101
|
Shi H, Han L, Zhao J, Wang K, Xu M, Shi J, Dong Z. Tumor stemness and immune infiltration synergistically predict response of radiotherapy or immunotherapy and relapse in lung adenocarcinoma. Cancer Med 2021; 10:8944-8960. [PMID: 34741449 PMCID: PMC8683560 DOI: 10.1002/cam4.4377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 09/12/2021] [Accepted: 10/07/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer stem cells (CSCs) have been shown to accelerate tumor recurrence, radiotherapy, and chemotherapy resistance. Immunotherapy is a powerful anticancer treatment that can significantly prolong the overall survival of patients with lung adenocarcinoma (LUAD). However, little is known about the function of genes related to tumor stemness and immune infiltration in LUAD. After integrating the tumor stemness index based on mRNA expression (mRNAsi), immune score, mRNA expression, and clinical information from the TCGA database, we screened 380 tumor stemness and immune (TSI)-related genes and constructed a five TSI-specific-gene (CPS1, CCR2, NT5E, ANLN, and ABCC2) signature (TSISig) using a machine learning method. Survival analysis indicated that TSISig could stably predict the prognosis of patients with LUAD. Comparison of mRNAsi and immune score between high- and low-TSISig groups suggested that TSISig characterized tumor stemness and immune infiltration. In addition, enrichment of immune subpopulations showed that the low-TSISig group held more immune subpopulations. GSEA revealed that TSISig had a strong association with the cell cycle and human immune response. Further analysis revealed that TSISig not only had a good predictive ability for prognosis but could also serve as an excellent predictor of tumor recurrence and response to radiotherapy and immunotherapy in LUAD patients. TSISig might regulate the development of LUAD by coordinating tumor stemness and immune infiltration. Finally, a connectivity map (CMap) analysis demonstrated that the HDAC inhibitor could target TSISig.
Collapse
Affiliation(s)
- Hongjie Shi
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linzhi Han
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinping Zhao
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kaijie Wang
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming Xu
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiajun Shi
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhe Dong
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
102
|
Liu M, Cai R, Wang T, Yang X, Wang M, Kuang Z, Xie Y, Zhang J, Zheng Y. LAMC2 promotes the proliferation of cancer cells and induce infiltration of macrophages in non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1392. [PMID: 34733944 PMCID: PMC8506528 DOI: 10.21037/atm-21-4507] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022]
Abstract
Background Non-small-cell lung cancer (NSCLC) is the most prevalent cancer worldwide. Tumor microenvironment (TME) plays a very important role in the cancer development. Thus, it is urgent to find the change of TME that contributes to NSCLC carcinogenesis and progression. Methods The bioinformatics analysis approach was applied to evaluate the change of TME and screen the differentially immune cells in NSCLC tissue based on The Cancer Genome Atlas (TCGA) data. Meanwhile, the association of differentially immune cells with tumor stage and prognosis of NSCLC was evaluated. Then, we screen the different expression genes between macrophages infiltration high group and low group. After that, the expression of LAMC2 was detected in 48 cases of NSCLC tissues and paired normal tissues. The function of LAMC2 was detected through cell experiments in vitro. Immunohistochemistry assay was used to detect the correlation between LAMC2 expression and macrophages infiltration in NSCLC tissue. LAMC2-related pathways were identified by gene set enrichment analysis. Results Compared with early stage, middle-advanced stage of NSCLC exhibited lower immune score. Macrophages were the main component of different immune cells and correlated with poor outcome. The results of immunohistochemistry indicated that the expression of LAMC2 in NSCLC tissues was higher than paired normal tissues. Down-regulation of LAMC2 inhibited the proliferation, migration and invasion of NSCLC cells in vitro. Overexpression of LAMC2 was positively associated with macrophages infiltration in NSCLC tissues. Inhibition of LAMC2 expression in NSCLC cells could reduce THP-1 infiltration, and LAMC2 protein could promote the infiltration of THP-1. The Gene Set Enrichment Analysis results showed that high expression of LAMC2 was correlated with focal adhesion and extracellular matrix receptor interaction. Conclusions Immune suppression and macrophages infiltration were correlated with poor outcomes in NSCLC. LAMC2 promoted macrophages infiltration and extracellular matrix remolding in NSCLC. Our studies suggested an oncogenic role of LAMC2 in NSCLC progression and it perhaps serve as a potential immune therapy target for NSCLC.
Collapse
Affiliation(s)
- Meiyuan Liu
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Rui Cai
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Tian Wang
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xia Yang
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Meng Wang
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhongsheng Kuang
- Department of Pathology, The First Affiliated Hospital of Guangzhou Traditional Chinese Medicine University, Guangzhou, China
| | - Yuhui Xie
- Department of Pathology, The First Affiliated Hospital of Guangzhou Traditional Chinese Medicine University, Guangzhou, China
| | - Jiren Zhang
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Yanfang Zheng
- Department of Medical Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
103
|
Wang ZH, Li Y, Zhang P, Xiang X, Wei XS, Niu YR, Ye LL, Peng WB, Zhang SY, Xue QQ, Zhou Q. Development and Validation of a Prognostic Autophagy-Related Gene Pair Index Related to Tumor-Infiltrating Lymphocytes in Early-Stage Lung Adenocarcinoma. Front Cell Dev Biol 2021; 9:719011. [PMID: 34616731 PMCID: PMC8488280 DOI: 10.3389/fcell.2021.719011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/30/2021] [Indexed: 01/07/2023] Open
Abstract
The role of autophagy in lung cancer is context-dependent and complex. Recent studies have reported the important role of autophagy in tumor immune escape. However, the association between autophagy and tumor-infiltrating lymphocytes (TILs) in early-stage lung adenocarcinoma (LUAD) remains unclear. In this study, we aimed to develop and validate the autophagy-related gene pair index (ATGPI) and autophagy clinical prognostic index (ACPI) in multiple LUAD cohorts, including The Cancer Genome Atlas (TCGA) cohort, Gene Expression Omnibus cohorts, and one cohort from Union Hospital, Wuhan (UH cohort), using a Cox proportional hazards regression model with the least absolute shrinkage and selection operator. Multivariate Cox regression analysis demonstrated that there was a significant difference in overall survival (OS) between patients with high and low ATGPI in the testing [hazard ratio (HR) = 1.97; P < 0.001] and TCGA validation (HR = 2.25; P < 0.001) cohorts. Time-dependent receiver operating characteristic curve analysis was also performed. We found that high ATGPI could accurately identify patients with early-stage LUAD with shorter OS, with the areas under the curve of 0.703 and 0.676 in the testing and TCGA validation cohorts, respectively. Concordance index (C-index) was used to evaluate the efficiency of ATGPI and ACPI. The C-index of ACPI was higher than that of ATGPI in the testing (0.71 vs. 0.66; P < 0.001), TCGA validation (0.69 vs. 0.65; P = 0.028), and UH (0.80 vs. 0.70; P = 0.015) cohorts. TIL analysis demonstrated that the proportions of tumor-infiltrating CD4+ T cells were lower in the high-ATGPI group than in the low-ATGPI group in both the TCGA validation and UH cohorts. These results indicate the potential clinical use of ATG signatures which are associated with TILs, in identifying patients with early-stage LUAD with different OS.
Collapse
Affiliation(s)
- Zi-Hao Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Li
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pei Zhang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Shan Wei
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Ran Niu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin-Lin Ye
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Bei Peng
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Yu Zhang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Xue
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiong Zhou
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
104
|
Yang Y, Li N, Wang TM, Di L. Natural Products with Activity against Lung Cancer: A Review Focusing on the Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms221910827. [PMID: 34639167 PMCID: PMC8509218 DOI: 10.3390/ijms221910827] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/05/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is one of the most prevalent malignancies worldwide. Despite the undeniable progress in lung cancer research made over the past decade, it is still the leading cause of cancer-related deaths and continues to challenge scientists and researchers engaged in searching for therapeutics and drugs. The tumor microenvironment (TME) is recognized as one of the major hallmarks of epithelial cancers, including the majority of lung cancers, and is associated with tumorigenesis, progression, invasion, and metastasis. Targeting of the TME has received increasing attention in recent years. Natural products have historically made substantial contributions to pharmacotherapy, especially for cancer. In this review, we emphasize the role of the TME and summarize the experimental proof demonstrating the antitumor effects and underlying mechanisms of natural products that target the TME. We also review the effects of natural products used in combination with anticancer agents. Moreover, we highlight nanotechnology and other materials used to enhance the effects of natural products. Overall, our hope is that this review of these natural products will encourage more thoughts and ideas on therapeutic development to benefit lung cancer patients.
Collapse
Affiliation(s)
| | - Ning Li
- Correspondence: (N.L.); (L.D.); Tel.: +86-551-6516-1115 (N.L.)
| | | | - Lei Di
- Correspondence: (N.L.); (L.D.); Tel.: +86-551-6516-1115 (N.L.)
| |
Collapse
|
105
|
Sinjab A, Han G, Treekitkarnmongkol W, Hara K, Brennan PM, Dang M, Hao D, Wang R, Dai E, Dejima H, Zhang J, Bogatenkova E, Sanchez-Espiridion B, Chang K, Little DR, Bazzi S, Tran LM, Krysan K, Behrens C, Duose DY, Parra ER, Raso MG, Solis LM, Fukuoka J, Zhang J, Sepesi B, Cascone T, Byers LA, Gibbons DL, Chen J, Moghaddam SJ, Ostrin EJ, Rosen D, Heymach JV, Scheet P, Dubinett SM, Fujimoto J, Wistuba II, Stevenson CS, Spira A, Wang L, Kadara H. Resolving the Spatial and Cellular Architecture of Lung Adenocarcinoma by Multiregion Single-Cell Sequencing. Cancer Discov 2021; 11:2506-2523. [PMID: 33972311 PMCID: PMC8487926 DOI: 10.1158/2159-8290.cd-20-1285] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/26/2021] [Accepted: 04/30/2021] [Indexed: 11/16/2022]
Abstract
Little is known of the geospatial architecture of individual cell populations in lung adenocarcinoma (LUAD) evolution. Here, we perform single-cell RNA sequencing of 186,916 cells from five early-stage LUADs and 14 multiregion normal lung tissues of defined spatial proximities from the tumors. We show that cellular lineages, states, and transcriptomic features geospatially evolve across normal regions to LUADs. LUADs also exhibit pronounced intratumor cell heterogeneity within single sites and transcriptional lineage-plasticity programs. T regulatory cell phenotypes are increased in normal tissues with proximity to LUAD, in contrast to diminished signatures and fractions of cytotoxic CD8+ T cells, antigen-presenting macrophages, and inflammatory dendritic cells. We further find that the LUAD ligand-receptor interactome harbors increased expression of epithelial CD24, which mediates protumor phenotypes. These data provide a spatial atlas of LUAD evolution, and a resource for identification of targets for its treatment. SIGNIFICANCE: The geospatial ecosystem of the peripheral lung and early-stage LUAD is not known. Our multiregion single-cell sequencing analyses unravel cell populations, states, and phenotypes in the spatial and ecologic evolution of LUAD from the lung that comprise high-potential targets for early interception.This article is highlighted in the In This Issue feature, p. 2355.
Collapse
Affiliation(s)
- Ansam Sinjab
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guangchun Han
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Warapen Treekitkarnmongkol
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kieko Hara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick M Brennan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dapeng Hao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ruiping Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Enyu Dai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hitoshi Dejima
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jiexin Zhang
- Department of Bioinformatics and Computer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elena Bogatenkova
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Beatriz Sanchez-Espiridion
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kyle Chang
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Danielle R Little
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer Bazzi
- Faculty of Arts and Sciences, University of Balamand, Koura, Lebanon
| | - Linh M Tran
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Kostyantyn Krysan
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Carmen Behrens
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dzifa Y Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luisa M Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Junya Fukuoka
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Jianjun Zhang
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Boris Sepesi
- Department of Cardiovascular and Thoracic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tina Cascone
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren Averett Byers
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Don L Gibbons
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Edwin J Ostrin
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Rosen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - John V Heymach
- Department of Thoracic, Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul Scheet
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven M Dubinett
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Avrum Spira
- Lung Cancer Initiative at Johnson and Johnson, Boston, Massachusetts
- Section of Computational Biomedicine, Boston University, Boston, Massachusetts
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
106
|
Hidden Treasures: Macrophage Long Non-Coding RNAs in Lung Cancer Progression. Cancers (Basel) 2021; 13:cancers13164127. [PMID: 34439281 PMCID: PMC8392679 DOI: 10.3390/cancers13164127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/16/2022] Open
Abstract
Ever since RNA sequencing of whole genomes and transcriptomes became available, numerous RNA transcripts without having the classic function of encoding proteins have been discovered. Long non-coding RNAs (lncRNAs) with a length greater than 200 nucleotides were considered as "junk" in the beginning, but it has increasingly become clear that lncRNAs have crucial roles in regulating a variety of cellular mechanisms and are often deregulated in several diseases, such as cancer. Lung cancer is the leading cause of cancer-related deaths and has a survival rate of less than 10%. Immune cells infiltrating the tumor microenvironment (TME) have been shown to have a great effect on tumor development with macrophages being the major cell type within the TME. Macrophages can inherit an inflammatory M1 or an anti-inflammatory M2 phenotype. Tumor-associated macrophages, which are predominantly polarized to M2, favor tumor growth, angiogenesis, and metastasis. In this review, we aimed to describe the complex roles and functions of lncRNAs in macrophages and their influence on lung cancer development and progression through the TME.
Collapse
|
107
|
Yu L, Cao S, Li J, Han B, Zhong H, Zhong R. Prognostic value and immune infiltration of a novel stromal/immune score-related P2RY12 in lung adenocarcinoma microenvironment. Int Immunopharmacol 2021; 98:107734. [PMID: 34175738 DOI: 10.1016/j.intimp.2021.107734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Increasing evidence highlights the clinical implications of P2RY12 that belongs to the family of G-protein coupled receptors in carcinogenesis. Here, this study was designed to explore the associations between P2RY12 and tumor immune microenvironment of lung adenocarcinoma (LUAD). METHODS Based on 352 LUAD samples from The Cancer Genome Atlas (TCGA), stromal and immune scores of each sample were estimated by ESTIMATE algorithm. Differential expression analysis was presented between stromal/immune high- and low-score groups. Protein-protein interaction (PPI) was then constructed by STRING database. Univariate and multivariate Cox regression analysis was utilized to screen prognosis-related factors. Co-expressed genes of P2RY12 were analyzed, followed by functional enrichment analysis. Furthermore, the correlation between P2RY12 and immune cell infiltrations was estimated using the TIMER database. P2RY12 expression was validated between 37 pairs of LUAD and normal tissues using RT-qPCR and immunohistochemistry. After overexpressing P2RY12, the proliferation and migration of A549 cells was detected by CCK-8 and scratch test. RESULTS 145 up- and 102 down-regulated stromal- and immune-related mRNAs were identified for LUAD. Based on 145 up-regulated mRNAs, a PPI network was conducted, consisting of 95 nodes and 210 relationship pairs. Ten hub genes were then identified. Among them, CCR8, CNR2, CXCR5, GPR18, GPR31 and P2RY12 were in association with overall survival of patients with LUAD. After adjusting other variables, P2RY12 expression was an independent prognostic factor for LUAD. 288 co-expressed genes of P2RY12 were determined and these co-expressed genes were primarily involved in immune-related biological processes or pathways. Moreover, P2RY12 was significantly correlated with M2 macrophage and dendritic cell infiltration. After validation, P2RY12 expression was significantly decreased in LUAD than normal tissues. Its overexpression distinctly suppressed proliferation and migration of A549 cells. CONCLUSION Our findings suggest that P2RY12 is an immune infiltration-related prognostic marker for LUAD.
Collapse
Affiliation(s)
- Lian Yu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Shuhui Cao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Jingwen Li
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Hua Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Runbo Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
108
|
Wu J, Li L, Zhang H, Zhao Y, Zhang H, Wu S, Xu B. A risk model developed based on tumor microenvironment predicts overall survival and associates with tumor immunity of patients with lung adenocarcinoma. Oncogene 2021; 40:4413-4424. [PMID: 34108619 DOI: 10.1038/s41388-021-01853-y] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/10/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022]
Abstract
Tumor microenvironment (TME) has been reported to exhibit a crucial effect in lung cancer. Therefore, this study was aimed to investigate the genes associated with TME and develop a risk score to predict the overall survival (OS) of patients with lung adenocarcinoma (LUAD) based on these genes. The immune and stromal scores were generated by the ESTIMATE algorithm for LUAD patients in The Cancer Genome Atlas (TCGA) database. Differentially expressed gene and weighted gene co-expression network analyses were used to derive immune- and stromal-related genes. The Least Absolute Shrinkage and Selection Operator (LASSO)-Cox regression was applied for further selection and the selected genes were inputted into stepwise regression to develop TME-related risk score (TMErisk) which was further validated in Gene Expression Omnibus (GEO) datasets. TMErisk-related biological phenotypes were analyzed in function enrichment, tumor immune signature, and tumor mutation signature. The patient's response to immunotherapy was inferred by the tumor immune dysfunction and exclusion (TIDE) score and immunophenoscore (IPS). According to our results, TMErisk was developed based on SERPINE1, CX3CR1, CD200R1, GBP1, IRF1, STAP1, LOX, and OR7E47P. Furthermore, high TMErisk was identified as a poor factor for OS in TCGA and GEO datasets, as well as in subgroup analysis with different gender, smoking status, age, race, anatomic site, therapies, and tumor-node-metastasis (TNM) stages. Higher TMErisk is also associated negatively with the abundance of B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and other stromal or immune cells. Several genes of the human leukocyte antigen (HLA) family and immune checkpoints were less expressed in the high-TMErisk group. Mutations of 19 genes occurred more frequently in the high-TMErisk group. These mutations may be associated with TME change and indicate patients' response to immunotherapy. According to our analyses, a lower TMErisk score may indicate better response and OS outcome of immunotherapy.
Collapse
Affiliation(s)
- Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Huibo Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yaqi Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Haohan Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyi Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
109
|
Saxena S, Singh RK. Chemokines orchestrate tumor cells and the microenvironment to achieve metastatic heterogeneity. Cancer Metastasis Rev 2021; 40:447-476. [PMID: 33959849 PMCID: PMC9863248 DOI: 10.1007/s10555-021-09970-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/22/2021] [Indexed: 01/26/2023]
Abstract
Chemokines, a subfamily of the cell cytokines, are low molecular weight proteins known to induce chemotaxis in leukocytes in response to inflammatory and pathogenic signals. A plethora of literature demonstrates that chemokines and their receptors regulate tumor progression and metastasis. With these diverse functionalities, chemokines act as a fundamental link between the tumor cells and their microenvironment. Recent studies demonstrate that the biology of chemokines and their receptor in metastasis is complex as numerous chemokines are involved in regulating site-specific tumor growth and metastasis. Successful treatment of disseminated cancer is a significant challenge. The most crucial problem for treating metastatic cancer is developing therapy regimes capable of overcoming heterogeneity problems within primary tumors and among metastases and within metastases (intralesional). This heterogeneity of malignant tumor cells can be related to metastatic potential, response to chemotherapy or specific immunotherapy, and many other factors. In this review, we have emphasized the role of chemokines in the process of metastasis and metastatic heterogeneity. Individual chemokines may not express the full potential to address metastatic heterogeneity, but chemokine networks need exploration. Understanding the interplay between chemokine-chemokine receptor networks between the tumor cells and their microenvironment is a novel approach to overcome the problem of metastatic heterogeneity. Recent advances in the understanding of chemokine networks pave the way for developing a potential targeted therapeutic strategy to treat metastatic cancer.
Collapse
Affiliation(s)
- Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
110
|
Li D, Zhao W, Zhang X, Lv H, Li C, Sun L. NEFM DNA methylation correlates with immune infiltration and survival in breast cancer. Clin Epigenetics 2021; 13:112. [PMID: 34001208 PMCID: PMC8130356 DOI: 10.1186/s13148-021-01096-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 05/02/2021] [Indexed: 11/25/2022] Open
Abstract
Background This study aims to determine whether NEFM (neurofilament medium) DNA methylation correlates with immune infiltration and prognosis in breast cancer (BRCA) and to explore NEFM-connected immune gene signature. Methods NEFM transcriptional expression was analyzed in BRCA and normal breast tissues using Oncomine and Tumor Immune Estimation Resource (TIMER) databases. The relationship between NEFM DNA methylation and NEFM transcriptional expression was investigated in TCGA. Potential influence of NEFM DNA methylation/expression on clinical outcome was evaluated using TCGA BRCA, The Human Protein Atlas and Kaplan–Meier plotter databases. Association of NEFM transcriptional expression/DNA methylation with cancer immune infiltration was investigated using TIMER and TISIDB databases. Results High expression of NEFM correlated with better overall survival (OS) and recurrence-free survival (RFS) in TCGA BRCA and Kaplan–Meier plotter, whereas NEFM DNA methylation with worse OS in TCGA BRCA. NEFM transcriptional expression negatively correlated with DNA methylation. NEFM DNA methylation significantly negatively correlated with infiltrating levels of B, CD8+ T/CD4+ T cells, macrophages, neutrophils and dendritic cells in TIMER and TISIDB. NEFM expression positively correlated with macrophage infiltration in TIMER and TISIDB. After adjusted with tumor purity, NEFM expression weekly negatively correlated with infiltration level of B cells, whereas positively correlated with CD8+ T cell infiltration in TIMER gene modules. NEFM expression/DNA methylation correlated with diverse immune markers in TCGA and TISIDB. Conclusions NEFM low-expression/DNA methylation correlates with poor prognosis. NEFM expression positively correlates with macrophage infiltration. NEFM DNA methylation strongly negatively correlates with immune infiltration in BRCA. Our study highlights novel potential functions of NEFM expression/DNA methylation in regulation of tumor immune microenvironment. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01096-4.
Collapse
Affiliation(s)
- Dandan Li
- Department of Radiotherapy Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenhao Zhao
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China
| | - Xinyu Zhang
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China
| | - Hanning Lv
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China
| | - Chunhong Li
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China.
| | - Lichun Sun
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
111
|
Wan Y, Wang X, Liu T, Fan T, Zhang Z, Wang B, Zhang B, Tian Z, Mao T, Gong Z, Zhang L. Prognostic value of CCR2 as an immune indicator in lung adenocarcinoma: A study based on tumor-infiltrating immune cell analysis. Cancer Med 2021; 10:4150-4163. [PMID: 33949150 PMCID: PMC8209599 DOI: 10.1002/cam4.3931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/21/2022] Open
Abstract
Background Prognostic indicators in lung adenocarcinoma (LUAD) have been seeking under database analysis, and remarkable advance is on the way. Methods This study calculated the scores of stromal and immune components of the tumor microenvironment (TME) in 551 LUAD samples using the ESTIMATE algorithm on The Cancer Genome Atlas (TCGA) database. R package ''limma'' was used to selected differentially expressed genes (DEG). We have analyzed the DEGs by means of Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichments. The protein‐protein network, univariate Cox analysis, and Lasso regression analysis were performed to selected survival‐related genes. Gene Set Enrichment Analysis (GSEA) represented the enriched pathway of CC chemokine receptor 2 (CCR2). The ratios of immune cells in the TME of each LUAD sample were obtained using the R package "limma" and CIBERSORT algorithm in R 4.0.2. Results The ImmuneScore was positively correlated with prognosis regarding survival rate, T classification of TNM stages, and clinicopathological staging characteristics. GO and KEGG enrichments showed DEGs were associated with immune‐related activities. Three genes of LUAD were selected from the PPI network and Cox proportional hazards regression analysis. CCR2 was the most survival correlated gene by Lasso regression analysis. GSEA results showed that C2 kegg gene sets in the CCR2 high‐expression group were mainly enriched in the B cell or T cell receptor signaling pathway and natural killer cell‐mediated cytotoxicity. Correlation of CCR2 expression with prognosis was conducted, implicating a positive correlation with the prognosis of survival rate and M classification, negative correlation with the prognosis of T and N classifications. The correlation between CCR2 and tumor‐infiltrating immune cells (TICs) was analyzed, and 14 kinds of TICs were found closely correlated with CCR2 expression through difference analysis. Conclusion Therefore, CCR2 has prognostic value as an immune indicator in LUAD.
Collapse
Affiliation(s)
- Yi Wan
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Xin Wang
- School of Stomatology, Qingdao University, Qingdao, Shandong Province, China
| | - Ting Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Tianyu Fan
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Zugui Zhang
- Value Institute, Christiana Care Health System, Newark, DE, USA
| | - Bin Wang
- Department of Specialty Medicine, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Bei Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Tao Mao
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Zheng Gong
- Sino-Cellbiomed Institutes of Medical Cell & Pharmaceutical Proteins, Qingdao University, Qingdao, Shandong Province, China
| | - Li Zhang
- Department of Immunology, School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
112
|
Fang J, Yang Z, Xie J, Li Z, Hu C, Yang M, Zhou X. Identification and validation of autophagy-related prognostic signature for head and neck squamous cell carcinoma. Transl Oncol 2021; 14:101094. [PMID: 33878525 PMCID: PMC8080081 DOI: 10.1016/j.tranon.2021.101094] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 01/07/2023] Open
Abstract
Identify novel autophagy-related signature for OS and DSS in HNSCC patients. The role of autophagy in tumor immune microenvironment. Provide promising targets that can enhance the efficacy of cancer immunotherapy.
Background Many studies have demonstrated that autophagy plays a significant role in regulating tumor growth and progression. However, the effect of autophagy-related genes (ARGs) on the prognosis have rarely been analyzed in head and neck squamous cell carcinoma (HNSCC). Methods We obtained differentially expressed ARGs from HNSCC mRNA data in The Cancer Genome Atlas (TCGA) database. And then we performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses to explore the autophagy-related biological functions. The overall survival (OS)-related and disease specific survival (DSS)-related ARGs were identified by univariate Cox regression analyses. With these genes, we established OS-related and DSS-related risk signature by LASSO regression method, respectively. We validated the reliability of the risk signature with receiver operating characteristic (ROC) analysis, Kaplan-Meier survival curves, clinical correlation analysis, and nomogram. Then we analyzed relationships between risk signature and immune cell infiltration. Results We established the prognostic signatures based on 14 ARGs for OS and 12 ARGs for DSS. The ROC curves, survival analysis, and nomogram validated the predictive accuracy of the models. Clinic correlation analysis showed that the risk group was closely related to Stage, pathological T stage, pathological N stage and human papilloma virus (HPV) subtype. Cox regression demonstrated that the risk score was an independent predictor for the prognosis of HNSCC patients. Furthermore, patients in low-risk score group exhibited higher immunescore and distinct immune cell infiltration than high-risk score group. And we further analysis revealed that the copy number alterations (CNAs) of ARGs-based signature affected the abundance of tumor-infiltrating immune cells. Conclusion In this study, we identified novel autophagy-related signature for the prediction of OS and DSS in patients with HNSCC. Meanwhile, our study provides a novel sight to understand the role of autophagy and elucidate the important role of autophagy in tumor immune microenvironment (TIME) of HNSCC.
Collapse
Affiliation(s)
- Jiayu Fang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Zhiqiang Yang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing Xie
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430000, China
| | - Ziang Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chang Hu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Minlan Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430000, China.
| | - Xuhong Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430000, China.
| |
Collapse
|
113
|
Fractionated Radiation Severely Reduces the Number of CD8+ T Cells and Mature Antigen Presenting Cells Within Lung Tumors. Int J Radiat Oncol Biol Phys 2021; 111:272-283. [PMID: 33865948 DOI: 10.1016/j.ijrobp.2021.04.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/12/2021] [Accepted: 04/08/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE The combination of standard-of-care radiation therapy (RT) with immunotherapy is moving to the mainstream of non-small cell lung cancer treatment. Multiple preclinical studies reported on the CD8+ T cell stimulating properties of RT, resulting in abscopal therapeutic effects. A literature search demonstrates that most preclinical lung cancer studies applied subcutaneous lung tumor models. Hence, in-depth immunologic evaluation of clinically relevant RT in orthotopic lung cancer models is lacking. METHODS AND MATERIALS We studied the therapeutic and immunologic effects of low-dose fractionated RT on lungs from C57BL/6 mice, challenged 2 weeks before with firefly luciferase expressing Lewis lung carcinoma cells via the tail vein. Low-dose fractionation was represented by 4 consecutive daily fractions of image guided RT at 3.2 Gy. RESULTS We showed reduced lung tumor growth upon irradiation using in vivo bioluminescence imaging and immunohistochemistry. Moreover, significant immunologic RT-induced changes were observed in irradiated lungs and in the periphery (spleen and blood). First, a significant decrease in the number of CD8+ T cells and trends toward more CD4+ and regulatory T cells were seen after RT in all evaluated tissues. Notably, only in the periphery did the remaining CD8+ T cells show a more activated phenotype. In addition, a significant expansion of neutrophils and monocytes was observed upon RT locally and systemically. Locally, RT increased the influx of tumor-associated macrophages and conventional type 2 dendritic cells, whereas the alveolar macrophages and conventional type 1 DCs dramatically decreased. Functionally, these antigen-presenting cells severely reduced their CD86 expression, suggesting a reduced capacity to induce potent immunity. CONCLUSIONS Our results imply that low-dose fractionated RT of tumor-bearing lung tissue shifts the immune cell balance toward an immature myeloid cell dominating profile. These data argue for myeloid cell repolarizing strategies to enhance the abscopal effects in patients with non-small cell lung cancer treated with fractionated RT.
Collapse
|
114
|
Wang Y, Gao R, Li J, Tang S, Li S, Tong Q, Li S. Downregulation of hsa_circ_0074854 Suppresses the Migration and Invasion in Hepatocellular Carcinoma via Interacting with HuR and via Suppressing Exosomes-Mediated Macrophage M2 Polarization. Int J Nanomedicine 2021; 16:2803-2818. [PMID: 33880025 PMCID: PMC8052130 DOI: 10.2147/ijn.s284560] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/11/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) have been identified as key factors in the development of hepatocellular carcinoma (HCC). However, the role and potential molecular mechanism of circRNAs in HCC remain largely unclear. In addition, exosomes are known as important messengers of the cross-talk between tumor cells and immune cells, while the role of extracellular circRNAs in the cell-to-cell communication of tumor cells and immune cells remains not unclear. METHODS The level of hsa_circ_0074854 in HCC cell lines and HCC cell-derived exosomes was assessed using RT-qPCR assay. In addition, CCK-8 and transwell assays were used to determine the viability, migration and invasion of HCC cells. RESULTS Hsa_circ_0074854 expression was upregulated in HCC tissues and HCC cell lines. Additionally, hsa_circ_0074854 knockdown was found to inhibit HCC growth in vitro and in vivo. Mechanistically, hsa_circ_0074854 knockdown inhibited the migration and invasion of HCC cells via interacting with human antigen R (HuR) to reduce its stability. Furthermore, hsa_circ_0074854 can be transferred from HCC cells to macrophages via exosomes. Exosomes with downregulated hsa_circ_0074854 suppressed macrophage M2 polarization, which in turn suppressing migration and invasion of HCC cells both in vitro and in vivo. CONCLUSION Downregulation of hsa_circ_0074854 suppresses the migration and invasion in hepatocellular carcinoma via interacting with HuR and via suppressing exosomes-mediated macrophage M2 polarization. Collectively, these findings may help to understand the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Rongfen Gao
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People’s Republic of China
| | - Jinpeng Li
- Department of Thyroid and Breast Surgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, Hubei, People’s Republic of China
| | - Shaotao Tang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Shuai Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| | - Shiwang Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People’s Republic of China
| |
Collapse
|
115
|
Haimour A, Abu-Shawer O, Abu-Shawer M, Al-Taji A, Altamimi T, Mansour R, Amarin JZ, Sultan H, Al-Hussaini M. The clinical potential of circulating immune cell counts in primary gastric lymphoma. J Gastrointest Oncol 2021; 12:365-376. [PMID: 34012632 DOI: 10.21037/jgo-20-383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background High neutrophil-lymphocyte ratio (NLR) is linked to poor overall survival (OS) in gastrointestinal tract cancers. This study explores the clinical value of NLR, in addition to absolute lymphocyte count (ALC) and other hematologic parameters in association with distant metastases and OS in primary gastric lymphoma (PGL) patients. Methods Clinical data of 139 PGL patients who received treatment at King Hussein Cancer Center (KHCC), Amman-Jordan were retrospectively evaluated. Using data from complete blood count (CBC) tests, the following hematologic parameters: absolute neutrophil count (ANC), ALC, absolute eosinophil count (AEC), absolute monocyte count (AMC), NLR, platelet-lymphocyte ratio (PLR), and monocyte-lymphocyte ratio (MLR) were assessed in association with the following clinical outcomes: presence or absence of baseline distant metastases and OS. We conducted univariate and multivariate analyses assessing the various hematologic parameters in association with distant metastases. Results Univariate and multivariate analyses indicated that patients with an elevated NLR (>3.14) displayed more baseline distant metastases compared to patients with a low NLR (≤3.14), (P value: 0.02 and 0.018, respectively). High baseline ALC (>1,819/µL) was associated with lower baseline distant metastases (P value: 0.04). In the OS analysis, high baseline ANC (>5,100/µL), NLR (>2.75), and PLR (>0.16) were associated with poor OS, (P value: 0.027, 0.016, and 0.011 respectively). Conclusions High NLR and ALC were associated with baseline distant metastases. High baseline ANC, NLR, and PLR were associated with poor OS. Hematologic parameters might be potentially helpful in assessing and correlating NLR with the response success to treatment in PGL.
Collapse
Affiliation(s)
- Ayman Haimour
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | | | | | - Ali Al-Taji
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Tamer Altamimi
- Internal Medicine Department, Rochester General Hospital, NY, USA
| | - Razan Mansour
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| | - Justin Z Amarin
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| | - Hala Sultan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Maysa Al-Hussaini
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
116
|
Sedighzadeh SS, Khoshbin AP, Razi S, Keshavarz-Fathi M, Rezaei N. A narrative review of tumor-associated macrophages in lung cancer: regulation of macrophage polarization and therapeutic implications. Transl Lung Cancer Res 2021; 10:1889-1916. [PMID: 34012800 PMCID: PMC8107755 DOI: 10.21037/tlcr-20-1241] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is the deadliest malignancy worldwide. An inflammatory microenvironment is a key factor contributing to lung tumor progression. Tumor-Associated Macrophages (TAMs) are prominent components of the cancer immune microenvironment with diverse supportive and inhibitory effects on growth, progression, and metastasis of lung tumors. Two main macrophage phenotypes with different functions have been identified. They include inflammatory or classically activated (M1) and anti-inflammatory or alternatively activated (M2) macrophages. The contrasting functions of TAMs in relation to lung neoplasm progression stem from the presence of TAMs with varying tumor-promoting or anti-tumor activities. This wide spectrum of functions is governed by a network of cytokines and chemokines, cell-cell interactions, and signaling pathways. TAMs are promising therapeutic targets for non-small cell lung cancer (NSCLC) treatment. There are several strategies for TAM targeting and utilizing them for therapeutic purposes including limiting monocyte recruitment and localization through various pathways such as CCL2-CCR2, CSF1-CSF1R, and CXCL12-CXCR4, targeting the activation of TAMs, genetic and epigenetic reprogramming of TAMs to antitumor phenotype, and utilizing TAMs as the carrier for anti-cancer drugs. In this review, we will outline the role of macrophages in the lung cancer initiation and progression, pathways regulating their function in lung cancer microenvironment as well as the role of these immune cells in the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Sahar Sadat Sedighzadeh
- Department of Biological Sciences, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amin Pastaki Khoshbin
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK
| |
Collapse
|
117
|
Wang K, Jiang L, Hu A, Sun C, Zhou L, Huang Y, Chen Q, Dong J, Zhou X, Zhang F. Vertebral-specific activation of the CX3CL1/ICAM-1 signaling network mediates non-small-cell lung cancer spinal metastasis by engaging tumor cell-vertebral bone marrow endothelial cell interactions. Am J Cancer Res 2021; 11:4770-4789. [PMID: 33754027 PMCID: PMC7978319 DOI: 10.7150/thno.54235] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Rationale: The spine is one of the most common metastatic sites of non-small cell lung cancer (NSCLC), and NSCLC spinal metastasis results in serious consequences. Metastatic extravasation of disseminated cancer cells including increased invasiveness, adhesion and transendothelial migration is crucial for tumor metastasis. This study aimed to investigate the mechanisms underlying NSCLC spinal metastasis based on the C-X3-C motif chemokine ligand 1- (CX3CL1) and intercellular adhesion molecule-1- (ICAM-1) mediated signaling network. Methods: Immunohistochemistry, western blotting, and reverse transcription-quantitative PCR were conducted to detect the distribution of CX3CL1/ICAM-1 in different organs. Transwell, adhesion, and transendothelial migration assays were performed to evaluate the regulatory effects of CX3CL1/ICAM-1 on NSCLC cell invasion, adhesion, and transendothelial migration in vitro. A spontaneous spinal metastasis mouse model was established via injection of NSCLC cells into the left cardiac ventricle of NOD/SCID mice. The effects of CX3CL1/ICAM-1 on NSCLC spinal metastasis in vivo were validated using bioluminescent, micro-computerized tomography, immunohistochemistry and histological analyses. Results: CX3CL1 expression was specifically higher in vertebral bone compared with limb bones and lung tissue, and was associated with NSCLC spinal metastasis. Mechanically, vertebral bone marrow endothelial cells (VBMECs) enhanced NSCLC cell invasion via CX3CL1 signaling-mediated activation of the PI3K/AKT pathway. Furthermore, we found that VBMECs effectively induced ICAM-1-dependent NSCLC cell adhesion in coordination with platelets through the CX3CL1/ICAM-1/LFA-1 pathway. Meanwhile, CX3CL1 enhanced NSCLC cell transendothelial migration by increasing permeability of VBMECs via ICAM-1-dependent activation of the Src/GEF-H1 pathway. Interestingly, NSCLC cells were indicated to promote CX3CL1 secretion of VBMECs through MAPK14/ADMA17-dependent CX3CL1 release and NF-κB-dependent CX3CL1 synthesis. Based on these findings, we revealed a novel feedback cycle between circulating NSCLC cells and VBMECs mediated by CX3CL1/ICAM-1 signaling. Further disengagement of the CX3CL1/ICAM-1-mediated feedback cycle in vivo significantly restricted metastasis and prolonged mouse survival. Conclusions: Our results indicated a unique feedback cycle between circulating NSCLC cells and VBMECs mediated by CX3CL1/ICAM-1 signaling, which is necessary for NSCLC spinal metastasis. This work provides a new perspective for underlying the mechanisms of NSCLC spinal metastasis and indicates potential novel targets for the prevention of NSCLC spinal metastasis.
Collapse
|
118
|
Xiang X, Wang J, Lu D, Xu X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther 2021; 6:75. [PMID: 33619259 PMCID: PMC7900181 DOI: 10.1038/s41392-021-00484-9] [Citation(s) in RCA: 512] [Impact Index Per Article: 128.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/30/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023] Open
Abstract
The current treatment strategies in advanced malignancies remain limited. Notably, immunotherapies have raised hope for a successful control of these advanced diseases, but their therapeutic responses are suboptimal and vary considerably among individuals. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are often correlated with poor prognosis and therapy resistance, including immunotherapies. Thus, a deeper understanding of the complex roles of TAMs in immunotherapy regulation could provide new insight into the TME. Furthermore, targeting of TAMs is an emerging field of interest due to the hope that these strategies will synergize with current immunotherapies. In this review, we summarize recent studies investigating the involvement of TAMs in immune checkpoint inhibition, tumor vaccines and adoptive cell transfer therapies, and discuss the therapeutic potential of targeting TAMs as an adjuvant therapy in tumor immunotherapies.
Collapse
Affiliation(s)
- Xiaonan Xiang
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
119
|
Xu T, Gao S, Liu J, Huang Y, Chen K, Zhang X. MMP9 and IGFBP1 Regulate Tumor Immune and Drive Tumor Progression in Clear Cell Renal Cell Carcinoma. J Cancer 2021; 12:2243-2257. [PMID: 33758602 PMCID: PMC7974879 DOI: 10.7150/jca.48664] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 01/13/2021] [Indexed: 01/20/2023] Open
Abstract
Immunotherapy is a novel approach and has been used in various diseases, especially in cancers. Recently, immunotherapy has gradually been used to treat advanced clear cell renal cell carcinoma (ccRCC) or metastatic ccRCC. However, the efficacy of immunotherapy is not satisfying due to the influence of the tumor microenvironment. In this study, we mainly focused on the abundance and function of tumor-infiltrating immune cells (TIICs). Monocyte and TNM stage were identified as independent prognostic factors via CIBERSORT and Cox regression analysis. Then, ccRCC patients were divided into high risk/TNMhighMonocyteslow cluster and low risk/TNMlowMonocyteshigh cluster. Further differential gene analysis, protein-protein interaction (PPI) network, and survival analysis screened nine hub genes between the above two clusters. MMP9 and IGFBP1 were selected for further study through sample validation. Moreover, gene set enrichment analysis revealed that MMP9 and IGFBP1 were involved in tumor immune via mediating cell surface receptor signal pathway, cytokine production pathway, or monocyte signal pathway. In conclusion, these findings suggested that monocyte acted as a protective factor and MMP9/IGFBP1 played a vital role in tumor immune, which might become potential novel biomarkers and therapeutic targets for immunotherapy in ccRCC.
Collapse
Affiliation(s)
- Tianbo Xu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
| | - Su Gao
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
- Institute of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
| | - Jingchong Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
| | - Yu Huang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
| | - Ke Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan 430022, China
| |
Collapse
|
120
|
Zha H, Jiang Y, Wang X, Shang J, Wang N, Yu L, Zhao W, Li Z, An J, Zhang X, Chen H, Zhu B, Li Z. Non-canonical PD-1 signaling in cancer and its potential implications in clinic. J Immunother Cancer 2021; 9:jitc-2020-001230. [PMID: 33593825 PMCID: PMC7888367 DOI: 10.1136/jitc-2020-001230] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death 1 (PD-1)-based immunotherapy has revolutionized the treatment of various cancers. However, only a certain group of patients benefit from PD-1 blockade therapy and many patients succumb to hyperprogressive disease. Although, CD8 T cells and conventional T cells are generally considered to be the primary source of PD-1 in cancer, accumulating evidence suggests that other distinct cell types, including B cells, regulatory T cells, natural killer cells, dendritic cells, tumor-associated macrophages and cancer cells, also express PD-1. Hence, the response of patients with cancer to PD-1 blockade therapy is a cumulative effect of anti-PD-1 antibodies acting on a myriad of cell types. Although, the contribution of CD8 T cells to PD-1 blockade therapy has been well-established, recent studies also suggest the involvement of non-canonical PD-1 signaling in blockade therapy. This review discusses the role of non-canonical PD-1 signaling in distinct cell types and explores how the available knowledge can improve PD-1 blockade immunotherapy, particularly in identifying novel biomarkers and combination treatment strategies.
Collapse
Affiliation(s)
- Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Ying Jiang
- Postgraduate Training Base in Rocket Army Special Medical Center of the PLA, Jinzhou Medical University, Jinzhou, P.R. China
| | - Xi Wang
- Otorhinolaryngology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Jin Shang
- Department of Health Service, Guard Bureau of the Joint Staff Department, Central Military Commission of PLA, Beijing, P.R. China
| | - Ning Wang
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Lei Yu
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Wei Zhao
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Zhihua Li
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Juan An
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Xiaochun Zhang
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Huoming Chen
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Zhaoxia Li
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| |
Collapse
|
121
|
Abstract
Mycobacterium tuberculosis, the pathogen of tuberculosis (TB), can survive in host macrophages and induce macrophages to M2 phenotype might result in latent MTB infection. During the latent phase, the expression of MTB heat-shock protein 16.3 (Hsp16.3) is markedly increased among most of bacterial proteins, but the role of Hsp16.3 in macrophage M2 polarization is not clear. In this work, we found that macrophages incubated with 100 ng/ml MTB Hsp16.3 increased the production of Arg-1, IL-10, TGF-beta, and CD206. These results showed that MTB Hsp16.3 may induce macrophage M2 phenotype. And the interaction of Hsp16.3 with macrophages was found to depend on chemokine receptors CCRL2 and CX3CR1. Additionally, we used overexpression and silencing techniques to further verify the effect of CCRL2 and CX3CR1 on MTB Hsp16.3-induced M2 polarization macrophages. Furthermore, we explored the downstream signaling molecules of CCRL2 and CX3CR1 and we found MTB Hsp16.3 altered the signal transduction of the AKT/ERK/p38-MAPK. Taken together, this study provides evidence that MTB Hsp16.3 promotes macrophages to M2 phenotype and explores its underlying mechanism.
Collapse
|
122
|
Salazar Y, Zheng X, Brunn D, Raifer H, Picard F, Zhang Y, Winter H, Guenther S, Weigert A, Weigmann B, Dumoutier L, Renauld JC, Waisman A, Schmall A, Tufman A, Fink L, Brüne B, Bopp T, Grimminger F, Seeger W, Pullamsetti SS, Huber M, Savai R. Microenvironmental Th9 and Th17 lymphocytes induce metastatic spreading in lung cancer. J Clin Invest 2021; 130:3560-3575. [PMID: 32229721 DOI: 10.1172/jci124037] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/24/2020] [Indexed: 01/10/2023] Open
Abstract
Immune microenvironment plays a critical role in lung cancer control versus progression and metastasis. In this investigation, we explored the effect of tumor-infiltrating lymphocyte subpopulations on lung cancer biology by studying in vitro cocultures, in vivo mouse models, and human lung cancer tissue. Lymphocyte conditioned media (CM) induced epithelial-mesenchymal transition (EMT) and migration in both primary human lung cancer cells and cell lines. Correspondingly, major accumulation of Th9 and Th17 cells was detected in human lung cancer tissue and correlated with poor survival. Coculturing lung cancer cells with Th9/Th17 cells or exposing them to the respective CM induced EMT in cancer cells and modulated the expression profile of genes implicated in EMT and metastasis. These features were reproduced by the signatory cytokines IL-9 and IL-17, with gene regulatory profiles evoked by these cytokines partly overlapping and partly complementary. Coinjection of Th9/Th17 cells with tumor cells in WT, Rag1-/-, Il9r-/-, and Il17ra-/- mice altered tumor growth and metastasis. Accordingly, inhibition of IL-9 or IL-17 cytokines by neutralizing antibodies decreased EMT and slowed lung cancer progression and metastasis. In conclusion, Th9 and Th17 lymphocytes induce lung cancer cell EMT, thereby promoting migration and metastatic spreading and offering potentially novel therapeutic strategies.
Collapse
Affiliation(s)
- Ylia Salazar
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Xiang Zheng
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - David Brunn
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Hartmann Raifer
- Institute for Medical Microbiology and.,CoreFacility Flow Cytometry, University of Marburg, Marburg, Germany
| | | | | | - Hauke Winter
- Translational Research Unit, Thoraxklinik at Heidelberg University, member of the DZL, Heidelberg, Germany
| | - Stefan Guenther
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Benno Weigmann
- Department of Medicine 1, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Laure Dumoutier
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | | | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Anja Schmall
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Amanda Tufman
- Respiratory Medicine and Thoracic Oncology, Internal Medicine V, Ludwig-Maximilians-University of Munich and Thoracic Oncology Centre, member of the DZL, Munich, Germany
| | - Ludger Fink
- Institute of Pathology and Cytology, Wetzlar, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany. Research Center for Immunotherapy and University Medical Center, Johannes Gutenberg-University, Mainz, Germany. German Cancer Consortium, Heidelberg, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany.,Institute or Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany
| | | | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, member of the German Center for Lung Research (DZL), member of Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.,Department of Internal Medicine, member of the DZL, member of CPI, Justus Liebig University, Giessen, Germany.,Institute or Lung Health (ILH), Justus Liebig University, Giessen, Germany
| |
Collapse
|
123
|
Bányai L, Trexler M, Kerekes K, Csuka O, Patthy L. Use of signals of positive and negative selection to distinguish cancer genes and passenger genes. eLife 2021; 10:e59629. [PMID: 33427197 PMCID: PMC7877913 DOI: 10.7554/elife.59629] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/10/2021] [Indexed: 12/14/2022] Open
Abstract
A major goal of cancer genomics is to identify all genes that play critical roles in carcinogenesis. Most approaches focused on genes positively selected for mutations that drive carcinogenesis and neglected the role of negative selection. Some studies have actually concluded that negative selection has no role in cancer evolution. We have re-examined the role of negative selection in tumor evolution through the analysis of the patterns of somatic mutations affecting the coding sequences of human genes. Our analyses have confirmed that tumor suppressor genes are positively selected for inactivating mutations, oncogenes, however, were found to display signals of both negative selection for inactivating mutations and positive selection for activating mutations. Significantly, we have identified numerous human genes that show signs of strong negative selection during tumor evolution, suggesting that their functional integrity is essential for the growth and survival of tumor cells.
Collapse
Affiliation(s)
- László Bányai
- Institute of Enzymology, Research Centre for Natural SciencesBudapestHungary
| | - Maria Trexler
- Institute of Enzymology, Research Centre for Natural SciencesBudapestHungary
| | - Krisztina Kerekes
- Institute of Enzymology, Research Centre for Natural SciencesBudapestHungary
| | - Orsolya Csuka
- Department of Pathogenetics, National Institute of OncologyBudapestHungary
| | - László Patthy
- Institute of Enzymology, Research Centre for Natural SciencesBudapestHungary
| |
Collapse
|
124
|
Yuanyuan L, Yakun L, Zhongyao L, Le Y, Weisong D, Moran G, Hui B, Chunyan L. Myeloid NEMO deficiency promotes tumor immunosuppression partly via MCP1-CCR2 axis. Exp Cell Res 2021; 399:112467. [PMID: 33428904 DOI: 10.1016/j.yexcr.2020.112467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 11/19/2022]
Abstract
Tumor-associated macrophages (TAM), which are found in the tumor microenvironment of solid tumors, not only mediate cancer immune evasion but also promote tumor growth. The transcription factor NF-κB, which is a crucial link between inflammation and tumors, can accelerate tumor occurrence and development. NEMO, the regulatory subunit of the IKK complex, plays a pivotal role in activating the NF-κB signaling pathway. However, the function of myeloid NEMO in the tumor microenvironment remains unclear. Here, we found that conditional knockout of NEMO in myeloid cells promoted tumor growth in a transplanted cancer mouse model. In Nemofl/fl lyz-cre+/- mice, the deletion of Nemo in myeloid cells increased the recruitment of M2 macrophages and myeloid-derived suppressor cells (MDSCs) into the tumor, reduced the expression of apoptosis-related proteins, and upregulated the expression of the chemokine receptor CCR2, thereby promoting tumor growth in vivo. Then, we showed that blocking the MCP1-CCR2 pathway could inhibit tumor growth, especially in mice with myeloid NEMO deletion. In this study, we examined the mechanism of NEMO in myeloid cells and explored the role of NEMO in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Li Yuanyuan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Liu Yakun
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Li Zhongyao
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Yi Le
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Duan Weisong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China; Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Guo Moran
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Bu Hui
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China; Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China
| | - Li Chunyan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People's Republic of China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, 050000, People's Republic of China; Institute of Cardiocerebrovascular Disease, Shijiazhuang, Hebei, 050000, People's Republic of China.
| |
Collapse
|
125
|
Interferon Regulatory Factor 9 Promotes Lung Cancer Progression via Regulation of Versican. Cancers (Basel) 2021; 13:cancers13020208. [PMID: 33430083 PMCID: PMC7827113 DOI: 10.3390/cancers13020208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Lung cancer is the leading cause of cancer-related deaths worldwide, accounting for more than 1.6 million deaths per year. The tumor microenvironment (TME) has been shown to play a crucial role in tumor progression and metastasis, and transcription factors link TME signaling to oncogenesis. Type I interferons (IFNs) are strong immune modulators that possess antiproliferative and proapoptotic properties. In this study, we investigated the role of the transcription factor interferon regulatory factor 9 (IRF9) in the IFN pathway in lung cancer. We performed in vitro and in vivo experiments to reveal the oncogenic properties of IRF9, which was highly upregulated in lung adenocarcinoma. For the first time, we showed that IRF9 binds to the promoter of the known oncogene versican, regulates its expression, and thereby promotes oncogenic activity. Abstract Transcription factors can serve as links between tumor microenvironment signaling and oncogenesis. Interferon regulatory factor 9 (IRF9) is recruited and expressed upon interferon stimulation and is dependent on cofactors that exert in tumor-suppressing or oncogenic functions via the JAK-STAT pathway. IRF9 is frequently overexpressed in human lung cancer and is associated with decreased patient survival; however, the underlying mechanisms remain to be elucidated. Here, we used stably transduced lung adenocarcinoma cell lines (A549 and A427) to overexpress or knockdown IRF9. Overexpression led to increased oncogenic behavior in vitro, including enhanced proliferation and migration, whereas knockdown reduced these effects. These findings were confirmed in vivo using lung tumor xenografts in nude mice, and effects on both tumor growth and tumor mass were observed. Using RNA sequencing, we identified versican (VCAN) as a novel downstream target of IRF9. Indeed, IRF9 and VCAN expression levels were found to be correlated. We showed for the first time that IRF9 binds at a newly identified response element in the promoter region of VCAN to regulate its transcription. Using an siRNA approach, VCAN was found to enable the oncogenic properties (proliferation and migration) of IRF9 transduced cells, perhaps with CDKN1A involvement. The targeted inhibition of IRF9 in lung cancer could therefore be used as a new treatment option without multimodal interference in microenvironment JAK-STAT signaling.
Collapse
|
126
|
Rivas-Fuentes S, Salgado-Aguayo A, Arratia-Quijada J, Gorocica-Rosete P. Regulation and biological functions of the CX3CL1-CX3CR1 axis and its relevance in solid cancer: A mini-review. J Cancer 2021; 12:571-583. [PMID: 33391453 PMCID: PMC7738983 DOI: 10.7150/jca.47022] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/11/2020] [Indexed: 12/13/2022] Open
Abstract
CX3CL1 is a transmembrane protein from which a soluble form can be generated by proteolytic shedding. Membranal and soluble forms of CX3CL1 exhibit different functions, although both bind to the CX3CR1 chemokine receptor. The CX3CL1-CX3CR1 axis mediates the adhesion of leukocytes and is also involved in cell survival and recruitment of immune cell subpopulations. The function of CX3CL1 is finely tuned by cytokines and transcription factors regulating its expression and post-translational modifications. On homeostasis, the CX3CL1-CX3CR1 axis participates in the removal of damaged neurons and neurogenesis, and it is also involved on several pathological contexts. The CX3CL1-CX3CR1 axis induces several cellular responses relevant to cancer such as proliferation, migration, invasion and apoptosis resistance. In this review, we address biological aspects of this molecular axis with important therapeutic potential, emphasizing its role in cancer, one of the most prevalent chronic diseases which significantly affect the quality of life and life expectancy of patients.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- Department of Research on Biochemistry, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Alfonso Salgado-Aguayo
- Laboratory of Research on Rheumatic Diseases, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Jenny Arratia-Quijada
- Department of Biomedical Sciences, Centro Universitario de Tonalá, Universidad de Guadalajara, Tonalá Jalisco, Mexico
| | - Patricia Gorocica-Rosete
- Department of Research on Biochemistry, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| |
Collapse
|
127
|
Ge Z, Ding S. The Crosstalk Between Tumor-Associated Macrophages (TAMs) and Tumor Cells and the Corresponding Targeted Therapy. Front Oncol 2020; 10:590941. [PMID: 33224886 PMCID: PMC7670061 DOI: 10.3389/fonc.2020.590941] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor microenvironment (TME) is composed of tumor cells and surrounding non-tumor stromal cells, mainly including tumor associated macrophages (TAMs), endothelial cells, and carcinoma-associated fibroblasts (CAFs). The TAMs are the major components of non-tumor stromal cells, and play an important role in promoting the occurrence and development of tumors. Macrophages originate from bone marrow hematopoietic stem cells and embryonic yolk sacs. There is close crosstalk between TAMs and tumor cells. With the occurrence of tumors, tumor cells secrete various chemokines to recruit monocytes to infiltrate tumor tissues and further promote their M2-type polarization. Importantly, M2-like TAMs can in turn accelerate tumor growth, promote tumor cell invasion and metastasis, and inhibit immune killing to promote tumor progression. Therefore, targeting TAMs in tumor tissues has become one of the principal strategies in current tumor immunotherapy. Current treatment strategies focus on reducing macrophage infiltration in tumor tissues and reprogramming TAMs to M1-like to kill tumors. Although these treatments have had some success, their effects are still limited. This paper mainly summarized the recruitment and polarization of macrophages by tumors, the support of TAMs for the growth of tumors, and the research progress of TAMs targeting tumors, to provide new treatment strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Zhe Ge
- School of Physical Education & Health Care, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Shuzhe Ding
- School of Physical Education & Health Care, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| |
Collapse
|
128
|
Wang W, Zheng F, Lin C, Zhang A. Changes in energy metabolism and macrophage polarization: Potential mechanisms of arsenic-induced lung injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 204:110948. [PMID: 32739672 DOI: 10.1016/j.ecoenv.2020.110948] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 06/11/2023]
Abstract
Exposure to arsenic is epidemiologically associated with increased lung disease. In detailing the mechanism by which arsenic exposure leads to disease, studies have emphasized that metabolic reprogramming and immune dysfunction are related to arsenic-induced lung injury. However, the association between the mechanisms listed above is not well understood. Thus, the current study aimed to investigate the interaction of energy metabolism and macrophage polarization, by which arsenic exposure adversely induced lung injury in both in vitro and human studies. First, we confirmed a shift to glycolytic metabolism resulting from mitochondrial dysfunction. This shift was accompanied by an increase in the levels of phosphorylated PDHE1α (S293) and PDK1 and a concomitant marked increase in several key markers of the HIF-1α signaling pathway (HIF-1α, p-PKM2, GLUT1 and HK-2). In addition, utilizing an in vitro model in which lung epithelial cells are cultured with macrophages, we determined that arsenic treatment polarizes macrophages towards the M2 phenotype through lactate. In the human study, the serum lactate and TGF-β levels were higher in arsenic-exposed subjects than that in reference subjects (t= 4.50, 6.24, both p < 0.05), while FVC and FEV1 were both lower (t= 5.47, 7.59, both p < 0.05). Pearson correlation analyses showed a significant negative correlation between the serum TGF-β and lactate levels and the lung function parameters (pcorrelation<0.05). In mediation analyses, lactate and TGF-β significantly mediated 24.3% and 9.0%, respectively, of the association between arsenic and FVC (pmediation<0.05), while lactate and TGF-β significantly mediated 22.2% and 12.5%, respectively, of the association between arsenic and FEV1 (pmediation<0.05). Together, the results of the in vitro and human studies indicated that there is complex communication between metabolic reprogramming and immune dysfunction, resulting in exacerbated effects in a feedback loop with increased arsenic-induced lung damage.
Collapse
Affiliation(s)
- Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Fanyan Zheng
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Changhu Lin
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, PR China.
| |
Collapse
|
129
|
He H, Xie C, Lu X. Injectable hydrogels for anti‐tumour treatment: a review. BIOSURFACE AND BIOTRIBOLOGY 2020. [DOI: 10.1049/bsbt.2020.0020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Huan He
- Key Lab of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong University610031ChengduSichuanPeople's Republic of China
| | - Chaoming Xie
- Key Lab of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong University610031ChengduSichuanPeople's Republic of China
| | - Xiong Lu
- Key Lab of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong University610031ChengduSichuanPeople's Republic of China
| |
Collapse
|
130
|
Larionova I, Tuguzbaeva G, Ponomaryova A, Stakheyeva M, Cherdyntseva N, Pavlov V, Choinzonov E, Kzhyshkowska J. Tumor-Associated Macrophages in Human Breast, Colorectal, Lung, Ovarian and Prostate Cancers. Front Oncol 2020; 10:566511. [PMID: 33194645 PMCID: PMC7642726 DOI: 10.3389/fonc.2020.566511] [Citation(s) in RCA: 259] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/21/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are major innate immune cells that constitute up to 50% of the cell mass of human tumors. TAMs are highly heterogeneous cells that originate from resident tissue-specific macrophages and from newly recruited monocytes. TAMs' variability strongly depends on cancer type, stage, and intratumor heterogeneity. Majority of TAMs are programmed by tumor microenvironment to support primary tumor growth and metastatic spread. However, TAMs can also restrict tumor growth and metastasis. In this review, we summarized the knowledge about the role of TAMs in tumor growth, metastasis and in the response to cancer therapy in patients with five aggressive types of cancer: breast, colorectal, lung, ovarian, and prostate cancers that are frequently metastasize into distant organs resulting in high mortality of the patients. Two major TAM parameters are applied for the evaluation of TAM correlation with the cancer progression: total amount of TAMs and specific phenotype of TAMs identified by functional biomarkers. We summarized the data generated in the wide range of international patient cohorts on the correlation of TAMs with clinical and pathological parameters of tumor progression including lymphatic and hematogenous metastasis, recurrence, survival, therapy efficiency. We described currently available biomarkers for TAMs that can be measured in patients' samples (tumor tissue and blood). CD68 is the major biomarker for the quantification of total TAM amounts, while transmembrane receptors (stabilin-1, CD163, CD206, CD204, MARCO) and secreted chitinase-like proteins (YKL-39, YKL-40) are used as biomarkers for the functional TAM polarization. We also considered that specific role of TAMs in tumor progression can depend on the localization in the intratumoral compartments. We have made the conclusion for the role of TAMs in primary tumor growth, metastasis, and therapy sensitivity for breast, colorectal, lung, ovarian, and prostate cancers. In contrast to other cancer types, majority of clinical studies indicate that TAMs in colorectal cancer have protective role for the patient and interfere with primary tumor growth and metastasis. The accumulated data are essential for using TAMs as biomarkers and therapeutic targets to develop cancer-specific immunotherapy and to design efficient combinations of traditional therapy and new immunomodulatory approaches.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Gulnara Tuguzbaeva
- Department of Pathophysiology, Bashkir State Medical University, Ufa, Russia
| | - Anastasia Ponomaryova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Marina Stakheyeva
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Nadezhda Cherdyntseva
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Valentin Pavlov
- Department of Urology, Bashkir State Medical University, Ufa, Russia
| | - Evgeniy Choinzonov
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg—Hessen, Mannheim, Germany
| |
Collapse
|
131
|
Zheng X, Mansouri S, Krager A, Grimminger F, Seeger W, Pullamsetti SS, Wheelock CE, Savai R. Metabolism in tumour-associated macrophages: a quid pro quo with the tumour microenvironment. Eur Respir Rev 2020; 29:29/157/200134. [PMID: 33004525 PMCID: PMC9488699 DOI: 10.1183/16000617.0134-2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the leading cause of death from cancer worldwide. Recent studies demonstrated that the tumour microenvironment (TME) is pivotal for tumour progression, providing multiple targeting opportunities for therapeutic strategies. As one of the most abundant stromal cell types in the TME, tumour-associated macrophages (TAMs) exhibit high plasticity. Malignant cells alter their metabolic profiles to adapt to the limited availability of oxygen and nutrients in the TME, resulting in functional alteration of TAMs. The metabolic features of TAMs are strongly associated with their functional plasticity, which further impacts metabolic profiling in the TME and contributes to tumourigenesis and progression. Here, we review the functional determination of the TME by TAM metabolic alterations, including glycolysis as well as fatty acid and amino acid metabolism, which in turn are influenced by environmental changes. Additionally, we discuss metabolic reprogramming of TAMs to a tumouricidal phenotype as a potential antitumoural therapeutic strategy. Tumour-associated macrophages (TAMs) display a high level of functional plasticity and altered metabolism symbolised by high sensitivity to the surrounding tumour microenvironment. The metabolism of TAMs provides novel therapeutic opportunities to treat cancer.https://bit.ly/31OqHhe
Collapse
Affiliation(s)
- Xiang Zheng
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,These authors contributed equally
| | - Siavash Mansouri
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,These authors contributed equally
| | - Annika Krager
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Friedrich Grimminger
- Dept of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Dept of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Soni S Pullamsetti
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Dept of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, Germany
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Dept of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany .,Dept of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt, Germany
| |
Collapse
|
132
|
El-Nikhely N, Karger A, Sarode P, Singh I, Weigert A, Wietelmann A, Stiewe T, Dammann R, Fink L, Grimminger F, Barreto G, Seeger W, Pullamsetti SS, Rapp UR, Savai R. Metastasis-Associated Protein 2 Represses NF-κB to Reduce Lung Tumor Growth and Inflammation. Cancer Res 2020; 80:4199-4211. [PMID: 32816854 DOI: 10.1158/0008-5472.can-20-1158] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/05/2020] [Accepted: 07/31/2020] [Indexed: 11/16/2022]
Abstract
Although NF-κB is known to play a pivotal role in lung cancer, contributing to tumor growth, microenvironmental changes, and metastasis, the epigenetic regulation of NF-κB in tumor context is largely unknown. Here we report that the IKK2/NF-κB signaling pathway modulates metastasis-associated protein 2 (MTA2), a component of the nucleosome remodeling and deacetylase complex (NuRD). In triple transgenic mice, downregulation of IKK2 (Sftpc-cRaf-IKK2DN) in cRaf-induced tumors in alveolar epithelial type II cells restricted tumor formation, whereas activation of IKK2 (Sftpc-cRaf-IKK2CA) supported tumor growth; both effects were accompanied by altered expression of MTA2. Further studies employing genetic inhibition of MTA2 suggested that in primary tumor growth, independent of IKK2, MTA2/NuRD corepressor complex negatively regulates NF-κB signaling and tumor growth, whereas later dissociation of MTA2/NuRD complex from the promoter of NF-κB target genes and IKK2-dependent positive regulation of MTA2 leads to activation of NF-κB signaling, epithelial-mesenchymal transition, and lung tumor metastasis. These findings reveal a previously unrecognized biphasic role of MTA2 in IKK2/NF-κB-driven primary-to-metastatic lung tumor progression. Addressing the interaction between MTA2 and NF-κB would provide potential targets for intervention of tumor growth and metastasis. SIGNIFICANCE: These findings strongly suggest a prominent role of MTA2 in primary tumor growth, lung metastasis, and NF-κB signaling modulatory functions.
Collapse
Affiliation(s)
- Nefertiti El-Nikhely
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Annika Karger
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Poonam Sarode
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Indrabahadur Singh
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Astrid Wietelmann
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Reinhard Dammann
- Institute for Genetics; member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ludger Fink
- Institute of Pathology and Cytology, UEGP, Wetzlar, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Guillermo Barreto
- Institute of Molecular Oncology, German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany.,Brain and Lung Epigenetics (BLUE), Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Soni S Pullamsetti
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Ulf R Rapp
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany. .,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| |
Collapse
|
133
|
Dai X, Lu L, Deng S, Meng J, Wan C, Huang J, Sun Y, Hu Y, Wu B, Wu G, Lovell JF, Jin H, Yang K. USP7 targeting modulates anti-tumor immune response by reprogramming Tumor-associated Macrophages in Lung Cancer. Theranostics 2020; 10:9332-9347. [PMID: 32802195 PMCID: PMC7415808 DOI: 10.7150/thno.47137] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/05/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Tumor associated macrophages (TAMs) have strong plasticity and if reprogrammed, can clear tumor cells and regulate the adaptive immune system for cancer immunotherapy. Deubiquitinating enzymes (DUBs), which can remove ubiquitin (Ub) from Ub-modified substrates, have been associated with oncogenic metabolism but are not well-known for regulating TAMs repolarization. Methods: The expression of DUB related genes in macrophages (MΦs) was detected by reverse transcription-PCR. Flow cytometry and immunofluorescence were used to detect the changes of immune cells in the tumor microenvironment and spleen, including M1 (CD11b+F4/80+CD86+CD206-), and M2 (CD11b+F4/80+CD86-CD206+) MΦs, and IFN-γ+CD8+T cells. A proliferation assay was used to determine the effect of M2 MΦs treated with a USP7 inhibitor on T cell proliferation. Western blotting was used to detect the expression of USP7 and the activation of the MAPK pathway. The TGCA database was used to assess the role of USP7 in the immune microenvironment of human lung adenocarcinoma (LUAD). Results: 51 DUB genes were screened and USP7 was identified as a highly expressed gene in M2 but not M1 MΦs. Specific silencing of USP7 using siRNA or USP7 inhibitors led to phenotypical and functional changes in M2 MΦs, favoring CD8+T cells proliferation in vitro. USP7 inhibitors delayed tumor growth in mice with Lewis lung carcinoma, and promoted tumor infiltration of M1 MΦs and IFN-γ+CD8+T cells. Depletion of TAMs attenuated these therapeutic effects. USP7 inhibition was shown to mediate MΦs reprogramming by activating the p38 MAPK pathway. Administration of USP7 inhibitors increased the expression of programmed cell death ligand 1 (PD-L1) in tumors, while blocking programmed cell death protein 1 (PD-1) provided an effective anti-tumor response. Clinical databases suggest that high expression of USP7 in LUAD was negatively correlated with innate and adaptive immunity. Conclusions: Taken together, these results provide evidence to suggest that therapeutic approaches targeting USP7, in combination with immunotherapy, should be considered for lung cancer treatment.
Collapse
|
134
|
Fibroblast Growth Factor-14 Acts as Tumor Suppressor in Lung Adenocarcinomas. Cells 2020; 9:cells9081755. [PMID: 32707902 PMCID: PMC7466013 DOI: 10.3390/cells9081755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 12/25/2022] Open
Abstract
Investigation of the molecular dynamics in lung cancer is crucial for the development of new treatment strategies. Fibroblast growth factor (FGF) 14 belongs to the FGF family, which might play a crucial role in cancer progression. We analyzed lung adenocarcinoma (LUAC) patients samples and found that FGF14 was downregulated, correlating with reduced survival and oncogenic mutation status. FGF14 overexpression in lung cancer cell lines resulted in decreased proliferation, colony formation, and migration, as well as increased expression of epithelial markers and a decreased expression of mesenchymal markers, indicating a mesenchymal to epithelial transition in vitro. We verified these findings using small interfering RNA against FGF14 and further confirmed the suppressive effect of FGF14 in a NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ immunodeficient xenograft tumor model. Moreover, FGF14 overexpressing tumor cell RNA sequencing data suggests that genes affected by FGF14 were related to the extracellular matrix, playing a role in proliferation and migration. Notably, newly identified FGF14 target genes, adenosine deaminase RNA specific B1 (ADARB1), collagen and calcium-binding epidermal growth factor domain-containing protein 1 (CCBE1), α1 chain of collagen XI (COL11A1), and mucin 16 (MUC16) expression was negatively correlated with overall survival when FGF14 was downregulated in LUAC. These findings led us to suggest that FGF14 regulates proliferation and migration in LUAC.
Collapse
|
135
|
Zheng X, Weigert A, Reu S, Guenther S, Mansouri S, Bassaly B, Gattenlöhner S, Grimminger F, Pullamsetti S, Seeger W, Winter H, Savai R. Spatial Density and Distribution of Tumor-Associated Macrophages Predict Survival in Non-Small Cell Lung Carcinoma. Cancer Res 2020; 80:4414-4425. [PMID: 32699134 DOI: 10.1158/0008-5472.can-20-0069] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/05/2020] [Accepted: 07/14/2020] [Indexed: 11/16/2022]
Abstract
The respective antitumoral and protumoral roles of M1 and M2 tumor-associated macrophages (TAM) typify the complexity of macrophage function in cancer. In lung cancer, density and topology of distinct TAM phenotypes at the tumor center (TC) versus the invasive margin (IM) are largely unknown. Here, we investigated TAM subtype density and distribution between TC and IM in human lung cancer and TAM associations with overall survival. Macrophages isolated from adjacent nontumor tissue (NM), the TC (TC-TAM), and the IM (IM-TAM) were analyzed with RNA-sequencing (RNA-seq). Lung tumor tissue microarrays from 104 patient samples were constructed. M1 and M2 TAMs were identified using multiplex immunofluorescence staining and a tumor cell-TAM proximity analysis was performed. RNA-seq identified marked differences among NM, TC-TAM, and IM-TAM. On the basis of a panel of five selected markers (CD68, IL12, CCR7, CD163, and ALOX15), M2 predominance over M1 and M2 proximity to tumor cells was observed, especially at IM. Tumor cell proximity to TAM was linked with tumor cell survival and hypoxia was associated with accumulation of M2 TAM. Notably, lower density of M1 TC-TAM and higher proximity of tumor cells to M2 IM-TAM or lower proximity to M1 IM-TAM were linked with poor survival. In addition, three novel molecules (UBXN4, MFSD12, and ACTR6) from RNA-seq served as potential prognostic markers for lung cancer, and M2 predominance and juxtaposition of M2 TAM near tumor cells were associated with poor survival. Together, our results reveal the marked heterogeneity of TAM populations in different tumor regions, with M2 TAM predominance, particularly at IM. SIGNIFICANCE: This study underlines the significance of the density, spatial distribution, and gene expression of TAM phenotypes as prognostic factors for overall survival in lung cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/20/4414/F1.large.jpg.
Collapse
Affiliation(s)
- Xiang Zheng
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Simone Reu
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Siavash Mansouri
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Birgit Bassaly
- Department of Pathology, Justus Liebig University, Giessen, Germany
| | | | - Friedrich Grimminger
- Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Hauke Winter
- Department of Thoracic Surgery, Translational Lung Research Center (TLRC) Thoraxklinik at the University Hospital Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany. .,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
136
|
Felsenstein M, Blank A, Bungert AD, Mueller A, Ghori A, Kremenetskaia I, Rung O, Broggini T, Turkowski K, Scherschinski L, Raggatz J, Vajkoczy P, Brandenburg S. CCR2 of Tumor Microenvironmental Cells Is a Relevant Modulator of Glioma Biology. Cancers (Basel) 2020; 12:cancers12071882. [PMID: 32668709 PMCID: PMC7408933 DOI: 10.3390/cancers12071882] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 07/10/2020] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) shows a high influx of tumor-associated macrophages (TAMs). The CCR2/CCL2 pathway is considered a relevant signal for the recruitment of TAMs and has been suggested as a therapeutic target in malignant gliomas. We found that TAMs of human GBM specimens and of a syngeneic glioma model express CCR2 to varying extents. Using a Ccr2-deficient strain for glioma inoculation revealed a 30% reduction of TAMs intratumorally. This diminished immune cell infiltration occurred with augmented tumor volumes likely based on increased cell proliferation. Remaining TAMs in Ccr2-/- mice showed comparable surface marker expression patterns in comparison to wildtype mice, but expression levels of inflammatory transcription factors (Stat3, Irf7, Cox2) and cytokines (Ifnβ, Il1β, Il12α) were considerably affected. Furthermore, we demonstrated an impact on blood vessel integrity, while vascularization of tumors appeared similar between mouse strains. The higher stability and attenuated leakiness of the tumor vasculature imply improved sustenance of glioma tissue in Ccr2-/- mice. Additionally, despite TAMs residing in the perivascular niche in Ccr2-/- mice, their pro-angiogenic activity was reduced by the downregulation of Vegf. In conclusion, lacking CCR2 solely on tumor microenvironmental cells leads to enhanced tumor progression, whereby high numbers of TAMs infiltrate gliomas independently of the CCR2/CCL2 signal.
Collapse
Affiliation(s)
- Matthäus Felsenstein
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Anne Blank
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Alexander D. Bungert
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Annett Mueller
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Adnan Ghori
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Irina Kremenetskaia
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Olga Rung
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Thomas Broggini
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Kati Turkowski
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Lea Scherschinski
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Jonas Raggatz
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| | - Peter Vajkoczy
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
- Department of Neurosurgery Charité, Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-560-002
| | - Susan Brandenburg
- Department of Experimental Neurosurgery Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.F.); (A.B.); (A.D.B.); (A.M.); (A.G.); (I.K.); (O.R.); (T.B.); (K.T.); (L.S.); (J.R.); (S.B.)
| |
Collapse
|
137
|
Liu Y, Ma H, Dong T, Yan Y, Sun L, Wang W. Clinical significance of expression level of CX3CL1-CX3CR1 axis in bone metastasis of lung cancer. Clin Transl Oncol 2020; 23:378-388. [PMID: 32638214 DOI: 10.1007/s12094-020-02431-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate the clinical significance of CX3 chemokine ligand 1(CX3CL1) and CX3CR1 in patients with bone metastasis from lung cancer. The expression levels of CX3CL1 and CX3CR1 mRNA and protein in primary lung cancer and lung cancer bone metastasis were detected by qRT-PCR and Western blot. METHODS One hundred patients with lung cancer were divided into a boneless metastasis group (50 patients with bone metastasis) and a bone metastasis group (50 patients without distant metastasis). The bone transfer component was graded by Soloway classification (0 to III). The expression levels of serum CX3CL1-CX3CR1 axis were detected by enzyme-linked immunosorbent assay (ELISA). RT-qPCR and Western Blot were used to verify the transfection efficiency. The scratching assay was used to detect the migration of CX3CL1 to 95-D cells after down-regulating the expression of CX3CR1. RESULTS The expression levels of CX3CL1 and CX3CR1 mRNA and protein in the primary lung cancer and lung cancer bone metastasis were significantly higher than those in the adjacent tissues (P < 0.0001). The levels of serum CX3CL1 and CX3CR1 in bone metastasis group were significantly higher than those in boneless metastasis group and healthy control group (P < 0.05). In the bone metastasis group, the levels of serum CX3CL1 and CX3CR1 were significantly positively correlated with the degree of disease progression (P < 0.01). CONCLUSION The expression level of serum CX3CL1-CX3CR1 axis is expected to be an auxiliary reference index for monitoring bone metastasis of lung cancer.
Collapse
Affiliation(s)
- Y Liu
- Test Room of Clinical Laboratory, The First Affiliated Hospital of Harbin Medical University, 199 Dongdazhi Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - H Ma
- Department of Human Resources, Xiamen Hospital of T.C.M, Xiamen, 361000, China
| | - T Dong
- Department of Obstetrics, Xiamen Hospital of T.C.M, Xiamen, 361000, China
| | - Y Yan
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - L Sun
- Ward 1, Department of Thoracic Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - W Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150001, China
| |
Collapse
|
138
|
Bölükbas DA, Datz S, Meyer-Schwickerath C, Morrone C, Doryab A, Gößl D, Vreka M, Yang L, Argyo C, van Rijt SH, Lindner M, Eickelberg O, Stoeger T, Schmid O, Lindstedt S, Stathopoulos GT, Bein T, Wagner DE, Meiners S. Organ-restricted vascular delivery of nanoparticles for lung cancer therapy. ADVANCED THERAPEUTICS 2020; 3:2000017. [PMID: 33884290 PMCID: PMC7610651 DOI: 10.1002/adtp.202000017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Indexed: 12/23/2022]
Abstract
Nanoparticle-based targeted drug delivery holds promise for treatment of cancers. However, most approaches fail to be translated into clinical success due to ineffective tumor targeting in vivo. Here, the delivery potential of mesoporous silica nanoparticles (MSN) functionalized with targeting ligands for EGFR and CCR2 is explored in lung tumors. The addition of active targeting ligands on MSNs enhances their uptake in vitro but fails to promote specific delivery to tumors in vivo, when administered systemically via the blood or locally to the lung into immunocompetent murine lung cancer models. Ineffective tumor targeting is due to efficient clearance of the MSNs by the phagocytic cells of the liver, spleen, and lung. These limitations, however, are successfully overcome using a novel organ-restricted vascular delivery (ORVD) approach. ORVD in isolated and perfused mouse lungs of Kras-mutant mice enables effective nanoparticle extravasation from the tumor vasculature into the core of solid lung tumors. In this study, ORVD promotes tumor cell-specific uptake of nanoparticles at cellular resolution independent of their functionalization with targeting ligands. Organ-restricted vascular delivery thus opens new avenues for optimized nanoparticles for lung cancer therapy and may have broad applications for other vascularized tumor types.
Collapse
Affiliation(s)
- Deniz A Bölükbas
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany; Lung Bioengineering and Regeneration, Dept of Experimental Medical Sciences, Stem Cell Centre, Wallenberg Center for Molecular Medicine, Lund University Cancer Centre (LUCC), Lund University, 22362 Lund, Sweden
| | - Stefan Datz
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU) 81377 Munich, Germany
| | - Charlotte Meyer-Schwickerath
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Carmela Morrone
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Ali Doryab
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Dorothee Gößl
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU) 81377 Munich, Germany
| | - Malamati Vreka
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany; Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, 26504 Patras, Greece
| | - Lin Yang
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Christian Argyo
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU) 81377 Munich, Germany
| | - Sabine H van Rijt
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Michael Lindner
- Center of Thoracic Surgery Munich, Asklepios Clinic Munich-Gauting, and Asklepios Biobank for Diseases of the Lung, Comprehensive Pneumology Center (CPC), Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), 82131 Gauting, Germany
| | - Oliver Eickelberg
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Tobias Stoeger
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Otmar Schmid
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Sandra Lindstedt
- Dept of Cardiothoracic Surgery, Heart and Lung Transplantation, Lund University Hospital 22242 Lund, Sweden
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany; Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, 26504 Patras, Greece
| | - Thomas Bein
- Department of Chemistry and Center for NanoScience (CeNS), University of Munich (LMU) 81377 Munich, Germany
| | - Darcy E Wagner
- Lung Bioengineering and Regeneration, Dept of Experimental Medical Sciences, Stem Cell Centre, Wallenberg Center for Molecular Medicine, Lund University Cancer Centre (LUCC), Lund University, 22362 Lund, Sweden
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital Ludwig-Maximilians University, and Helmholtz Zentrum München, Munich, Germany. Member of the German Center for Lung Research (DZL), 81377 Munich, Germany
| |
Collapse
|
139
|
He M, Yu W, Chang C, Miyamoto H, Liu X, Jiang K, Yeh S. Estrogen receptor α promotes lung cancer cell invasion via increase of and cross-talk with infiltrated macrophages through the CCL2/CCR2/MMP9 and CXCL12/CXCR4 signaling pathways. Mol Oncol 2020; 14:1779-1799. [PMID: 32356397 PMCID: PMC7400793 DOI: 10.1002/1878-0261.12701] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 02/14/2020] [Accepted: 04/24/2020] [Indexed: 12/13/2022] Open
Abstract
Data analysis of clinical samples suggests that higher estrogen receptor α (ERα) expression could be associated with worse overall survival in some patients with non‐small‐cell lung cancer (NSCLC). Immunofluorescence results further showed that higher ERα expression was linked to larger numbers of infiltrated macrophages in NSCLC tissues. However, the detailed mechanisms underlying this phenomenon remain unclear. Results from in vitro studies with multiple cell lines revealed that, in NSCLC cells, ERα can activate the CCL2/CCR2 axis to promote macrophage infiltration, M2 polarization, and MMP9 production, which can then increase NSCLC cell invasion. Mechanistic studies using chromatin immunoprecipitation and promoter luciferase assays demonstrated that ERα could bind to estrogen response elements (EREs) on the CCL2 promoter to increase CCL2 expression. Furthermore, ERα‐increased macrophage infiltration can induce a positive feedback mechanism to increase lung cancer cell ERα expression via the up‐regulation of the CXCL12/CXCR4 pathway. Targeting these newly identified pathways, NSCLC ERα‐increased macrophage infiltration or the macrophage‐to‐NSCLC CXCL12/CXCR4/ERα signal, with anti‐estrogens or CCR2/CXCR4 antagonists, may help in the development of new alternative therapies to better treat NSCLC.
Collapse
Affiliation(s)
- Miao He
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China.,George Whipple Lab for Cancer Research, Departments of Urology and Pathology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Weiwei Yu
- George Whipple Lab for Cancer Research, Departments of Urology and Pathology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Chawnshang Chang
- George Whipple Lab for Cancer Research, Departments of Urology and Pathology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Hiroshi Miyamoto
- George Whipple Lab for Cancer Research, Departments of Urology and Pathology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaohong Liu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shuyuan Yeh
- George Whipple Lab for Cancer Research, Departments of Urology and Pathology and the Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
140
|
Sarode P, Zheng X, Giotopoulou GA, Weigert A, Kuenne C, Günther S, Friedrich A, Gattenlöhner S, Stiewe T, Brüne B, Grimminger F, Stathopoulos GT, Pullamsetti SS, Seeger W, Savai R. Reprogramming of tumor-associated macrophages by targeting β-catenin/FOSL2/ARID5A signaling: A potential treatment of lung cancer. SCIENCE ADVANCES 2020; 6:eaaz6105. [PMID: 32548260 PMCID: PMC7274802 DOI: 10.1126/sciadv.aaz6105] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/27/2020] [Indexed: 05/03/2023]
Abstract
Tumor-associated macrophages (TAMs) influence lung tumor development by inducing immunosuppression. Transcriptome analysis of TAMs isolated from human lung tumor tissues revealed an up-regulation of the Wnt/β-catenin pathway. These findings were reproduced in a newly developed in vitro "trained" TAM model. Pharmacological and macrophage-specific genetic ablation of β-catenin reprogrammed M2-like TAMs to M1-like TAMs both in vitro and in various in vivo models, which was linked with the suppression of primary and metastatic lung tumor growth. An in-depth analysis of the underlying signaling events revealed that β-catenin-mediated transcriptional activation of FOS-like antigen 2 (FOSL2) and repression of the AT-rich interaction domain 5A (ARID5A) drive gene regulatory switch from M1-like TAMs to M2-like TAMs. Moreover, we found that high expressions of β-catenin and FOSL2 correlated with poor prognosis in patients with lung cancer. In conclusion, β-catenin drives a transcriptional switch in the lung tumor microenvironment, thereby promoting tumor progression and metastasis.
Collapse
Affiliation(s)
- Poonam Sarode
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
| | - Xiang Zheng
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
| | - Georgia A. Giotopoulou
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, 26504, Greece and Lung Carcinogenesis Laboratory, Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich 81377, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt 60323, Germany
| | - Carste Kuenne
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
| | - Aleksandra Friedrich
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
| | - Stefan Gattenlöhner
- Department of Pathology, Member of the DZL, Justus Liebig University, Giessen 35390, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University Marburg, Member of the DZL, Marburg 35043, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt 60323, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, 35392 Giessen, Germany
| | - Georgios T. Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, Rio, 26504, Greece and Lung Carcinogenesis Laboratory, Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), University Hospital, Ludwig-Maximilians University and Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich 81377, Germany
| | - Soni Savai Pullamsetti
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
- Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, 35392 Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
- Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, 35392 Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim 61231, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, 60596 Frankfurt am Main, Germany
- Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, 35392 Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
- Corresponding author.
| |
Collapse
|
141
|
Fractalkine/CX3CL1 in Neoplastic Processes. Int J Mol Sci 2020; 21:ijms21103723. [PMID: 32466280 PMCID: PMC7279446 DOI: 10.3390/ijms21103723] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Fractalkine/CX3C chemokine ligand 1 (CX3CL1) is a chemokine involved in the anticancer function of lymphocytes-mainly NK cells, T cells and dendritic cells. Its increased levels in tumors improve the prognosis for cancer patients, although it is also associated with a poorer prognosis in some types of cancers, such as pancreatic ductal adenocarcinoma. This work focuses on the 'hallmarks of cancer' involving CX3CL1 and its receptor CX3CR1. First, we describe signal transduction from CX3CR1 and the role of epidermal growth factor receptor (EGFR) in this process. Next, we present the role of CX3CL1 in the context of cancer, with the focus on angiogenesis, apoptosis resistance and migration and invasion of cancer cells. In particular, we discuss perineural invasion, spinal metastasis and bone metastasis of cancers such as breast cancer, pancreatic cancer and prostate cancer. We extensively discuss the importance of CX3CL1 in the interaction with different cells in the tumor niche: tumor-associated macrophages (TAM), myeloid-derived suppressor cells (MDSC) and microglia. We present the role of CX3CL1 in the development of active human cytomegalovirus (HCMV) infection in glioblastoma multiforme (GBM) brain tumors. Finally, we discuss the possible use of CX3CL1 in immunotherapy.
Collapse
|
142
|
Iurca I, Tirpe A, Zimta AA, Moldovan C, Gulei D, Slabý O, Condorelli G, Berindan-Neagoe I. Macrophages Interaction and MicroRNA Interplay in the Modulation of Cancer Development and Metastasis. Front Immunol 2020; 11:870. [PMID: 32477352 PMCID: PMC7235377 DOI: 10.3389/fimmu.2020.00870] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/15/2020] [Indexed: 12/24/2022] Open
Abstract
Advancement in cancer research has shown that the tumor microenvironment plays a crucial role in the installation, progression, and dissemination of cancer cells. Among the heterogeneous panel of cells within the malignant microenvironment are tumor-associated macrophages that are sustaining the malignant cells through strict feedback mechanisms and spatial distribution. Considering that the presence of metastasis is one of the main feature associated with decreased survival rates among patients, in the present article we briefly present the involvement of tumor-associated macrophages in the hallmarks of metastasis and their microRNA-related regulation with a focus on lung cancer in order to coordinate the vast information under one pathology. As shown, these cells have emerged as coordinators of immunosuppression, angiogenesis and lymphangiogenesis, vessel intravasation and extravasation of cancer cells, and premetastatic niche formation, transforming the macrophages in potential therapeutic targets and also prognostic markers according to their density within the tumor and polarization phenotype. An indirect therapeutic approach on tumor-associated macrophages can be also represented by regulation of microRNAs involved in their polarization and implicit oncogenic features. Examples of these microRNAs consist in the highly studied miR-21 and miR-155, but also other microRNA with less feedback in the literature: miR-1207-5p, miR-193b, miR-320a, and others.
Collapse
Affiliation(s)
- Ioana Iurca
- Tumor Biology Department, The Oncology Institute "Prof. Dr. Ion Chiricuta", Cluj-Napoca, Romania.,Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alexandru Tirpe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristian Moldovan
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ondřej Slabý
- Centre for Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk Memorial Cancer Institute, Masaryk University, Brno, Czech Republic
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Research Center for Advanced Medicine-Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta," Cluj-Napoca, Romania
| |
Collapse
|
143
|
Sarode P, Schaefer MB, Grimminger F, Seeger W, Savai R. Macrophage and Tumor Cell Cross-Talk Is Fundamental for Lung Tumor Progression: We Need to Talk. Front Oncol 2020; 10:324. [PMID: 32219066 PMCID: PMC7078651 DOI: 10.3389/fonc.2020.00324] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Regardless of the promising results of certain immune checkpoint blockers, current immunotherapeutics have met a bottleneck concerning response rate, toxicity, and resistance in lung cancer patients. Accumulating evidence forecasts that the crosstalk between tumor and immune cells takes center stage in cancer development by modulating tumor malignancy, immune cell infiltration, and immune evasion in the tumor microenvironment (TME). Cytokines and chemokines secreted by this crosstalk play a major role in cancer development, progression, and therapeutic management. An increased infiltration of Tumor-associated macrophages (TAMs) was observed in most of the human cancers, including lung cancer. In this review, we emphasize the role of cytokines and chemokines in TAM-tumor cell crosstalk in the lung TME. Given the role of cytokines and chemokines in immunomodulation, we propose that TAM-derived cytokines and chemokines govern the cancer-promoting immune responses in the TME and offer a new immunotherapeutic option for lung cancer treatment.
Collapse
Affiliation(s)
- Poonam Sarode
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Martina Barbara Schaefer
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
144
|
Dai X, Meng J, Deng S, Zhang L, Wan C, Lu L, Huang J, Hu Y, Zhang Z, Li Y, Lovell JF, Wu G, Yang K, Jin H. Targeting CAMKII to reprogram tumor-associated macrophages and inhibit tumor cells for cancer immunotherapy with an injectable hybrid peptide hydrogel. Am J Cancer Res 2020; 10:3049-3063. [PMID: 32194854 PMCID: PMC7053188 DOI: 10.7150/thno.42385] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Simultaneously targeted treatment of tumor cells and their surrounding growth-supporting immune cells is a promising strategy to reshape immunosuppressive tumor microenvironment (TME) and potentiate host innate and adaptive antitumor immune responses. Methods: We designed a series of melittin-(RADA)n hybrid peptide sequences with varying self-assembling motifs of RADA and screened out a melittin-(RADA)6 peptide that has an optimal gel-formation ability and in vitro antitumor activity. Results: The formed melittin-(RADA)6 (MR52) hydrogel scaffold could be loaded with a specific Ca2+/calmodulin-dependent protein kinase II (CAMKII) inhibitor, KN93, originally found to have both direct tumoricidal activity and macrophages-reprogramming ability, for potent immunotherapy against melanoma and hepatoma ascites in mice models. Our MR52 hydrogel has an interweaving nanofiber-like structure, possesses direct antitumor and controlled drug release properties, and promotes the enhanced intracellular uptake of loaded cargo. Compared to free KN93, the MR52-KN93 hydrogel (MRK) improved the killing effects and levels of immunogenic cell death (ICD) on tumor cells significantly. Due to the dual role of KN93, the injection of the MRK hydrogel retarded the growth of subcutaneous melanoma tumors dramatically and resulted in a high number of mature dendritic cells of draining lymph nodes, significantly enhancing the portion of cytotoxic T cells and reduced number of M2-like tumor-associated macrophages (TAMs) in tumors. Using a mouse model of malignant ascites (MAs), where traditional therapy was ineffective, we demonstrated that the MRK hydrogel treatment offered a significantly prolonged survival compared to controls. Following treatment with the MRK hydrogel, macrophages had elevated programmed cell death protein ligand-1 (PD-L1) expression, promising follow-up combined anti-PD-1 therapy that confers a cure rate of approximately 30% against MAs in mice models. Conclusion: Thus, the MRK hydrogel may serve as a prospective platform for antitumor applications.
Collapse
|
145
|
Deng S, Clowers MJ, Velasco WV, Ramos-Castaneda M, Moghaddam SJ. Understanding the Complexity of the Tumor Microenvironment in K-ras Mutant Lung Cancer: Finding an Alternative Path to Prevention and Treatment. Front Oncol 2020; 9:1556. [PMID: 32039025 PMCID: PMC6987304 DOI: 10.3389/fonc.2019.01556] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022] Open
Abstract
Kirsten rat sarcoma viral oncogene (K-ras) is a well-documented, frequently mutated gene in lung cancer. Since K-ras regulates numerous signaling pathways related to cell survival and proliferation, mutations in this gene are powerful drivers of tumorigenesis and confer prodigious survival advantages to developing tumors. These malignant cells dramatically alter their local tissue environment and in the process recruit a powerful ally: inflammation. Inflammation in the context of the tumor microenvironment can be described as either antitumor or protumor (i.e., aiding or restricting tumor progression, respectively). Many current treatments, like immune checkpoint blockade, seek to augment antitumor inflammation by alleviating inhibitory signaling in cytotoxic T cells; however, a burgeoning area of research is now focusing on ways to modulate and mitigate protumor inflammation. Here, we summarize the interplay of tumor-promoting inflammation and K-ras mutant lung cancer pathogenesis by exploring the cytokines, signaling pathways, and immune cells that mediate this process.
Collapse
Affiliation(s)
- Shanshan Deng
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Michael J Clowers
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Walter V Velasco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marco Ramos-Castaneda
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
146
|
Activated macrophages are crucial during acute PM2.5 exposure-induced angiogenesis in lung cancer. Oncol Lett 2020; 19:725-734. [PMID: 31897188 PMCID: PMC6924157 DOI: 10.3892/ol.2019.11133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 10/22/2019] [Indexed: 12/04/2022] Open
Abstract
The importance of ambient particulate matter (PM2.5) in lung cancer progression is well established; however, the precise mechanisms by which PM2.5 modulates lung cancer development have not yet been determined. The present study demonstrated increased mRNA and protein expression levels of vascular endothelial growth factor in PM2.5-induced macrophages. However, no significant changes to the expression levels of angiogenic cytokines (vascular endothelial growth factor A, matric metallopeptidase 9, basic fibroblast growth factor and platelet-derived growth factor) were observed in the Lewis lung carcinoma (LLC) cell line in response to acute PM2.5 exposure. PM2.5-induced activated macrophages were revealed to upregulate angiogenic cytokine expression following the acute exposure of LLC cells to PM2.5-induced macrophage supernatant. In vivo, the pro-angiogenic and macrophage accumulation functions of PM2.5 were supported by the establishment of a polyvinyl alcohol sponge implantation mouse model. Furthermore, PM2.5 was demonstrated to increase angiogenesis and macrophage recruitment in mice that were subcutaneously injected with LLCs. These results indicated that PM2.5 increases angiogenesis, and macrophages are crucial mediators of PM2.5-induced angiogenesis in lung cancer. These findings may provide novel insights for the development of lung cancer treatment strategies.
Collapse
|
147
|
CX3CL1 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1231:1-12. [PMID: 32060841 DOI: 10.1007/978-3-030-36667-4_1] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
CX3CL1 (Fractalkine) is a multifunctional inflammatory chemokine with a single receptor CX3CR1. The biological effects elicited by CX3CL1 on surrounding cells vary depending on a number of factors including its structure, the expression pattern of CX3CR1, and the cell type. For instance, the transmembrane form of CX3CL1 primarily serves as an adhesion molecule, but when cleaved to a soluble form, CX3CL1 predominantly functions as a chemotactic cytokine (Fig. 1.1). However, the biological functions of CX3CL1 also extend to immune cell survival and retention. The pro-inflammatory nature of CX3CR1-expressing immune cells place the CX3CL1:CX3CR1 axis as a central player in multiple inflammatory disorders and position this chemokine pathway as a potential therapeutic target. However, the emerging role of this chemokine pathway in the maintenance of effector memory cytotoxic T cell populations implicates it as a key chemokine in anti-viral and anti-tumor immunity, and therefore an unsuitable therapeutic target in inflammation. The reported role of CX3CL1 as a key regulator of cytotoxic T cell-mediated immunity is supported by several studies that demonstrate CX3CL1 as an important TIL-recruiting chemokine and a positive prognostic factor in colorectal, breast, and lung cancer. Such reports are conflicting with an overwhelming number of studies demonstrating a pro-tumorigenic and pro-metastatic role of CX3CL1 across multiple blood and solid malignancies.This chapter will review the unique structure, function, and biology of CX3CL1 and address the diversity of its biological effects in the immune system and the tumor microenvironment. Overall, this chapter highlights how we have just scratched the surface of CX3CL1's capabilities and suggests that further in-depth and mechanistic studies incorporating all CX3CL1 interactions must be performed to fully appreciate its role in cancer and its potential as a therapeutic target.
Collapse
|
148
|
Venosa A, Smith LC, Murray A, Banota T, Gow AJ, Laskin JD, Laskin DL. Regulation of Macrophage Foam Cell Formation During Nitrogen Mustard (NM)-Induced Pulmonary Fibrosis by Lung Lipids. Toxicol Sci 2019; 172:344-358. [PMID: 31428777 PMCID: PMC6876262 DOI: 10.1093/toxsci/kfz187] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nitrogen mustard (NM) is a vesicant known to target the lung, causing acute injury which progresses to fibrosis. Evidence suggests that activated macrophages contribute to the pathologic response to NM. In these studies, we analyzed the role of lung lipids generated following NM exposure on macrophage activation and phenotype. Treatment of rats with NM (0.125 mg/kg, i.t.) resulted in a time-related increase in enlarged vacuolated macrophages in the lung. At 28 days postexposure, macrophages stained positively for Oil Red O, a marker of neutral lipids. This was correlated with an accumulation of oxidized phospholipids in lung macrophages and epithelial cells and increases in bronchoalveolar lavage fluid (BAL) phospholipids and cholesterol. RNA-sequencing and immunohistochemical analysis revealed that lipid handling pathways under the control of the transcription factors liver-X receptor (LXR), farnesoid-X receptor (FXR), peroxisome proliferator-activated receptor (PPAR)-ɣ, and sterol regulatory element-binding protein (SREBP) were significantly altered following NM exposure. Whereas at 1-3 days post NM, FXR and the downstream oxidized low-density lipoprotein receptor, Cd36, were increased, Lxr and the lipid efflux transporters, Abca1 and Abcg1, were reduced. Treatment of naïve lung macrophages with phospholipid and cholesterol enriched large aggregate fractions of BAL prepared 3 days after NM exposure resulted in upregulation of Nos2 and Ptgs2, markers of proinflammatory activation, whereas large aggregate fractions prepared 28 days post NM upregulated expression of the anti-inflammatory markers, Il10, Cd163, and Cx3cr1, and induced the formation of lipid-laden foamy macrophages. These data suggest that NM-induced alterations in lipid handling and metabolism drive macrophage foam cell formation, potentially contributing to the development of pulmonary fibrosis.
Collapse
Affiliation(s)
- Alessandro Venosa
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Ley Cody Smith
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Alexa Murray
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Tanvi Banota
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| | - Jeffrey D Laskin
- Department of Environmental and Occupational Health, School of Public Health, Rutgers University, Piscataway, New Jersey 08854
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy
| |
Collapse
|
149
|
Fu C, Jiang L, Hao S, Liu Z, Ding S, Zhang W, Yang X, Li S. Activation of the IL-4/STAT6 Signaling Pathway Promotes Lung Cancer Progression by Increasing M2 Myeloid Cells. Front Immunol 2019; 10:2638. [PMID: 31798581 PMCID: PMC6863933 DOI: 10.3389/fimmu.2019.02638] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence shows that signal transducer and activator of transcription 6 (STAT6) plays critical roles in tumor development. We previously found high-level expression of STAT6 in human lung adenocarcinoma and squamous cell carcinoma, specifically in infiltrated immune cells located in the lung interstitium. Nevertheless, the role of STAT6 signaling in lung carcinogenesis and lung cancer proliferation and its underlying mechanisms remain unclear. This study aimed to investigate the role of STAT6 and the interaction between STAT6 and the tumor microenvironment in pulmonary tumorigenesis. We established a murine model of primary lung carcinogenesis in STAT6-deficient (STAT6−/−) and STAT6 wild-type (WT) BALB/c mice using the carcinogen urethane. Two-month-old male mice were intraperitoneally injected with urethane (1 g/kg) dissolved in phosphate buffered saline (PBS). Primary tumors were monitored in vivo by positron emission tomography scanning. At 4, 6, and 9 months after urethane injection, lung tumors were harvested from the STAT6−/− and WT mice for analysis. Small interfering RNA was used to downregulate the expression of STAT6 in tumor cells. Fluorescence activated cell sorting analysis was used to analyze fluorescence-conjugated cell markers. Transwell assays were used in coculturing experiments. STAT6 protein expression was detected by Western blotting, immunohistochemistry, and immunofluorescence. STAT6 mRNA expression was detected by quantitative real time-polymerase chain reaction. Cell Counting Kit-8 and colony formation assays were performed to evaluate cell proliferation. We detected high expression of STAT6 in CD11b+ cells of lung carcinoma. Our results indicate that STAT6 deficiency inhibits carcinogen-induced tumor growth and improves prognosis. STAT6 deficiency also decreased the mobilization and differentiation of CD11b+ cells. STAT6 deficiency in CD11b+ cells but not tumor cells decreased interleukin (IL)-4 secretion and the differentiation of CD11b+ cells into M2 macrophage cells. In conclusion, our findings indicate that IL-4/STAT6 signaling in CD11b+ cells promotes lung cancer progression by triggering an IL-4 positive feedback loop and increasing M2 myeloid cells. STAT6 may be a new therapeutic target for the prevention and treatment of lung cancer.
Collapse
Affiliation(s)
- Cuiping Fu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liyan Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shengyu Hao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suling Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiwei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangdong Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
150
|
de Sousa JR, Da Costa Vasconcelos PF, Quaresma JAS. Functional aspects, phenotypic heterogeneity, and tissue immune response of macrophages in infectious diseases. Infect Drug Resist 2019; 12:2589-2611. [PMID: 31686866 PMCID: PMC6709804 DOI: 10.2147/idr.s208576] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/05/2019] [Indexed: 12/13/2022] Open
Abstract
Macrophages are a functionally heterogeneous group of cells with specialized functions depending not only on their subgroup but also on the function of the organ or tissue in which the cells are located. The concept of macrophage phenotypic heterogeneity has been investigated since the 1980s, and more recent studies have identified a diverse spectrum of phenotypic subpopulations. Several types of macrophages play a central role in the response to infectious agents and, along with other components of the immune system, determine the clinical outcome of major infectious diseases. Here, we review the functions of various macrophage phenotypic subpopulations, the concept of macrophage polarization, and the influence of these cells on the evolution of infections. In addition, we emphasize their role in the immune response in vivo and in situ, as well as the molecular effectors and signaling mechanisms used by these cells. Furthermore, we highlight the mechanisms of immune evasion triggered by infectious agents to counter the actions of macrophages and their consequences. Our aim here is to provide an overview of the role of macrophages in the pathogenesis of critical transmissible diseases and discuss how elucidation of this relationship could enhance our understanding of the host-pathogen association in organ-specific immune responses.
Collapse
Affiliation(s)
- Jorge Rodrigues de Sousa
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
| | - Pedro Fernando Da Costa Vasconcelos
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
| | - Juarez Antonio Simões Quaresma
- Tropical Medicine Center, Federal University of Pará, Belém, PA, Brazil
- Evandro Chagas Institute, Ministry of Health, Ananindeua, PA, Brazil
- Center of Biological and Health Sciences, State University of Pará, Belém, PA, Brazil
- School of Medicine, São Paulo University, São Paulo, SP, Brazil
| |
Collapse
|