1
|
Sun M, Yu Y, Zhu H, Yao Y, Zhou X, Wang X, Zhang Y, Xu X, Zhuang J, Sun C. Hepatic Growth Factor as a Potential Biomarker for Lung Adenocarcinoma: A Multimodal Study. Curr Issues Mol Biol 2025; 47:208. [PMID: 40136462 PMCID: PMC11941628 DOI: 10.3390/cimb47030208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
(1) Background: Despite previous studies linking inflammatory cytokines to lung adenocarcinoma (LUAD), their causal mechanisms remain unclear. This study aims to explore the causal relationship between inflammatory cytokines and LUAD to fill this knowledge gap. (2) Methods: This study employs a comprehensive approach, integrating Mendelian randomization (MR) analysis, single-cell RNA sequencing (scRNA-seq), and transcriptomic sequencing (RNA-seq) data to investigate the relationship between inflammatory cytokines and LUAD. (3) Results: In forward MR analysis, elevated levels of hepatocyte growth factor (HGF), interleukin-1 receptor antagonist (IL-1RA), IL-5, monocyte chemoattractant protein-3, and monokine induced by interferon-γ were causally associated with an increased risk of LUAD. In reverse MR analysis, LUAD exhibited a positive causal relationship with the levels of regulated upon activation normal T cell expressed and secreted factor (RANTES) and stromal cell-derived factor-1α. The scRNA-seq data further identified specific cell populations that may influence LUAD onset and progression through the expression of particular inflammatory genes and intercellular communication. RNA-seq data analysis highlighted the role of the HGF gene in LUAD diagnosis, demonstrating its strong correlation with patient prognosis and immune cell infiltration within the tumor microenvironment. (4) Conclusions: The findings reveal a causal relationship between inflammatory cytokines and LUAD, with HGF emerging as a potential biomarker of significant clinical relevance. This study provides new insights into the molecular mechanisms underlying LUAD and lays the foundation for future therapeutic strategies.
Collapse
Affiliation(s)
- Mengxuan Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
| | - Hanci Zhu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
| | - Yan Yao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
| | - Xintong Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
| | - Xue Wang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261000, China; (X.W.); (X.X.)
| | - Yubao Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
| | - Xiaowei Xu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261000, China; (X.W.); (X.X.)
| | - Jing Zhuang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261000, China; (X.W.); (X.X.)
| | - Changgang Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (M.S.); (H.Z.); (Y.Y.); (X.Z.); (Y.Z.)
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang 261000, China; (X.W.); (X.X.)
| |
Collapse
|
2
|
Arandhara A, Bhuyan P, Das BK. Exploring lung cancer microenvironment: pathways and nanoparticle-based therapies. Discov Oncol 2025; 16:159. [PMID: 39934547 DOI: 10.1007/s12672-025-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Lung cancer stands out as a significant global health burden, with staggering incidence and mortality rates primarily linked to smoking and environmental carcinogens. The tumor microenvironment (TME) emerges as a critical determinant of cancer progression and treatment outcomes, comprising a complex interplay of cells, signaling molecules, and extracellular matrix. Through a comprehensive literature review, we elucidate current research trends and therapeutic prospects, aiming to advance our understanding of TME modulation strategies and their clinical implications for lung cancer treatment. Dysregulated immune responses within the TME can facilitate tumor evasion, limiting the efficacy of immune checkpoint inhibitors (ICI). Consequently, TME modulation strategies have become potential avenues to enhance therapeutic responses. However, conventional TME-targeted therapies often face challenges. In contrast, nanoparticle (NP)-based therapies offer promising prospects for improved drug delivery and reduced toxicity, leveraging the enhanced permeability and retention (EPR) effect. Despite NP design and delivery advancements, obstacles like poor tumor cell uptake and off-target effects persist, necessitating further optimization. This review underscores the pivotal role of TME in lung cancer management, emphasizing the synergistic potential of immunotherapy and nano-therapy.
Collapse
Affiliation(s)
- Arunabh Arandhara
- Assam Pharmacy Institute, Titabar, Amgurikhat, Jorhat, Assam, 785632, India
| | - Pallabi Bhuyan
- School of Pharmacy, The Assam Kaziranga University, Koraikhowa, Jorhat, Assam, 785006, India
| | - Bhrigu Kumar Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Azara, Guwahati, Assam, 781017, India.
| |
Collapse
|
3
|
Wu J, Wang H, Yang R, Liu D, Li W. Prognostic significance of the modified Glasgow Prognostic Score in NSCLC patients undergoing immune checkpoint inhibitor therapy: a meta-analysis. Front Oncol 2024; 14:1449853. [PMID: 39464717 PMCID: PMC11502296 DOI: 10.3389/fonc.2024.1449853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 09/25/2024] [Indexed: 10/29/2024] Open
Abstract
Background The modified Glasgow Prognosis Score (mGPS), which considers both inflammatory response and nutritional status, has been linked to the prognosis of various tumors. The relationship between mGPS and non-small cell lung cancer (NSCLC) patients receiving immune checkpoint inhibitors (ICIs) is still a subject of debate. This meta-analysis aims to comprehensively assess the association between mGPS and survival in NSCLC treated with ICIs. Methods A thorough review of studies from PubMed, Web of Science, Scopus, and Embase was conducted up to June 4, 2024. Fixed-effect or random-effect models were employed, combining hazard ratios (HRs) and 95% confidence intervals (CI), to assess the prognostic value of mGPS for OS and PFS in patients with NSCLC receiving immunotherapy. Results A total of 1,022 patients from 11 studies were recruited. Combined results showed that mGPS elevation was significantly associated with poor OS (HR = 1.63, 95%CI: 1.42-1.87, P < 0.01) and PFS (HR = 1.71, 95%CI: 1.31-2.24, P < 0.01). Subgroup analysis and sensitivity analysis further determined the predictive effect of elevated mGPS on OS and PFS deterioration in NSCLC patients receiving immunotherapy. Conclusion mGPS can be used as a good noninvasive biomarker to demonstrate prognostic and clinical significance in patients with NSCLC undergoing immunotherapy. Systematic review registration http://www.crd.york.ac.uk/prospero/ PROSPERO, identifier CRD42023432661.
Collapse
Affiliation(s)
- Jiaxuan Wu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Haoyu Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruiyuan Yang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
- Institute of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Ludwig N, Cucinelli S, Hametner S, Muckenthaler MU, Schirmer L. Iron scavenging and myeloid cell polarization. Trends Immunol 2024; 45:625-638. [PMID: 39054114 DOI: 10.1016/j.it.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Myeloid cells that populate all human organs and blood are a versatile class of innate immune cells. They are crucial for sensing and regulating processes as diverse as tissue homeostasis and inflammation and are frequently characterized by their roles in either regulating or promoting inflammation. Recent studies in cultured cells and mouse models highlight the role of iron in skewing the functional properties of myeloid cells in tissue damage and repair. Here, we review certain emerging concepts on how iron influences and determines myeloid cell polarization in the context of its uptake, storage, and metabolism, including in conditions such as multiple sclerosis (MS), sickle cell disease, and tumors.
Collapse
Affiliation(s)
- Natalie Ludwig
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Stefania Cucinelli
- Department of Paediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany; Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory and University of Heidelberg, Heidelberg, Germany
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria; Medical Neuroscience Cluster, Medical University of Vienna, Vienna, Austria
| | - Martina U Muckenthaler
- Department of Paediatric Hematology, Oncology, and Immunology, University of Heidelberg, Heidelberg, Germany; Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory and University of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Partner site Heidelberg/Mannheim, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany; Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
5
|
Lin S, Yong J, Zhang L, Chen X, Qiao L, Pan W, Yang Y, Zhao H. Applying image features of proximal paracancerous tissues in predicting prognosis of patients with hepatocellular carcinoma. Comput Biol Med 2024; 173:108365. [PMID: 38537563 DOI: 10.1016/j.compbiomed.2024.108365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Most of the methods using digital pathological image for predicting Hepatocellular carcinoma (HCC) prognosis have not considered paracancerous tissue microenvironment (PTME), which are potentially important for tumour initiation and metastasis. This study aimed to identify roles of image features of PTME in predicting prognosis and tumour recurrence of HCC patients. METHODS We collected whole slide images (WSIs) of 146 HCC patients from Sun Yat-sen Memorial Hospital (SYSM dataset). For each WSI, five types of regions of interests (ROIs) in PTME and tumours were manually annotated. These ROIs were used to construct a Lasso Cox survival model for predicting the prognosis of HCC patients. To make the model broadly useful, we established a deep learning method to automatically segment WSIs, and further used it to construct a prognosis prediction model. This model was tested by the samples of 225 HCC patients from the Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC). RESULTS In predicting prognosis of the HCC patients, using the image features of manually annotated ROIs in PTME achieved C-index 0.668 in the SYSM testing dataset, which is higher than the C-index 0.648 reached by the model only using image features of tumours. Integrating ROIs of PTME and tumours achieved C-index 0.693 in the SYSM testing dataset. The model using automatically segmented ROIs of PTME and tumours achieved C-index of 0.665 (95% CI: 0.556-0.774) in the TCGA-LIHC samples, which is better than the widely used methods, WSISA (0.567), DeepGraphSurv (0.593), and SeTranSurv (0.642). Finally, we found the Texture SumAverage Skew HV on immune cell infiltration and Texture related features on desmoplastic reaction are the most important features of PTME in predicting HCC prognosis. We additionally used the model in prediction HCC recurrence for patients from SYSM-training, SYSM-testing, and TCGA-LIHC datasets, indicating the important roles of PTME in the prediction. CONCLUSIONS Our results indicate image features of PTME is critical for improving the prognosis prediction of HCC. Moreover, the image features related with immune cell infiltration and desmoplastic reaction of PTME are the most important factors associated with prognosis of HCC.
Collapse
Affiliation(s)
- Siying Lin
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China; Department of Pathology, Department of Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Juanjuan Yong
- Department of Pathology, Department of Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Lei Zhang
- Department of Pancreatic-Hepato-Biliary-Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Xiaolong Chen
- Department of Hepatic Surgery, Liver Transplantation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Weidong Pan
- Department of Pancreatic-Hepato-Biliary-Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Huiying Zhao
- Department of Pathology, Department of Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Horvat N, Chocarro S, Marques O, Bauer TA, Qiu R, Diaz-Jimenez A, Helm B, Chen Y, Sawall S, Sparla R, Su L, Klingmüller U, Barz M, Hentze MW, Sotillo R, Muckenthaler MU. Superparamagnetic Iron Oxide Nanoparticles Reprogram the Tumor Microenvironment and Reduce Lung Cancer Regrowth after Crizotinib Treatment. ACS NANO 2024; 18:11025-11041. [PMID: 38626916 PMCID: PMC11064219 DOI: 10.1021/acsnano.3c08335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 05/01/2024]
Abstract
ALK-positive NSCLC patients demonstrate initial responses to ALK tyrosine kinase inhibitor (TKI) treatments, but eventually develop resistance, causing rapid tumor relapse and poor survival rates. Growing evidence suggests that the combination of drug and immune therapies greatly improves patient survival; however, due to the low immunogenicity of the tumors, ALK-positive patients do not respond to currently available immunotherapies. Tumor-associated macrophages (TAMs) play a crucial role in facilitating lung cancer growth by suppressing tumoricidal immune activation and absorbing chemotherapeutics. However, they can also be programmed toward a pro-inflammatory tumor suppressive phenotype, which represents a highly active area of therapy development. Iron loading of TAMs can achieve such reprogramming correlating with an improved prognosis in lung cancer patients. We previously showed that superparamagnetic iron oxide nanoparticles containing core-cross-linked polymer micelles (SPION-CCPMs) target macrophages and stimulate pro-inflammatory activation. Here, we show that SPION-CCPMs stimulate TAMs to secrete reactive nitrogen species and cytokines that exert tumoricidal activity. We further show that SPION-CCPMs reshape the immunosuppressive Eml4-Alk lung tumor microenvironment (TME) toward a cytotoxic profile hallmarked by the recruitment of CD8+ T cells, suggesting a multifactorial benefit of SPION-CCPM application. When intratracheally instilled into lung cancer-bearing mice, SPION-CCPMs delay tumor growth and, after first line therapy with a TKI, halt the regrowth of relapsing tumors. These findings identify SPIONs-CCPMs as an adjuvant therapy, which remodels the TME, resulting in a delay in the appearance of resistant tumors.
Collapse
Affiliation(s)
- Natalie
K. Horvat
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Sara Chocarro
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Oriana Marques
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Tobias A. Bauer
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ruiyue Qiu
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Alberto Diaz-Jimenez
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Barbara Helm
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
| | - Yuanyuan Chen
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Stefan Sawall
- X-ray
Imaging and CT, German Cancer Research Center
(DKFZ), Im Neuenheimer
Feld 280, 69120, Heidelberg, Germany
| | - Richard Sparla
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Lu Su
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ursula Klingmüller
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Matthias Barz
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
- Department
of Dermatology, University Medical Center
of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias W. Hentze
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Meyerhofstr.1, 69117, Heidelberg, Germany
| | - Rocío Sotillo
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Martina U. Muckenthaler
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, 69120, Heidelberg/Mannheim, Germany
| |
Collapse
|
7
|
Huang K, Zhang Y, Shi X, Yin Z, Zhao W, Huang L, Wang F, Zhou X. Cell-type-specific alternative polyadenylation promotes oncogenic gene expression in non-small cell lung cancer progression. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:816-831. [PMID: 37675185 PMCID: PMC10477688 DOI: 10.1016/j.omtn.2023.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023]
Abstract
Disrupted alternative polyadenylation (APA) is frequently involved in tumorigenesis and cancer progression by regulating the gene expression of oncogenes and tumor suppressors. However, limited knowledge of tumor-type- and cell-type-specific APA events may lead to novel APA events and their functions being overlooked. Here, we compared APA events across different cell types in non-small cell lung cancer (NSCLC) and normal tissues and identified functionally related APA events in NSCLC. We found several cell-specific 3'-UTR alterations that regulate gene expression changes showed prognostic value in NSCLC. We further investigated the function of APA-mediated 3'-UTR shortening through loss of microRNA (miRNA)-binding sites, and we identified and experimentally validated several oncogene-miRNA-tumor suppressor axes. According to our analyses, we found SPARC as an APA-regulated oncogene in cancer-associated fibroblasts in NSCLC. Knockdown of SPARC attenuates lung cancer cell invasion and metastasis. Moreover, we found high SPARC expression associated with resistance to several drugs except cisplatin. NSCLC patients with high SPARC expression could benefit more compared to low-SPARC-expression patients with cisplatin treatment. Overall, our comprehensive analysis of cell-specific APA events shed light on the regulatory mechanism of cell-specific oncogenes and provided opportunities for combination of APA-regulated therapeutic target and cell-specific therapy development.
Collapse
Affiliation(s)
- Kexin Huang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
- West China Biomedical Big Data Centre, West China Hospital of Sichuan University, Chengdu 610041, China
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yun Zhang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Xiaorui Shi
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Zhiqin Yin
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Weiling Zhao
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Liyu Huang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
| | - Fu Wang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710071, China
- School of Pharmacy, Shaanxi Institute of International Trade and Commerce, Xianyang, Shaanxi 712046, China
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
8
|
Chang W, Zhu J, Yang D, Shang A, Sun Z, Quan W, Li D. Plasma versican and plasma exosomal versican as potential diagnostic markers for non-small cell lung cancer. Respir Res 2023; 24:140. [PMID: 37259101 DOI: 10.1186/s12931-023-02423-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/16/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND AND AIMS This study aimed to investigate the expression of plasma versican and plasma exosomal versican in non-small cell lung cancer (NSCLC) and its correlation with clinicopathological features, and to evaluate its diagnostic performance in NSCLC and its predictive function for NSCLC incidence and metastasis risk. MATERIALS AND METHODS There were 110 instances of NSCLC, 42 cases of benign lung disease, and 55 healthy controls from September 2018 to October 2020 at Tongji Hospital Affiliated to Tongji University. Blood was collected and plasma was separated before surgery, and plasma exosomes were extracted by ExoQuick kit. Morphological and molecular phenotype identification of exosomes was performed by transmission electron microscopy, Nanosight particle tracking analysis, and western blotting. Plasma versican and plasma exosomal versican were detected in all subjects to assess their expression levels and diagnostic value in NSCLC. Clinicopathological data were collected to explore correlations between abnormal plasma versican and plasma exosomal versican expression and clinicopathological parameters. Receiver operating characteristic (ROC) curve was used to judge its diagnostic performance in NSCLC, and binary logistic regression analysis was used to predict the risk of NSCLC incidence and metastasis. RESULTS Plasma versican and plasma exosomal versican expression in NSCLC patients was significantly upregulated and was significantly higher in T3 + T4 patients compared with T1 + T2 patients (P < 0.05); the levels of plasma versican and plasma exosomal versican were positively correlated with lymph node metastasis, distant metastases (e.g., brain, bone), and mutation(e.g., EGFR,ALK)in NSCLC patients (all P < 0.05). Furthermore, ROC curve analysis showed that plasma versican and plasma exosomal versican had higher AUC values than NSE, CYFRA21-1, and SCC, and better diagnostic performance in NSCLC patients. However, the AUC and diagnostic performances of plasma versican and plasma exosomal versican in advanced-stage NSCLC patients were not shown to be significantly better than CEA. The results of binary logistic regression analysis showed that high levels of plasma exosomal versican had higher predictive value for lung cancer incidence, while high levels of plasma versican had higher predictive value for lung cancer metastasis. CONCLUSION Our findings showed that plasma versican and plasma exosomal versican might be potential diagnostic markers for NSCLC. High plasma exosomal versican expression can be used as a predictor of NSCLC risk and high plasma versican expression can be used as a predictor of NSCLC metastasis risk.
Collapse
Grants
- 81902984, 81974314, 82072362, 82002223 National Natural Science Foundation of China
- 81902984, 81974314, 82072362, 82002223 National Natural Science Foundation of China
- 81902984, 81974314, 82072362, 82002223 National Natural Science Foundation of China
- 81902984, 81974314, 82072362, 82002223 National Natural Science Foundation of China
- GWV-10.1-XK04 Shanghai Public Health System Construction Three-Year Action Plan
- 20204Y0070 Shanghai Municipal Health and Family Planning Commission
Collapse
Affiliation(s)
- Wenjing Chang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Jichao Zhu
- Department of Laboratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou Normal University, Huzhou, 313003, China
| | - Dianyu Yang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Anquan Shang
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Zujun Sun
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Wenqiang Quan
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Dong Li
- Department of Laboratory Medicine, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
9
|
Kantarci A, Kansal S, Hasturk H, Stephens D, Van Dyke TE. Resolvin E1 Reduces Tumor Growth in a Xenograft Model of Lung Cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1470-1484. [PMID: 35944728 PMCID: PMC9552033 DOI: 10.1016/j.ajpath.2022.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/05/2022] [Accepted: 07/12/2022] [Indexed: 05/14/2023]
Abstract
Inflammation plays a significant role in carcinogenesis and tumor growth. The current study was designed to test the hypothesis that resolvin E1 (RvE1) and overexpression of the receptor for RvE1 (ERV1) will prevent and/or reverse tumor generation in a gain-of-function mouse model of tumor seeding with lung cancer cells. To measure the impact of enhanced resolution of inflammation on cancer pathogenesis, ERV1-overexpressing transgenic (TG) and wild-type FVB mice were given an injection of 1 × 106 LA-P0297 cells subcutaneously and were treated with RvE1 (100 ng; intraperitoneally) or placebo. To assess the impact of RvE1 as an adjunct to chemotherapy, ERV1-TG and wild-type FVB mice were treated with cisplatin or cisplatin + RvE1. RvE1 significantly prevented tumor growth and reduced tumor size, cyclooxygenase-2, NF-κB, and proinflammatory cytokines in TG animals as compared to wild-type animals. A significant decrease in Ki-67, vascular endothelial growth factor, angiopoietin (Ang)-1, and Ang-2 was also observed in TG animals as compared to wild-type animals. Tumor-associated neutrophils and macrophages were significantly reduced by RvE1 in transgenics (P < 0.001). RvE1 administration with cisplatin led to a significant reduction of tumor volume and reduced cyclooxygenase-2, NF-κB, vascular endothelial growth factor-A, Ang-1, and Ang-2. These data suggest that RvE1 prevents inflammation and vascularization, reduces tumor seeding and tumor size, and, when used as an adjunct to chemotherapy, enhances tumor reduction at significantly lower doses of cisplatin.
Collapse
|
10
|
Zhou X, Wang X, Sun Q, Zhang W, Liu C, Ma W, Sun C. Natural compounds: A new perspective on targeting polarization and infiltration of tumor-associated macrophages in lung cancer. Biomed Pharmacother 2022; 151:113096. [PMID: 35567987 DOI: 10.1016/j.biopha.2022.113096] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 11/02/2022] Open
Abstract
With the development in tumor immunology, people are gradually understanding the complexity and diversity of the tumor microenvironment immune status and its important effect on tumors. Tumor-associated macrophages (TAMs), an important part of the tumor immune microenvironment, have a double effect on tumor growth and metastasis. Many studies have focused on lung cancer, especially non-small cell lung cancer and other "hot tumors" with typical inflammatory characteristics. The polarization and infiltration of TAMs is an important mechanism in the occurrence and development of malignant tumors, such as lung cancer, and in the tumor immune microenvironment. Therapeutic drugs designed for these reasons are key to targeting TAMs in the treatment of lung cancer. A large number of reports have suggested that natural compounds have a strong potential of affecting immunity by targeting the polarization and infiltration of TAMs to improve the immune microenvironment of lung cancer and exert a natural antitumor effect. This paper discusses the infiltration and polarization effects of natural compounds on lung cancer TAMs, provides a detailed classification and systematic review of natural compounds, and summarizes the bias of different kinds of natural compounds by affecting their antitumor mechanism of TAMs, with the aim of providing new perspectives and potential therapeutic drugs for targeted macrophages in the treatment of lung cancer.
Collapse
Affiliation(s)
- Xintong Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaomin Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qi Sun
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenfeng Zhang
- School of Traditional Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China; College of Chinese Medicine, Weifang Medical University, Weifang, China; Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.
| |
Collapse
|
11
|
Liang W, Chen W, Wei J, Yao H, Shi J, Hou X, Deng Y, Ou M. Zinc finger C3H1-type containing serves as a novel prognostic biomarker in human pan-cancer. Gene X 2022; 820:146251. [PMID: 35131366 DOI: 10.1016/j.gene.2022.146251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/21/2021] [Accepted: 01/21/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Zinc finger C3H1 domain-containing protein (ZFC3H1) is differentially expressed between primary tumor and the normal in most cancers. Additionally, a recent study has suggested that ZFC3H1 could serve as a novel marker for the prognosis of prostate adenocarcinoma (PRAD). However, the relationship between ZFC3H1 expression and the prognostic values in most tumors remains unclear. Our study is mainly for exploring the prognosis of ZFC3H1 in pan-cancer and for further discovering a potential therapeutics target. METHODS Based on the clinical big data, we performed a pan-cancer analysis of ZFC3H1, including gene expression, survival prognosis, genetic alteration, protein phosphorylation, immune infiltration and enrichment analysis. In addition, Real-Time PCR and Western Blot were used to further confirm the role of ZFC3H1 in the colorectal cancer. RESULTS We found that ZFC3H1 expression was connected with the prognosis of multiple malignant tumors. Furthermore, we also observed that ZFC3H1 was highly expressed in colorectal cancer through Real-Time PCR and Western Blot. The primary tumors presented higher phosphorylation level of the S655 site in lung adenocarcinoma, colon adenocarcinoma and uterine corpus endometrial carcinoma. ZFC3H1 expression was positively correlated with the immune infiltration of Cancer-associated fibroblasts (CAFs) in some tumors, such as liver hepatocellular carcinoma. And RNA surveillance pathways may be closely associated with the occurrence of tumors. CONCLUSIONS Our study first reveals that ZFC3H1 could serve as a novel prognostic biomarker of pan-cancer, especially colorectal cancer.
Collapse
Affiliation(s)
- Wenken Liang
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China; College of Life Science, Guangxi Normal University, Guilin 541000, China.
| | - Wei Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China.
| | - Jianfen Wei
- College of Life Science, Guangxi Normal University, Guilin 541000, China.
| | - Hongbing Yao
- Biliary Hepatopancreatic Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China.
| | - Jianling Shi
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China.
| | - Xianliang Hou
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China.
| | - Yecheng Deng
- College of Life Science, Guangxi Normal University, Guilin 541000, China.
| | - Minglin Ou
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin 541000, China.
| |
Collapse
|
12
|
Hu C, Wu J, Liu Y, Zhou J, Wang W, Wang X, Guo J, Wang Q, Zhang X, Li D, Xie J, Ding X, Xing Y, Hu D. Relationship Between Neutrophil-To-Lymphocyte Ratio and Brain Metastasis in Non-Small Cell Lung Cancer Patients. Cancer Control 2022; 29:10732748221076805. [PMID: 35209734 PMCID: PMC8883297 DOI: 10.1177/10732748221076805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective To investigate the relationship between the neutrophil-to-lymphocyte ratio (NLR) of patients with non-small cell lung cancer (NSCLC) and their risk of developing brain metastases after adjusting for confounding factors. Methods A retrospective observational study of the general data of patients with NSCLC diagnosed from January 2016 to December 2020. Multivariate logistic regression was used to calculate the dominance ratio (OR) with 95% confidence interval (CI) for NLR and NSCLC brain metastases with subgroup analysis. Generalized summation models and smoothed curve fitting were used to identify whether there was a nonlinear relationship between them. Results In all 3 models, NLR levels were positively correlated with NSCLC brain metastasis (model 1: OR: 1.12, 95% CI: 1.01-1.23, P = .025; model 2: OR: 1.16, 95% CI: 1.04-1.29, P = .007; model 3: OR: 1.20, 95% CI: 1.05-1.37, P = .006). Stratified analysis showed that this positive correlation was present in patients with adenocarcinoma (LUAD) and female patients (LUAD: OR: 1.30, 95% CI: 1.10-1.54, P = .002; female: OR: 1.52, 95% CI: 1.05-2.20, P = .026), while there was no significant correlation in patients with squamous carcinoma (LUSC) and male patients (LUSC: OR:0.76,95% CI:0.38- 1.53, P = .443; male: OR:1.13, 95% CI:0.95-1.33, P = .159). Conclusion This study showed that elevated levels of NLR were independently associated with an increased risk of developing brain metastases in patients with NSCLC, and that this correlation varied by TYPE and SEX, with a significant correlation in female patients and patients with LUAD.
Collapse
Affiliation(s)
- Chunxiao Hu
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Jing Wu
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China.,Anhui Province Engineering Laboratory of Occupational Health and Safety, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Yafeng Liu
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Jiawei Zhou
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Wenyang Wang
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Xueqin Wang
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Jianqiang Guo
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Qingsen Wang
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Xin Zhang
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Danting Li
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Jun Xie
- Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Xuansheng Ding
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China.,Affiliated Cancer Hospital, 91594Anhui University of Science and Technology, Huainan, P.R. China.,School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yingru Xing
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China.,Affiliated Cancer Hospital, 91594Anhui University of Science and Technology, Huainan, P.R. China
| | - Dong Hu
- School of Medicine, 91594Anhui University of Science and Technology, Huainan, P.R. China.,Anhui Province Engineering Laboratory of Occupational Health and Safety, 91594Anhui University of Science and Technology, Huainan, P.R. China.,Key Laboratory of Industrial Dust Prevention and Control & Occupational Safety and Health of the Ministry of Education, 91594Anhui University of Science and Technology, Huainan, P.R. China
| |
Collapse
|
13
|
Tantipaiboonwong P, Chaiwangyen W, Suttajit M, Kangwan N, Kaowinn S, Khanaree C, Punfa W, Pintha K. Molecular Mechanism of Antioxidant and Anti-Inflammatory Effects of Omega-3 Fatty Acids in Perilla Seed Oil and Rosmarinic Acid Rich Fraction Extracted from Perilla Seed Meal on TNF-α Induced A549 Lung Adenocarcinoma Cells. Molecules 2021; 26:6757. [PMID: 34833849 PMCID: PMC8622939 DOI: 10.3390/molecules26226757] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Industrially, after the removal of oil from perilla seeds (PS) by screw-type compression, the large quantities of residual perilla seed meal (PSM) becomes non-valuable waste. Therefore, to increase the health value and price of PS and PSM, we focused on the biological effects of perilla seed oil (PSO) and rosmarinic acid-rich fraction (RA-RF) extracted from PSM for their role in preventing oxidative stress and inflammation caused by TNF-α exposure in an A549 lung adenocarcinoma culture model. The A549 cells were pretreated with PSO or RA-RF and followed by TNF-α treatment. We found that PSO and RA-RF were not toxic to TNF-α-induced A549 cells. Both extracts significantly decreased the generation of reactive oxygen species (ROS) in this cell line. The mRNA expression levels of IL-1β, IL-6, IL-8, TNF-α, and COX-2 were significantly decreased by the treatment of PSO and RA-RF. The Western blot indicated that the expression of MnSOD, FOXO1, and NF-κB and phosphorylation of JNK were also significantly diminished by PSO and RA-RF treatment. The results demonstrated that PSO and RA-RF act as antioxidants to scavenge TNF-α induced ROS levels, resulting in decreased the expression of MnSOD, FOXO1, NF-κB and JNK signaling pathway in a human lung cell culture exposed to TNF-α.
Collapse
Affiliation(s)
- Payungsak Tantipaiboonwong
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (P.T.); (W.C.); (M.S.)
| | - Wittaya Chaiwangyen
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (P.T.); (W.C.); (M.S.)
| | - Maitree Suttajit
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (P.T.); (W.C.); (M.S.)
| | - Napapan Kangwan
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand;
| | - Sirichat Kaowinn
- Department of General Science and Liberal Arts, King Mongkut’s Institute of Technology Ladkrabang Prince of Chumphon Campus, Pathiu, Chumphon 86160, Thailand;
| | - Chakkrit Khanaree
- School of Traditional and Alternative Medicine, Chiang Rai Rajabhat University, Chiang Rai 57100, Thailand; (C.K.); (W.P.)
| | - Wanisa Punfa
- School of Traditional and Alternative Medicine, Chiang Rai Rajabhat University, Chiang Rai 57100, Thailand; (C.K.); (W.P.)
| | - Komsak Pintha
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (P.T.); (W.C.); (M.S.)
| |
Collapse
|
14
|
An Inflammation-Related Nine-Gene Signature to Improve Prognosis Prediction of Lung Adenocarcinoma. DISEASE MARKERS 2021; 2021:9568057. [PMID: 34580602 PMCID: PMC8464410 DOI: 10.1155/2021/9568057] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/26/2021] [Indexed: 11/18/2022]
Abstract
Background A novel predictive model was rarely reported based on inflammation-related genes to explore clinical outcomes of lung adenocarcinoma (LUAD) patients. Methods Using TCGA database, we screened nine inflammation-related genes with a prognostic value, and LASSO regression was applied for model construction. The predictive value of the prognostic signature developed from inflammation-related genes was assessed by survival assays and multivariate assays. PCA and t-SNE analysis were performed to demonstrate clustering abilities of risk scores. Results Thirteen inflammation-related genes (BTG2, CCL20, CD69, DCBLD2, GPC3, IL7R, LAMP3, MMP14, NMUR1, PCDH7, PIK3R5, RNF144B, and TPBG) with prognostic values were finally identified. LASSO regression further screened nine candidates (BTG2, CCL20, CD69, IL7R, MMP14, NMUR1, PCDH7, RNF144B, and TPBG). Then, a prognostic prediction model using the above nine genes was constructed. A reliable clustering ability of risk score was demonstrated by PCA and t-SNE assays in 500 LUAD patients. The survival assays revealed that the overall survivals of the high-risk group were distinctly poorer than those of the low-risk group with 1-, 3-, and 5-year AUC values of 0.695, 0.666, and 0.694, respectively. Finally, multivariate assays demonstrated the scoring system as an independent prognostic factor for overall survival. Conclusions Our study shows that the signature of nine inflammation-related genes can be used as a prognostic marker for LUAD.
Collapse
|
15
|
Kim Y, Yoo G, Lee DH, Lee CS, Chung C. Spontaneous regression of incidentally diagnosed bronchial squamous cell lung carcinoma after severe bronchitis: A case report. SAGE Open Med Case Rep 2021; 9:2050313X211010639. [PMID: 34035913 PMCID: PMC8132097 DOI: 10.1177/2050313x211010639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/23/2021] [Indexed: 11/17/2022] Open
Abstract
Spontaneous regression of lung cancer is exceptionally rare. But there have been several intriguing cases reported in early and even advanced stages of lung cancer. Although the exact mechanism remains to be elucidated, the inflammation and immunologic response have been suggested as one of the means of spontaneous regression. Chronic inflammation is generally known to induce and aggravate tumorigenesis, but the relationship between cancer and inflammation highly depends on the contexts. Here, we present a case of a 60-year-old male ex-smoker who complained of recurrent hemoptysis, cough, and purulent sputum. The initial chest CT scan revealed diffuse bronchial thickening and an endobronchial mass-like lesion in the left lingular segment. The bronchoscopic and pathological findings also suggested a diagnosis of squamous cell carcinoma with severe mucosal inflammation. He was treated with antibiotics for the bronchitis during the first 1 week and his symptoms markedly improved. After 3 weeks, he underwent a follow-up examination. Chest computed tomography and bronchoscopy revealed the significant improvement of the bronchial narrowing and mucosal edema. Biopsy was performed several times around the lesion where the tissue was initially taken. However, the pathological results showed only chronic inflammation of bronchi, not cancer cells. Fortunately, there was no recurrence of lung cancer in follow-up chest computed tomography or bronchoscopy for almost 5 years. In this case, the incidentally diagnosed bronchial squamous cell carcinoma disappeared after severe inflammatory reaction of the bronchial wall. The clinician should remind the risk of early lung cancer accompanied with bronchitis in high-risk patients of lung cancer and also be aware that although it is very rare, the lesions could spontaneously regress.
Collapse
Affiliation(s)
- Yoonjoo Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Geon Yoo
- Clinical Research Division, National Institute of Food and Drug Safety Evaluation, Cheongju-si, Republic of Korea
| | - Da-Hye Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Choong-Sik Lee
- Department of Pathology, Konyang University Hospital, Daejeon, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
16
|
Liang ZW, Ge XX, Xu MD, Qin H, Wu MY, Shen M, Zhang Y, Liu XM, Chen K, Li W, Duan W, Qin S. Tumor-associated macrophages promote the metastasis and growth of non-small-cell lung cancer cells through NF-κB/PP2Ac-positive feedback loop. Cancer Sci 2021; 112:2140-2157. [PMID: 33609307 PMCID: PMC8177805 DOI: 10.1111/cas.14863] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/29/2020] [Accepted: 01/24/2021] [Indexed: 01/22/2023] Open
Abstract
Non‐small‐cell lung cancer (NSCLC), with its aggressive biological behavior, is one of the most diagnosed cancers. Tumor‐associated inflammatory cells play important roles in the interaction between chronic inflammation and lung cancer, however the mechanisms involved are far from defined. In the present study, by developing an orthotopic NSCLC mouse model based on chronic inflammation, we proved that an inflammatory microenvironment accelerated the growth of orthotopic xenografts in vivo. Tumor‐associated macrophages, the most abundant population of inflammatory cells, were identified. Treatment with macrophage‐conditioned medium (MCM) promoted the growth and migration of NSCLC cells. Using bioinformatics analysis, we identified downregulated PP2Ac expression in NSCLC cells upon treatment with MCM. We further confirmed that this downregulation was executed in an NF‐κB pathway‐dependent manner. As IκB kinase (IKK) has been proved to be a substrate of PP2Ac, inhibition on PP2Ac could result in amplification of NF‐κB pathway signaling. Overexpression of PP2Ac, or the dominant‐negative forms of IKK or IκB, attenuated the acceleration of growth and metastasis by MCM. Using bioinformatics analysis, we further identified that CXCL1 and COL6A1 could be downstream of NF‐κB/PP2Ac pathway. Luciferase assay and ChIP assay further confirmed the location of response elements on the promoter regions of CXCL1 and COL6A1. Elevated CXCL1 facilitated angiogenesis, whereas upregulated COL6A1 promoted proliferation and migration.
Collapse
Affiliation(s)
- Zhan-Wen Liang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xin-Xin Ge
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Dan Xu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hualong Qin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng-Yao Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiao-Meng Liu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiming Duan
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Songbing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
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
|
18
|
Zabłocka-Słowińska K, Prescha A, Płaczkowska S, Porębska I, Kosacka M, Pawełczyk K. Serum and Whole Blood Cu and Zn Status in Predicting Mortality in Lung Cancer Patients. Nutrients 2020; 13:nu13010060. [PMID: 33375477 PMCID: PMC7824662 DOI: 10.3390/nu13010060] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022] Open
Abstract
Alterations in circulating Cu and Zn are negative predictors of survival in neoplastic patients and are known during lung cancer. However, no data on predicting mortality of lung cancer patients based on the level of these elements in the blood have been presented to date. The aims of this prospective cohort study were as follows: (i) To evaluate the disturbances in serum and whole blood Cu and Zn, (ii) to assess the relationships between serum and whole blood Cu and Zn status and clinical, sociodemographic, and nutritional data, and (iii) to investigate the association of Cu and Zn status with all-cause mortality in lung cancer. Naïve-treatment lung cancer patients (n = 167) were characterized in terms of sociodemographic, clinical, and anthropometric data and dietary intake and compared with sex-matched control subjects (n = 48). Whole blood and serum Cu and Zn status was determined by atomic absorption spectrometry. Cox proportional hazards models adjusted for multiple confounders/mediators were used to estimate the association between all-cause death and Cu and Zn status. Sex, cardiovascular disease, chronic obstructive pulmonary disease, clinical stage, and hemoglobin, platelet, and glucose concentrations significantly differentiated Cu and Zn status. All-cause mortality in lung cancer patients was positively associated with serum Cu levels, Cu:Zn ratio, and whole blood Zn levels. However, an advanced clinical stage of disease was the strongest predictor of all-cause mortality. Circulatory status of Cu and Zn might be included in routine clinical characteristics of patients with lung cancer patients as additional prognostic variables, but only after further more detail studies.
Collapse
Affiliation(s)
- Katarzyna Zabłocka-Słowińska
- Department of Food Science and Dietetics, Wroclaw Medical University, ul. Borowska 211, 50-556 Wroclaw, Poland;
- Correspondence:
| | - Anna Prescha
- Department of Food Science and Dietetics, Wroclaw Medical University, ul. Borowska 211, 50-556 Wroclaw, Poland;
| | - Sylwia Płaczkowska
- Diagnostics Laboratory for Teaching and Research, Department of Laboratory Diagnostics Wroclaw Medical University, ul. Borowska 211a, 50-556 Wroclaw, Poland;
| | - Irena Porębska
- Department and Clinic of Pulmonology and Lung Cancers, Wroclaw Medical University, ul. Grabiszynska 105, 53-439 Wroclaw, Poland; (I.P.); (M.K.)
| | - Monika Kosacka
- Department and Clinic of Pulmonology and Lung Cancers, Wroclaw Medical University, ul. Grabiszynska 105, 53-439 Wroclaw, Poland; (I.P.); (M.K.)
| | - Konrad Pawełczyk
- Department and Clinic of Thoracic Surgery, Wroclaw Medical University, ul. Grabiszynska 105, 53-439 Wroclaw, Poland;
| |
Collapse
|
19
|
Teng J, Zhao Y, Jiang Y, Wang Q, Zhang Y. [Correlation between Gut Microbiota and Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:909-915. [PMID: 32798442 PMCID: PMC7583874 DOI: 10.3779/j.issn.1009-3419.2020.101.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Gene-environment interactions underlie cancer susceptibility and progression. The human body is exposed to and affected by the microenvironment seiscasts of various microorganisms and their metabolites, such as the microenvironment of gut microbiota. The relative abundance of some intestinal microbes in lung cancer patients was significantly different from that in the control group. These studies suggest that gut microbiota may be associated with lung cancer through some ways. At the same time, gut microbiota is relatively manageable environmental variables compared to the external environment we are exposed to, as they are highly quantifiable and relatively stable in the individual. Just as some measures of diagnosis, intervention and treatment of lung cancer targeting gut microbiota have achieved some results in clinical practice. In this review, we mainly discuss the role of gut microbiota and its metabolites in the progression and treatment of lung cancer through certain ways, such as regulation of metabolism, inflammation, and immune response. Finally, based on current research progress, it is inferred that research on gut microbiota may be an effective approach to the precise and personalized medical treatment of lung cancer.
.
Collapse
Affiliation(s)
- Jun Teng
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanfen Zhao
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yunning Jiang
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Wang
- Department of Respiratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| | - Yongsheng Zhang
- Department of Respiratory, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China
| |
Collapse
|
20
|
Mishra SR, Mahapatra KK, Behera BP, Bhol CS, Praharaj PP, Panigrahi DP, Patra S, Singh A, Patil S, Dhiman R, Patra SK, Bhutia SK. Inflammasomes in cancer: Effect of epigenetic and autophagic modulations. Semin Cancer Biol 2020; 83:399-412. [PMID: 33039557 DOI: 10.1016/j.semcancer.2020.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022]
Abstract
Tumour-promoting inflammation is a critical hallmark in cancer development, and inflammasomes are well-known regulators of inflammatory processes within the tumour microenvironment. Different inflammasome components along with the adaptor, apoptosis-associated speck-like protein containing caspase activation and recruitment domain (ASC), and the effector, caspase-1, have a significant influence on tumorigenesis but in a tissue-specific and stage-dependent manner. The downstream products of inflammasome activation, that is the proinflammatory cytokines such as IL-1β and IL-18, regulate tissue homeostasis and induce antitumour immune responses, but in contrast, they can also favour cancer growth and proliferation by directing various oncogenic signalling pathways in cancer cells. Moreover, different epigenetic mechanisms, including DNA methylation, histone modification and noncoding RNAs, control inflammasomes and their components by regulating gene expression during cancer progression. Furthermore, autophagy, a master controller of cellular homeostasis, targets inflammasome-induced carcinogenesis by maintaining cellular homeostasis and removing potential cancer risk factors that promote inflammasome activation in support of tumorigenesis. Here, in this review, we summarize the effect of inflammasome activation in cancers and discuss the role of epigenetic and autophagic regulatory mechanisms in controlling inflammasomes. A proper understanding of the interactions among these key processes will be useful for developing novel therapeutic regimens for targeting inflammasomes in cancer.
Collapse
Affiliation(s)
- Soumya Ranjan Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Bishnu Prasad Behera
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Chandra Sekhar Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Amruta Singh
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Shankargouda Patil
- Department of Maxillofacial Surgery and Diagnostic Sciences, Division of Oral Pathology, College of Dentistry, Jazan University, Saudi Arabia
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
21
|
Sun S, Qu Y, Wen F, Yu H. Initial neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio as prognostic markers in patients with inoperable locally advanced non-small-cell lung cancer. Biomark Med 2020; 14:1341-1352. [PMID: 32808809 DOI: 10.2217/bmm-2019-0583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Aim: To determine whether pretreatment of neutrophil-to-lymphocyte ratio (NLR) or platelet-to-lymphocyte ratio (PLR) has a prognostic value in patients with inoperable locally advanced non-small-cell lung cancer. Materials & methods: A total of 167 patients between 2013 and 2016 were analyzed retrospectively. Results: Appropriate cut-off values for initial NLR (3.06) and PLR (168.03) were determined by receiver operating characteristic curves. High NLR (p < 0.001 and p < 0.001) was related to poor overall survival (OS) and progression-free survival (PFS) via univariate analysis. Multivariable analysis showed that NLR can independently influence OS (hazard ratio: 1.570; p = 0.012) and PFS (hazard ratio: 1.471; p = 0.023). PLR did not correlate with OS or PFS. Conclusion: Pretreatment of NLR could independently predict the prognosis of inoperable locally advanced non-small-cell lung cancer patients, while pretreatment of PLR does not have prognostic value.
Collapse
Affiliation(s)
- Siyu Sun
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District Shenyang, Liaoning Province 110042, PR China
| | - Yanli Qu
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District Shenyang, Liaoning Province 110042, PR China
| | - Fengyun Wen
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District Shenyang, Liaoning Province 110042, PR China
| | - Hong Yu
- Radiation Oncology Department of Thoracic Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District Shenyang, Liaoning Province 110042, PR China
| |
Collapse
|
22
|
Jury D, Daugaard I, Sanders KJ, Hansen LL, Agalliu D, Pedersen IM. miR-151a enhances Slug dependent angiogenesis. Oncotarget 2020; 11:2160-2171. [PMID: 32577162 PMCID: PMC7289531 DOI: 10.18632/oncotarget.27331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 10/19/2019] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miRs) are small non-coding RNAs, that modulate cognate gene expression either by inducing mRNA degradation or by blocking translation, and play crucial and complex roles in tissue homeostasis and during disease initiation and progression. The sprouting of new blood vessels by angiogenesis is critical in vascular development and homeostasis and aberrant angiogenesis is associated with pathological conditions such as ischemia and cancer. We have previously established that miR-151a functions as an onco-miR in non-small cell lung cancer (NSCLC) cells by inducing partial EMT and enhancing tumor growth. Here, we identify anti-miR-151a as a molecule that promotes endothelial cell contacts and barrier properties, suggesting that miR-151a regulates cell-cell junctions. We find that induced miR-151a expression enhances endothelial cell motility and angiogenesis and these functions depend on miR-151a-induced Slug levels. Moreover, we show that miR-151a overexpression enhances tumor-associated angiogenesis in 3D vascularized tumor spheroid assays. Finally, we verify that miR-151a is expressed in the vasculature of normal lung and NSCLC tissue. Our results suggest that miR-151a plays multi-faceted roles in the lung, by regulating multiple functions (cell growth, motility, partial EMT and angiogenesis) in distinct cell types.
Collapse
Affiliation(s)
- Douglas Jury
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Iben Daugaard
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA 92697, USA.,Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark.,Department of Pathology, Aarhus University Hospital, Aarhus DK-8200, Denmark
| | - Katie J Sanders
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Lise Lotte Hansen
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Dritan Agalliu
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Irene Munk Pedersen
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA 92697, USA.,Scintillon Institute, San Diego, CA 92121, USA
| |
Collapse
|
23
|
Zhang Y, Roth JA, Yu H, Ye Y, Xie K, Zhao H, Chang DW, Huang M, Li H, Qu J, Wu X. A 5-microRNA signature identified from serum microRNA profiling predicts survival in patients with advanced stage non-small cell lung cancer. Carcinogenesis 2020; 40:643-650. [PMID: 30428030 DOI: 10.1093/carcin/bgy132] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/11/2018] [Indexed: 12/19/2022] Open
Abstract
Circulating microRNAs (miRNAs) are potential biomarkers for cancer diagnosis, screening and prognosis. This study aimed to identify serum miRNAs as predictors of survival in patients with advanced non-small cell lung cancer (NSCLC). We profiled serum miRNAs in a pilot set of four patients with good survival (>24 months) and four patients with poor survival (<6 months). We selected 140 stably detectable miRNAs and 42 miRNAs reported in literature for further analysis. Expression of these 182 miRNAs was measured using high-throughput polymerase chain reaction assay, and their association with 3-year survival in the discovery (n = 345) and validation (n = 177) cohorts was assessed. Five serum miRNAs (miR-191, miR-28-3p, miR-145, miR-328 and miR-18a) were significantly associated with 3-year overall survival in both cohorts. A combined 5-miRNA risk score was created to assess the cumulative impact of these miRNAs on risk of death. Quartile analysis of the risk score showed significant association with 3-year death risk, with a 4.6-, 6.8- and 9.3-month reduction in median survival time for the second, third and fourth quartiles, respectively. Survival tree analysis also identified distinct risk groups with different 3-year survival durations. Data from The Cancer Genome Atlas revealed all five miRNAs were differentially expressed (P < 0.0001) in paired tumor and normal tissues. Pathway analysis indicated that target genes of these five miRNAs were mainly enriched in inflammatory/immune response pathways and pathways implicated in resistance to chemoradiotherapy and/or targeted therapy. Our results suggested that the 5-miRNA signature could serve as a prognostic predictor in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Yajie Zhang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hao Yu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuanqing Ye
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kunlin Xie
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hua Zhao
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David W Chang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maosheng Huang
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xifeng Wu
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Yu S, Choi HH, Kim IW, Kim TJ. Conditioned medium from asbestos-exposed fibroblasts affects proliferation and invasion of lung cancer cell lines. PLoS One 2019; 14:e0222160. [PMID: 31491033 PMCID: PMC6730856 DOI: 10.1371/journal.pone.0222160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 08/22/2019] [Indexed: 12/28/2022] Open
Abstract
The importance of the role of fibroblasts in cancer microenvironment is well-recognized. However, the relationship between fibroblasts and asbestos-induced lung cancer remains underexplored. To investigate the effect of the asbestos-related microenvironment on lung cancer progression, lung cancer cells (NCI-H358, Calu-3, and A549) were cultured in media derived from IMR-90 lung fibroblasts exposed to 50 mg/L asbestos (chrysotile, amosite, and crocidolite) for 24 h. The kinetics and migration of lung cancer cells in the presence of asbestos-exposed lung fibroblast media were monitored using a real-time cell analysis system. Proliferation and migration of A549 cells increased in the presence of media derived from asbestos-exposed lung fibroblasts than in the presence of media derived from normal lung fibroblasts. We observed no increase in proliferation and migration in lung cancer cells cultured in asbestos-exposed lung cancer cell medium. In contrast, increased proliferation and migration in lung cancer cells exposed to media from asbestos-exposed lung fibroblasts was observed for all types of asbestos. Media derived from lung fibroblasts exposed to other stressors, such as hydrogen peroxide and UV radiation didn't show as similar effect as asbestos exposure. An enzyme-linked immunosorbent assay (ELISA)-based cytokine array identified interleukin (IL)-6 and IL-8, which show pleiotropic regulatory effects on lung cancer cells, to be specifically produced in higher amounts by the three types of asbestos-exposed lung fibroblasts than normal lung fibroblasts. Thus, the present study demonstrated that interaction of lung fibroblasts with asbestos may support the growth and metastasis of lung cancer cells and that chrysotile exposure can lead to lung cancer similar to that caused by amphibole asbestos (amosite and crocidolite).
Collapse
Affiliation(s)
- Seunghye Yu
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Chemical Engineering, Soongsil University, Seoul, Korea
| | - Hee-Hyun Choi
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Il Won Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Korea
| | - Tae-Jung Kim
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail:
| |
Collapse
|
25
|
Chen Y, Chen JQ, Ge MM, Zhang Q, Wang XQ, Zhu JY, Xie CF, Li XT, Zhong CY, Han HY. Sulforaphane inhibits epithelial-mesenchymal transition by activating extracellular signal-regulated kinase 5 in lung cancer cells. J Nutr Biochem 2019; 72:108219. [PMID: 31473507 DOI: 10.1016/j.jnutbio.2019.108219] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/06/2019] [Accepted: 07/24/2019] [Indexed: 12/27/2022]
Abstract
Epithelial-mesenchymal transition (EMT) contributes to the initiation, invasion, metastasis and drug resistance of cancer. The function of extracellular signal-regulated kinase 5 (ERK5) in lung cancer progression remains elusive. In this study, we investigated the effect of sulforaphane (SFN) on lung cancer EMT and the role of ERK5 in its effect. Wound healing and Transwell assays were applied to examine the migratory and invasive capacity in vitro. Quantitative real-time polymerase chain reaction and immunoblotting analysis were performed to investigate the expression of mRNA and protein levels. Small-interfering RNA was used to silence ERK5. Xenograft model was used to confirm the effect of SFN in vivo. Enhanced EMT and decreased ERK5 activation were observed in lung cancer cells in comparison with normal human bronchial epithelial cells. SFN diminished the migratory and invasive capacity of lung cancer cells. Additionally, significantly increased expression of epithelial markers (E-cadherin and ZO-1), decreased expression of mesenchymal markers (N-cadherin and Snail1) and activation of ERK5 were observed after SFN treatment. The inhibitory effect of SFN on lung cancer cell EMT was attenuated by ERK5 silencing. SFN-induced EMT suppression and ERK5 activation were further confirmed in lung cancer xenograft mouse model. The present study illustrated for the first time that ERK5 activation mediates SFN suppression of lung cancer cell EMT. These findings could provide new insights into the function of ERK5 in EMT regulation and the potential therapeutic application of SFN in cancer intervention.
Collapse
Affiliation(s)
- Yue Chen
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jia-Qi Chen
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Miao-Miao Ge
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qi Zhang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xue-Qi Wang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jian-Yun Zhu
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China; Suzhou Digestive Diseases and Nutrition Research Center, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Chun-Feng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiao-Ting Li
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Cai-Yun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Hong-Yu Han
- Department of Clinical Nutrition, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
26
|
陈 浩, 薛 昊, 刘 文, 吴 方, 王 一, 高 红. [Meta-analysis of Platelet Lymphocyte Ratio as A Prognostic Factor for
Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:289-298. [PMID: 31109438 PMCID: PMC6533188 DOI: 10.3779/j.issn.1009-3419.2019.05.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/19/2019] [Accepted: 04/03/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Current research shows that platelet to lymphocyte ratio (PLR) has important prognostic value in renal cell carcinoma, esophageal cancer, gastric cancer, liver cancer and colon cancer. The aim of the study is to evaluate the prognostic value of PLR in non-small cell lung cancer (NSCLC) through meta-analysis. METHODS Literature search for PubMed, EMBASE, Web of Science, Medline, Cochrane Library, China National Knowledge Internet (CNKI), China Biomedical Medicine disc (CBMdisc), VIP, Wanfang Database using computer electronic system to study the association between PLR and overall survival (OS) and disease-free survival (DFS). Each eligible study data is extracted and a meta-analysis is performed using the hazard risk (HR) and 95% confidence interval (95%CI) to assess the prognostic value of PLR, the time limit for the search is to build the library until November 2018. RESULTS We include a total of 15 research literatures involving 5,524 patients for meta-analysis. According to the results of the meta-analysis: The OS of the higher PLR group is significantly lower than that of the lower PLR group (HR=1.69, 95%CI: 1.45-1.97, P<0.000,01, I²=46.2%, Pheterogeneity=0.026); the DFS of the higher PLR group is significantly lower than that of the lower PLR group (HR=1.41, 95%CI: 1.14-1.74, P=0.001, I²=46.2%, Pheterogeneity=0.026). Subgroup analysis show that the OS of the higher PLR group is still significantly lower than the lower PLR group (P<0.05) after grouping by ethnicity, sample size, PLR cutoff value and treatment. CONCLUSIONS Increased PLR is associated with poor prognosis in NSCLC, so PLR may be an important biological predictive marker for NSCLC patients, however, its clinical application still needs to be verified through more research in the future.
Collapse
Affiliation(s)
- 浩然 陈
- 100850 北京,军事科学院军事医学研究院研究生部Academy of Military Medical Sciences, 100850 Beijing, China
| | - 昊 薛
- 100850 北京,军事科学院军事医学研究院研究生部Academy of Military Medical Sciences, 100850 Beijing, China
| | - 文静 刘
- 100071 北京,解放军总医院第五医学中心肺部肿瘤内科Department of Lung Neoplasms, Fifth Medical Center of the Chinese PLA General Hospital, Beijing 100071, China
| | - 方方 吴
- 100071 北京,解放军总医院第五医学中心肺部肿瘤内科Department of Lung Neoplasms, Fifth Medical Center of the Chinese PLA General Hospital, Beijing 100071, China
| | - 一托 王
- 100700 北京,解放军总医院第七医学中心放射科Department of Radiology, Seventh Medical Center of the Chinese PLA General Hospital, Beijing 100700, China
| | - 红军 高
- 100071 北京,解放军总医院第五医学中心肺部肿瘤内科Department of Lung Neoplasms, Fifth Medical Center of the Chinese PLA General Hospital, Beijing 100071, China
| |
Collapse
|
27
|
Huang L, Zhang P, Duan S, Shao H, Gao M, Zhang Q, Feng F. The comparison of two mouse models of inflammation-related lung tumorigenesis induced by benzo(a)pyrene and lipopolysaccharide. Exp Anim 2019; 68:301-306. [PMID: 30867368 PMCID: PMC6699972 DOI: 10.1538/expanim.18-0159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Inflammation-related animal model is necessary to better understanding the association of
inflammation with tumorigenesis. Although mouse models of inflammation-related lung
tumorigenesis on A/J mice strain have been set up in previous study, there is no report on
the model on C57BL/6J mice. In this study, C57BL/6J mice were randomly divided into two
groups and instilled with benzo(a)pyrene [B(a)p] plus lipopolysaccharide (LPS) with
different treatments. Mice in Group I were instilled intratracheally with B(a)p (1
mg/mouse) and LPS (5 µg/mouse), once a week for 4 times, on Tuesday and
Friday, respectively [the week of the last time of B(a)p treatment named Week 0]. At Week
4, mice continued to be treated with LPS, once every four weeks for 5 times. Mice in Group
II were exposed to B(a)p (1 mg/mouse, once a week for 4 times) and 3 weeks later instilled
intratracheally with LPS (2.5 µg/mouse) once every three weeks for 5
times. At Week 30, the incidence, number, size and histopathology of lung tumor in two
models were compared. The tumor incidence (96.97%) and mean tumor count (13.0 ± 12.4) of
mice in Group II were significantly increased compared with those in Group I (69.23%, 4.9
± 5.1), respectively. In addition, smaller tumors (≤1 mm) were more abundant in Group II
than Group I. Histopathological examination found the tumors induced by B(a)p plus LPS in
Group II were more advanced tumors. In conclusion, a better mouse model of
inflammation-related lung tumorigenesis induced by B(a)p plus LPS in C57BL/6J mice was set
up successfully.
Collapse
Affiliation(s)
- Li Huang
- Department of Toxicology, College of Public Health, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Peng Zhang
- Department of Bone and soft tissue cancer, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), No. 127 Dongming Road, Zhengzhou, Henan 450008, China
| | - Shuyin Duan
- Department of Toxicology, College of Public Health, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hua Shao
- Department of Toxicology, College of Public Health, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Min Gao
- Department of Toxicology, College of Public Health, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Qiao Zhang
- Department of Toxicology, College of Public Health, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, No. 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| |
Collapse
|
28
|
Huang L, Duan S, Shao H, Zhang A, Chen S, Zhang P, Wang N, Wang W, Wu Y, Wang J, Liu H, Yao W, Zhang Q, Feng F. NLRP3 deletion inhibits inflammation-driven mouse lung tumorigenesis induced by benzo(a)pyrene and lipopolysaccharide. Respir Res 2019; 20:20. [PMID: 30696442 PMCID: PMC6352353 DOI: 10.1186/s12931-019-0983-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/13/2019] [Indexed: 12/13/2022] Open
Abstract
Background Inflammatory micro-environment has been proposed to play a critical role in lung tumorigenesis. NLRP3 is known as an intracellular receptor involving inflammation and has been reported which is increasingly associated with tumor development, but the role in inflammation-driven lung cancer has not been fully clarified. In this study, we investigated whether lipopolysaccharide (LPS)-induced pulmonary inflammation could contribute to lung tumorigenesis induced by benzo(a)pyrene [B(a)p] in C57BL/6J mice and the role of NLRP3 in the pathogenesis. Methods NLRP3−/− mice and C57BL/6J mice (wide-type, WT) were instilled intratracheally with B(a)p (1 mg/mouse) once a week for 4 times [the week of the last time of B(a)p treatment named Week 0], and mice were then instilled intratracheally with LPS at Week 3, 2.5 μg/mouse, once every three weeks for 5 times. At Week 30, the incidence, number, size and histopathology of lung tumor were analyzed. Results Mice exposed to B(a)p or B(a)p plus LPS could induce lung tumors, whereas LPS or vehicles treatment could not induce lung tumorigenesis. In WT mice, B(a)p plus LPS exposure significantly increased tumor incidence, mean tumor count and tumor size of visible tumors of lungs compared with B(a)p treatment alone, and NLRP3 deletion inhibited lung tumorigenesis induced by B(a)p or B(a)p plus LPS. Histopathological examination found LPS-induced pulmonary inflammatory changes enhanced lung tumorigenesis induced by B(a)p in WT mice, deletion of NLRP3 improved the inflammatory changes induced by LPS and the number and size of pathological tumor nests induced by B(a)p or B(a)p plus LPS. In addition, we found B(a)p treatment and B(a)p plus LPS treatment predominately induced the development of adenoma. Conclusion LPS enhanced B(a)p-induced lung tumorigenesis in WT and NLRP3−/− mice of C57BL/6J strain, and NLRP3 deletion inhibits lung tumorigenesis induced by B(a)p or B(a)p plus LPS.
Collapse
Affiliation(s)
- Li Huang
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Shuyin Duan
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Hua Shao
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Aihua Zhang
- Department of Nephrology, the Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuang Chen
- Institute of Pediatrics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Zhang
- Department of Bone and Soft Tissue Cancer, Cancer, the Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, Henan, China
| | - Na Wang
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Wei Wang
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Yongjun Wu
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Jing Wang
- Department of Pulmonary Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hong Liu
- Department of Pulmonary Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wu Yao
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Qiao Zhang
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, No.100 Kexue Avenue, Zhengzhou, 450001, Henan province, China.
| |
Collapse
|
29
|
Belgodere JA, King CT, Bursavich JB, Burow ME, Martin EC, Jung JP. Engineering Breast Cancer Microenvironments and 3D Bioprinting. Front Bioeng Biotechnol 2018; 6:66. [PMID: 29881724 PMCID: PMC5978274 DOI: 10.3389/fbioe.2018.00066] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) is a critical cue to direct tumorigenesis and metastasis. Although two-dimensional (2D) culture models have been widely employed to understand breast cancer microenvironments over the past several decades, the 2D models still exhibit limited success. Overwhelming evidence supports that three dimensional (3D), physiologically relevant culture models are required to better understand cancer progression and develop more effective treatment. Such platforms should include cancer-specific architectures, relevant physicochemical signals, stromal-cancer cell interactions, immune components, vascular components, and cell-ECM interactions found in patient tumors. This review briefly summarizes how cancer microenvironments (stromal component, cell-ECM interactions, and molecular modulators) are defined and what emerging technologies (perfusable scaffold, tumor stiffness, supporting cells within tumors and complex patterning) can be utilized to better mimic native-like breast cancer microenvironments. Furthermore, this review emphasizes biophysical properties that differ between primary tumor ECM and tissue sites of metastatic lesions with a focus on matrix modulation of cancer stem cells, providing a rationale for investigation of underexplored ECM proteins that could alter patient prognosis. To engineer breast cancer microenvironments, we categorized technologies into two groups: (1) biochemical factors modulating breast cancer cell-ECM interactions and (2) 3D bioprinting methods and its applications to model breast cancer microenvironments. Biochemical factors include matrix-associated proteins, soluble factors, ECMs, and synthetic biomaterials. For the application of 3D bioprinting, we discuss the transition of 2D patterning to 3D scaffolding with various bioprinting technologies to implement biophysical cues to model breast cancer microenvironments.
Collapse
Affiliation(s)
- Jorge A. Belgodere
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Connor T. King
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Jacob B. Bursavich
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Matthew E. Burow
- Department of Medicine, Section Hematology/Oncology, Tulane University, New Orleans, LA, United States
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Jangwook P. Jung
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
30
|
Fregni G, Quinodoz M, Möller E, Vuille J, Galland S, Fusco C, Martin P, Letovanec I, Provero P, Rivolta C, Riggi N, Stamenkovic I. Reciprocal modulation of mesenchymal stem cells and tumor cells promotes lung cancer metastasis. EBioMedicine 2018; 29:128-145. [PMID: 29503225 PMCID: PMC5925622 DOI: 10.1016/j.ebiom.2018.02.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/14/2018] [Accepted: 02/19/2018] [Indexed: 12/15/2022] Open
Abstract
Metastasis is a multi-step process in which direct crosstalk between cancer cells and their microenvironment plays a key role. Here, we assessed the effect of paired tumor-associated and normal lung tissue mesenchymal stem cells (MSCs) on the growth and dissemination of primary human lung carcinoma cells isolated from the same patients. We show that the tumor microenvironment modulates MSC gene expression and identify a four-gene MSC signature that is functionally implicated in promoting metastasis. We also demonstrate that tumor-associated MSCs induce the expression of genes associated with an aggressive phenotype in primary lung cancer cells and selectively promote their dissemination rather than local growth. Our observations provide insight into mechanisms by which the stroma promotes lung cancer metastasis. Distinct gene expression profiles distinguish normal lung and tumor-associated MSCs. MSCs induce EMT- and hypoxia-related genes in primary tumor cells and promote their metastatic potential. A 4-gene T-MSC signature is involved in MSC-induced metastasis promotion.
The tumor microenvironment, which includes mesenchymal stem cells (MSCs) among many other stromal cell types, plays a fundamental role in cancer metastasis. Although MSCs are suggested to participate in tumor progression, most studies thus far have been performed on bone marrow-derived MSCs and cancer cell lines. Using primary human pulmonary MSCs and paired lung cancer cells, we show that tumor cells modulate MSCs to acquire properties, including a four-gene signature, which allow them to promote tumor dissemination. Our results provide insight into the mutual cancer cell-stromal cell modulation that drives tumor dissemination.
Collapse
Affiliation(s)
- Giulia Fregni
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Mathieu Quinodoz
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, 1011, Switzerland
| | - Emely Möller
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Joanna Vuille
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Sabine Galland
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Carlo Fusco
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Patricia Martin
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Igor Letovanec
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Paolo Provero
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, IRCCS, Milan20132, Italy; Dept. of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy
| | - Carlo Rivolta
- Department of Computational Biology, Unit of Medical Genetics, University of Lausanne, 1011, Switzerland; Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 9HN, UK
| | - Nicolo Riggi
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland
| | - Ivan Stamenkovic
- Experimental Pathology Service, CHUV and University of Lausanne, 1011, Switzerland.
| |
Collapse
|
31
|
Metastatic Lung Lesions as a Preferred Resection Site for Immunotherapy With Tumor Infiltrating Lymphocytes. J Immunother 2018; 39:218-22. [PMID: 27163742 DOI: 10.1097/cji.0000000000000124] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Adoptive cell therapy with tumor infiltrating lymphocytes (TIL) yields 50% response rates in metastatic melanoma and shows promising clinical results in other solid tumors. Autologous TIL cultures are isolated from resected tumor tissue, expanded ex vivo to large numbers and reinfused to the preconditioned patient. In this prospective study, we validate the origin of the tumor biopsy and its effect on T-cell function and clinical response. One hundred forty-four patients underwent surgery and 79 patients were treated with TIL adoptive cell therapy. Cultures from lung tissue were compared with other origins. The success rate of establishing TIL culture from lung tissue was significantly higher compared with nonlung tissue (94% vs. 72%, respectively, P≤0.003). Lung-derived TIL cultures gave rise to higher cell numbers (P≤0.011) and exhibited increased in vitro antitumor reactivity. The average fold expansion for lung-derived TIL during a rapid expansion procedure was 1349±557 compared with 1061±473 for nonlung TIL (P≤0.038). Patients treated with TIL cultures of lung origin (compared with nonlung) had prolonged median overall survival (29 vs. 9.5 mo; P≤0.065). Given the remarkable advancement in minimally invasive thoracic surgery and the results of this study, we suggest efforts should be taken to resect lung metastasis rather than other sites to generate TIL cultures for clinical use.
Collapse
|
32
|
Ma XM, Sun X, Yang GW, Yu MW, Zhang GL, Yu J, Zhang Y, Wang XM. The platelet-to-lymphocyte ratio as a predictor of patient outcomes in ovarian cancer: a meta-analysis. Climacteric 2017; 20:448-455. [PMID: 28569074 DOI: 10.1080/13697137.2017.1326894] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The platelet-to-lymphocyte ratio (PLR) is a predictive clinical biomarker for different cancers. However, the results of several studies investigating the association between the PLR and the prognosis of ovarian cancer have been inconclusive. Therefore, there is a need to conduct a meta-analysis to estimate the prognostic value of the PLR in ovarian cancer. METHODS We searched the EMBASE, Medline, PubMed, and Web of Science databases to identify clinical studies that had evaluated the association between the PLR and ovarian cancer prognosis. Outcomes evaluated included overall survival (OS) and progression-free survival (PFS). We also analyzed PLR differences between malignant ovarian masses and the controls. RESULTS Twelve relevant studies that comprised 2340 patients were selected for the meta-analysis. The results revealed that elevated PLR was significantly associated with poor OS (hazard ratio (HR) 1.63, 95% confidence interval (CI) 1.05-2.56, p < 0.01) and PFS (HR 1.61, 95% CI 1.03-2.51, p < 0.01). The PLRs in malignant cases were higher than in controls (mean difference = 63.57, 95% CI 39.47-87.66, p < 0.00001). CONCLUSION An elevated PLR is associated with poor prognosis in patients with ovarian cancer. The PLR could be employed as a prognostic marker in patients with ovarian cancer.
Collapse
Affiliation(s)
- X-M Ma
- a School of Graduates , Beijing University of Chinese Medicine , Beijing , China.,b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - X Sun
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - G-W Yang
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - M-W Yu
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - G-L Zhang
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - J Yu
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - Y Zhang
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| | - X-M Wang
- b Department of Oncology , Beijing Hospital of Traditional Chinese Medicine Affiliated to Capital Medical University , Beijing , China
| |
Collapse
|
33
|
Critical roles of mucin-1 in sensitivity of lung cancer cells to tumor necrosis factor-alpha and dexamethasone. Cell Biol Toxicol 2017; 33:361-371. [DOI: 10.1007/s10565-017-9393-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 04/18/2017] [Indexed: 12/16/2022]
|
34
|
Yang D, Cao X, Wang F, Jiang H, Feng D, Guo H, Du L, Jin Y, Chen Y, Yin X, Li C. LFG-500, a novel synthetic flavonoid, suppresses epithelial-mesenchymal transition in human lung adenocarcinoma cells by inhibiting NLRP3 in inflammatory microenvironment. Cancer Lett 2017; 400:137-148. [PMID: 28461245 DOI: 10.1016/j.canlet.2017.04.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/17/2022]
Abstract
Increasing evidence indicates that inflammatory microenvironment facilitates tumor metastasis. Here, we found that LFG-500, a novel synthetic flavonoid, significantly inhibited epithelial-mesenchymal transition (EMT) in human lung adenocarcinoma A549 and H1299 cells co-cultured with LPS-challenged THP-1 cells or cultured in THP-1 cell-derived conditioned medium. Moreover, we found that TNF-α is a direct and decisive factor for promoting EMT and LFG-500 suppressed TNF-α-induced EMT and cell motility. NLRP3 knockdown inactivated NLRP3 inflammasome, which subsequently inhibited EMT and blocked cell migration, indicating that TNF-α-induced EMT requires the NLRP3 inflammasome. LFG-500 inhibited the activation of the NLRP3 inflammasome, thus inhibiting EMT. Moreover, LFG-500 treatment significantly inhibited metastasis in vivo by downregulating NLRP3 expression. Importantly, we found that NLRP3 was highly expressed in high-grade lung adenocarcinoma and that its expression was correlated with lymph node metastasis. NLRP3 and vimentin levels were significantly increased in matched metastatic lymph nodes. Moreover, a significant positive correlation was observed between their levels. Together, these results suggest that LFG-500 markedly suppresses EMT by inhibiting the NLRP3 inflammasome in the inflammatory microenvironment and that NLRP3 is a potential biomarker of lung adenocarcinoma metastasis.
Collapse
Affiliation(s)
- Dan Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Xin Cao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Fan Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Haijing Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Dingding Feng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Hao Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China
| | - Yingliang Jin
- School of Public Health, Xuzhou Medical University, Xuzhou, PR China
| | - Yansu Chen
- School of Public Health, Xuzhou Medical University, Xuzhou, PR China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China.
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, PR China.
| |
Collapse
|
35
|
Wang L, Zhu B, Zhang M, Wang X. Roles of immune microenvironment heterogeneity in therapy-associated biomarkers in lung cancer. Semin Cell Dev Biol 2017; 64:90-97. [DOI: 10.1016/j.semcdb.2016.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022]
|
36
|
Zhu HX, Shi L, Zhang Y, Zhu YC, Bai CX, Wang XD, Zhou JB. Myocyte enhancer factor 2D provides a cross-talk between chronic inflammation and lung cancer. J Transl Med 2017; 15:65. [PMID: 28340574 PMCID: PMC5366127 DOI: 10.1186/s12967-017-1168-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/19/2017] [Indexed: 01/11/2023] Open
Abstract
Background Lung cancer is the leading cause of cancer-related morbidity and mortality worldwide. Patients with chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD), are exposed to a higher risk of developing lung cancer. Chronic inflammation may play an important role in the lung carcinogenesis among those patients. The present study aimed at identifying candidate biomarker predicting lung cancer risk among patients with chronic respiratory diseases. Methods We applied clinical bioinformatics tools to analyze different gene profile datasets with a special focus on screening the potential biomarker during chronic inflammation-lung cancer transition. Then we adopted an in vitro model based on LPS-challenged A549 cells to validate the biomarker through RNA-sequencing, quantitative real time polymerase chain reaction, and western blot analysis. Results Bioinformatics analyses of the 16 enrolled GSE datasets from Gene Expression Omnibus online database showed myocyte enhancer factor 2D (MEF2D) level significantly increased in COPD patients coexisting non-small-cell lung carcinoma (NSCLC). Inflammation challenge increased MEF2D expression in NSCLC cell line A549, associated with the severity of inflammation. Extracellular signal-regulated protein kinase inhibition could reverse the up-regulation of MEF2D in inflammation-activated A549. MEF2D played a critical role in NSCLC cell bio-behaviors, including proliferation, differentiation, and movement. Conclusions Inflammatory conditions led to increased MEF2D expression, which might further contribute to the development of lung cancer through influencing cancer microenvironment and cell bio-behaviors. MEF2D might be a potential biomarker during chronic inflammation-lung cancer transition, predicting the risk of lung cancer among patients with chronic respiratory diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1168-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hai-Xing Zhu
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Respiratory Research Institute, Shanghai, China
| | - Lin Shi
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yong Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Respiratory Research Institute, Shanghai, China
| | - Yi-Chun Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chun-Xue Bai
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Respiratory Research Institute, Shanghai, China
| | - Xiang-Dong Wang
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China.,Fudan University Center for Clinical Bioinformatics, Shanghai, China
| | - Jie-Bai Zhou
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Respiratory Research Institute, Shanghai, China.
| |
Collapse
|
37
|
Jian H, Fangrong S, Haitao H, Chunhua L, Guangbo Z. Th1high in tumor microenvironment is an indicator of poor prognosis for patients with NSCLC. Oncotarget 2017; 8:13116-13125. [PMID: 28061450 PMCID: PMC5355081 DOI: 10.18632/oncotarget.14471] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/20/2016] [Indexed: 02/07/2023] Open
Abstract
CD4+Th subsets play an important role in tumor progression but their expression characteristics and clinical significance in human tumor microenvironment remains unclear. In this study, we aim to analyze the expression and clinical significance of tissue-infiltrating Th1, Th2 and Th17 in lung cancer by flow cytometry. We found that the frequency of CD3+CD4+IFN-γ+Th1 in tumor nest was significantly lower than that in tumor boundary, adjacent normal lung tissue or corresponding lymph node tissue; the frequency of CD3+CD4+IL-4+Th2 in tumor nest was significantly higher than that in tumor boundary, adjacent normal lung tissue or corresponding lymph node tissue; the frequency of CD3+CD4+IL-17+Th17 in tumor nest was significantly lower than that in tumor boundary, but not adjacent normal tissue or corresponding lymph node tissue. Survival analysis of 2-years survival after surgery showed that Th1high group was significantly lower compared with Th1low group; Th2high and Th17low is a good prognosis index compared with the Th2low and Th17high groups respectively, but this difference failed to significance. In addition, we also found that PD-1 expression showed a high level on lung tumor tissues and adjacent non- tumor tissue infiltrating T cells, and no significant difference was found between the two groups. However PD-L1 on CD45+CD14+mononcytes/macrophages in tumor tissue show a significantly higher level compared with that in adjacent nontumor tissues. In vitro stimulation experiments showed that IFN-γ could significantly increase PD-L1 expression on monocyte. In conclusion, we for the first time found Th1high is a poor indicator for prognosis of lung cancer.
Collapse
Affiliation(s)
- Huang Jian
- Department of respiratory, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
- Department of Emergency, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Shen Fangrong
- Clinical Immunology Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Huang Haitao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Ling Chunhua
- Department of respiratory, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Zhang Guangbo
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215007, China
| |
Collapse
|
38
|
PPAR Agonists for the Prevention and Treatment of Lung Cancer. PPAR Res 2017; 2017:8252796. [PMID: 28316613 PMCID: PMC5337885 DOI: 10.1155/2017/8252796] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/08/2016] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the most common and most fatal of all malignancies worldwide. Furthermore, with more than half of all lung cancer patients presenting with distant metastases at the time of initial diagnosis, the overall prognosis for the disease is poor. There is thus a desperate need for new prevention and treatment strategies. Recently, a family of nuclear hormone receptors, the peroxisome proliferator-activated receptors (PPARs), has attracted significant attention for its role in various malignancies including lung cancer. Three PPARs, PPARα, PPARβ/δ, and PPARγ, display distinct biological activities and varied influences on lung cancer biology. PPARα activation generally inhibits tumorigenesis through its antiangiogenic and anti-inflammatory effects. Activated PPARγ is also antitumorigenic and antimetastatic, regulating several functions of cancer cells and controlling the tumor microenvironment. Unlike PPARα and PPARγ, whether PPARβ/δ activation is anti- or protumorigenic or even inconsequential currently remains an open question that requires additional investigation. This review of current literature emphasizes the multifaceted effects of PPAR agonists in lung cancer and discusses how they may be applied as novel therapeutic strategies for the disease.
Collapse
|
39
|
Zhang Y, Wang DC, Shi L, Zhu B, Min Z, Jin J. Genome analyses identify the genetic modification of lung cancer subtypes. Semin Cancer Biol 2017; 42:20-30. [DOI: 10.1016/j.semcancer.2016.11.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/08/2016] [Indexed: 12/15/2022]
|
40
|
Wang DC, Shi L, Zhu Z, Gao D, Zhang Y. Genomic mechanisms of transformation from chronic obstructive pulmonary disease to lung cancer. Semin Cancer Biol 2017; 42:52-59. [DOI: 10.1016/j.semcancer.2016.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/08/2016] [Indexed: 01/17/2023]
|
41
|
Lu J, Wang W, Xu M, Li Y, Chen C, Wang X. A global view of regulatory networks in lung cancer: An approach to understand homogeneity and heterogeneity. Semin Cancer Biol 2016; 42:31-38. [PMID: 27894849 DOI: 10.1016/j.semcancer.2016.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022]
Abstract
A number of new biotechnologies are used to identify potential biomarkers for the early detection of lung cancer, enabling a personalized therapy to be developed in response. The combinatorial cross-regulation of hundreds of biological function-specific transcription factors (TFs) is defined as the understanding of regulatory networks of molecules within the cell. Here we integrated global databases with 537 patients with lung adenocarcinoma (ADC), 140 with lung squamous carcinoma (SCC), 9 with lung large-cell carcinoma (LCC), 56 with small-cell lung cancer (SCLC), and 590 without cancer with the understanding of TF functions. The present review aims at the homogeneity or heterogeneity of gene expression profiles among subtypes of lung cancer. About 5, 136, 52, or 16 up-regulated or 19, 24, 122, or 97down-regulated type-special TF genes were identified in ADC, SCC, LCC or SCLC, respectively. DNA-binding and transcription regulator activity associated genes play a dominant role in the differentiation of subtypes in lung cancer. Subtype-specific TF gene regulatory networks with elements should be an alternative for diagnostic and therapeutic targets for early identification of lung cancer and can provide insightful clues to etiology and pathogenesis.
Collapse
Affiliation(s)
- Jiapei Lu
- Department of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - William Wang
- Department of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Menglin Xu
- Department of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuping Li
- Department of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Department of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiangdong Wang
- Department of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
42
|
Wang L, Wang H, Song D, Xu M, Liebmen M. New strategies for targeting drug combinations to overcome mutation-driven drug resistance. Semin Cancer Biol 2016; 42:44-51. [PMID: 27840276 DOI: 10.1016/j.semcancer.2016.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022]
Abstract
Targeted therapies are suggested as an effective alternative for patients with cancer that harbor mutations, but treatment outcomes are frequently limited by primary or acquired drug resistance. The present review describes potential mechanisms of primary or acquired drug resistances to provide a resource for considering how to be overcome. We focus on strategies of targeted drug combinations to minimize the development of drug resistance within the context how resistance develops. Strategies benefit from the combined use of "omics" technologies, i.e., high-throughput functional genomics data, pharmacogenomics, or genome-wide CRISPR-Cas9 screening, to analyze and design targeted drug combinations for mutation-driven drug resistance. We also introduce new insights towards pathway-centric combined therapies as an alternative to overcome the heterogeneity and benefit patient prognoses.
Collapse
Affiliation(s)
- Linyan Wang
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China.
| | - Haiyun Wang
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Dongli Song
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China
| | - Menglin Xu
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China
| | - Michael Liebmen
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China.
| |
Collapse
|
43
|
Regulatory roles of epigenetic modulators, modifiers and mediators in lung cancer. Semin Cancer Biol 2016; 42:4-12. [PMID: 27840279 DOI: 10.1016/j.semcancer.2016.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/08/2016] [Indexed: 12/19/2022]
Abstract
Lung cancer as the leading cause of cancer-related deaths can be initiated and progressed by the interaction between dynamically genetic and epigenetic elements, although mechanisms mediating lung cancer development and progression remain unclear. Tumor progenitor genes may contribute to lung carcinogenesis and cancer progression, are epigenetically disrupted at the early stages of malignancies even before mutations, and alter cell differentiation throughout tumor evolution. The present review explores potential roles and mechanisms of epigenetic modulators, modifiers and mediators in the development of lung cancer. We also overviewed potential mechanisms by which epigenetic modulators, modifiers and mediators control and regulate 3D nuclear architectures, and discussed translational efforts to epigenetic modifications for treatment of lung cancer. Deep understanding of epigenetic modulators, modifiers and mediators will benefit the discovery and development of new diagnostics and therapies for lung cancer.
Collapse
|
44
|
Wang DC, Wang X. Systems heterogeneity: An integrative way to understand cancer heterogeneity. Semin Cell Dev Biol 2016; 64:1-4. [PMID: 27552921 DOI: 10.1016/j.semcdb.2016.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/19/2016] [Indexed: 12/14/2022]
Abstract
The concept of systems heterogeneity was firstly coined and explained in the Special Issue, as a new alternative to understand the importance and complexity of heterogeneity in cancer. Systems heterogeneity can offer a full image of heterogeneity at multi-dimensional functions and multi-omics by integrating gene or protein expression, epigenetics, sequencing, phosphorylation, transcription, pathway, or interaction. The Special Issue starts with the roles of epigenetics in the initiation and development of cancer heterogeneity through the interaction between permanent genetic mutations and dynamic epigenetic alterations. Cell heterogeneity was defined as the difference in biological function and phenotypes between cells in the same organ/tissue or in different organs, as well as various challenges, as exampled in telocytes. The single cell heterogeneity has the value of identifying diagnostic biomarkers and therapeutic targets and clinical potential of single cell systems heterogeneity in clinical oncology. A number of signaling pathways and factors contribute to the development of systems heterogeneity. Proteomic heterogeneity can change the strategy and thinking of drug discovery and development by understanding the interactions between proteins or proteins with drugs in order to optimize drug efficacy and safety. The association of cancer heterogeneity with cancer cell evolution and metastasis was also overviewed as a new alternative for diagnostic biomarkers and therapeutic targets in clinical application.
Collapse
Affiliation(s)
- Diane Catherine Wang
- Minghang Hospital of Fudan University, Shanghai Medical College, Shanghai, China
| | - Xiangdong Wang
- Minghang Hospital of Fudan University, Shanghai Medical College, Shanghai, China.
| |
Collapse
|
45
|
Geng S, Zheng Y, Meng M, Guo Z, Cao N, Ma X, Du Z, Li J, Duan Y, Du G. Gingerol Reverses the Cancer-Promoting Effect of Capsaicin by Increased TRPV1 Level in a Urethane-Induced Lung Carcinogenic Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:6203-6211. [PMID: 27436516 DOI: 10.1021/acs.jafc.6b02480] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Both gingerol and capsaicin are agonists of TRPV1, which can negatively control tumor progression. This study observed the long-term effects of oral administration of 6-gingerol alone or in combination with capsaicin for 20 weeks in a urethane-induced lung carcinogenic model. We showed that lung carcinoma incidence and multiplicity were 70% and 21.2 ± 3.6, respectively, in the control versus 100% and 35.6 ± 5.2 in the capsaicin group (P < 0.01) and 50% and 10.8 ± 3.1 in the 6-gingerol group (P < 0.01). The combination of 6-gingerol and capsaicin reversed the cancer-promoting effect of capsaicin (carcinoma incidence of 100% versus 20% and multiplicity of 35.6 ± 5.2 versus 4.7 ± 2.3; P < 0.001). The cancer-promoting effect of capsaicin was due to increased epidermal growth-factor receptor (EGFR) level by decreased transient receptor potential vanilloid type-1 (TRPV1) level (P < 0.01) . The capsaicin-decreased EGFR level subsequently reduced levels of nuclear factor-κB (NF-κB) and cyclin D1 that favored enhanced lung epithelial proliferation and epithelial-mesenchymal transition (EMT) during lung carcinogenesis (P < 0.01). In contrast, 6-gingerol promoted TRPV1 level and drastically decreased the levels of EGFR, NF-κB, and cyclin D1 that favored reduced lung epithelial proliferation and EMT (P < 0.01). This study provides valuable information for the long-term consumption of chili-pepper-rich diets to decrease the risk of cancer development.
Collapse
Affiliation(s)
- Shengnan Geng
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Yaqiu Zheng
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Mingjing Meng
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Zhenzhen Guo
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Ning Cao
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Xiaofang Ma
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Zhenhua Du
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Jiahuan Li
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| | - Yongjian Duan
- Department of Oncology, The First Hospital Affiliated to Henan University , Kaifeng, Henan 475001, China
| | - Gangjun Du
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming District, Kaifeng, Henan 475004, China
| |
Collapse
|
46
|
Kim SK, Wu CC, Horowitz DP. Stereotactic body radiotherapy for the pancreas: a critical review for the medical oncologist. J Gastrointest Oncol 2016; 7:479-86. [PMID: 27284482 DOI: 10.21037/jgo.2015.10.01] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
With recent advances in imaging modalities and radiation therapy, stereotactic body radiotherapy (SBRT) has allowed for the delivery of high doses of radiation with accuracy and precision. As such, SBRT has generated favorable results in the treatment of several cancers. Although the role of radiation has been controversial for the treatment of pancreatic ductal adenocarcinoma (PDAC) due to rather lackluster results in clinical trials, SBRT may offer improved outcomes, enhance the quality of life, and aid in palliative care settings for PDAC patients. This review delineates the role of SBRT in the treatment of PDAC, presents the defining principles of radiation biology and the radiation oncology work flow, and discusses the prospects of new treatment regimens involving tumor immunology and radiation therapy.
Collapse
Affiliation(s)
- Samuel K Kim
- Department of Radiation Oncology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY 10032, USA
| | - Cheng-Chia Wu
- Department of Radiation Oncology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY 10032, USA
| | - David P Horowitz
- Department of Radiation Oncology, New York Presbyterian Hospital, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
47
|
Liao CG, Yao L, Xie W, Liu L, Wu SD, Lu N, Huang JG, Kong LM, Zhang HL. Basigin-2 upregulated by receptor activator of NF-κB ligand enhances lung cancer-induced osteolytic lesions. Cancer Cell Int 2016; 16:28. [PMID: 27042161 PMCID: PMC4818914 DOI: 10.1186/s12935-016-0302-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Lung cancer bone metastasis causes poor prognosis. Basigin-2, a novel cancer-associated biomarker, is upregulated in lung cancer and has been linked with tumor progression. But little is known about the role of basigin-2 in lung cancer bone metastasis and osteolytic lesion. METHODS Basigin-2 expression was evaluated in biopsy tissue specimens of 20 lung cancer patients with bone metastases via immunohistochemistry. Invasion assay and MTT proliferation assay were performed to test the invasion and proliferation of lung cancer cell after modulated basigin-2 expression. The osteoclastic activity of basigin-2 was detected in tibia cancer model by injected of lung cancer cells. The regulation role of receptor activator of NF-κB ligand (RANKL) on basigin-2 and its downstream molecules were measured by real-time quantitative RT-PCR, gelatin zymography and western blot analysis. RESULTS We found that basigin-2 was highly expressed in lung cancer bone metastases. Then, we demonstrated that basigin-2 could promote lung cancer cells invasion, metastasis and proliferation through upregulating metalloproteinases-2 (MMP-2), MMP-9 and vascular endothelial growth factor (VEGF) expression. The lung cancer cells overexpressing basigin-2 strongly induced the osteolytic lesions in immunodeficient mice, which were reduced by treatment with basigin-2 blocking antibody. Furthermore, we explored the enhanced basigin-2 molecular mechanism in lung cancer bone metastasis. Our results indicated the RANKL, pivotal for the control of bone resorption, could increase basigin-2 and its downstream molecules MMP-2, MMP-9 and VEGF expression in vitro. CONCLUSIONS Basigin-2 upregulated by RANKL induces MMPs and VEGF, which may increase lung cancer cell metastasis ability and support osteoclastic activity. Thus, our data suggest important roles for basigin-2 in lung cancer-induced osteolytic lesion and implicate this protein potential application as a target for lung cancer bone metastasis therapy.
Collapse
Affiliation(s)
- Cheng-Gong Liao
- Department of Oncology, Tangdu Hospital, Cancer Institute, Fourth Military Medical University, Xi'an, 710038 People's Republic of China.,Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Li Yao
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038 People's Republic of China
| | - Wei Xie
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Lili Liu
- Department of Oncology, Tangdu Hospital, Cancer Institute, Fourth Military Medical University, Xi'an, 710038 People's Republic of China
| | - Sheng-Da Wu
- Cadet Brigade, Fourth Military Medical University, Xi'an, 710032 People's Republic of China
| | - Ning Lu
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Jian-Guo Huang
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Ling-Min Kong
- Cell Engineering Research Center and Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032 People's Republic of China
| | - He-Long Zhang
- Department of Oncology, Tangdu Hospital, Cancer Institute, Fourth Military Medical University, Xi'an, 710038 People's Republic of China
| |
Collapse
|
48
|
Qiang G, Liang C, Xiao F, Yu Q, Wen H, Song Z, Tian Y, Shi B, Guo Y, Liu D. Prognostic significance of platelet-to-lymphocyte ratio in non-small-cell lung cancer: a meta-analysis. Onco Targets Ther 2016; 9:869-76. [PMID: 26955285 PMCID: PMC4768894 DOI: 10.2147/ott.s96804] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background The platelet-to-lymphocyte ratio (PLR) is a useful predictive factor in several cancers. However, the prognostic value of PLR in patients with non-small-cell lung cancer (NSCLC) is still indistinct. Therefore, it was necessary for us to perform a meta-analysis to assess the prognostic value of PLR in patients with NSCLC. Methods A systematic literature search was performed by using PubMed, EMBASE, and Web of Science databases for relevant studies until May 2015. Published studies investigating the association between PLR and overall survival (OS) and disease-free survival (DFS) were selected. Data from each eligible study were extracted. A meta-analysis was performed to analyze the prognostic value of PLR by using the hazard ratio (HR) and 95% confidence intervals (95% CI). Results A total of seven studies involving 1,554 patients were included in our meta-analysis. Our pooled results demonstrated that high PLR was associated with poor OS (HR: 1.60, 95% CI: 1.34–1.90, I2=22.3%, Pheterogeneity = 0.259) and DFS (HR: 1.38, 95% CI: 1.11–1.73, I2=0%, Pheterogeneity = 0.482). Subgroup analysis between PLR and OS was performed in a further investigation. When the patients were segregated according to ethnicity, sample size, cutoff value, stage, and treatment modality, high PLR was also significantly correlated with OS. There was no significant heterogeneity among included studies. Conclusion High PLR is associated with poor prognosis in patients with NSCLC. PLR may be a significant predictive biomarker in patients with NSCLC.
Collapse
Affiliation(s)
- Guangliang Qiang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Chaoyang Liang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Qiduo Yu
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Huanshun Wen
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Zhiyi Song
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Yanchu Tian
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Bin Shi
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Yongqing Guo
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Deruo Liu
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, People's Republic of China
| |
Collapse
|
49
|
Potential roles of telocytes in lung diseases. Semin Cell Dev Biol 2016; 55:31-9. [PMID: 26855021 DOI: 10.1016/j.semcdb.2016.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 12/20/2022]
Abstract
Telocytes (TCs) are a unique type of interstitial cells with specific, extremely long prolongations named telopodes (Tps), as shown by immune-positive staining against CD34, c-kit and vimentin. They were found in many organs of mammals, with potential biological functions, including the trachea and lung, even though the exact function remains unclear. Here, we give a historical overview of the TCs research field and summarize the latest findings associated with TCs, with a special focus on the recent progress about TCs specific gene and protein profiles that has been made in understanding that TCs may play a potential, but important, role in the pathogenesis of lung diseases.
Collapse
|
50
|
Guo L, Zhang Y, Zhang L, Huang F, Li J, Wang S. MicroRNAs, TGF-β signaling, and the inflammatory microenvironment in cancer. Tumour Biol 2016; 37:115-25. [PMID: 26563372 PMCID: PMC4841843 DOI: 10.1007/s13277-015-4374-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 11/03/2015] [Indexed: 12/19/2022] Open
Abstract
Inflammatory cells and mediators form a major part of the tumor microenvironment and play important roles in the regulation of cancer initiation, tumor cell proliferation, and metastasis. MicroRNAs (miRNAs) play important roles in several physiological and pathological processes, including the regulation of the inflammatory microenvironment in cancer. Transforming growth factor-β (TGF-β) is an inflammation-related cytokine that functions in both tumor suppression and promotion; however, its underlying molecular mechanisms remain unclear. Recent evidence indicates an association between miRNAs and TGF-β signaling, providing new insight into the nature of the inflammatory microenvironment in cancer. The present review is an overview of the interaction between miRNAs and inflammatory cytokines, with emphasis on the cross talk between TGF-β signaling and miRNAs and their influence on cancer cell behavior. The emerging roles of miRNAs in cancer-related inflammation and the potential to target miRNA signaling pathways for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Lingling Guo
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Yongsheng Zhang
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lifeng Zhang
- Department of Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Fengbo Huang
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Jinfan Li
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Shouli Wang
- Department of Pathology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China.
- Institute of Radiology and Oncology, Soochow University, Suzhou, 215006, China.
| |
Collapse
|