Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 341
Filter
1.
J Transl Med ; 22(1): 833, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256832

ABSTRACT

BACKGROUND: Family with sequence similarity 109, member B (FAM109B) is involved in endocytic transport and affects genetic variation in brain methylation. It is one of the important genes related to immune cell-associated diseases. In the tumor immune system, methylation can regulate tumor immunity and influence the maturation and functional response of immune cells. Whether FAM109B is involved in tumor progression and its correlation with the tumor immune microenvironment has not yet been disclosed. METHODS: A comprehensive pan-cancer analysis of FAM109B expression, prognosis, immunity, and TMB was conducted. The expression, clinical features, and prognostic value of FAM109B in low-grade gliomas (LGG) were evaluated using TCGA, CGGA, and Gravendeel databases. The expression of FAM109B was validated by qRT-PCR, immunohistochemistry (IHC), and Western blotting (WB). The relationship between FAM109B and methylation, Copy Number Variation (CNV), prognosis, immune checkpoints (ICs), and common chemotherapy drug sensitivity in LGG was explored through Cox regression, Kaplan-Meier curves, and Spearman correlation analysis. FAM109B levels and their distribution were studied using the TIMER database and single-cell analysis. The potential role of FAM109B in gliomas was further investigated through in vitro and in vivo experiments. RESULTS: FAM109B was significantly elevated in various tumor types and was associated with poor prognosis. Its expression was related to aggressive progression and poor prognosis in low-grade glioma patients, serving as an independent prognostic marker for LGG. Glioma grade was negatively correlated with FAM109B DNA promoter methylation. Immune infiltration and single-cell analysis showed significant expression of FAM109B in tumor-associated macrophages (TAMs). The expression of FAM109B was closely related to gene mutations, immune checkpoints (ICs), and chemotherapy drugs in LGG. In vitro studies showed increased FAM109B expression in LGG, closely related to cell proliferation. In vivo studies showed that mice in the sh-FAM109B group had slower tumor growth, slower weight loss, and longer survival times. CONCLUSIONS: FAM109B, as a novel prognostic biomarker for low-grade gliomas, exhibits specific overexpression in TAMs and may be a potential therapeutic target for LGG patients.


Subject(s)
Brain Neoplasms , DNA Methylation , Gene Expression Regulation, Neoplastic , Glioma , Neoplasm Grading , Tumor-Associated Macrophages , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Humans , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Tumor-Associated Macrophages/immunology , DNA Methylation/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/immunology , Prognosis , Carcinogenesis/genetics , Carcinogenesis/pathology , DNA Copy Number Variations/genetics , Tumor Microenvironment , Cell Line, Tumor , Female , Male , Mice, Nude , Mice , Kaplan-Meier Estimate , Databases, Genetic
2.
Acta Neuropathol Commun ; 12(1): 133, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39148129

ABSTRACT

Tumor-associated macrophages (TAMs) residing in the tumor microenvironment (TME) are characterized by their pivotal roles in tumor progression, antitumor immunity, and TME remodeling. However, a thorough comparative characterization of tumor-TAM crosstalk across IDH-defined categories of glioma remains elusive, likely contributing to mixed outcomes in clinical trials. We delineated the phenotypic heterogeneity of TAMs across IDH-stratified gliomas. Notably, two TAM subsets with a mesenchymal phenotype were enriched in IDH-WT glioblastoma (GBM) and correlated with poorer patient survival and reduced response to anti-PD-1 immune checkpoint inhibitor (ICI). We proposed SLAMF9 receptor as a potential therapeutic target. Inference of gene regulatory networks identified PPARG, ELK1, and MXI1 as master transcription factors of mesenchymal BMD-TAMs. Our analyses of reciprocal tumor-TAM interactions revealed distinct crosstalk in IDH-WT tumors, including ANXA1-FPR1/3, FN1-ITGAVB1, VEGFA-NRP1, and TNFSF12-TNFRSF12A with known contribution to immunosuppression, tumor proliferation, invasion and TAM recruitment. Spatially resolved transcriptomics further elucidated the architectural organization of highlighted communications. Furthermore, we demonstrated significant upregulation of ANXA1, FN1, NRP1, and TNFRSF12A genes in IDH-WT tumors using bulk RNA-seq and RT-qPCR. Longitudinal expression analysis of candidate genes revealed no difference between primary and recurrent tumors indicating that the interactive network of malignant states with TAMs does not drastically change upon recurrence. Collectively, our study offers insights into the unique cellular composition and communication of TAMs in glioma TME, revealing novel vulnerabilities for therapeutic interventions in IDH-WT GBM.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Transcriptome , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Tumor Microenvironment/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Isocitrate Dehydrogenase/genetics , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Single-Cell Analysis
3.
J Biochem Mol Toxicol ; 38(8): e23801, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39132772

ABSTRACT

Lung cancer (LC) is a major inducer of cancer-related death. We aim to reveal the effect of Calsequestrin2 (CASQ2) on macrophage polarization and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway in LC. Hub genes were determined from protein-protein interaction networks based on GSE21933 and GSE1987 data sets using bioinformatic analysis. Expression of hub genes was verified by real-time quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, wound-healing, colony formation, and transwell assays were performed to assess the impact of CASQ2 on LC cells. A xenograft mouse model was evaluated using hematoxylin-eosin, immunohistochemistry, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining to investigate the effect of CASQ2 on LC. The role of CASQ2 in regulating macrophage polarization and JAK/STAT pathway was evaluated by western blot andRT-qPCR. We screened out 155 common differentially expressed genes in GSE21933 and GSE1987 data sets. Myomesin-2, tyrosine kinase, sex determining region Y-box 2, platelet and endothelial cell adhesion molecule 1, matrix metallopeptidase 9, claudin-5, caveolin-1, CASQ2, recombinant ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 (ATP2A2), and ankyrin repeat domain 1 were identified as the hub genes with high prediction value. CASQ2 was selected as a pivotal regulator of LC. In vitro experiments and xenograft models revealed that CASQ2 overexpression suppressed proliferation, colony formation, migration, invasion of LC cells, and tumor growth in vivo. Additionally, overexpression of CASQ2 promoted the expression of M1 macrophage markers (cluster of differentiation 80 [CD80], interleukin [IL]-12, inducible nitric oxide synthase [iNOS]), while decreasing the expression of M2 macrophage markers (CD163, IL-10, Arg1) in tumor-associated macrophages and xenograft tissues. Finally, we found that overexpression of CASQ2 inhibited JAK/STAT pathway. CASQ2 is a novel biomarker, which can alleviate LC via inhibiting M2 tumor-associated macrophage polarization and JAK/STAT pathway.


Subject(s)
Janus Kinases , Lung Neoplasms , STAT Transcription Factors , Tumor-Associated Macrophages , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Humans , Animals , Mice , Janus Kinases/metabolism , Janus Kinases/genetics , STAT Transcription Factors/metabolism , STAT Transcription Factors/genetics , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Signal Transduction , Mice, Nude , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Cell Line, Tumor
4.
Sci Rep ; 14(1): 18617, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127769

ABSTRACT

Endometrial cancer (EC), one of the most prevalent carcinomas in females, is associated with increasing mortality. We identified the CHD4 R975H mutation as a high-frequency occurrence in EC patients through a comprehensive survey of EC databases. Computational predictions suggest that this mutation profoundly impacts the structural and functional integrity of CHD4. Functional assays revealed that the CHD4 R975H mutation enhances EC cell invasion, proliferation, and colony formation, promoting a cancer stem cell (CSC)-like phenotype. RNA-seq analysis of cells expressing CHD4 R975H mutant revealed a transcriptomic landscape marked by the activation of several cancer-promoting signaling pathways, including TNF-α signaling via NF-κB, KRAS, P53, mTOR, TGF-ß, EGFR, Myc and growth factor signaling. Validation assays confirmed the activation of these pathways, further demonstrating that CHD4 R975H mutation induces stemness in EC cells and M2-like polarization of tumor-associated macrophages (TAMs). Our study elucidated the oncogenic role of CHD4 R975H mutation, highlighting its dual impact on facilitating cancer stemness and transforming TAMs into an immunosuppressive subtype. These findings contribute valuable insights into the molecular mechanisms driving EC progression and open avenues for targeted therapeutic interventions.


Subject(s)
Endometrial Neoplasms , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Neoplastic Stem Cells , Signal Transduction , Animals , Female , Humans , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
5.
Oncogene ; 43(36): 2737-2749, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112517

ABSTRACT

The proneural-mesenchymal (PN-MES) transformation of glioma stem cells (GSCs) can significantly increase proliferation, invasion, chemotherapy tolerance, and recurrence. M2-like polarization of tumor-associated macrophages (TAMs) has a strong immunosuppressive effect, promoting tumor malignancy and angiogenesis. There is limited understanding on the interactions between GSCs and TAMs as well as their associated molecular mechanisms. In the present study, bioinformatics analysis, GSC and TAM co-culture, determination of TAM polarization phenotypes, and other in vitro experiments confirmed that CCL2 secreted by MES-GSCs promotes TAM-M2 polarization via the IKZF1-CD84-SHP2 pathway and PN-MES transformation of GSCs via the IKZF1-LRG1 pathway in TAMs. IKZF1 inhibitors could significantly reduce tumor volumes in animal glioma models and improve survival, as well as suppress TAM-M2 polarization and the GSC malignant phenotype. The results of this study indicate the important interaction between TAMs and GSCs in the glioma microenvironment as well as its role in tumor progression. The findings also suggest a novel target for follow-up clinical transformation research on the regulation of TAM function and GSCs malignant phenotype.


Subject(s)
Chemokine CCL2 , Glioma , Ikaros Transcription Factor , Neoplastic Stem Cells , Tumor Microenvironment , Tumor-Associated Macrophages , Glioma/pathology , Glioma/genetics , Glioma/metabolism , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Animals , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Humans , Mice , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/metabolism , Tumor Microenvironment/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction/genetics , Cell Line, Tumor , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Antigens, CD/metabolism , Antigens, CD/genetics , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Macrophages/pathology
6.
Cell Death Dis ; 15(8): 633, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198393

ABSTRACT

Long non-coding RNAs (lncRNAs) play an important role in breast cancer progression, but the function of lncRNAs in regulating tumor-associated macrophages (TAMs) remains unclear. As carriers of lncRNAs, exosomes play an important role as mediators in the communication between cancer cells and the tumor microenvironment. In this study, we found that lncRNA HAGLROS was highly expressed in breast cancer tissues and plasma exosomes, and its high expression was related to the poor prognosis of breast cancer patients. Functionally, breast cancer cell-derived exosomal lncRNA HAGLROS promotes breast cancer cell proliferation, migration, epithelial-mesenchymal transition (EMT) process and angiogenesis by inducing TAM/M2 polarization. Mechanistically, lncRNA HAGLROS competitively binds to miR-135-3p to prevent the degradation of its target gene COL10A1. Collectively, these results indicated that the lncRNA HAGLROS/miR-135b-3p/COL10A1 axis promoted breast cancer progression, and revealed the interactive communication mechanism between breast cancer cells and TAMs, suggesting that lncRNA HAGLROS may be a potential biomarker and therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms , Exosomes , MicroRNAs , RNA, Long Noncoding , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Exosomes/metabolism , Exosomes/genetics , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
7.
Article in English | MEDLINE | ID: mdl-39098442

ABSTRACT

OBJECTIVE: The current study aimed to investigate the characteristics of tumor-associated macrophages (TAMs) and their association with microvascular density (MVD) in tumor progression in different grades of orofacial squamous cell carcinoma (OSCC) in the Pakistani population. STUDY DESIGN: This prospective study included 234 patients with oral cancer reported at different hospitals in Pakistan diagnosed with OSCC. Tumors were graded on the Anneroth grading system and the association between the frequency of TAMs and MVD was examined in vivo. The macrophages visible through immunohistochemistry for CD68 and the microvessels observed through immunohistochemistry for CD34 were manually counted in 3 high-power fields. RESULTS: The CD68 and CD34 counts were significantly lower in well-differentiated squamous cell carcinoma compared to poorly differentiated squamous cell carcinoma. Linear regression analysis revealed a positive correlation between the area percentage of CD68 immunoreactivity and the grade of the tumor (r = 0.776). Vice versa, a positive correlation also existed between the area percentage of CD34 immunoreactivity and the grade of the tumor (r = 0.690). Pearson correlation revealed a positive association between the TAMs and MVD (r = 0.680; P < .001). CONCLUSIONS: There was an increased population of tumor-associated macrophages and tumor angiogenesis with the increasing grade of orofacial squamous cell carcinoma. (Oral Surg Oral Med Oral Pathol Oral Radiol YEAR;VOL:page range).


Subject(s)
Antigens, Differentiation, Myelomonocytic , Carcinoma, Squamous Cell , Disease Progression , Immunohistochemistry , Microvascular Density , Mouth Neoplasms , Neoplasm Grading , Tumor-Associated Macrophages , Humans , Male , Female , Prospective Studies , Middle Aged , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/blood supply , Mouth Neoplasms/pathology , Mouth Neoplasms/blood supply , Tumor-Associated Macrophages/pathology , Aged , Adult , Antigens, CD/analysis , Neovascularization, Pathologic/pathology , Antigens, CD34 , CD68 Molecule
8.
Pathol Res Pract ; 261: 155474, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067172

ABSTRACT

BACKGROUND: C-X-C motif chemokine ligand 5 (CXCL5) is a chemokine molecule that is secreted by immune cells in attracting granulocytes. Studies showed that CXCL5 was related to the progression of papillary thyroid carcinoma (PTC) tumor cells. However, the in vivo effects of CXCL5 on PTC tumor cells and their microenvironment have not been elucidated. The present study aimed to investigate the biological effects of CXCL5 on tumor cells, microenvironment, and clinical progression of PTC. MATERIALS AND METHODS: The PTC patients from The Human Cancer Genome Atlas (TCGA) - thyroid carcinoma (THCA) were retrieved. There were a total of 500 patients who met the criteria of our study. Differential expression (DEA) and pathway analyses were used to explore the biological effects of CXCL5 gene expression. RESULTS: In DEA, we found that CXCL5 was mostly associated with PBPP, SLC11A1, and MRC1 (adjusted p<0.001). Samples with CXCL5 FPKM≥1 were related to a different immune profile (p<0.001). In pathway analyses, samples with higher CXCL5 expression possessed higher activities of RAS-RAF, NF-kB, PRC2, IL2, IL5, and Wnt pathways (adjusted p<0.001). In microenvironment analysis, CXCL5 was highly correlated with the activity of macrophage (Rho=0.76; adjusted p<0.001). Clinically, high level of CXCL5 expression was an indicator of tumor stages (p<0.001), nodal metastasis (AUC=0.68), and prognosis (p=0.001). CONCLUSION: CXCL5 was a significant biomarker of PTC. CXCL5 was highly associated with tumor immunology and microenvironment. Samples with higher CXCL5 expression had more advanced disease status and worse prognosis. CXCL5 target therapy is potentially helpful in advanced PTC.


Subject(s)
Chemokine CXCL5 , Thyroid Cancer, Papillary , Thyroid Neoplasms , Tumor Microenvironment , Humans , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Tumor Microenvironment/immunology , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/immunology , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/immunology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Male , Female , Middle Aged , Macrophages/metabolism , Macrophages/immunology , Macrophages/pathology , Adult , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Signal Transduction , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
9.
Mol Carcinog ; 63(9): 1827-1841, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39016636

ABSTRACT

LAIR1, a receptor found on immune cells, is capable of binding to collagen and is involved in immune-related diseases. However, the precise contribution of LAIR1 expressed on hepatocellular carcinoma (HCC) cells to tumor microenvironment is still unclear. In our study, bioinformatics analysis and immunofluorescence were employed to study the correlation between LAIR1 levels and clinical indicators. Transwell and scratch tests were used to evaluate how LAIR1 affected the migration and invasion of HCC cells. The chemotactic capacity and alternative activation of macrophages were investigated using RT-qPCR, transwell, and immunofluorescence. To investigate the molecular mechanisms, transcriptome sequencing analysis, Western blot, nucleus/cytoplasm fractionation, ELISA, and cytokine microarray were employed. We revealed a significant correlation between the presence of LAIR1 and an unfavorable outcome in HCC. We indicated that LAIR1 promoted migration and invasion of HCC cells through the AKT-IKKß-p65 axis. Additionally, the alternative activation and infiltration of tumor-associated macrophages induced by LAIR1 were reliant on the upregulation of IL6 and CCL5 within this axis, respectively. In conclusion, blocking LAIR1 was found to be an effective approach in combating the cancerous advancement of HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell Movement , Liver Neoplasms , Proto-Oncogene Proteins c-akt , Receptors, Immunologic , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , I-kappa B Kinase/metabolism , I-kappa B Kinase/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Cell Line, Tumor , Tumor Microenvironment , Gene Expression Regulation, Neoplastic , Signal Transduction , Macrophages/metabolism , Macrophages/pathology , Cell Proliferation , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Tumor-Associated Macrophages/immunology , Neoplasm Invasiveness
10.
Cell Death Dis ; 15(7): 498, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997297

ABSTRACT

The tumor microenvironment is a complex space comprised of normal, cancer and immune cells. The macrophages are considered as the most abundant immune cells in tumor microenvironment and their function in tumorigenesis is interesting. Macrophages can be present as M1 and M2 polarization that show anti-cancer and oncogenic activities, respectively. Tumor-associated macrophages (TAMs) mainly have M2 polarization and they increase tumorigenesis due to secretion of factors, cytokines and affecting molecular pathways. Hepatocellular carcinoma (HCC) is among predominant tumors of liver that in spite of understanding its pathogenesis, the role of tumor microenvironment in its progression still requires more attention. The presence of TAMs in HCC causes an increase in growth and invasion of HCC cells and one of the reasons is induction of glycolysis that such metabolic reprogramming makes HCC distinct from normal cells and promotes its malignancy. Since M2 polarization of TAMs stimulates tumorigenesis in HCC, molecular networks regulating M2 to M1 conversion have been highlighted and moreover, drugs and compounds with the ability of targeting TAMs and suppressing their M2 phenotypes or at least their tumorigenesis activity have been utilized. TAMs increase aggressive behavior and biological functions of HCC cells that can result in development of therapy resistance. Macrophages can provide cell-cell communication in HCC by secreting exosomes having various types of biomolecules that transfer among cells and change their activity. Finally, non-coding RNA transcripts can mainly affect polarization of TAMs in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Tumor-Associated Macrophages/immunology , Animals , Macrophages/metabolism
11.
Asian Pac J Cancer Prev ; 25(7): 2515-2527, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39068587

ABSTRACT

BACKGROUND: Cluster of differentiation 47 (CD47) has been identified as a new immune checkpoint. The exact role of CD47 in prognosis of breast cancer remains unclear. This study aims to evaluate immunohistochemical (IHC) expression of CD47 in breast cancer, and to measure the density of tumor associated macrophages (TAMs) infiltration by CD68 IHC staining. Furthermore, assessing the relations of CD47 and CD68 expression to different clinicopathological variables and evaluating the prognostic role of CD47 and CD68 in breast cancer cases. METHODS: This retrospective cohort study included 200 diagnosed primary breast cancer cases who underwent surgical resection at the Oncology Center of Mansoura University (OCMU), Faculty of Medicine, Egypt. Clinicopathological and survival data were collected. IHC for CD47 and CD68 was performed. RESULTS: Among 200 breast cancer cases, high CD47 expression was detected in 89 cases (44.5%). CD47 high expression was significantly associated with presence of distant metastasis (P=0.04), advanced TNM stage (P=0.02), ER & PR negativity (P=0.04 & 0.004 respectively), and molecular subtype (P=0.03). Their was a statistically significant association between CD47 and CD68 expression (P=0.002). CD47 high expression was found to predict poor overall survival, but it is not considered alone as independent poor prognostic factor by multivariate analysis. Multivariate analysis spotted combined high expression of CD47 and CD68 as an independent prognostic predictor for shorter OS in breast cancer patients (P=0.002). CONCLUSION: CD47 high expression is related to poor prognosis in breast cancer patients especially when associated with high CD68+TAMs infiltration. Therefore, CD47 is a promising prognostic and therapeutic target in breast carcinoma that may direct selection of patients for immunotherapy.


Subject(s)
Antigens, CD , Antigens, Differentiation, Myelomonocytic , Biomarkers, Tumor , Breast Neoplasms , CD47 Antigen , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , CD47 Antigen/metabolism , Prognosis , Retrospective Studies , Middle Aged , Biomarkers, Tumor/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Adult , Follow-Up Studies , Survival Rate , Immunohistochemistry , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , CD68 Molecule
12.
Sci Rep ; 14(1): 17412, 2024 07 29.
Article in English | MEDLINE | ID: mdl-39075108

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal cancer that accounts for over 90% of all pancreatic cancer cases. With a 5-year survival rate of only 13%, PDAC has proven to be extremely desmoplastic and immunosuppressive to most current therapies, including chemotherapy and surgical resection. In recent years, focus has shifted to understanding the tumor microenvironment (TME) around PDAC, enabling a greater understanding of biological pathways and intercellular interactions that can ultimately lead to potential for future drug targets. In this study, we leverage a combination of single-cell and spatial transcriptomics to further identify cellular populations and interactions within the highly heterogeneous TME. We demonstrate that SPP1+APOE+ tumor-associated macrophages (TAM) and CTHRC1+GREM1+ cancer-associated myofibroblasts (myCAF) not only act synergistically to promote an immune-suppressive TME through active extracellular matrix (ECM) deposition and epithelial mesenchymal transition (EMT), but are spatially colocalized and correlated, leading to worse prognosis. Our results highlight the crosstalk between stromal and myeloid cells as a significant area of study for future therapeutic targets to treat cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Macrophages , Pancreatic Neoplasms , Tumor Microenvironment , Tumor Microenvironment/immunology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/immunology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/immunology , Humans , Macrophages/metabolism , Macrophages/immunology , Animals , Mice , Fibroblasts/metabolism , Fibroblasts/pathology , Epithelial-Mesenchymal Transition , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Cell Line, Tumor , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Prognosis
13.
Mol Cancer ; 23(1): 150, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068459

ABSTRACT

Tumor-associated macrophages (TAMs) are pivotal in cancer progression, influencing tumor growth, angiogenesis, and immune evasion. This review explores the spatial and temporal heterogeneity of TAMs within the tumor microenvironment (TME), highlighting their diverse subtypes, origins, and functions. Advanced technologies such as single-cell sequencing and spatial multi-omics have elucidated the intricate interactions between TAMs and other TME components, revealing the mechanisms behind their recruitment, polarization, and distribution. Key findings demonstrate that TAMs support tumor vascularization, promote epithelial-mesenchymal transition (EMT), and modulate extracellular matrix (ECM) remodeling, etc., thereby enhancing tumor invasiveness and metastasis. Understanding these complex dynamics offers new therapeutic targets for disrupting TAM-mediated pathways and overcoming drug resistance. This review underscores the potential of targeting TAMs to develop innovative cancer therapies, emphasizing the need for further research into their spatial characteristics and functional roles within the TME.


Subject(s)
Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Tumor Microenvironment/immunology , Neoplasms/pathology , Neoplasms/immunology , Neoplasms/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Animals , Epithelial-Mesenchymal Transition , Neovascularization, Pathologic/pathology
14.
J Cancer Res Clin Oncol ; 150(6): 320, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38914803

ABSTRACT

PURPOSE: Tumor-associated macrophages (TAMs) play a critical role in hepatocellular carcinoma (HCC) progression and metastasis. Systematic investigation of the cross-talk between TAMs and HCC may help in searching for the critical target to guard against HCC metastasis. METHODS AND RESULTS: Herein, we found that TREM1 highly expressed in HCC tissue by analyzing the data obtain from GEO database. Interestingly, the results indicated that TREM1 was primarily expressed by monocytes. Immune infiltration studies further validated that TREM1 expression was positively related with increased infiltration of macrophages in HCC tissues. In vitro, we observed that TREM1 knockdown significantly abrogated the effect of TAMs in promoting the metastasis and epithelial-mesenchymal transition (EMT) of HCC cells. Additionally, cytokine array detection identified CCL7 as the main responsive cytokine following with TREM1 knockdown in TAMs. CONCLUSION: Taken together, our findings strongly suggested that high expression of TREM1 was positively associated with metastasis and poor prognosis of HCC. Furthermore, TAMs expressing TREM1 contribute to EMT-based metastasis through secreting CCL7. These results provide a novel insight into the potential development of targeting the TREM1/CCL7 pathway for preventing metastatic HCC.


Subject(s)
Carcinoma, Hepatocellular , Epithelial-Mesenchymal Transition , Liver Neoplasms , Triggering Receptor Expressed on Myeloid Cells-1 , Female , Humans , Male , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Neoplasm Metastasis , Prognosis , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/genetics , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
15.
BMC Cancer ; 24(1): 698, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849760

ABSTRACT

BACKGROUND: Tumor-associated macrophages (TAMs) constitute a substantial part of human hepatocellular carcinoma (HCC). The present study was devised to explore TAM diversity and their roles in HCC progression. METHODS: Through the integration of multiple 10 × single-cell transcriptomic data derived from HCC samples and the use of consensus nonnegative matrix factorization (an unsupervised clustering algorithm), TAM molecular subtypes and expression programs were evaluated in detail. The roles played by these TAM subtypes in HCC were further probed through pseudotime, enrichment, and intercellular communication analyses. Lastly, vitro experiments were performed to validate the relationship between CD63, which is an inflammatory TAM expression program marker, and tumor cell lines. RESULTS: We found that the inflammatory expression program in TAMs had a more obvious interaction with HCC cells, and CD63, as a marker gene of the inflammatory expression program, was associated with poor prognosis of HCC patients. Both bulk RNA-seq and vitro experiments confirmed that higher TAM CD63 expression was associated with the growth of HCC cells as well as their epithelial-mesenchymal transition, metastasis, invasion, and the reprogramming of lipid metabolism. CONCLUSIONS: These analyses revealed that the TAM inflammatory expression program in HCC is closely associated with malignant tumor cells, with the hub gene CD63 thus representing an ideal target for therapeutic intervention in this cancer type.


Subject(s)
Carcinoma, Hepatocellular , Disease Progression , Epithelial-Mesenchymal Transition , Liver Neoplasms , Tetraspanin 30 , Tumor-Associated Macrophages , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Tetraspanin 30/metabolism , Tetraspanin 30/genetics , Lipid Metabolism/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Prognosis , Cellular Reprogramming/genetics
16.
FASEB J ; 38(13): e23762, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38923643

ABSTRACT

Exosomes play significant roles in the communications between tumor cells and tumor microenvironment. However, the specific mechanisms by which exosomes modulate tumor development under hypoxia in pancreatic neuroendocrine tumors (pNETs) are not well understood. This study aims to investigate these mechanisms and made several important discoveries. We found that hypoxic exosomes derived from pNETs cells can activate tumor-associated macrophages (TAM) to the M2 phenotype, in turn, the M2-polarized TAM, facilitate the migration and invasion of pNETs cells. Further investigation revealed that CEACAM5, a protein highly expressed in hypoxic pNETs cells, is enriched in hypoxic pNETs cell-derived exosomes. Hypoxic exosomal CEACAM5 was observed to induce M2 polarization of TAM through activation of the MAPK signaling pathway. Coculturing pNETs cells with TAM or treated with hypoxic exosomes enhanced the metastatic capacity of pNETs cells. In conclusion, these findings suggest that pNETs cells generate CEACAM5-rich exosomes in a hypoxic microenvironment, which in turn polarize TAM promote malignant invasion of pNETs cells. Targeting exosomal CEACAM5 could potentially serve as a diagnostic and therapeutic strategy for pNETs.


Subject(s)
Antigens, CD , Exosomes , GPI-Linked Proteins , Matrix Metalloproteinase 9 , Neuroendocrine Tumors , Pancreatic Neoplasms , Tumor Microenvironment , Tumor-Associated Macrophages , Exosomes/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Humans , Animals , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Matrix Metalloproteinase 9/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Mice , Cell Line, Tumor , Antigens, CD/metabolism , GPI-Linked Proteins/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Neoplasm Metastasis , Mice, Nude , Hypoxia/metabolism , Cell Hypoxia/physiology , Carcinoembryonic Antigen
17.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892411

ABSTRACT

Breast cancers (BCs) are solid tumors composed of heterogeneous tissues consisting of cancer cells and an ever-changing tumor microenvironment (TME). The TME includes, among other non-cancer cell types, immune cells influencing the immune context of cancer tissues. In particular, the cross talk of immune cells and their interactions with cancer cells dramatically influence BC dissemination, immunoediting, and the outcomes of cancer therapies. Tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) represent prominent immune cell populations of breast TMEs, and they have important roles in cancer immunoescape and dissemination. Therefore, in this article we review the features of TILs, TAMs, and MDSCs in BCs. Moreover, we highlight the mechanisms by which these immune cells remodel the immune TME and lead to breast cancer metastasis.


Subject(s)
Breast Neoplasms , Lymphocytes, Tumor-Infiltrating , Myeloid-Derived Suppressor Cells , Neoplasm Metastasis , Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Tumor Microenvironment/immunology , Breast Neoplasms/pathology , Breast Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Female , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , T-Lymphocytes/immunology , Animals
18.
Cell Rep Methods ; 4(6): 100800, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38889689

ABSTRACT

The tumor microenvironment harbors a variety of different cell types that differentially impact tumor biology. In this issue of Cell Reports Methods, Raffo-Romero et al. standardized and optimized 3D tumor organoids to model the interactions between tumor-associated macrophages and tumor cells in vitro.


Subject(s)
Organoids , Tumor Microenvironment , Humans , Organoids/pathology , Neoplasms/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology , Animals
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167276, 2024 10.
Article in English | MEDLINE | ID: mdl-38844114

ABSTRACT

The role of hypoxia in the tumor microenvironment of intrahepatic cholangiocarcinoma (iCCA) remains unclear. Here, we generated a comprehensive atlas of the entire tumor microenvironment and delineated the multifaceted cell-cell interactions to decipher hypoxia-induced pro-tumor immune suppression. We discovered hypoxia is significantly associated with iCCA progression via the activation of HIF1A expression. Moreover, hypoxia-dependent PPARγ-mediated fatty acid oxidation in APOE+ TAMs promoted M2 macrophage polarization by activating the HIF1A-PPARG-CD36 axis. These polarized APOE+ TAMs recruited Treg cell infiltration via the CCL3-CCR5 pair to form an immunosuppressive microenvironment. APOE+ TAMs tended to co-localize spatially with Treg cells in the malignant tissue based on spatial transcriptome data and immunofluorescence analysis results. We identified tumor-reactive CXCL13+ CD8-PreTex with specific high expression of ENTPD1 and ITGAE, which acted as precursors of CD8-Tex and had higher cytotoxicity, lower exhaustion, and more vigorous proliferation. Consequently, CXCL13+ CD8-PreTex functioned as a positive regulator of antitumor immunity by expressing the pro-inflammatory cytokines IFNG and TNF, associated with a better survival outcome. Our study reveals the mechanisms involved in hypoxia-induced immunosuppression and suggests that targeting precursor-exhausted CXCL13+CD8+ T cells might provide a pratical immunotherapeutic approach.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Single-Cell Analysis , Tumor Microenvironment , Cholangiocarcinoma/immunology , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Tumor Microenvironment/immunology , Humans , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/metabolism , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Mice , Cell Line, Tumor , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
20.
Hum Pathol ; 150: 29-35, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38914168

ABSTRACT

Discoidin Domain Receptor 2 (DDR2) is a receptor tyrosine kinase for collagen, stimulating epithelial-mesenchymal transition and stiffness in breast cancer. Here, we investigated levels of DDR2 in breast tumor cells in relation to vascular invasion, TIL subsets, macrophages, molecular tumor subtypes, modes of detection and prognosis. This retrospective, population-based series of invasive breast carcinomas from the Norwegian Screening Program in Vestfold County (Norway), period 2004-2009, included 200 screening patients and 82 cases detected in screening intervals. DDR2 was examined on core needle biopsies using a semi-quantitative, immunohistochemical staining index and dichotomized as low or high DDR2 expression. Counts of macrophages and TIL subsets were dichotomized based on immunohistochemistry using TMA. We also recorded blood or lymphatic vessel invasion (BVI or LVI) as present or absent by immunohistochemistry. High expression of DDR2 in tumor cells showed significant relation with high counts of CD163+ macrophages (p < 0.001) and FOXP3 TILs (p = 0.011), presence of BVI (p = 0.028), high tumor cell proliferation by Ki67 (p = 0.033), ER negativity (p = 0.001), triple-negative cases (p = 0.038), basal-like features (p < 0.001) as well as interval detection (p < 0.001). By multivariate analysis, high DDR2 expression was related to reduced recurrence-free survival (HR, 2.3, p = 0.017), when examined together with histologic grading, lymph node assessment, tumor diameter, BVI, and molecular tumor subtype. This study supports a link between high DDR2 expression, high counts of macrophages by CD163 (tumor associated) and regulatory T cells by FOXP3 together with the presence of BVI, possibly indicating increased tumor motility and intravasation in aggressive breast tumors.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Discoidin Domain Receptor 2 , Immunohistochemistry , Neoplasm Invasiveness , T-Lymphocytes, Regulatory , Tumor-Associated Macrophages , Humans , Female , Breast Neoplasms/pathology , Retrospective Studies , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , Biomarkers, Tumor/analysis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Middle Aged , Aged , Lymphocytes, Tumor-Infiltrating/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Norway , Prognosis , Receptors, Cell Surface/analysis , Kaplan-Meier Estimate , Antigens, CD , Antigens, Differentiation, Myelomonocytic/analysis , Antigens, Differentiation, Myelomonocytic/metabolism , Biopsy, Large-Core Needle , Proportional Hazards Models , Predictive Value of Tests , Forkhead Transcription Factors/analysis , Macrophages/pathology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL