in the GSE42572 data set. Finally, the correlation between immunocyte infiltration and key genes was analyzed by CIBERSORT. RESULTS: The regression analysis results of LASSO and SVM are the following three DEGs: FK501 binding protein 51 (FKBP5), C-C motif chemokine ligand 5 (CCL5), complement component 1 Q subcomponent B chain (C1QB). We evaluated the diagnostic performance of three biomarkers (FKBP5, CCL5 and C1QB) for osteosarcoma using receiver operating characteristic (ROC) analysis. In the training group, the area under the curve (AUC) of FKBP5, CCL5 and C1QB was 0.907, 0.874 and 0.676, respectively. In the validation group, the AUC of FKBP5, CCL5 and C1QB was 0.618, 0.932 and 0.895, respectively. It is noteworthy that these genes were more expressed in tumor tissues than in normal tissues by various immune cell types, such as plasma cells, CD8+ T cells, T regulatory cells (Tregs), activated NK cells, activated dendritic cells and activated mast cells. These immune cell types are also associated with the expression levels of the three diagnostic genes that we identified. CONCLUSION: We found that CCL5 can be considered an early diagnostic gene of osteosarcoma, and CCL5 interacts with immune cells to influence tumor occurrence and development. These findings have important implications for the early detection of osteosarcoma and the identification of novel therapeutic targets.


Sujet(s)
Tumeurs osseuses , Ostéosarcome , Humains , Ligands , Ostéosarcome/diagnostic , Ostéosarcome/génétique , Ostéosarcome/thérapie , Marqueurs biologiques , Immunothérapie , Tumeurs osseuses/diagnostic , Tumeurs osseuses/génétique , Tumeurs osseuses/thérapie , Chimiokine CCL5/génétique
19.
J Gene Med ; 26(1): e3630, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37985959

RÉSUMÉ

BACKGROUND: Cholangiocarcinoma (CCA) stands as an aggressive malignancy of the biliary tract. The interplay between the tumor and immune system plays a pivotal role in disease progression and treatment outcomes. Hence, the present study aimed to extensively explore the immunogenomic landscape of CCA, with the objective of unveiling unique molecular and immunological signatures that could guide personalized therapeutic approaches. METHODS: The study collected data from The Cancer Genome Atlas databases, performed gene set variation analysis for the chemokine ligand 5 (CCL5) high/low expression group, conducted principal component analysis, gene set enrichment analysis enrichment and mutation pattern analysis, generated a heatmap, and performed cox regression analysis. RESULTS: The two discrete subpopulations were found to exhibit contrasting mutational and immunogenomic characteristics, emphasizing the heterogeneity of CCA. These subsets also showed pronounced discrepancies in the infiltration of immune cells, indicating diverse interactions with the tumor immune microenvironment. Furthermore, the dissimilarities in mutational patterns were observed within the two CCA subgroups, with PBRM1 and BAP1 emerging as the most frequently mutated genes. In addition, a prognostic framework was formulated and validated utilizing the expression profiles of COX16 and RSAD2 genes, effectively segregating patients into high-risk and low-risk cohorts. Furthermore, the connections between immune-related parameters and these risk groups were identified, underscoring the potential significance of the immune microenvironment in patient prognosis. In vitro experiments have shown that COX16 promotes the proliferation and metastasis of CCA cells, whereas RSAD2 inhibits it. CONCLUSIONS: The present study provides an intricate depiction of the immunogenomic landscape of CCA based on CCL5 expression, thereby paving the way for novel immunotherapy strategies and prognostic assessment.


Sujet(s)
Tumeurs des canaux biliaires , Cholangiocarcinome , Humains , Pronostic , Ligands , Tumeurs des canaux biliaires/génétique , Tumeurs des canaux biliaires/thérapie , Cholangiocarcinome/génétique , Cholangiocarcinome/thérapie , Cholangiocarcinome/anatomopathologie , Conduits biliaires intrahépatiques/anatomopathologie , Microenvironnement tumoral/génétique , Chimiokine CCL5/génétique
20.
Int J Mol Sci ; 24(22)2023 Nov 10.
Article de Anglais | MEDLINE | ID: mdl-38003358

RÉSUMÉ

The imbalance that occurs in bone remodeling induced by irradiation (IR) is the disruption of the balance between bone formation and bone resorption. In this study, primary osteocytes (OCYs) of femoral and tibial origin were cultured and irradiated. It was observed that irradiated OCY showed extensive DNA damage, which led to the initiation of a typical phenotype of cellular senescence, including the secretion of senescence-associated secretory phenotype (SASP), especially the C-C motif chemokine ligand 5 (CCL5). In order to explore the regulation of osteoclastogenic potential by IR-induced senescent OCYs exocytosis factor CCL5, the conditioned medium (CM) of OCYs was co-cultured with RAW264.7 precursor cells. It was observed that in the irradiated OCY co-cultured group, the migration potential increased compared with the vehicle culture group, accompanied by an enhancement of typical mature OCs; the expression of the specific function of enzyme tartrate-resistant acid phosphatase (TRAP) increased; and the bone-destructive function was enhanced. However, a neutralizing antibody to CCL5 could reverse the extra-activation of osteoclastogenesis. Accordingly, the overexpression of p-STAT3 in irradiated OCY was accompanied by CCL5. It was concluded that CCL5 is a potential key molecule and the interventions targeting CCL5 could be a potential strategy for inhibiting osteoclastogenesis and restoring bone remodeling.


Sujet(s)
Résorption osseuse , Ostéogenèse , Humains , Remodelage osseux , Résorption osseuse/métabolisme , Vieillissement de la cellule/génétique , Chimiokine CCL5/génétique , Chimiokine CCL5/métabolisme , Ligands , Ostéoclastes/métabolisme , Ostéogenèse/génétique , Ligand de RANK/métabolisme , Animaux , Souris
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE