Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.366
Filter
1.
J Transl Med ; 22(1): 502, 2024 May 26.
Article in English | MEDLINE | ID: mdl-38797830

ABSTRACT

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.


Subject(s)
Chemokine CCL5 , Familial Primary Pulmonary Hypertension , Killer Cells, Natural , RNA-Seq , Single-Cell Analysis , T-Lymphocytes , Animals , Humans , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/pathology , Familial Primary Pulmonary Hypertension/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Male , Cell Communication/genetics , Rats, Sprague-Dawley , Lung/pathology , Rats , Gene Regulatory Networks , Monocrotaline , Protein Interaction Maps/genetics , Computational Biology
2.
J Immunother Cancer ; 12(5)2024 May 07.
Article in English | MEDLINE | ID: mdl-38719543

ABSTRACT

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.


Subject(s)
Chemokine CCL5 , Colorectal Neoplasms , Receptors, CCR5 , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Neoplasm Metastasis
3.
J Immunother ; 47(6): 195-204, 2024.
Article in English | MEDLINE | ID: mdl-38654631

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes , Chemokine CCL5 , Colorectal Neoplasms , Interferon-alpha , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2 , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Animals , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Humans , Cell Line, Tumor , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Tumor Microenvironment/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Gene Expression Regulation, Neoplastic/drug effects , Female
4.
Am J Physiol Cell Physiol ; 326(5): C1320-C1333, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38497114

ABSTRACT

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.


Subject(s)
Adipocytes , Chemokine CCL5 , Myokines , Obesity , Animals , Mice , Chemokine CCL5/genetics , Chemokine CCL5/pharmacology , Ligands , Mice, Obese , Muscle, Skeletal/metabolism , Receptors, CCR/metabolism , Adipocytes/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/pathology
5.
Clin Cancer Res ; 30(9): 1934-1944, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38372707

ABSTRACT

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.


Subject(s)
Dendritic Cells , Drug Resistance, Neoplasm , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , Humans , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Microenvironment/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Disease Models, Animal , Neoadjuvant Therapy/methods , Female , Cell Line, Tumor
6.
Sci China Life Sci ; 67(6): 1226-1241, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38300441

ABSTRACT

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.


Subject(s)
Chemokine CCL5 , Cisplatin , Epoxide Hydrolases , Ovarian Neoplasms , Tumor Microenvironment , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Humans , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects , Cisplatin/therapeutic use , Cisplatin/pharmacology , Epoxide Hydrolases/metabolism , Epoxide Hydrolases/genetics , Cell Line, Tumor , Prognosis , Gene Expression Regulation, Neoplastic , Arachidonic Acid/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Mice
7.
Medicine (Baltimore) ; 103(8): e36897, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38394497

ABSTRACT

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.


Subject(s)
Chemokine CCL11 , Chemokine CCL17 , Chemokine CCL5 , Dermatitis, Atopic , Humans , Chemokine CCL11/genetics , Chemokine CCL17/genetics , Chemokine CCL5/genetics , Dermatitis, Atopic/genetics , Genetic Predisposition to Disease , Genotype , Ligands , Polymorphism, Single Nucleotide , Risk Factors
8.
Hypertension ; 81(4): 776-786, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38240165

ABSTRACT

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.


Subject(s)
Aldosterone , Chemokine CCL5 , Hypertension , Receptors, CCR5 , Animals , Mice , Aldosterone/pharmacology , Endothelial Cells/metabolism , Hypertension/chemically induced , Hypertension/metabolism , Inflammation , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism
9.
J Immunother Cancer ; 12(1)2024 01 11.
Article in English | MEDLINE | ID: mdl-38212126

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Receptors, Chemokine , Humans , B7-H1 Antigen/genetics , Ligands , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokines/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression , Tumor Microenvironment , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
10.
Curr Cancer Drug Targets ; 24(3): 308-318, 2024.
Article in English | MEDLINE | ID: mdl-37581517

ABSTRACT

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.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Ligands , Osteosarcoma/diagnosis , Osteosarcoma/genetics , Osteosarcoma/therapy , Biomarkers , Immunotherapy , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Chemokine CCL5/genetics
11.
J Gene Med ; 26(1): e3630, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37985959

ABSTRACT

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.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Prognosis , Ligands , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/therapy , Cholangiocarcinoma/pathology , Bile Ducts, Intrahepatic/pathology , Tumor Microenvironment/genetics , Chemokine CCL5/genetics
12.
Cancer Res ; 84(2): 276-290, 2024 01 16.
Article in English | MEDLINE | ID: mdl-37890164

ABSTRACT

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.


Subject(s)
Breast Neoplasms , DNA-Binding Proteins , Humans , Female , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Breast Neoplasms/pathology , Heat Shock Transcription Factors/genetics , Cell Line, Tumor , Transcription Factors/genetics , Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism
13.
Int J Mol Sci ; 24(22)2023 Nov 10.
Article in English | MEDLINE | ID: mdl-38003358

ABSTRACT

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.


Subject(s)
Bone Resorption , Osteogenesis , Humans , Bone Remodeling , Bone Resorption/metabolism , Cellular Senescence/genetics , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Ligands , Osteoclasts/metabolism , Osteogenesis/genetics , RANK Ligand/metabolism , Animals , Mice
14.
J Cancer Res Clin Oncol ; 149(19): 17335-17346, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37831273

ABSTRACT

PURPOSE: The CCR5/CCL5 axis is essential for interactions between malignant cells and microenvironment components, promoting tumor progression in oral squamous cell carcinoma (OSCC). This study aims to evaluate the association of CCL5 and CCR5 with the behavior of oral cancer and assess the therapeutic potential of a CCR5 antagonist. METHODS: A retrospective study to analyze CCR5 and CCL5 expression on paraffin-embedded tissues was performed. In cell lines, rhCCL5 was added to induce CCR5-related pathways, and Maraviroc and shRNA against CCR5 were used to neutralize the receptor. Finally, an in vivo murine orthotopic xenograft model of tongue cancer was used to evaluate Maraviroc as an oncologic therapy. After 15 days, the mice were killed, and the primary tumors and cervical lymph nodes were analyzed. RESULTS: The expression of CCR5 was associated with clinical stage and metastasis, and CCL5 was related to overall survival. Adding rhCCL5 induced cell proliferation, while shRNA and Maraviroc reduced it in a dose-dependent manner. Maraviroc treatment also increased apoptosis and modified cytoskeletal organization. In vivo, Maraviroc reduced neck metastasis. CONCLUSIONS: The effects of CCR5 antagonists in OSCC have been poorly studied, and this study reports in vitro and in vivo evidence for the effects of Maraviroc in OSCC. Our results suggest that the CCR5/CCL5 axis plays a role in oral cancer behavior, and that its inhibition is a promising new therapy alternative.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Animals , Mice , Maraviroc/pharmacology , Carcinoma, Squamous Cell/drug therapy , Squamous Cell Carcinoma of Head and Neck , Retrospective Studies , Cell Line, Tumor , Mouth Neoplasms/drug therapy , RNA, Small Interfering/metabolism , Tumor Microenvironment , Chemokine CCL5/genetics , Chemokine CCL5/metabolism
15.
Viruses ; 15(9)2023 09 20.
Article in English | MEDLINE | ID: mdl-37766364

ABSTRACT

Genetic polymorphisms in genes that encode natural ligands of CCR5 (the main human HIV coreceptor), such as CCL5/RANTES, can alter the levels of secretion of these peptides. This article sought to review the relationship between single nucleotide polymorphisms (SNPs) of CCL5/RANTES and HIV-1 disease susceptibility. A meta-analysis was conducted through 17 articles found from January 1999 to December 2022 in the PUBMED, Science Direct, Medline, and SciELO databases. A total of three SNPs were identified and investigated under their dominant genotypic model and through a fixed-effects model. In terms of the SNP rs2107538 (G > A), in Africa and Asia, it has a protective role (OR = 0.56; 95% CI = 0.41-0.76; p = 0.0002, and OR = 0.88; 95% CI = 0.76-1.02; p = 0.08, respectively). In terms of the SNP rs2280788 (C > G), in Europe and America, it shows a higher risk role (OR = 1.92; 95% CI = 1.06-3.47; p = 0.03, and OR = 0.94; 95% CI = 0.94-1.11; p = 0.04, respectively), but in the population of Asia, with its mutant allele, it has a protective role (OR = 0.76; 95% CI = 0.63-0.93; p = 0.007). In terms of the SNP rs2280789 (T > C), no significant associations were found. Both SNPs rs2107538 and rs2280788 have a positive transcriptional effect on the RANTES/CCL5 gene, while SNP rs2280789 causes a decrease in gene expression levels. This study suggests that there is an association between the increased expression of CCL5/RANTES and a lower risk of AIDS. Therefore, further studies are needed to arrive at a definitive conclusion, and these results may help establish scientific bases for effective HIV/AIDS control strategies.


Subject(s)
Acquired Immunodeficiency Syndrome , HIV Infections , HIV Seropositivity , HIV-1 , Humans , Polymorphism, Single Nucleotide , HIV-1/genetics , HIV Infections/genetics , Chemokine CCL5/genetics
16.
Am J Rhinol Allergy ; 37(6): 730-738, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37525517

ABSTRACT

BACKGROUND: Neurokinin-1 receptor (NK-1R) and normal T cell expressed and secreted (RANTES) have been shown to play important roles in allergic rhinitis (AR). However, whether the regulating effect of NK-1R in AR is achieved via RANTES remains unknown. METHODS: In the present study, Sprague-Dawley rats were sensitized and challenged with ovalbumin to make AR models. During the challenge period, the rats were treated intranasally with NK-1R-specific small interfering RNA (siRNA) for NKR group, negative siRNA for NCS group, rats in NSAR group and NS group were given saline. The amount of nasal secretion and the numbers of nose rubs and sneezes were measured in each rat. The levels of NK-1R and RANTES in the nasal mucosal tissues were determined through real-time fluorescence quantitative RT-PCR and immunohistochemical staining. The numbers of eosinophils in the collected nasal lavage fluid (NLF) were counted, and the concentration of RANTES in NLF was determined by enzyme-linked immunosorbent assay. RESULTS: Compared with that in the NS group, the expression of NK-1R and RANTES was significantly higher in the nasal mucosa of NSAR and NCS group rats. The sneezing and nose rubbing counts and the amount of nasal secretions were increased significantly in the NSAR and NCS groups. Rats in the NKR group experienced greater relief from AR symptoms than rats in the NSAR and NCS groups. Furthermore, knockdown of NK-1R expression also significantly eliminated RANTES expression and eosinophil infiltration in the nasal mucosa of NKR group rats. CONCULSION: For the first time, we show that intranasal treatment with NK-1R-specific siRNA can significantly decrease RANTES expression, AR-related symptoms, and eosinophil inflammation, suggesting that the regulating effect of NK-1R in the development of AR occurs via alteration of RANTES expression.


Subject(s)
Chemokine CCL5 , RNA, Small Interfering , Receptors, Neurokinin-1 , Rhinitis, Allergic , Animals , Rats , Disease Models, Animal , Nasal Mucosa/metabolism , Ovalbumin , Rats, Sprague-Dawley , Receptors, Neurokinin-1/genetics , Rhinitis, Allergic/genetics , RNA, Small Interfering/genetics , Sneezing , Gene Knockdown Techniques , Chemokine CCL5/genetics
17.
Cell Rep ; 42(6): 112654, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37342906

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive subtype with limited effective therapies because of the absence of definitive targets. Here, we demonstrate that the expression of ZNF451, a poorly characterized vertebrate zinc-finger protein, is upregulated in TNBC and associated with a poor prognosis. Elevated ZNF451 expression facilitates TNBC progression by interacting with and enhancing the activity of the transcriptional activator snail family transcriptional repressor 2 (SLUG). Mechanistically, the ZNF451-SLUG complex preferentially recruits the acetyltransferase p300/CBP-associated factor (PCAF) to the CCL5 promoter, selectively facilitating CCL5 transcription by enhancing the acetylation of SLUG and local chromatin, leading to recruitment and activation of tumor-associated macrophages (TAMs). Disturbing the ZNF451-SLUG interaction using a peptide suppresses TNBC progression by reducing CCL5 expression and counteracting the migration and activation of TAMs. Collectively, our work provides mechanistic insights into the oncogene-like functions of ZNF451 and suggests that ZNF451 is a potential target for development of effective therapies against TNBC.


Subject(s)
Chemokine CCL5 , Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Chemokine CCL5/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Zinc Fingers
18.
Oncol Rep ; 50(1)2023 07.
Article in English | MEDLINE | ID: mdl-37264954

ABSTRACT

Ovarian serous carcinoma (OC) is a common cause of mortality among gynecological malignancies. Although tumor­infiltrating CD8 T cells are associated with a favorable prognosis of OC, the underlying mechanisms are not clearly understood. The present study identified the key genes and potential molecular mechanisms associated with CD8 T­cell infiltration in OC. The score of CD8 T cells in The Cancer Genome Atlas dataset (376 samples from patients with OC) was estimated using the quanTIseq and MCP­counter algorithms. Thereafter, a protein­protein interaction network of differentially expressed genes was constructed and the hub genes were identified using cytoHubba in Cytoscape. The results revealed that signal transducer and activator of transcription 4 (STAT4) was strongly correlated with CD8 T­cell infiltration in OC. Furthermore, the prognostic value of STAT4 in OC was verified by Kaplan­Meier curve, and univariate and multivariate analyses. The biological functions of STAT4 were determined by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses, which revealed that STAT4 is closely related to cytokines in OC. Moreover, Spearman correlation analysis suggested that STAT4 was most positively correlated with CC chemokine ligand 5 (CCL5). CCL5 was revealed to be critical for orchestrating T­cell infiltration in tumors. Moreover, immunohistochemistry and reverse transcription­quantitative PCR showed that STAT4, CCL5 and CD8A (a marker for CD8 T cells) were closely related in OC. Moreover, in vitro analysis revealed that STAT4 knockdown led to a decrease in CCL5 expression and CD8 T­cell migration. Taken together, the present study suggested that STAT4 may regulate CD8 T­cell infiltration in OC tissues by inducing CCL5 secretion. Furthermore, STAT4 may be considered a promising prognostic biomarker for OC.


Subject(s)
Carcinoma , Ovarian Neoplasms , Humans , Female , Chemokines, CC/metabolism , Ligands , Prognosis , CD8-Positive T-Lymphocytes/metabolism , Carcinoma/pathology , Ovarian Neoplasms/pathology , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/metabolism
19.
Mol Cancer Res ; 21(7): 741-752, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37027010

ABSTRACT

Cancer-associated fibroblasts (CAF) can promote tumor growth, metastasis, and therapeutic resistance in esophageal squamous cell carcinoma (ESCC), but the mechanisms of action remain elusive. Our objective was to identify secreted factor(s) that mediate the communication between CAFs and ESCC tumor cells with the aim of identifying potential druggable targets. Through unbiased cytokine arrays, we have identified CC motif chemokine ligand 5 (CCL5) as a secreted factor that is increased upon co-culture of ESCC cells and CAFs, which we replicated in esophageal adenocarcinoma (EAC) with CAFs. Loss of tumor-cell-derived CCL5 reduces ESCC cell proliferation in vitro and in vivo and we propose this is mediated, in part, by a reduction in ERK1/2 signaling. Loss of tumor-derived CCL5 reduces the percentage of CAFs recruited to xenograft tumors in vivo. CCL5 is a ligand for the CC motif receptor 5 (CCR5), for which a clinically approved inhibitor exists, namely Maraviroc. Maraviroc treatment reduced tumor volume, CAF recruitment, and ERK1/2 signaling in vivo, thus, mimicking the effects observed with genetic loss of CCL5. High CCL5 or CCR5 expression is associated with worse prognosis in low-grade esophageal carcinomas. IMPLICATIONS: These data highlight the role of CCL5 in tumorigenesis and the therapeutic potential of targeting the CCL5-CCR5 axis in ESCC.


Subject(s)
Cancer-Associated Fibroblasts , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Proliferation , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CCL5/pharmacology , Chemokines/metabolism , Chemokines/pharmacology , Chemokines/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Fibroblasts/metabolism , Ligands , Maraviroc/metabolism , Maraviroc/pharmacology , Maraviroc/therapeutic use , Animals
20.
Mol Carcinog ; 62(8): 1091-1106, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37067401

ABSTRACT

Aberrant N7 -methylguanosine (m7G) levels closely correlate with tumor genesis and progression. NCBP2 and EIF4E3 are two important m7G-related cap-binding genes. This study aimed to identify the relationship between the EIF4E3/NCBP2 function and immunological characteristics of head and neck squamous cell carcinoma (HNSCC). Hierarchical clustering was employed in classifying HNSCC patients into two groups based on the expressions of NCBP2 and EIF4E3. The differentially expressed genes were identified between the two groups, and GO functional enrichment was subsequently performed. Weighted gene co-expression network analysis was conducted to identify the hub genes related to EIF4E3/NCBP2 expression and immunity. The differential infiltration of immune cells and the response to immunotherapy were compared between the two groups. Single-cell sequence and trajectory analyses were performed to predict cell differentiation and display the expression of EIF4E3/NCBP2 in each state. In addition, quantitative real-time PCR, spatial transcriptome analysis, transwell assay, and western blotting were conducted to verify the biological function of EIF4E3/NCBP2. Here, group A showed a higher EIF4E3 expression and a lower NCBP2 expression, which had higher immune scores, proportion of most immune cells, immune activities, expression of immunomodulatory targets, and a better response to cancer immunotherapy. Besides, 56 hub molecules with notable immune regulation significance were identified. A risk model containing 17 hub genes and a prognostic nomogram was successfully established. Moreover, HNSCC tissues had a lower EIF4E3 expression and a higher NCBP2 expression than normal tissues. NCBP2 and EIF4E3 played a vital role in the differentiation of monocytes. Furthermore, the expression of CCL4/CCL5 can be regulated via EIF4E3 overexpression and NCBP2 knockdown. Collectively, NCBP2 and EIF4E3 can affect downstream gene expression, as well as immune contexture and response to immunotherapy, which could induce "cold-to-hot" tumor transformation in HNSCC patients.


Subject(s)
Chemokine CCL4 , Chemokine CCL5 , Eukaryotic Initiation Factor-4E , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms , Squamous Cell Carcinoma of Head and Neck , Humans , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/physiopathology , Head and Neck Neoplasms/therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/physiopathology , Squamous Cell Carcinoma of Head and Neck/therapy , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Nuclear Cap-Binding Protein Complex/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic/genetics , Cell Line, Tumor , Male , Female , Middle Aged , Aged , Immunotherapy , Models, Statistical , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...