1
|
Araújo TG, Mota STS, Ferreira HSV, Ribeiro MA, Goulart LR, Vecchi L. Annexin A1 as a Regulator of Immune Response in Cancer. Cells 2021; 10:2245. [PMID: 34571894 PMCID: PMC8464935 DOI: 10.3390/cells10092245] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 01/01/2023] Open
Abstract
Annexin A1 is a 37 kDa phospholipid-binding protein that is expressed in many tissues and cell types, including leukocytes, lymphocytes and epithelial cells. Although Annexin A1 has been extensively studied for its anti-inflammatory activity, it has been shown that, in the cancer context, its activity switches from anti-inflammatory to pro-inflammatory. Remarkably, Annexin A1 shows pro-invasive and pro-tumoral properties in several cancers either by eliciting autocrine signaling in cancer cells or by inducing a favorable tumor microenvironment. Indeed, the signaling of the N-terminal peptide of AnxA1 has been described to promote the switching of macrophages to the pro-tumoral M2 phenotype. Moreover, AnxA1 has been described to prevent the induction of antigen-specific cytotoxic T cell response and to play an essential role in the induction of regulatory T lymphocytes. In this way, Annexin A1 inhibits the anti-tumor immunity and supports the formation of an immunosuppressed tumor microenvironment that promotes tumor growth and metastasis. For these reasons, in this review we aim to describe the role of Annexin A1 in the establishment of the tumor microenvironment, focusing on the immunosuppressive and immunomodulatory activities of Annexin A1 and on its interaction with the epidermal growth factor receptor.
Collapse
Affiliation(s)
- Thaise Gonçalves Araújo
- Laboratory of Genetics and Biotechnology, Federal University of Uberlandia, Patos de Minas 387400-128, MG, Brazil; (T.G.A.); (S.T.S.M.); (H.S.V.F.); (M.A.R.)
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil;
| | - Sara Teixeira Soares Mota
- Laboratory of Genetics and Biotechnology, Federal University of Uberlandia, Patos de Minas 387400-128, MG, Brazil; (T.G.A.); (S.T.S.M.); (H.S.V.F.); (M.A.R.)
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil;
| | - Helen Soares Valença Ferreira
- Laboratory of Genetics and Biotechnology, Federal University of Uberlandia, Patos de Minas 387400-128, MG, Brazil; (T.G.A.); (S.T.S.M.); (H.S.V.F.); (M.A.R.)
| | - Matheus Alves Ribeiro
- Laboratory of Genetics and Biotechnology, Federal University of Uberlandia, Patos de Minas 387400-128, MG, Brazil; (T.G.A.); (S.T.S.M.); (H.S.V.F.); (M.A.R.)
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil;
| | - Lara Vecchi
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlandia 38400-902, MG, Brazil;
| |
Collapse
|
2
|
Tan YQ, Li YT, Yan TF, Xu Y, Liu BH, Yang JA, Yang X, Chen QX, Zhang HB. Six Immune Associated Genes Construct Prognostic Model Evaluate Low-Grade Glioma. Front Immunol 2020; 11:606164. [PMID: 33408717 PMCID: PMC7779629 DOI: 10.3389/fimmu.2020.606164] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/18/2020] [Indexed: 12/18/2022] Open
Abstract
Background The immunotherapy of Glioma has always been a research hotspot. Although tumor associated microglia/macrophages (TAMs) proves to be important in glioma progression and drug resistance, our knowledge about how TAMs influence glioma remains unclear. The relationship between glioma and TAMs still needs further study. Methods We collected the data of TAMs in glioma from NCBI Gene Expression Omnibus (GEO) that included 20 glioma samples and 15 control samples from four datasets. Six genes were screened from the Differential Expression Gene through Gene ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein-protein interaction (PPI) network and single-cell sequencing analysis. A risk score was then constructed based on the six genes and patients' overall survival rates of 669 patients from The Cancer Genome Atlas (TCGA). The efficacy of the risk score in prognosis and prediction was verified in Chinese Glioma Genome Atlas (CGGA). Results Six genes, including CD163, FPR3, LPAR5, P2ry12, PLAUR, SIGLEC1, that participate in signal transduction and plasma membrane were selected. Half of them, like CD163, FPR3, SIGLEC1, were mainly expression in M2 macrophages. FPR3 and SIGLEC1 were high expression genes in glioma associated with grades and IDH status. The overall survival rates of the high risk score group was significantly lower than that of the low risk score group, especially in LGG. Conclusion Joint usage of the 6 candidate genes may be an effective method to diagnose and evaluate the prognosis of glioma, especially in Low-grade glioma (LGG).
Collapse
Affiliation(s)
- Yin Qiu Tan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yun Tao Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Teng Feng Yan
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bao Hui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji An Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Xue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong Bo Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Guangzhou, China
| |
Collapse
|
3
|
Application of small molecule FPR1 antagonists in the treatment of cancers. Sci Rep 2020; 10:17249. [PMID: 33057069 PMCID: PMC7560711 DOI: 10.1038/s41598-020-74350-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
Abstract
The formylpeptide receptor-1 (FPR1) is a member of the chemotactic GPCR-7TM formyl peptide receptor family, whose principle function is in trafficking of various leukocytes into sites of bacterial infection and inflammation. More recently, FPR1 has been shown to be expressed in different types of cancer and in this context, plays a significant role in their expansion, resistance and recurrence. ICT12035 is a selective and potent (30 nM in calcium mobilisation assay) small molecule FPR1 antagonist. Here, we demonstrate the efficacy of ICT12035, in a number of 2D and 3D proliferation and invasion in vitro assays and an in vivo model. Our results demonstrate that targeting FPR1 by a selective small molecule antagonist, such as ICT12035, can provide a new avenue for the treatment of cancers.
Collapse
|
4
|
Hu Y, Li Y, Wu C, Zhou L, Han X, Wang Q, Xie X, Zhou Y, Du Z. MicroRNA-140-5p inhibits cell proliferation and invasion by regulating VEGFA/MMP2 signaling in glioma. Tumour Biol 2017; 39:1010428317697558. [PMID: 28443475 DOI: 10.1177/1010428317697558] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Glioma is the most common primary malignant tumor of the central nervous system, which results in both a poor prognosis and outcome because of the aggressive progression of disease, growth and resistance to surgery, chemotherapy, and radiotherapy. MiR-140-5p is a small, non-coding single-stranded RNA molecule, which was previously studied in the settings of human tongue cancer, hepatocellular carcinoma, and colorectal cancer. However, detailed data that formally demonstrate the contribution of miR-140-5p to glioma development are missing. Similarly, relatively little is known about the relationship of miR-140-5p, vascular endothelial growth factor A, and matrix metalloproteinase-2 in glioma progression. In this study, we found that miR-140-5p expression was significantly decreased in glioma tissues and in the glioma cell-lines U87 and U251 as compared with non-cancerous brain tissues by quantitative real-time polymerase chain reaction. In addition, miR-140-5p inhibited glioma cell proliferation and invasion and promoted glioma cell apoptosis both in vivo and in vitro. Interestingly, while the expression levels of miR-140-5p were higher in glioma cells, the messenger RNA or protein expression levels of vascular endothelial growth factor A and matrix metalloproteinase-2 were lower in glioma cells as determined by quantitative real-time polymerase chain reaction, western blot assay, and immunohistochemistry. By contrast, downregulation in the expression levels of miR-140-5p augmented the messenger RNA and protein expression levels of both vascular endothelial growth factor A and matrix metalloproteinase-2. These findings suggested that miR-140-5p inhibited glioma proliferation and invasion by regulating the vascular endothelial growth factor A/matrix metalloproteinase-2 signaling pathway both in vitro and in vivo.
Collapse
Affiliation(s)
- Yuan Hu
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yanyan Li
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Chun Wu
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Liang Zhou
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiaoxiao Han
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Qingyue Wang
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xueshun Xie
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Youxin Zhou
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ziwei Du
- Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
5
|
Zhao T, Zhao C, Zhou Y, Zheng J, Gao S, Lu Y. HIF-1α binding to AEG-1 promoter induced upregulated AEG-1 expression associated with metastasis in ovarian cancer. Cancer Med 2017; 6:1072-1081. [PMID: 28401704 PMCID: PMC5430094 DOI: 10.1002/cam4.1053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/26/2017] [Accepted: 02/08/2017] [Indexed: 12/13/2022] Open
Abstract
Ovarian cancer with the highest mortality rate among gynecological malignancies is one of common cancers among female cancer patients. As reported in recent years, AEG‐1 was associated with the occurrence, development, and metastasis of ovarian cancer, but the mechanisms remain unclear. In the current study, invasion capabilities of ovarian cancer OVCAR3 cells were measured by viral infection and transwell assay. Western blot analysis was used to evaluate the expression levels of β‐catenin, E‐cadherin, MMP2, and MMP9. With qRT‐PCR analysis, AEG‐1 and HIF‐1α gene expression were detected. We used luciferase reporter gene to measure AEG‐1 promoter activity under normoxia/hypoxia in OVCAR3 cells. Our work demonstrated that AEG‐1 significantly enhanced invasion capabilities of OVCAR3 cells and the expression levels of β‐catenin, E‐cadherin, MMP2, and MMP9 associated with invasion capabilities of OVCAR3 cells were upregulated. Furthermore, hypoxia enhanced invasion capabilities of OVCAR3 cells and induced AEG‐1 high gene expression, which was reversed by AEG‐1 knockdown lentivirus. HIF‐1α expression upregulation was induced in OVCAR3 cells after hypoxia. HIF‐1α knockdown lentivirus induced downregulated expression of AEG‐1 and invasion capabilities of OVCAR3 cells were also inhibited. Wild‐type AEG‐1 promoter activity under hypoxic conditions was significantly higher than that AEG‐1 mutation under normoxic conditions in the absence of hypoxia response. Our results suggested that HIF‐1α binds to AEG‐1 promoter to upregulate its expression, which was correlated with metastasis in ovarian cancer by inducing the expression of MMP2 and MMP9 as well as inhibiting expression of E‐cadherin and β‐catenin.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chenyan Zhao
- Department of Pathology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yanting Zhou
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jing Zheng
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shujun Gao
- The Diagnosis and Treatment Center of Cervical Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yuan Lu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
6
|
Snapkov I, Öqvist CO, Figenschau Y, Kogner P, Johnsen JI, Sveinbjørnsson B. The role of formyl peptide receptor 1 (FPR1) in neuroblastoma tumorigenesis. BMC Cancer 2016; 16:490. [PMID: 27432059 PMCID: PMC4950242 DOI: 10.1186/s12885-016-2545-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 07/11/2016] [Indexed: 12/26/2022] Open
Abstract
Background Formyl peptide receptor 1 (FPR1) is a G protein-coupled receptor mainly expressed by the cells of myeloid origin, where it mediates the innate immune response to bacterial formylated peptides. High expression of FPR1 has been detected in various cancers but the function of FPR1 in tumorigenesis is poorly understood. Methods Expression of FPR1 in neuroblastoma cell lines and primary tumors was studied using RT-PCR, western blotting, immunofluorescence and immunohistochemistry. Calcium mobilization assays and western blots with phospho-specific antibodies were used to assess the functional activity of FPR1 in neuroblastoma. The tumorigenic capacity of FPR1 was assessed by xenografting of neuroblastoma cells expressing inducible FPR1 shRNA, FPR1 cDNA or control shRNA in nude mice. Results FPR1 is expressed in neuroblastoma primary tumors and cell lines. High expression of FPR1 corresponds with high-risk disease and poor patient survival. Stimulation of FPR1 in neuroblastoma cells using fMLP, a selective FPR1 agonist, induced intracellular calcium mobilization and activation of MAPK/Erk, PI3K/Akt and P38-MAPK signal transduction pathways that were inhibited by using Cyclosporin H, a selective receptor antagonist for FPR1. shRNA knock-down of FPR1 in neuroblastoma cells conferred a delayed xenograft tumor development in nude mice, whereas an ectopic overexpression of FPR1 promoted augmented tumorigenesis in nude mice. Conclusion Our data demonstrate that FPR1 is involved in neuroblastoma development and could represent a therapy option for the treatment of neuroblastoma. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2545-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Igor Snapkov
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.
| | - Carl Otto Öqvist
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Yngve Figenschau
- Endocrinology Research Group, Department of Clinical Medicine, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.,Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway.,Department of Laboratory Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Baldur Sveinbjørnsson
- Molecular Inflammation Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø, Norway
| |
Collapse
|
7
|
Shao W, Gu J, Huang C, Liu D, Huang H, Huang Z, Lin Z, Yang W, Liu K, Lin D, Ji T. Malignancy-associated metabolic profiling of human glioma cell lines using 1H NMR spectroscopy. Mol Cancer 2014; 13:197. [PMID: 25163530 PMCID: PMC4158044 DOI: 10.1186/1476-4598-13-197] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 08/21/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ambiguity in malignant transformation of glioma has made prognostic diagnosis very challenging. Tumor malignant transformation is closely correlated with specific alterations of the metabolic profile. Exploration of the underlying metabolic alterations in glioma cells of different malignant degree is therefore vital to develop metabolic biomarkers for prognosis monitoring. METHODS We conducted (1)H nuclear magnetic resonance (NMR)-based metabolic analysis on cell lines (CHG5, SHG44, U87, U118, U251) developed from gliomas of different malignant grades (WHO II and WHO IV). Several methods were applied to analyze the (1)H-NMR spectral data of polar extracts of cell lines and to identify characteristic metabolites, including principal component analysis (PCA), partial least squares discriminant analysis (PLS-DA), fuzzy c-means clustering (FCM) analysis and orthogonal projection to latent structure with discriminant analysis (OPLS-DA). The expression analyses of glial fibrillary acidic protein (GFAP) and matrix metal proteinases (MMP-9) were used to assess malignant behaviors of cell lines. GeneGo pathway analysis was used to associate characteristic metabolites with malignant behavior protein markers GFAP and MMP-9. RESULTS Stable and distinct metabolic profiles of the five cell lines were obtained. The metabolic profiles of the low malignancy grade group (CHG5, SHG44) were clearly distinguished from those of the high malignancy grade group (U87, U118, U251). Seventeen characteristic metabolites were identified that could distinguish the metabolic profiles of the two groups, nine of which were mapped to processes related to GFAP and MMP-9. Furthermore, the results from both quantitative comparison and metabolic correlation analysis indicated that the significantly altered metabolites were primarily involved in perturbation of metabolic pathways of tricarboxylic acid (TCA) cycle anaplerotic flux, amino acid metabolism, anti-oxidant mechanism and choline metabolism, which could be correlated with the changes in the glioma cells' malignant behaviors. CONCLUSIONS Our results reveal the metabolic heterogeneity of glioma cell lines with different degrees of malignancy. The obtained metabolic profiles and characteristic metabolites are closely associated with the malignant features of glioma cells, which may lay the basis for both determining the molecular mechanisms underlying glioma malignant transformation and exploiting non-invasive biomarkers for prognosis monitoring.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Donghai Lin
- Chenggong Hospital and College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | | |
Collapse
|
8
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
9
|
Differential expression of the G-protein-coupled formyl Peptide receptor in melanoma associates with aggressive phenotype. Am J Dermatopathol 2013; 35:184-90. [PMID: 23147350 DOI: 10.1097/dad.0b013e31825b2506] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Melanoma, due to its metastatic rate, is among the most aggressive forms of skin cancer. Human formyl peptide receptor (FPR) and its variant FPR-like 1 (FPRL1) have been associated with cell migration and invasiveness in neoplasms. We have studied the in situ expression of these receptors in a large series of melanocytic lesions and correlated the expression with clinicopathological features and prognosis. Tissue microarray blocks of 141 cases including nevi (31 cases), primary (84 cases), and metastatic melanomas (26 cases) were semiquantitatively evaluated by immunohistochemistry for the expression of FPR and FPRL1 proteins. A significant association was observed regarding diagnosis and percentage of cells showing expression of FPR (P = 0.0311) and FPRL1 (P = 0.0053). A gain of FPR immunoreactivity was observed in the lesions having ulceration (P = 0.0194) and Breslow thickness (P = 0.044). Also, high FPRL1 cytoplasmic immunoreactivity was seen in lesions without tumor regression (P = 0.04). In addition, in patients with increased cytoplasmic staining for FPR, the probability of disease-specific survival was significantly lower (log rank test, P = 0.0089). Our findings reveal that FPR and FPRL1 are overexpressed in primary melanoma and correlate with aggressive tumor characteristics, underscoring them as potential therapeutic targets.
Collapse
|
10
|
Yang J, Wang Q, Zhao R, Sun B, Wang L, Hou Y, Li X, Wu C. Identification of oligomer proanthocyanidins (F2) isolated from grape seeds as a formyl peptide receptor 1 partial agonist. Int Immunopharmacol 2013; 15:756-63. [PMID: 23523627 DOI: 10.1016/j.intimp.2013.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/01/2013] [Accepted: 03/07/2013] [Indexed: 11/27/2022]
Abstract
Formyl peptide receptor 1 (FPR1) plays an important role in the rapid progression of glioblastoma and has been considered as a molecular target for the treatment. Previously, we have shown that oligomer proanthocyanidins (F2, degree of polymerization 2-15), isolated from grape seeds, inhibited FPR1-mediated chemotaxis of U-87 glioblastoma cells. In the present study, we investigated the capacity of F2 to interact with FPR1. The cross attenuation of chemotaxis revealed that F2 shared FPR1 with formyl-methionyl-leucyl-phenylalanine (fMLF), which is a prototype agonist of FPR1. F2 was chemotactic for U-87 cells, and the chemotactic response was abolished when FPR1 gene was silenced or FPR1 was competitively occupied. We further show that F2 specifically blocked the binding of fluorescent agonist to FPR1. Interestingly, F2 exhibited the characteristic of a partial agonist for FPR1, as shown by its capacity to activate FPR1-mediated PI3K-PKC-MAPK pathways. Meanwhile, F2 also attenuated fMLF-triggered MAPK activation, suggesting that F2 could antagonize the effect of an agonist. Furthermore, F2 abolished the invasion of U-87 cells induced by fMLF. Thus, we have identified F2 as a novel, partial agonist for FPR1, which may be useful for glioblastoma therapy.
Collapse
Affiliation(s)
- Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, 110016, Shenyang, Engineering Technology and Research Center for Plant Polyphenols of Liaoning Province, 110016 Shenyang, PR China
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Ye XZ, Xu SL, Xin YH, Yu SC, Ping YF, Chen L, Xiao HL, Wang B, Yi L, Wang QL, Jiang XF, Yang L, Zhang P, Qian C, Cui YH, Zhang X, Bian XW. Tumor-associated microglia/macrophages enhance the invasion of glioma stem-like cells via TGF-β1 signaling pathway. THE JOURNAL OF IMMUNOLOGY 2012; 189:444-53. [PMID: 22664874 DOI: 10.4049/jimmunol.1103248] [Citation(s) in RCA: 343] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The invasion of malignant glioma cells into the surrounding normal brain tissues is crucial for causing the poor outcome of this tumor type. Recent studies suggest that glioma stem-like cells (GSLCs) mediate tumor invasion. However, it is not clear whether microenvironment factors, such as tumor-associated microglia/macrophages (TAM/Ms), also play important roles in promoting GSLC invasion. In this study, we found that in primary human gliomas and orthotopical transplanted syngeneic glioma, the number of TAM/Ms at the invasive front was correlated with the presence of CD133(+) GSLCs, and these TAM/Ms produced high levels of TGF-β1. CD133(+) GSLCs isolated from murine transplanted gliomas exhibited higher invasive potential after being cocultured with TAM/Ms, and the invasiveness was inhibited by neutralization of TGF-β1. We also found that human glioma-derived CD133(+) GSLCs became more invasive upon treatment with TGF-β1. In addition, compared with CD133(-) committed tumor cells, CD133(+) GSLCs expressed higher levels of type II TGF-β receptor (TGFBR2) mRNA and protein, and downregulation of TGFBR2 with short hairpin RNA inhibited the invasiveness of GSLCs. Mechanism studies revealed that TGF-β1 released by TAM/Ms promoted the expression of MMP-9 by GSLCs, and TGFBR2 knockdown reduced the invasiveness of these cells in vivo. These results demonstrate that TAM/Ms enhance the invasiveness of CD133(+) GSLCs via the release of TGF-β1, which increases the production of MMP-9 by GSLCs. Therefore, the TGF-β1 signaling pathway is a potential therapeutic target for limiting the invasiveness of GSLCs.
Collapse
Affiliation(s)
- Xian-zong Ye
- Key Laboratory of Tumor Immunopathology, Third Military Medical University, Ministry of Education of China, Chongqing 400038, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Stimulation of human formyl peptide receptors by calpain inhibitors: homology modeling of receptors and ligand docking simulation. Arch Biochem Biophys 2011; 516:121-7. [PMID: 22005393 DOI: 10.1016/j.abb.2011.09.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 09/30/2011] [Accepted: 09/30/2011] [Indexed: 01/14/2023]
Abstract
Calpain inhibitors, including peptide aldehydes (N-acetyl-Leu-Leu-Nle-CHO and N-acetyl-Leu-Leu-Met-CHO) and α-mercapto-acrylic acid derivatives (PD150606 and PD151746), have been shown to stimulate phagocyte functions via activation of human formyl peptide receptor (hFPR) and/or hFPR-like 1 (hFPRL1). Using the homology modeling of the receptors and the ligand docking simulation, here we show that these calpain inhibitors could bind to the putative N-formyl-Met-Leu-Phe (fMLF) binding site on hFPR and/or hFPRL1. The studies with HEK-293 cells stably expressing hFPR or hFPRL1 showed that the concentrations of calpain inhibitors required to induce an increase in cytoplasmic free Ca(2+) ([Ca(2+)](i)) was much higher (>100 folds) than those of fMLF and Trp-Lys-Tyr-Met-Val-D-Met (WKYMVm). HEK-293 cells expressing hFPR or hFPRL1 with the mutated fMLF binding site never exhibited the [Ca(2+)](i) response to calpain inhibitors. When the optimal concentrations of each stimulus were used, pretreatment of cells with fMLF or WKYMVm abolished an increase in [Ca(2+)](i) induced by calpain inhibitors as well as the same stimulus, whereas pretreatment of cells with calpain inhibitors significantly suppressed, but never abolished, the [Ca(2+)](i) response induced by fMLF or WKYMVm, suggesting that the binding affinity of the inhibitors to the putative fMLF binding site may be lower than that of fMLF or WKYMVm.
Collapse
|
13
|
Yang L, Ping YF, Yu X, Qian F, Guo ZJ, Qian C, Cui YH, Bian XW. Gastric cancer stem-like cells possess higher capability of invasion and metastasis in association with a mesenchymal transition phenotype. Cancer Lett 2011; 310:46-52. [PMID: 21782323 DOI: 10.1016/j.canlet.2011.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 06/03/2011] [Accepted: 06/03/2011] [Indexed: 12/11/2022]
Abstract
Cancer stem cells have been isolated from various types of cancer including leukemia and solid tumors. However, the methods for isolating gastric cancer stem-like cells (GCSCs) have not been well established. As a consequence, the biological behavior and the significance of these cells to cancer progression remains to be clarified. In this study, we isolated and characterized GCSCs from a gastric cancer cell line SGC7901 and found their enhanced capabilities of invasion in vitro and metastasis in vivo. We further studied the expression of molecules related to epithelial-mesenchymal and invasion in GCSCs and found there were decreased E-cadherin, but increased vimentin and matrix metalloproteinase 2 (MMP-2), in these cells. Our results suggest that decreased E-cadherin and increased MMP-2 may be associated with the capacity of GCSCs to metastasize.
Collapse
Affiliation(s)
- Lang Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | |
Collapse
|