251
|
Ye MY, Chen MY, Chang YH, Huang JS, Huang TT, Wong TY, Hong TM, Chen YL. Growth-regulated oncogene-α from oral submucous fibrosis fibroblasts promotes malignant transformation of oral precancerous cells. J Oral Pathol Med 2018; 47:880-886. [PMID: 30035347 DOI: 10.1111/jop.12768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 06/28/2018] [Accepted: 07/19/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a common human malignancy and is usually preceded by the oral precancerous lesions. Oral submucous fibrosis (OSF) is one of the oral precancerous lesions with high incidence of malignant transformation. In addition to cancer cells, cancer-associated fibroblasts in the tumor microenvironment are correlated with cancer progression, but the role of fibroblasts from OSF in tumorigenesis and progression is still unknown. Growth-regulated oncogene-α (GRO-α), a member of CXC chemokine family, is related to tumorigenesis in several cancers. In this study, we would like to explore the role of GRO-α from OSF-associated fibroblasts in oral cancer progression. METHODS We isolated primary culture fibroblasts of normal, precancerous, and tumor tissues from patients with OSCC accompanied with OSF. A cytokine array was used to screen cytokine secretions in the conditioned media of the fibroblasts. A wound healing migration assay, WST-1 cell proliferation assay, rhodamine-phalloidin staining, and soft agar colony formation assay were used to investigate the effects of GRO-α on a dysplastic oral keratinocyte cell line (DOK) cell migration, growth, and anchorage-independent growth. RESULTS GRO-α was identified to be increased in conditioned media of OSF-associated fibroblasts. GRO-α promotes DOK cells proliferation, migration, and anchorage-independent growth through enhancing the EGFR/ERK signaling pathway, F-actin rearrangement, and stemness properties, respectively. Moreover, GRO-α neutralizing antibodies downregulated the conditioned medium-induced cell proliferation and migration of DOK. CONCLUSION GRO-α from OSF-associated fibroblasts paracrinally promotes oral malignant transformation and significantly contributes to OSCC development.
Collapse
Affiliation(s)
- Mei-Yin Ye
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Yen Chen
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Han Chang
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jehn-Shyun Huang
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tze-Ta Huang
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tung-Yiu Wong
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tse-Ming Hong
- Institutes of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuh-Ling Chen
- Institutes of Oral Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
252
|
Yu Y, Liu D, Liu Z, Li S, Ge Y, Sun W, Liu B. The inhibitory effects of COL1A2 on colorectal cancer cell proliferation, migration, and invasion. J Cancer 2018; 9:2953-2962. [PMID: 30123364 PMCID: PMC6096367 DOI: 10.7150/jca.25542] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/23/2018] [Indexed: 12/12/2022] Open
Abstract
Purpose: Collagen type I alpha 2 chain (COL1A2) has been shown to participate in the development of various human malignancies. However, the role of COL1A2 in human colorectal cancer (CRC) remains unknown. This study investigated the expression pattern of COL1A2 in primary CRC tissues as well as the correlation of COL1A2 expression with clinicopathological features and prognosis of CRC. The function of COL1A2 in CRC cell proliferation, migration, and invasion as well as the possible mechanisms were also examined. Methods: Real-time PCR and immunohistochemical analysis were performed to determine the expression of COL1A2 in primary cancer tissues and adjacent normal tissues from CRC patients. A COL1A2-expressing lentiviral vector was transfected into CRC cells, and cell counting kit-8 and Transwell assays were used to explore the effects of COL1A2 on CRC cell proliferation, migration, and invasion. Microarray-based mRNA expression profile screening was performed to reveal the possible signaling pathways involved in COL1A2-regulated cell behaviors. Results: COL1A2 was significantly downregulated in primary CRC tissues. The mRNA levels of COL1A2 in CRC tissues were correlated with tumor differentiation, invasion, and lymph node metastasis. Overexpression of COL1A2 inhibited proliferation, migration, and invasion of CRC cell lines (SW480 and SW620). The microarray analysis showed that COL1A2 overexpression regulated numerous oncogenes and cancer-related signaling pathways. Among them, altered expression of ten representative cancer-related genes in these pathways were further confirmed by western blotting. Conclusions: Our study identified COL1A2 as a novel tumor suppressor in CRC and provided a potential therapeutic approach to treat CRC.
Collapse
Affiliation(s)
- Yifan Yu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dongliang Liu
- Department of Ear-nose-throat department, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenghao Liu
- Department of Graduate School, China Medical University, Shenyang, China
| | - Shuqiang Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Ge
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Sun
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Baolin Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
253
|
Chemokine (C-X-C motif) ligand 1 is associated with tumor progression and poor prognosis in patients with colorectal cancer. Biosci Rep 2018; 38:BSR20180580. [PMID: 29784873 PMCID: PMC6028751 DOI: 10.1042/bsr20180580] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/15/2018] [Accepted: 05/21/2018] [Indexed: 12/21/2022] Open
Abstract
Chemokine (C-X-C motif) ligand 1 (CXCL1) is a chemotactic cytokine known to regulate cancer progression and invasion. However, the prognostic significance of CXCL1 expression in colorectal cancer (CRC) has not been fully characterized. The present study explored the clinicopathological significance and potential role of CXCL1 in the carcinogenesis and progression of CRC. The protein expression of CXCL1 was measured immunohistochemically in tissue microarrays constructed from 276 CRC patients. CXCL1 expression levels and their associations with clinicopathological characteristics and patient survival were evaluated. The effect of CXCL1 on glycolysis was also examined. High CXCL1 expression was detected in 165 (59.8%) cases. CXCL1 expression was correlated with tumor diameter (P=0.002), T stage (P=0.044), N stage (P=0.005), M stage (P=0.001), lymphovascular invasion (P=0.010), and carcinoembryonic antigen status (P=0.019). High CXCL1 expression was validated as an independent prognostic factor for overall survival (OS) and disease-free survival (DFS) by both univariate and multivariate Cox regression analyses (both P<0.05). Experimentally, expression of CXCL1 was knocked down by stable transfected short hairpin RNA, resulting in a significantly decreased rate of glycolysis both in in vitro assays and in patients' samples (P<0.05). Silencing the expression of CXCL1 decreased the levels of the glycolytic enzymes GLUT1, HK2, and LDHA. In conclusion, by inducing glycolysis, CXCL1 plays a crucial role in both cancer progression and metastasis in CRC patients. The CXCL1 expression level is an independent prognostic factor for both OS and DFS. Moreover, CXCL1 may serve as a new biomarker and potential therapeutic target for CRC treatment.
Collapse
|
254
|
Hand F, Harmon C, Elliott LA, Caiazza F, Lavelle A, Maguire D, Hoti E, Nolan N, Geoghegan JG, Ryan EJ, O'Farrelly C. Depleted polymorphonuclear leukocytes in human metastatic liver reflect an altered immune microenvironment associated with recurrent metastasis. Cancer Immunol Immunother 2018; 67:1041-1052. [PMID: 29572702 PMCID: PMC11028161 DOI: 10.1007/s00262-018-2149-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hepatic immunity, normally protective against neoplasia, is subverted in colorectal liver metastasis (CRLM). Here, we compare the inflammatory microenvironment of CRLM-bearing liver tissue to donor liver. METHODS Twenty-five patients undergoing resection for CRLM were recruited, 13 of whom developed intrahepatic recurrence within 18 months. Biopsies were obtained from tumour and normal liver tissue adjacent to and distal from, the tumour. Donor liver biopsies were obtained during transplantation. Biopsies were cultured and conditioned media (CM) screened for 102 inflammatory mediators. Twelve of these were validated by Luminex assay. Transwell assays measured cancer cell chemotaxis. Polymorphonuclear leukocytes (PMN) and lymphocytes were quantified in H&E sections. RESULTS Fewer periportal tissue-resident PMN were present in metastatic liver compared to donor liver. Patients with the fewest PMN in liver tissue distal to their tumour had a shorter time to intrahepatic recurrence (P < 0.001). IL-6, CXCL1, CXCL5, G-CSF, GM-CSF, VEGF, LIF, and CCL3 were higher in liver-bearing CRLM compared to donor tissue. Consequently, cancer cells migrated equally towards CM of all regions of metastatic liver but not towards donor liver CM. CONCLUSIONS The local inflammatory environment may affect both immune cell infiltration and cancer cell migration contributing to recurrence following resection for CRLM.
Collapse
Affiliation(s)
- Fiona Hand
- National Liver Unit, St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
- School of Biochemistry and Immunology and School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 154-160 Pearse Street, Dublin 2, D02 R590, Ireland
| | - Cathal Harmon
- School of Biochemistry and Immunology and School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 154-160 Pearse Street, Dublin 2, D02 R590, Ireland
| | - Louise A Elliott
- Centre for Colorectal Disease, School of Medicine, University College Dublin and St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Francesco Caiazza
- Centre for Colorectal Disease, School of Medicine, University College Dublin and St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Aonghus Lavelle
- Centre for Colorectal Disease, School of Medicine, University College Dublin and St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Donal Maguire
- National Liver Unit, St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Emir Hoti
- National Liver Unit, St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Niamh Nolan
- National Liver Unit, St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Justin G Geoghegan
- National Liver Unit, St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Elizabeth J Ryan
- Centre for Colorectal Disease, School of Medicine, University College Dublin and St. Vincent's Hospital, Elm Park, Dublin 4, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology and School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, 154-160 Pearse Street, Dublin 2, D02 R590, Ireland.
| |
Collapse
|
255
|
Shimizu D, Kanda M, Kodera Y. Emerging evidence of the molecular landscape specific for hematogenous metastasis from gastric cancer. World J Gastrointest Oncol 2018; 10:124-136. [PMID: 29988904 PMCID: PMC6033711 DOI: 10.4251/wjgo.v10.i6.124] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 03/23/2018] [Accepted: 04/20/2018] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed cancers in the world. Most GC patients are diagnosed when the cancer is in an advanced stage, and consequently, some develop metastatic lesions that generally cause cancer-related death. Metastasis establishment is affected by various conditions, such as tumor location, hemodynamics and organotropism. While digestive cancers may share a primary site, certain cases develop hematogenous metastasis with the absence of peritoneal metastasis, and vice versa. Numerous studies have revealed the clinicopathological risk factors for hematogenous metastasis from GC, such as vascular invasion, advanced age, differentiation, Borrmann type 1 or 2 and expansive growth. Recently, molecular mechanisms that contribute to metastatic site determination have been elucidated by advanced molecular biological techniques. Investigating the molecules that specifically participate in metastasis establishment in distinct secondary organs will lead to the development of novel biomarkers for patient stratification according to their metastatic risk and strategies for preventing and treating distinct metastases. We reviewed articles related to the molecular landscape of hematogenous metastasis from GC.
Collapse
Affiliation(s)
- Dai Shimizu
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Mitsuro Kanda
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery (Surgery II), Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
256
|
Abstract
Chronic inflammation is a risk factor for gastrointestinal cancer and other diseases. Most studies have focused on cytokines and chemokines as mediators connecting chronic inflammation to cancer, whereas the involvement of lipid mediators, including prostanoids, has not been extensively investigated. Prostanoids are among the earliest signaling molecules released in response to inflammation. Multiple lines of evidence suggest that prostanoids are involved in gastrointestinal cancer. In this Review, we discuss how prostanoids impact gastrointestinal cancer development. In particular, we highlight recent advances in our understanding of how prostaglandin E2 induces the immunosuppressive microenvironment in gastrointestinal cancers.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Raymond N DuBois
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Research and Division of Gastroenterology, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
257
|
Feng PH, Chen KY, Huang YC, Luo CS, Wu SM, Chen TT, Lee CN, Yeh CT, Chuang HC, Han CL, Lin CF, Lee WH, Kuo CH, Lee KY. Bevacizumab Reduces S100A9-Positive MDSCs Linked to Intracranial Control in Patients with EGFR-Mutant Lung Adenocarcinoma. J Thorac Oncol 2018; 13:958-967. [PMID: 29684573 DOI: 10.1016/j.jtho.2018.03.032] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 10/17/2022]
Abstract
INTRODUCTION In vitro models have demonstrated immune-modulating effects of bevacizumab (BEV). Combinations of an EGFR tyrosine kinase inhibitor (TKI) with BEV improve progression-free survival (PFS) in patients with EGFR-mutated lung adenocarcinoma. How BEV confers this clinical effect and the underlying mechanisms of its effect are not clear. METHODS A total of 55 patients with stage 4 EGFR-mutated lung adenocarcinoma were enrolled. Myeloid-derived suppressor cells (MDSCs), type 1 and type 2 helper T cells, and cytotoxic T lymphocytes were analyzed by flow cytometry. Clinical data were collected for analysis. RESULT In all, 25 patients received EGFR TKI and BEV combination therapy (the BEV/TKI group) and 30 patients received EGFR TKI monotherapy (the TKI-only group). The BEV/TKI group had longer PFS (23.0 versus 8.6 months [p = 0.001]) and, in particular, better intracranial control rates (80.0% versus 43.0% [p = 0.03]), a longer time to intracranial progression (49.1 versus 12.9 months [p = 0.002]), and fewer new brain metastases (38.0% versus 71.0% [p = 0.03]) than the TKI-only group did. The BEV/TKI group had a lower percentage of circulating MDSCs (20.4% ± 6.5% before treatment versus 12.8% ± 6.6% after treatment, respectively [p = 0.02]), and higher percentages of type 1 helper T cells (22.9% ± 15.3% versus 33.2% ± 15.6% [p < 0.01]) and cytotoxic T lymphocytes (15.5% ± 7.2% versus 21.2% ± 5.6% [p < 0.01]) after treatment, changes that were not seen in the TKI-only group. Pretreatment percentage of MDSCs was correlated with PFS, with this correlation attenuated after BEV/TKI treatment. Percentage of MDSCs was also associated with shorter time to intracranial progression. CONCLUSION Combining a EGFR TKI with BEV extended PFS and protected against brain metastasis. Those effects were probably due to the reduction of circulating S100A9-positive MDSCs by BEV, which leads to restoration of effective antitumor immunity. Our data also support the rationale for a BEV-immune checkpoint inhibitor combination.
Collapse
Affiliation(s)
- Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China
| | - Yu-Chen Huang
- Division of Pulmonary Medicine, Department of Internal Medicine, Chang Gung Medical Foundation, Linko Branch, Taoyuan, Republic of China
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China
| | - Shen Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China
| | - Chun-Nin Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China
| | - Chi-Tai Yeh
- Department of Medical Research and Education, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Republic of China
| | - Chia-Li Han
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Republic of China
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China
| | - Wei-Hwa Lee
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China
| | - Chih-Hsi Kuo
- Division of Pulmonary Medicine, Department of Internal Medicine, Chang Gung Medical Foundation, Linko Branch, Taoyuan, Republic of China
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Republic of China; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Republic of China.
| |
Collapse
|
258
|
Hong SS, Zhang MX, Zhang M, Yu Y, Chen J, Zhang XY, Xu CJ. Follicle-stimulating hormone peptide-conjugated nanoparticles for targeted shRNA delivery lead to effective gro-α silencing and antitumor activity against ovarian cancer. Drug Deliv 2018; 25:576-584. [PMID: 29461120 PMCID: PMC6058603 DOI: 10.1080/10717544.2018.1440667] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The distinct hormone molecules and receptors, such as follicle-stimulating hormone receptor (FSHR) in ovarian cancer, provide opportunities for more precisely targeted therapy. We previously developed FSHR-mediated nanoparticles and found that FSH peptides on the surface of nanoparticles improved the delivery of short interfering RNA (siRNA) into ovarian cancer cells. However, the high toxicity of the nanoparticles and the transient silencing of the siRNA in vivo limited further study. Here, we developed FSH peptide-conjugated nanoparticles with an increased amount of polyethylene glycol (PEG) grafting and encapsulated short hairpin RNA (shRNA) to silence the target gene, growth-regulated oncogene α (gro-α). The nanoparticle complexes exhibited good stability over three weeks. Expression of the target gene, gro-α, was significantly down-regulated by gro-α shRNA-loaded nanoparticles conjugated with FSH peptides (FSH33-G-NP) in FSHR-positive HEY cells. Cell proliferation, migration, and invasion were also inhibited by FSH33-G-NP. Tumor growth was delayed significantly in the mice treated with FSH33-G-NP. No significant loss of body weight or severe toxic effects were observed in any groups. In conclusion, gro-α shRNA-loaded nanoparticles conjugated with FSH peptides overcame the drawbacks of the in vivo application of RNAi therapeutics and polymer-based nanocarriers and showed safe antitumor efficacy. Our study might contribute to the application of FSHR-based targeted therapy and imaging in cancer.
Collapse
Affiliation(s)
- Shan-Shan Hong
- a Obstetrics and Gynecology Hospital , Fudan University , Shanghai , China
| | - Ming-Xing Zhang
- a Obstetrics and Gynecology Hospital , Fudan University , Shanghai , China
| | - Meng Zhang
- a Obstetrics and Gynecology Hospital , Fudan University , Shanghai , China
| | - Yi Yu
- a Obstetrics and Gynecology Hospital , Fudan University , Shanghai , China
| | - Jun Chen
- b Departments of Pharmaceutics, School of Pharmacy , Fudan University , Shanghai , China
| | - Xiao-Yan Zhang
- a Obstetrics and Gynecology Hospital , Fudan University , Shanghai , China.,c Department of Obstetrics and Gynecology of Shanghai Medical School , Fudan University , Shanghai , China.,d Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , China
| | - Cong-Jian Xu
- a Obstetrics and Gynecology Hospital , Fudan University , Shanghai , China.,c Department of Obstetrics and Gynecology of Shanghai Medical School , Fudan University , Shanghai , China.,d Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , China
| |
Collapse
|
259
|
Gabrusiewicz K, Li X, Wei J, Hashimoto Y, Marisetty AL, Ott M, Wang F, Hawke D, Yu J, Healy LM, Hossain A, Akers JC, Maiti SN, Yamashita S, Shimizu Y, Dunner K, Zal MA, Burks JK, Gumin J, Nwajei F, Rezavanian A, Zhou S, Rao G, Sawaya R, Fuller GN, Huse JT, Antel JP, Li S, Cooper L, Sulman EP, Chen C, Geula C, Kalluri R, Zal T, Heimberger AB. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018; 7:e1412909. [PMID: 29632728 PMCID: PMC5889290 DOI: 10.1080/2162402x.2017.1412909] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/22/2022] Open
Abstract
Exosomes can mediate a dynamic method of communication between malignancies, including those sequestered in the central nervous system and the immune system. We sought to determine whether exosomes from glioblastoma (GBM)-derived stem cells (GSCs) can induce immunosuppression. We report that GSC-derived exosomes (GDEs) have a predilection for monocytes, the precursor to macrophages. The GDEs traverse the monocyte cytoplasm, cause a reorganization of the actin cytoskeleton, and skew monocytes toward the immune suppresive M2 phenotype, including programmed death-ligand 1 (PD-L1) expression. Mass spectrometry analysis demonstrated that the GDEs contain a variety of components, including members of the signal transducer and activator of transcription 3 (STAT3) pathway that functionally mediate this immune suppressive switch. Western blot analysis revealed that upregulation of PD-L1 in GSC exosome-treated monocytes and GBM-patient-infiltrating CD14+ cells predominantly correlates with increased phosphorylation of STAT3, and in some cases, with phosphorylated p70S6 kinase and Erk1/2. Cumulatively, these data indicate that GDEs are secreted GBM-released factors that are potent modulators of the GBM-associated immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Konrad Gabrusiewicz
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xu Li
- Institute of Biology, Westlake Institute for Advanced Study, Westlake University, Hangzhou, Zhejiang Province, China
| | - Jun Wei
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuuri Hashimoto
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anantha L Marisetty
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Martina Ott
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fei Wang
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Hawke
- Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Yu
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luke M Healy
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Anwar Hossain
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Johnny C Akers
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Sourindra N Maiti
- Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shinji Yamashita
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuzaburo Shimizu
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Dunner
- Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Anna Zal
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared K Burks
- Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joy Gumin
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Felix Nwajei
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aras Rezavanian
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Shouhao Zhou
- Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganesh Rao
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raymond Sawaya
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory N Fuller
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason T Huse
- Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack P Antel
- Neuroimmunology Unit, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Shulin Li
- Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurence Cooper
- Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erik P Sulman
- Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clark Chen
- Center for Theoretical and Applied Neuro-Oncology, University of California, San Diego, CA, USA
| | - Changiz Geula
- Laboratory for Cognitive and Molecular Morphometry, Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Raghu Kalluri
- Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomasz Zal
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy B Heimberger
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
260
|
Chen SC, Chen FW, Hsu YL, Kuo PL. Systematic Analysis of Transcriptomic Profile of Renal Cell Carcinoma under Long-Term Hypoxia Using Next-Generation Sequencing and Bioinformatics. Int J Mol Sci 2017; 18:ijms18122657. [PMID: 29215599 PMCID: PMC5751259 DOI: 10.3390/ijms18122657] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/25/2017] [Accepted: 12/04/2017] [Indexed: 01/28/2023] Open
Abstract
Patients with clear cell renal cell carcinoma (ccRCC) are often diagnosed with both von Hippel-Lindau (VHL) mutations and the constitutive activation of hypoxia-inducible factor-dependent signaling. In this study, we investigated the effects of long-term hypoxia in 786-O, a VHL-defective renal cell carcinoma cell line, to identify potential genes and microRNAs associated with tumor malignancy. The transcriptomic profiles of 786-O under normoxia, short-term hypoxia and long-term hypoxia were analyzed using next-generation sequencing. The results showed that long-term hypoxia promoted the ability of colony formation and transwell migration compared to normoxia. In addition, the differentially expressed genes induced by long-term hypoxia were involved in various biological processes including cell proliferation, the tumor necrosis factor signaling pathway, basal cell carcinoma and cancer pathways. The upregulated (L1CAM and FBN1) and downregulated (AUTS2, MAPT, AGT and USH1C) genes in 786-O under long-term hypoxia were also observed in clinical ccRCC samples along with malignant grade. The expressions of these genes were significantly correlated with survival outcomes in patients with renal cancer. We also found that long-term hypoxia in 786-O resulted in decreased expressions of hsa-mir-100 and hsa-mir-378 and this effect was also observed in samples of metastatic ccRCC compared to samples of non-metastatic ccRCC. These findings may provide a new direction for the study of potential molecular mechanisms associated with the progression of ccRCC.
Collapse
Affiliation(s)
- Szu-Chia Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Feng-Wei Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
261
|
Zhou X, An D, Liu X, Jiang M, Yuan C, Hu J. TNFα induces tolerant production of CXC chemokines in colorectal cancer HCT116 cells via A20 inhibition of ERK signaling. Int Immunopharmacol 2017; 54:296-302. [PMID: 29175508 DOI: 10.1016/j.intimp.2017.11.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/21/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022]
Abstract
Ubiquitin editing enzyme A20 functions as a tumor suppressor in various cancer. However, the mechanism for A20 regulation of cancer progress is not fully understood. In this study, we found that in human colorectal cancer HCT116 cells, TNFα induced a tolerant production of CXC chemokines, including CXCL1, 2, and 8 in a dose and time dependent manner. TNFα pre-treatment of HCT116 cells down-regulated the chemokine production induced by TNFα re-treatment. TNFα induced the phosphorylation of MAPKs ERK, JNK, P38 and NF-κB P65, but only ERK inhibition decreased TNFα-induced chemokine production. Both RT-PCR and FACS results showed that TNFα treatment did not regulate the expression of TNF receptors. However, TNFα up-regulated the expression of A20 at both mRNA and protein levels significantly. TNFα pre-treatment inhibited the signal transduction of MAPKs induced by TNFα re-stimulation, and A20 over-expression decreased the signal transduction of ERK and P38. Meanwhile, A20 inhibition by RNA interference reversed chemokine down-regulation induced by TNFα re-stimulation after TNFα pre-treatment. Taken together, these results suggested that in human colorectal cancer cells, A20 may function to inhibit cancer progression via down-regulation of TNFα-induced chemokine production by suppression of ERK signaling.
Collapse
Affiliation(s)
- Xin Zhou
- Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China; Graduate School, University of South China, Hengyang, Hunan 421001, China
| | - Dongjian An
- Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China.
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Chuang Yuan
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China.
| |
Collapse
|
262
|
TAMeless traitors: macrophages in cancer progression and metastasis. Br J Cancer 2017; 117:1583-1591. [PMID: 29065107 PMCID: PMC5729447 DOI: 10.1038/bjc.2017.356] [Citation(s) in RCA: 456] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/14/2017] [Accepted: 09/19/2017] [Indexed: 12/28/2022] Open
Abstract
Macrophages are conventionally classified into M1 and M2 subtypes according to their differentiation status and functional role in the immune system. However, accumulating evidence suggests that this binary classification system is insufficient to account for the remarkable plasticity of macrophages that gives rise to an immense diversity of subtypes. This diverse spectrum of macrophage subtypes play critical roles in various homeostatic and immune functions, but remain far from being fully characterised. In addition to their roles in normal physiological conditions, macrophages also play crucial roles in disease conditions such as cancer. In this review, we discuss the roles tumour-associated macrophages (TAMs) play in regulating different steps of tumour progression and metastasis, and the opportunities to target them in the quest for cancer prevention and treatment.
Collapse
|
263
|
Kasashima H, Yashiro M, Nakamae H, Masuda G, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Nakane T, Hino M, Hirakawa K, Ohira M. Clinicopathologic significance of the CXCL1-CXCR2 axis in the tumor microenvironment of gastric carcinoma. PLoS One 2017; 12:e0178635. [PMID: 28575019 PMCID: PMC5456266 DOI: 10.1371/journal.pone.0178635] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 05/16/2017] [Indexed: 12/21/2022] Open
Abstract
Purpose It was reported that the chemokine (C-X-C motif) ligand 1 (CXCL1) from cancer cells stimulated the recruitment of bone marrow-derived mesenchymal cells (BM-MCs) into tumor stroma via chemokine (C-X-C motif) receptor 2 (CXCR2) signaling. We conducted this retrospective study to determine the clinicopathologic significance of the CXCL1-CXCR2 axis in human gastric cancer. Methods The correlations between the clinicopathological features of 270 primary gastric carcinomas and CXCL1 in cancer cells and CXCR2 in stromal cells were analyzed in immunohistochemical studies. The effect of gastric cancer cells on the expression of CXCR2 in BM-MCs was examined using diffuse-type gastric cancer cell lines in vitro. Results The expression of CXCL1 in cancer cells was correlated with T invasion (T2–T4), lymph node metastasis, lymphatic invasion, venous invasion, peritoneal cytology, peritoneal metastasis and CXCR2 expression in stromal cells. The expression of CXCR2 in stromal cells was correlated with macroscopic type-4 cancers, histological type, T invasion (T2–T4), lymph node metastasis, lymphatic invasion, infiltration, peritoneal cytology, peritoneal metastasis and CD271 expression in stromal cells. The overall survival of patients with CXCL1 and CXCR2-positive cancer was poorer than that of the patients with negative cancer. Both CXCL1 expression in cancer cells and CXCR2 expression in stromal cells were independent prognostic factors for gastric cancer patients. Conclusion The expressions of CXCL1 in cancer cells and CXCR2 in stromal cells are useful prognostic factors for gastric cancer patients.
Collapse
Affiliation(s)
- Hiroaki Kasashima
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan.,Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Go Masuda
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Haruhito Kinoshita
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tamami Morisaki
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tatsunari Fukuoka
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tsuyoshi Hasegawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takahiko Nakane
- Department of Hematology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masayuki Hino
- Department of Hematology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kosei Hirakawa
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|