1
|
Chemokines and NSCLC: Emerging role in prognosis, heterogeneity, and therapeutics. Semin Cancer Biol 2022; 86:233-246. [PMID: 35787939 DOI: 10.1016/j.semcancer.2022.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022]
Abstract
Lung cancer persists to contribute to one-quarter of cancer-associated deaths. Among the different histologies, non-small cell lung cancer (NSCLC) alone accounts for 85% of the cases. The development of therapies involving immune checkpoint inhibitors and angiogenesis inhibitors has increased patients' survival probability and reduced mortality rates. Developing targeted therapies against essential genetic alterations also translates to better treatment strategies. But the benefits still seem farfetched due to the development of drug resistance and refractory tumors. In this review, we have highlighted the interplay of different tumor microenvironment components, essentially discussing the chemokine families (CC, CXC, C, and CX3C) that regulate the tumor biology in NSCLC and promote tumor growth, metastasis, and associated heterogeneity. The development of therapeutics and prognostic markers is a complex and multipronged approach. However, some essential chemokines can act as critical players for being considered potential prognostic markers and therapeutic targets.
Collapse
|
2
|
Ghasemi M, Bakhshi B, Khashei R, Soudi S. Mesoporous silica nano-adjuvant triggers pro-inflammatory responses in Caco-2/peripheral blood mononuclear cell (PBMC) co-cultures. Nanobiomedicine (Rij) 2022; 9:18495435221088374. [PMID: 35677573 PMCID: PMC9168868 DOI: 10.1177/18495435221088374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 02/06/2022] [Indexed: 11/28/2022] Open
Abstract
The aim of this study was to evaluate the cytotoxicity and immune-stimulatory
effect of Mesoporous silica nanoparticle (MSN) Nano-adjuvant on pro-inflammatory
cytokines and pattern recognition receptors (PRR) genes expression in
Caco-2/PBMC co-culture model. MSNs were synthesized and characterized by
scanning electron microscope (SEM), Brunauer Emmett Teller (BET) and Barrett
Joyner Halenda (BJH) techniques. The BET specific surface area of MSNs was
around 947 m2/g and the total pore volume and average pore diameter
were 1.5 cm3/g and 8.01 nm, respectively. At the concentration of
10 µg/mL, MSN showed a low and time-dependent cytotoxicity on Caco-2 cells,
while no cytotoxic effect was observed for 0.1 and 1 µg/mL concentrations after
24, 48 and 72 h. The expression of pro-inflammatory cytokines genes (IL-1, IL-8
and TNF-α) in co-cultures treated with different concentrations of MSN showed a
dose-dependent significant increase up to 17.44, 2.722 and 4.34 folds,
respectively, while the expression augmentation of IL-1 gene was significantly
higher than the others. This indicates slight stimulation of intestinal
inflammation. Different concentrations of MSN significantly increased TLR4 and
NOD2 expression to 4.14 and 2.14 folds, respectively. NOD1 was not affected
significantly. It can be concluded that MSN might increase protective immune
responses against antigens as a vaccine adjuvant candidate. It seems that
stimulation of TNF-α, IL-1, and IL-8 expression in enterocytes probably
transpires through the agonistic activity of MSN for TLRs including TLR4, while
NOD2-associated signaling pathways are also involved. This study provides an
overall picture of MSN as a novel and potent oral adjuvant for mucosal
immunity.
Collapse
Affiliation(s)
- Maryam Ghasemi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Khashei
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Mohamad SA, Safwat MA, Elrehany M, Maher SA, Badawi AM, Mansour HF. A novel nasal co-loaded loratadine and sulpiride nanoemulsion with improved downregulation of TNF-α, TGF-β and IL-1 in rabbit models of ovalbumin-induced allergic rhinitis. Drug Deliv 2021; 28:229-239. [PMID: 33501873 PMCID: PMC7850330 DOI: 10.1080/10717544.2021.1872741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 10/28/2022] Open
Abstract
PURPOSE The work aimed to develop a co-loaded loratadine and sulpiride nasal nanoemulsion for allergic rhinitis management. METHODS Compatibility studies were conducted adopting differential scanning calorimetry and Fourier transform infrared spectroscopy. Nanoemulsion formulations were prepared using soybean lecithin, olive oil and tween 80. Sodium cholate and glycerol were employed as co-surfactants. Nanoemulsions were assessed for viscosity, pH, droplet size, polydispersity index, zeta potential, electrical conductivity, entrapment, In vitro drug release and corresponding kinetics. Stability of the selected formulation was investigated. The biological effectiveness was evaluated in rabbit models of ovalbumin-induced allergic rhinitis by measuring TNF-α, TGF-β and IL-1. RESULTS Compatibility studies revealed absence of drug/drug interactions. Nanoemulsions exhibited > 90% entrapment efficiency. The selected nanoemulsion demonstrated small droplet size (85.2 ± 0.2 nm), low PDI (0.35 ± 0.0) and appropriate Zeta Potential (-23.3 ± 0.2) and stability. It also displayed enhanced in vitro drug release following the Higuashi Diffusion and Baker-Lonsdale models. The mean relative mRNA expression of TNF-α, IL-1 and TGF-β significantly decreased from 9.59 ± 1.06, 4.15 ± 0.02 and 4.15 ± 0.02 to 1.28 ± 0.02, 1.93 ± 0.06 and 1.56 ± 0.02 respectively after treatment with the selected nanoemulsion formulation. CONCLUSION The results reflected a promising potent effect of the combined loratadine and sulpiride nasal nanoemulsion in managing the symptoms of allergic rhinitis.
Collapse
Affiliation(s)
- Soad A. Mohamad
- Department of Pharmaceutics, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Mohamed A. Safwat
- Department of Pharmaceutics, Faculty of Pharmacy, South Valley University, Qena, Egypt
| | - Mahmoud Elrehany
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, Minia, Egypt
- Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Sherif A. Maher
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, Minia, Egypt
| | - Ahmed M. Badawi
- Department of Otorhinolaryngology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Heba F. Mansour
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
4
|
Martens B, Drebert Z. Glucocorticoid-mediated effects on angiogenesis in solid tumors. J Steroid Biochem Mol Biol 2019; 188:147-155. [PMID: 30654109 DOI: 10.1016/j.jsbmb.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/13/2019] [Accepted: 01/13/2019] [Indexed: 12/28/2022]
Abstract
Angiogenesis is essential in tumor development to maintain the oxygen and nutrient supply. Glucocorticoids have shown both direct and indirect angiostatic properties in various types of solid cancers. In most of the reported cases glucocorticoid-mediated actions involved suppression of multiple pro-angiogenic factors expression by cancer cells. The anti-angiogenic properties of glucocorticoids correlated with diminished tumor vasculature and reduced tumor growth in multiple in vivo studies. However, when glucocorticoid treatment is considered, possible adverse events should be taken into account. Additional research is needed to further test the use of these steroidal drugs in cancer therapy.
Collapse
Affiliation(s)
- Broes Martens
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - Zuzanna Drebert
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology & Experimental Cancer Research, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
5
|
Rivas-Fuentes S, Salgado-Aguayo A, Pertuz Belloso S, Gorocica Rosete P, Alvarado-Vásquez N, Aquino-Jarquin G. Role of Chemokines in Non-Small Cell Lung Cancer: Angiogenesis and Inflammation. J Cancer 2015; 6:938-52. [PMID: 26316890 PMCID: PMC4543754 DOI: 10.7150/jca.12286] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/23/2015] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of aggressive cancer. The tumor tissue, which shows an active angiogenesis, is composed of neoplastic and stromal cells, and an abundant inflammatory infiltrate. Angiogenesis is important to support tumor growth, while infiltrating cells contribute to the tumor microenvironment through the secretion of growth factors, cytokines and chemokines, important molecules in the progression of the disease. Chemokines are important in development, activation of the immune response, and physiological angiogenesis. Chemokines have emerged as important regulators in the pathophysiology of cancer. These molecules are involved in the angiogenesis/angiostasis balance and in the recruitment of tumor infiltrating hematopoietic cells. In addition, chemokines promote tumor cell survival, as well as the directing and establishment of tumor cells to metastasis sites. The findings summarized here emphasize the central role of chemokines as modulators of tumor angiogenesis and their potential role as therapeutic targets in the inflammatory process of NSCLC angiogenesis.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfonso Salgado-Aguayo
- 2. Laboratory of Research on Rheumatic Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Silvana Pertuz Belloso
- 3. Department of Comparative Biology, Faculty of Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Patricia Gorocica Rosete
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Noé Alvarado-Vásquez
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Guillermo Aquino-Jarquin
- 4. Laboratory of Research on Genomics, Genetics and Bioinformatics. Tower of Haemato-oncology, Children´s Hospital of Mexico “Federico Gomez”, Mexico City, Mexico
| |
Collapse
|
6
|
Illeperuma RP, Kim DK, Park YJ, Son HK, Kim JY, Kim J, Lee DY, Kim KY, Jung DW, Tilakaratne WM, Kim J. Areca nut exposure increases secretion of tumor-promoting cytokines in gingival fibroblasts that trigger DNA damage in oral keratinocytes. Int J Cancer 2015; 137:2545-57. [PMID: 26076896 PMCID: PMC4744697 DOI: 10.1002/ijc.29636] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 05/28/2015] [Accepted: 05/28/2015] [Indexed: 02/06/2023]
Abstract
Molecular crosstalk between cancer cells and fibroblasts has been an emerging hot issue in understanding carcinogenesis. As oral submucous fibrosis (OSF) is an inflammatory fibrotic disease that can potentially transform into squamous cell carcinoma, OSF has been considered to be an appropriate model for studying the role of fibroblasts during early stage carcinogenesis. In this sense, this study aims at investigating whether areca nut (AN)‐exposed fibroblasts cause DNA damage of epithelial cells. For this study, immortalized hNOF (hTERT‐hNOF) was used. We found that the levels of GRO‐α, IL‐6 and IL‐8 increased in AN‐exposed fibroblasts. Cytokine secretion was reduced by antioxidants in AN‐exposed fibroblasts. Increase in DNA double strand breaks (DSB) and 8‐oxoG FITC‐conjugate was observed in immortalized human oral keratinocytes (IHOK) after the treatment of cytokines or a conditioned medium derived from AN‐exposed fibroblasts. Cytokine expression and DNA damage were also detected in OSF tissues. The DNA damage was reduced by neutralizing cytokines or antioxidant treatment. Generation of reactive oxygen species (ROS) and DNA damage response, triggered by cytokines, were abolished when NADPH oxidase (NOX) 1 and 4 were silenced in IHOK, indicating that cytokine‐triggered DNA damage was caused by ROS generation through NOX1 and NOX4. Taken together, this study provided strong evidence that blocking ROS generation might be a rewarding approach for cancer prevention and intervention in OSF. What's new? Fibroblasts in the tumor microenvironment influence tumor initiation and growth and are of particular interest in oral submucous fibrosis (OSF), a progressive fibrotic disease of malignant potential. This study shows that the release of tumor‐promoting cytokines by fibroblasts exposed to areca nut, the primary cause of OSF, induce DNA damage in oral keratinocytes. The findings suggest that fibroblasts indirectly promote epithelial transformation in OSF by secreting cytokines, whereby DNA damage of epithelial cells is inflicted by reactive oxygen species generated via NADPH oxidases. These insights could inform the development of new therapeutic approaches for OSF.
Collapse
Affiliation(s)
- Rasika P Illeperuma
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea.,Faculty of Allied Health Sciences, Department of Medical Laborotary Science, University of Peradeniya, Sri Lanka
| | - Do Kyeong Kim
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea.,BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Young Jin Park
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea.,BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Hwa Kyung Son
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea.,Department of Dental Hygiene, Yeungnam University College, Daegu, Korea
| | - Jue Young Kim
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea.,BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Jinmi Kim
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea
| | - Doo Young Lee
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea
| | - Ki-Yeol Kim
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea
| | - Da-Woon Jung
- Department of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | | | - Jin Kim
- Oral Cancer Research Institute, Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea.,BK 21 PLUS Project, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
7
|
Shen SC, Wu MS, Lin HY, Yang LY, Chen YH, Chen YC. Reactive oxygen species-dependent nitric oxide production in reciprocal interactions of glioma and microglial cells. J Cell Physiol 2014; 229:2015-26. [PMID: 24777714 DOI: 10.1002/jcp.24659] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/25/2014] [Indexed: 01/30/2023]
Abstract
Conditioned mediums (CMs) from glioma cells U87, GBM-8401, and C6 significantly induced iNOS protein and NO production by microglial cells BV-2 but without altering the cell viability or cell-cycle progression of BV2 microglia. Significant increases in intracellular peroxide by U87-CM and C6-CM were detected by a DCHF-DA assay, and vitamin (Vit) C and N-acetyl cysteine (NAC)-reduced intracellular peroxide levels elicited by CMs lead to inhibition of iNOS/NO production The extracellular signal-regulated kinase (ERK) inhibitor, U0126, and c-Jun N-terminal kinase (JNK) inhibitor, SP600125, suppressed U87-CM- and C6-CM-induced iNOS/NO production by respectively blocking phosphorylated ERK (pERK) and JNK (pJNK) protein expressions stimulated by U87-CM and C6-CM. Increased migration of U87 and C6 glioma cells by a co-culture with BV-2 microglial cells or adding the nitric oxide (NO) donor, sodium nitroprusside (SNP) was observed, and that was blocked by adding an NO synthase (NOS) inhibitor, N-nitro L-arginine methyl ester (NAME). Contributions of ROS, pERK, and pJNK to the migration of glioma cells was further demonstrated in a transwell coculture system of U87 and C6 gliomas with BV-2 microglial cells. Furthermore, expressions of tumor necrosis factor (TNF)-α and monocyte chemoattractant protein (MCP)-1 messenger (m)RNA in U87 and C6 cells were detected by an RT-PCR, and TNF-α and MCP-1 induced iNOS protein expression in time- and concentration-dependent manners. Neutralization of TNF-α or MCP-1 in U87-CM and C6-CM using a TNF-α or MCP-1 antibody inhibited iNOS protein expression, and increased intracellular peroxide by TNF-α or MCP-1 was identified in BV-2 cells. The reciprocal activation of glioma cells and microglia via ROS-dependent iNOS/NO elevation at least partially mediated by TNF-α and MCP-1 is elucidated.
Collapse
Affiliation(s)
- Shing-Chuan Shen
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
8
|
Riabov V, Gudima A, Wang N, Mickley A, Orekhov A, Kzhyshkowska J. Role of tumor associated macrophages in tumor angiogenesis and lymphangiogenesis. Front Physiol 2014; 5:75. [PMID: 24634660 PMCID: PMC3942647 DOI: 10.3389/fphys.2014.00075] [Citation(s) in RCA: 451] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 02/06/2014] [Indexed: 12/12/2022] Open
Abstract
Tumor angiogenesis is an essential process for supplying rapidly growing malignant tissues with essential nutrients and oxygen. An angiogenic switch allows tumor cells to survive and grow, and provides them access to vasculature resulting in metastatic disease. Monocyte-derived macrophages recruited and reprogrammed by tumor cells serve as a major source of angiogenic factors boosting the angiogenic switch. Tumor endothelium releases angiopoietin-2 and further facilitates recruitment of TIE2 receptor expressing monocytes (TEM) into tumor sites. Tumor-associated macrophages (TAM) sense hypoxia in avascular areas of tumors, and react by production of angiogenic factors such as VEGFA. VEGFA stimulates chemotaxis of endothelial cells (EC) and macrophages. In some tumors, TAM appeared to be a major source of MMP9. Elevated expression of MMP9 by TAM mediates extracellular matrix (ECM) degradation and the release of bioactive VEGFA. Other angiogenic factors released by TAM include basic fibroblast growth factor (bFGF), thymidine phosphorylase (TP), urokinase-type plasminogen activator (uPA), and adrenomedullin (ADM). The same factors used by macrophages for the induction of angiogenesis [like vascular endothelial growth factor A (VEGF-A) and MMP9] support lymphangiogenesis. TAM can express LYVE-1, one of the established markers of lymphatic endothelium. TAM support tumor lymphangiogenesis not only by secretion of pro-lymphangiogenic factors but also by trans-differentiation into lymphatic EC. New pro-angiogenic factor YKL-40 belongs to a family of mammalian chitinase-like proteins (CLP) that act as cytokines or growth factors. Human CLP family comprises YKL-40, YKL-39, and SI-CLP. Production of all three CLP in macrophages is antagonistically regulated by cytokines. It was recently established that YKL-40 induces angiogenesis in vitro and in animal tumor models. YKL-40-neutralizing monoclonal antibody blocks tumor angiogenesis and progression. The role of YKL-39 and SI-CLP in tumor angiogenesis and lymphangiogenesis remains to be investigated.
Collapse
Affiliation(s)
- Vladimir Riabov
- Department of Dermatology, University Medical Center and Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg Mannheim, Germany ; Department of Nanopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia
| | - Alexandru Gudima
- Department of Dermatology, University Medical Center and Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg Mannheim, Germany ; Department of Innate Immunity and Tolerance, University Medical Center and Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg Mannheim, Germany
| | - Nan Wang
- Department of Dermatology, University Medical Center and Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg Mannheim, Germany
| | - Amanda Mickley
- Department of Dermatology, University Medical Center and Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg Mannheim, Germany ; Department of Innate Immunity and Tolerance, University Medical Center and Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg Mannheim, Germany
| | - Alexander Orekhov
- Department of Nanopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia
| | - Julia Kzhyshkowska
- Department of Dermatology, University Medical Center and Medical Faculty Mannheim, Ruprecht-Karls University of Heidelberg Mannheim, Germany ; Department of Nanopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences Moscow, Russia ; Department of Innate Immunity and Tolerance, University Medical Center and Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Ruprecht-Karls University of Heidelberg Mannheim, Germany
| |
Collapse
|
9
|
Lee CY, Tsai YT, Loh SH, Liu JC, Chen TH, Chao HH, Cheng TH, Chen JJ. Urotensin II induces interleukin 8 expression in human umbilical vein endothelial cells. PLoS One 2014; 9:e90278. [PMID: 24587311 PMCID: PMC3931834 DOI: 10.1371/journal.pone.0090278] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 01/28/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Urotensin II (U-II), an 11-amino acid peptide, exerts a wide range of actions in cardiovascular systems. Interleukin-8 (IL-8) is secreted by endothelial cells, thereby enhancing endothelial cell survival, proliferation, and angiogenesis. However, the interrelationship between U-II and IL-8 as well as the detailed intracellular mechanism of U-II in vascular endothelial cells remain unclear. The aim of this study was to investigate the effect of U-II on IL-8 expression and to explore its intracellular mechanism in human umbilical vein endothelial cells. METHODS/PRINCIPAL FINDINGS Primary human umbilical vein endothelial cells were used. Expression of IL-8 was determined by real-time quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, and luciferase reporter assay. Western blot analyses and experiments with specific inhibitors were performed to reveal the downstream signaling pathways as concerned. U-II increased the mRNA/protein levels of IL-8 in human umbilical vein endothelial cells. The U-II effects were significantly inhibited by its receptor antagonist [Orn(5)]-URP. Western blot analyses and experiments with specific inhibitors indicated the involvement of phosphorylation of p38 mitogen-activated protein kinase and extracellular signal-regulated kinase in U-II-induced IL-8 expression. Luciferase reporter assay further revealed that U-II induces the transcriptional activity of IL-8. The site-directed mutagenesis indicated that the mutation of AP-1 and NF-kB binding sites reduced U-II-increased IL-8 promoter activities. Proliferation of human umbilical vein endothelial cells induced by U-II could be inhibited significantly by IL-8 RNA interference. CONCLUSION/SIGNIFICANCE The results show that U-II induces IL-8 expression in human umbilical vein endothelial cells via p38 mitogen-activated protein kinase and extracellular signal-regulated kinase signaling pathways and IL-8 is involved in the U-II-induced proliferation of human umbilical vein endothelial cells.
Collapse
Affiliation(s)
- Chung-Yi Lee
- Department of Cardiovascular Surgery, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
| | - Yi-Tin Tsai
- Department of Cardiovascular Surgery, Tri-Service General Hospital, Taipei, Taiwan, Republic of China
| | - Shih-Hurng Loh
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Ju-Chi Liu
- Department of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Tso-Hsiao Chen
- Department of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Hung-Hsing Chao
- Department of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
- Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Tzu-Hurng Cheng
- Department of Biochemistry, School of Medicine, China Medical University, Taichung, Taiwan, Republic of China
| | - Jin-Jer Chen
- Graduate Institute of Clinical Medicine, College of Medicine, China Medical University, Taichung, Taiwan, Republic of China
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
10
|
Kuo CH, Chen KF, Chou SH, Huang YF, Wu CY, Cheng DE, Chen YW, Yang CJ, Hung JY, Huang MS. Lung tumor-associated dendritic cell-derived resistin promoted cancer progression by increasing Wolf-Hirschhorn syndrome candidate 1/Twist pathway. Carcinogenesis 2013; 34:2600-9. [PMID: 23955539 DOI: 10.1093/carcin/bgt281] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The interaction between tumors and their microenvironments leads to a vicious cycle, which strengthens both immune suppression and cancer progression. The present study demonstrates for the first time that tumor-associated dendritic cells (TADCs) are a source of resistin, which is responsible for increasing lung cancer epithelial-to-mesenchymal transition. In addition, large amounts of resistin in the condition medium (CM) of TADCs increase cell migration and invasion, as well as the osteolytic bone metastatic properties of lung cancer cells. Neutralization of resistin from TADC-CM prevents the advanced malignancy-inducing features of TADC-CM. Significantly elevated levels of resistin have been observed in mice transplanted with lung cancer cells, tumor-infiltrating CD11c(+) DCs in human lung cancer samples and lung cancer patients' sera. Induction of lung cancer progression by TADC-derived resistin is associated with increased expression of Wolf-Hirschhorn syndrome candidate 1 (WHSC1), a histone methyltransferase. Resistin-induced WHSC1 increases the dimethylation of histone 3 at lysine 36 and decreases the trimethylation of histone 3 at lysine 27 on the promoter of Twist, resulting in an enhancement of the expression of Twist. Knockdown of WHSC1 by small interfering RNA transfection significantly decreases resistin-mediated cancer progression by decreasing the upregulation of Twist, suggesting that WHSC1 plays a critical role in the regulation of Twist by epigenetic modification. Furthermore, mice that received antiresistin antibodies showed a decreased incidence of cancer development and metastasis. These findings suggest that TADC-derived resistin may be a novel candidate in promoting the development of lung cancer.
Collapse
Affiliation(s)
- Chih-Hsin Kuo
- The Affiliated Senior High School of National Kaohsiung Normal University, Kaohsiung 802, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Myeloid cell RelA/p65 promotes lung cancer proliferation through Wnt/β-catenin signaling in murine and human tumor cells. Oncogene 2013; 33:1239-48. [PMID: 23563178 DOI: 10.1038/onc.2013.75] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 01/22/2013] [Accepted: 01/25/2013] [Indexed: 12/18/2022]
Abstract
Smoking is the most important risk factor for both lung cancer (LC) and chronic obstructive pulmonary disease. The aim of this study was to investigate the role of myeloid cell nuclear factor-κB in the regulation of tumor cell growth signaling. We subjected mice lacking myeloid RelA/p65 (rela(Δ-/-)) to a metastatic LC model. Cigarette smoke (CS) exposure significantly increased the proliferation of Lewis lung carcinoma cell tumors in wild-type mice. In CS-exposed rela(Δ-/-) mice, the tumor growth was largely inhibited. Transcriptome and pathway analysis of cancer tissue revealed a fundamental impact of myeloid cells on various growth signaling pathways, including the Wnt/β-catenin pathway. In conclusion, myeloid RelA/p65 is necessary to link smoke-induced inflammation with LC growth and has a role in the activation of Wnt/β-catenin signaling in tumor cells.
Collapse
|
12
|
FOX SIMONA, LOH SUZANNES, MAHENDRAN SATHISHK, GARLEPP MICHAELJ. Regulated chemokine gene expression in mouse mesothelioma and mesothelial cells: TNF-α upregulates both CC and CXC chemokine genes. Oncol Rep 2012; 28:707-13. [DOI: 10.3892/or.2012.1809] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 03/23/2012] [Indexed: 11/06/2022] Open
|
13
|
Sunaga N, Imai H, Shimizu K, Shames DS, Kakegawa S, Girard L, Sato M, Kaira K, Ishizuka T, Gazdar AF, Minna JD, Mori M. Oncogenic KRAS-induced interleukin-8 overexpression promotes cell growth and migration and contributes to aggressive phenotypes of non-small cell lung cancer. Int J Cancer 2011; 130:1733-44. [PMID: 21544811 DOI: 10.1002/ijc.26164] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 04/21/2011] [Indexed: 12/21/2022]
Abstract
The CXC chemokine interleukin-8 (IL-8) is an angiogenic growth factor that is overexpressed in various cancers, including non-small cell lung cancer (NSCLC). Previously, IL-8 was shown as a transcriptional target of RAS signaling, raising the possibility of its role in oncogenic KRAS-driven NSCLC. Using microarray analysis, we identified IL-8 as the most downregulated gene by shRNA-mediated KRAS knockdown in NCI-H1792 NSCLC cells where IL-8 is overexpressed. NSCLC cell lines harboring KRAS or EGFR mutations overexpressed IL-8, while IL-8 levels were more prominent in KRAS mutants compared to EGFR mutants. IL-8 expression was downregulated by shRNA-mediated KRAS knockdown in KRAS mutants or by treatment with EGFR tyrosine kinase inhibitors and EGFR siRNAs in EGFR mutants. In our analysis of the relationship of IL-8 expression with clinical parameters and mutation status of KRAS or EGFR in 89 NSCLC surgical specimens, IL-8 expression was shown to be significantly higher in NSCLCs of males, smokers, and elderly patients and those with pleural involvement and KRAS mutated adenocarcinomas. In KRAS mutant cells, the MEK inhibitor markedly decreased IL-8 expression, while the p38 inhibitor increased IL-8 expression. Attenuation of IL-8 function by siRNAs or a neutralizing antibody inhibited cell proliferation and migration of KRAS mutant/IL-8 overexpressing NSCLC cells. These results indicate that activating mutations of KRAS or EGFR upregulate IL-8 expression in NSCLC; IL-8 is highly expressed in NSCLCs from males, smokers, elderly patients, NSCLCs with pleural involvement, and KRAS-mutated adenocarcinomas; and IL-8 plays a role in cell growth and migration in oncogenic KRAS-driven NSCLC.
Collapse
Affiliation(s)
- Noriaki Sunaga
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi, Gunma, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lin CW, Shen SC, Ko CH, Lin HY, Chen YC. Reciprocal activation of macrophages and breast carcinoma cells by nitric oxide and colony-stimulating factor-1. Carcinogenesis 2010; 31:2039-48. [PMID: 20876703 DOI: 10.1093/carcin/bgq172] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Induction of inducible nitric oxide synthase (iNOS) gene expression, nitric oxide (NO) production and migration of RAW264.7 macrophages by coculture with breast cancer MDA-MB-231 cells or the addition of conditioned medium derived from MDA-MB-231 cells (MDA-CM) was identified. Increased iNOS/NO induction and migration of macrophages by MDA-CM were significantly blocked by adding the c-Jun-N-terminal protein kinase (JNK) inhibitor, SP600125, the nuclear factor-kappa B (NF-κB) inhibitor, BAY117082 and pyrrolidine dithiocarbamic acid and a dominant-negative JNK. The addition of an NO donor, Diethylenetriamine-NONOate, significantly activated expressions of MMP-9 and VEGF-A genes in breast carcinoma MDA-MD-231 cells and invasion of MDA-MB-231 cells in coculture with RAW264.7 macrophages as determined using Transwell systems, but that was inhibited by adding SP600125, BAY117082 and the nitric oxide synthase inhibitor, NG-nitro-L-arginine methyl ester. Induction of heme oxygenase-1 in macrophages reduced MDA-CM-induced iNOS/NO, JNK and NF-κB activations in accordance with inhibiting VEGF-A and MMP-9 gene expressions by MDA-MB-231 cells via Transwell assays. Furthermore, VEGF, sRANKL, TNF-α, IL-1α, TGF-β, CSF-1 and MCP-1 were applied, and CSF-1 showed the most potent stimulation of iNOS/NO production and migration of macrophages. MCF-7 cells with lower CSF-1 expression than MDA-MB-231 cells showed a poor stimulatory effect on iNOS/NO production and migration of macrophages. Neutralization of CSF-1 in MDA-CM using CSF-1 antibody inhibited MDA-CM-induced iNOS protein expression and migration of macrophages, and CSF-1-induced iNOS protein and migration was blocked by adding JNK inhibitor SP and NF-κB inhibitor BAY. The reciprocal activation of breast cancer and macrophages via NO-CSF-1 is first elucidated herein.
Collapse
Affiliation(s)
- Cheng-Wei Lin
- Graduate Institute of Pharmacy, School of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | | | | | | | | |
Collapse
|
15
|
Gerber PA, Hippe A, Buhren BA, Müller A, Homey B. Chemokines in tumor-associated angiogenesis. Biol Chem 2010; 390:1213-23. [PMID: 19804363 DOI: 10.1515/bc.2009.144] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor growth is dependent on several key factors. Apart from immune escape and an efficient blockade of apoptotic signals, tumors require oxygen and nutrients to grow past a diameter of 2 microm. Therefore, it is of vital importance for the tumor to facilitate tumor-associated angiogenesis, e.g., the de novo formation of new blood vessels. In addition to established and key angiogenic factors, such as vascular endothelial growth factor, chemokines, a superfamily of cytokine-like proteins that bind to seven transmembrane-spanning G-protein-coupled receptors, have been associated with angiogenesis under homeostatic conditions. Chemokines were initially identified as key factors that control the directional migration of leukocytes, stem cells and cancer cells in vitro and which critically regulate their trafficking in vivo. Recently their role in establishing a favorable microenvironment for tumor-associated angiogenesis, a process that requires complex bidirectional interactions of the tumor and associated vessels, has been the focus of research. Chemokine-promoted angiogenesis not only facilitates tumor growth by supplying nutrients and oxygen but it is also a prerequisite to tumor metastasis. Hence, the pharmacologic control of tumor angiogenesis presents a promising strategy for novel anticancer therapeutics. Here, we discuss the current pathogenetic concepts of tumor-associated angiogenesis in the context of chemokines and their receptors and highlight promising therapeutic strategies.
Collapse
Affiliation(s)
- Peter Arne Gerber
- Department of Dermatology, University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
16
|
Abstract
Tumor growth is restricted to approximately 2 microm diameters by simple dissociation of nutrients and oxygen. Hence, tumors require the formation of new blood vessels for further growth progression. This process is referred to as tumor neo-angiogenesis. The process of tumor neo-angiogenesis is directed by complex bidirectional interactions between the tumor and the vessels, and creates a favorable microenvironment for angiogenesis. The tumor vessel system not only facilitates tumor growth by providing nutrients and oxygen but also functions as a convenient route for metastasis. A group of small cytokine-like molecules called chemokines have been shown to participate in angiogenesis under homeostatic and neoplastic conditions. This review summarizes their role in tumor-associated angiogenesis.
Collapse
|
17
|
Zhang B, Wang J, Gao J, Guo Y, Chen X, Wang B, Gao J, Rao Z, Chen Z. Alternatively activated RAW264.7 macrophages enhance tumor lymphangiogenesis in mouse lung adenocarcinoma. J Cell Biochem 2009; 107:134-43. [PMID: 19241443 DOI: 10.1002/jcb.22110] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor-associated macrophages (TAMs) have been implicated in promoting tumor progression and invasion. The onset and maintenance of tumor angiogenesis and lymphangiogenesis also seem to be partly driven by a group of polarized alternatively activated macrophages (aaMphi) in lung adenocarcinoma. Here, the aaMphi and classically activated macrophages (caMphi) were obtained using RAW264.7 cells via IL-4 and IFN-gamma + LPS treatment, respectively. Co-inoculation of aaMphi with Lewis lung carcinoma (LLC) cells promoted tumor growth, increased lymph node metastasis, and reduced the survival in C57BL/6 mice bearing LLC. Furthermore, the effects of the activated macrophages on the lymphangiogenesis-related properties of lymphatic endothelial cells (LECs) were investigated in vitro. When LECs were cultured in macrophages conditioned medium or in a co-culture system of macrophages and LECs, aaMphi significantly promoted proliferation, migration, and tube-like formation of LECs. We identified high VEGF-C expression in aaMphi and low expression in caMphi as well as unactivated macrophages by ELISA and Western blotting. In LECs, co-culture with aaMphi resulted in a significant increase of mRNA levels of specific lymphatic marker VEGF receptor-3 and the homeobox gene Prox-1, as well as lymphangiogenic factor VEGF-C rather than VEGF-D by quantitative RT-PCR. Furthermore, enhanced LECs migration and capillary formation by co-culture with aaMphi were significantly inhibited by rVEGF receptor-3/Fc chimera. In conclusion, these data show that aaMphi play a critical role in tumor-induced lymphangiogenesis through up-regulating VEGF-C and increasing lymphangiogenesis-related behavior of LECs, which may contribute to lymphatic invasion in lung adenocarcinoma.
Collapse
Affiliation(s)
- Bicheng Zhang
- Department of Oncology, Wuhan General Hospital of Guangzhou Command PLA, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Chiang YY. Hepatocyte growth factor induces hypoxia-related interleukin-8 expression in lung adenocarcinoma cells. Mol Carcinog 2009; 48:662-70. [PMID: 19184985 DOI: 10.1002/mc.20521] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Rapid growth of cancer cells often creates insufficient supply of oxygen and nutrients in the tumour nest. The frequent detection of hypoxia-inducible factor (HIF) and interleukin-8 (IL-8) in afflicted tissues suggests that IL-8 expression could be associated with elevated levels of HIF. Recently, we found that hypoxia also upregulated the expression of hepatocyte growth factor (HGF) in lung adenocarcinoma (LAD) cells. However, the relationship between HGF and IL-8 has not been investigated in LAD cells. In this study, we found that HGF induced IL-8 expression in LAD. Interestingly, hypoxia also increased the level of prostaglandin F(2alpha) (PGF(2alpha)), a product of dihydrodiol dehydrogenase (DDH). When expression of DDH was suppressed by siRNA, the levels of PGF(2alpha), HGF and IL-8 were reduced; however, their levels returned to normal after DDH was reintroduced. These data suggest that hypoxia induces biosynthesis of PGF(2alpha), which then activates HGF and IL-8 expression. The results provide a reasonable explanation of how PGF(2alpha), HGF and IL-8 exert their effects on cancer cell metastasis.
Collapse
Affiliation(s)
- Yung-Yen Chiang
- Department of Dental Laboratory Technology, Central Taiwan University of Science and Technology, Taichung, Taiwan
| |
Collapse
|
19
|
Induced interleukin-8 expression in gliomas by tumor-associated macrophages. J Neurooncol 2009; 93:289-301. [PMID: 19156359 DOI: 10.1007/s11060-008-9786-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2008] [Accepted: 12/30/2008] [Indexed: 11/26/2022]
|
20
|
Kodama T, Kanazawa H, Tochino Y, Kyoh S, Asai K, Hirata K. A technological advance comparing epithelial lining fluid from different regions of the lung in smokers. Respir Med 2009; 103:35-40. [DOI: 10.1016/j.rmed.2008.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 08/19/2008] [Accepted: 09/03/2008] [Indexed: 11/25/2022]
|
21
|
Lee CY, Sher HF, Chen HW, Liu CC, Chen CH, Lin CS, Yang PC, Tsay HS, Chen JJW. Anticancer effects of tanshinone I in human non-small cell lung cancer. Mol Cancer Ther 2008; 7:3527-38. [PMID: 19001436 DOI: 10.1158/1535-7163.mct-07-2288] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tanshinones are the major bioactive compounds of Salvia miltiorrhiza Bunge (Danshen) roots, which are used in many therapeutic remedies in Chinese traditional medicine. We investigated the anticancer effects of tanshinones on the highly invasive human lung adenocarcinoma cell line, CL1-5. Tanshinone I significantly inhibited migration, invasion, and gelatinase activity in macrophage-conditioned medium-stimulated CL1-5 cells in vitro and also reduced the tumorigenesis and metastasis in CL1-5-bearing severe combined immunodeficient mice. Unlike tanshinone IIA, which induces cell apoptosis, tanshinone I did not have direct cytotoxicity. Real-time quantitative PCR, luciferase reporter assay, and electrophoretic mobility shift assay revealed that tanshinone I reduces the transcriptional activity of interleukin-8, the angiogenic factor involved in cancer metastasis, by attenuating the DNA-binding activity of activator protein-1 and nuclear factor-kappaB in conditioned medium-stimulated CL1-5 cells. Microarray and pathway analysis of tumor-related genes identified the differentially expressed genes responding to tanshinone I, which may be associated with the Ras-mitogen-activated protein kinase and Rac1 signaling pathways. These results suggest that tanshinone I exhibits anticancer effects both in vitro and in vivo and that these effects are mediated at least partly through the interleukin-8, Ras-mitogen-activated protein kinase, and Rac1 signaling pathways. Although tanshinone I has a remarkable anticancer action, its potential anticoagulant effect should be noted and evaluated.
Collapse
Affiliation(s)
- Chen-Yu Lee
- Institutes of Biomedical Sciences and Molecular Biology, National Chung-Hsing University, Taichung, Taiwan 40227, Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Murphy DM, Forrest IA, Corris PA, Johnson GE, Small T, Jones D, Fisher AJ, Egan JJ, Cawston TE, Lordan JL, Ward C. Azithromycin attenuates effects of lipopolysaccharide on lung allograft bronchial epithelial cells. J Heart Lung Transplant 2008; 27:1210-6. [PMID: 18971093 DOI: 10.1016/j.healun.2008.07.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 07/15/2008] [Accepted: 07/29/2008] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The bronchial epithelium is a source of mediators that may play a role in the airway inflammation and remodeling of post-transplant obliterative bronchiolitis (OB). Traditional strategies have failed to have an impact on OB. Recent studies have suggested a role for azithromycin in managing the condition. In this study we aimed to determine the effect of azithromycin on LPS-mediated epithelial release of factors relevant to airway neutrophilia and remodeling in a unique population of primary bronchial epithelial cells (PBECs) derived from stable lung allografts. METHODS PBECs were established from bronchial brushings of stable lung transplant recipients and treated with lipopolysaccharide (LPS, 0.1, 1 and 10 microg/ml) for 48 hours. Interleukin-8 (IL-8), granulocyte macrophage colony-stimulating factor (GM-CSF) and vascular endothelial growth factor (VEGF) protein levels were measured by Luminex analyzer. PBECs were then incubated with LPS and azithromycin, and protein levels were again determined. RESULTS LPS caused a significant increase in IL-8 and GM-CSF at concentrations of 1 and 10 microg/ml, with no effect on VEGF release. Azithromycin caused a significant decrease in the LPS-stimulated IL-8 and GM-CSF release. CONCLUSIONS LPS upregulates release of IL-8 and GM-CSF from PBECs derived from stable lung allografts. Sub-microbicidal concentrations of azithromycin attenuate this and may, therefore, alleviate infection-driven neutrophilic airway inflammation and remodeling in the allograft airway.
Collapse
Affiliation(s)
- Desmond M Murphy
- Applied Immunobiology and Transplantation Research Group, Institute of Cellular Medicine, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Boost KA, Sadik CD, Bachmann M, Zwissler B, Pfeilschifter J, Mühl H. IFN-gamma impairs release of IL-8 by IL-1beta-stimulated A549 lung carcinoma cells. BMC Cancer 2008; 8:265. [PMID: 18801189 PMCID: PMC2556346 DOI: 10.1186/1471-2407-8-265] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Accepted: 09/18/2008] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Production of interferon (IFN)-gamma is key to efficient anti-tumor immunity. The present study was set out to investigate effects of IFNgamma on the release of the potent pro-angiogenic mediator IL-8 by human A549 lung carcinoma cells. METHODS A549 cells were cultured and stimulated with interleukin (IL)-1beta alone or in combination with IFNgamma. IL-8 production by these cells was analyzed with enzyme linked immuno sorbent assay (ELISA). mRNA-expression was analyzed by real-time PCR and RNase protection assay (RPA), respectively. Expression of inhibitor-kappa Balpha, cellular IL-8, and cyclooxygenase-2 was analyzed by Western blot analysis. RESULTS Here we demonstrate that IFNgamma efficiently reduced IL-8 secretion under the influence of IL-1beta. Surprisingly, real-time PCR analysis and RPA revealed that the inhibitory effect of IFNgamma on IL-8 was not associated with significant changes in mRNA levels. These observations concurred with lack of a modulatory activity of IFNgamma on IL-1beta-induced NF-kappaB activation as assessed by cellular IkappaB levels. Moreover, analysis of intracellular IL-8 suggests that IFNgamma modulated IL-8 secretion by action on the posttranslational level. In contrast to IL-8, IL-1beta-induced cyclooxygenase-2 expression and release of IL-6 were not affected by IFNgamma indicating that modulation of IL-1beta action by this cytokine displays specificity. CONCLUSION Data presented herein agree with an angiostatic role of IFNgamma as seen in rodent models of solid tumors and suggest that increasing T helper type 1 (Th1)-like functions in lung cancer patients e.g. by local delivery of IFNgamma may mediate therapeutic benefit via mechanisms that potentially include modulation of pro-angiogenic IL-8.
Collapse
Affiliation(s)
- Kim A Boost
- Klinik für Anaesthesiologie, Ludwig-Maximilians-University Munich, Marchioninistr. 15, 81377 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Ho CC, Liao WY, Wang CY, Lu YH, Huang HY, Chen HY, Chan WK, Chen HW, Yang PC. TREM-1 Expression in Tumor-associated Macrophages and Clinical Outcome in Lung Cancer. Am J Respir Crit Care Med 2008; 177:763-70. [DOI: 10.1164/rccm.200704-641oc] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
25
|
|
26
|
Abstract
Infection and chronic inflammation contribute to about 1 in 4 of all cancer cases. Mediators of the inflammatory response, e.g., cytokines, free radicals, prostaglandins and growth factors, can induce genetic and epigenetic changes including point mutations in tumor suppressor genes, DNA methylation and post-translational modifications, causing alterations in critical pathways responsible for maintaining the normal cellular homeostasis and leading to the development and progression of cancer. Recent discovery of an interaction between microRNAs and innate immunity during inflammation has further strengthened the association between inflammation and cancer.
Collapse
Affiliation(s)
- S Perwez Hussain
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, MD, USA
| | | |
Collapse
|
27
|
Tew GW, Lorimer EL, Berg TJ, Zhi H, Li R, Williams CL. SmgGDS regulates cell proliferation, migration, and NF-kappaB transcriptional activity in non-small cell lung carcinoma. J Biol Chem 2007; 283:963-76. [PMID: 17951244 DOI: 10.1074/jbc.m707526200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is promoted by the increased activities of several small GTPases, including K-Ras4B, Rap1A, Rap1B, RhoC, and Rac1. SmgGDS is an unusual guanine nucleotide exchange factor that activates many of these small GTPases, and thus may promote NSCLC development or progression. We report here that SmgGDS protein levels are elevated in NSCLC tumors, compared with normal lung tissue from the same patients or from individuals without cancer. To characterize SmgGDS functions in NSCLC, we tested the effects of silencing SmgGDS expression by transfecting cultured NSCLC cells with SmgGDS small interfering RNA (siRNA). Cells with silenced SmgGDS expression form fewer colonies in soft agar, do not proliferate in culture due to an arrest in G(1) phase, and exhibit disrupted myosin organization and reduced cell migration. The transcriptional activity of NF-kappaB in NSCLC cells is diminished by transfecting the cells with SmgGDS siRNA, and enhanced by transfecting the cells with a cDNA encoding SmgGDS. Because RhoA is a major substrate for SmgGDS, we investigated whether diminished RhoA expression mimics the effects of diminished SmgGDS expression. Silencing RhoA expression with RhoA siRNA disrupts myosin organization, but only moderately decreases cell proliferation and does not inhibit migration. Our finding that the aggressive NSCLC phenotype is more effectively suppressed by silencing SmgGDS than by silencing RhoA is consistent with the ability of SmgGDS to regulate multiple small GTPases in addition to RhoA. These results demonstrate that SmgGDS promotes the malignant NSCLC phenotype and is an intriguing therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Gaik Wei Tew
- Department of Pharmacology and Toxicology, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, 53226, USA
| | | | | | | | | | | |
Collapse
|
28
|
Seike M, Yanaihara N, Bowman ED, Zanetti KA, Budhu A, Kumamoto K, Mechanic LE, Matsumoto S, Yokota J, Shibata T, Sugimura H, Gemma A, Kudoh S, Wang XW, Harris CC. Use of a cytokine gene expression signature in lung adenocarcinoma and the surrounding tissue as a prognostic classifier. J Natl Cancer Inst 2007; 99:1257-1269. [PMID: 17686824 DOI: 10.1093/jnci/djm083] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND A 17-cytokine gene expression signature in noncancerous hepatic tissue from patients with metastatic hepatocellular carcinoma (HCC) was recently found to predict HCC metastasis and recurrence. We examined whether the cytokine gene expression profile of noncancerous lung tissue could predict the metastatic capability of adjacent lung adenocarcinoma. METHODS We analyzed a 15-cytokine gene expression profile in noncancerous lung tissue and corresponding lung tumor tissue from 80 US lung adenocarcinoma patients using real-time quantitative reverse transcription-polymerase chain reaction. We then used unsupervised hierarchical clustering and Prediction Analysis of Microarray classification to test the prognostic ability of the 15-cytokine gene profile in the US patients and in an independent validation set comprising 50 Japanese patients with stage I disease. Survival was analyzed by the Kaplan-Meier method using the log-rank test, and univariate and multivariable Cox proportional hazards modeling were used to analyze the association of clinical variables with patient survival. All statistical tests were two-sided. RESULTS A 15-cytokine gene signature in noncancerous lung tissue primarily reflected the lymph node status of 80 lung adenocarcinoma patients, whereas the gene signature of the corresponding lung tumor tissue was associated with prognosis independent of lymph node status. Cytokine Lung Adenocarcinoma Survival Signature of 11 genes (CLASS-11), a refined 11-gene signature, accurately classified patients, including those with stage I disease, according to risk of death from adenocarcinoma. CLASS-11 prognostic classification was statistically significantly associated with survival and was an independent prognostic factor for stage I patients (hazard ratio for death in the high-risk CLASS-11 group compared with the low-risk CLASS-11 reference group = 7.46, 95% confidence interval = 2.14 to 26.05; P = .002). CLASS-11 also classified patients in the validation set according to risk of recurrence. CONCLUSION CLASS-11, which consists of genes for pro- and anti-inflammatory cytokines, identifies stage I lung adenocarcinoma patients who have a poor prognosis.
Collapse
Affiliation(s)
- Masahiro Seike
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4258, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Raman D, Baugher PJ, Thu YM, Richmond A. Role of chemokines in tumor growth. Cancer Lett 2007; 256:137-65. [PMID: 17629396 PMCID: PMC2065851 DOI: 10.1016/j.canlet.2007.05.013] [Citation(s) in RCA: 449] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Revised: 05/21/2007] [Accepted: 05/21/2007] [Indexed: 12/11/2022]
Abstract
Chemokines play a paramount role in the tumor progression. Chronic inflammation promotes tumor formation. Both tumor cells and stromal cells elaborate chemokines and cytokines. These act either by autocrine or paracrine mechanisms to sustain tumor cell growth, induce angiogenesis and facilitate evasion of immune surveillance through immunoediting. The chemokine receptor CXCR2 and its ligands promote tumor angiogenesis and leukocyte infiltration into the tumor microenvironment. In harsh acidic and hypoxic microenvironmental conditions tumor cells up-regulate their expression of CXCR4, which equips them to migrate up a gradient of CXCL12 elaborated by carcinoma-associated fibroblasts (CAFs) to a normoxic microenvironment. The CXCL12-CXCR4 axis facilitates metastasis to distant organs and the CCL21-CCR7 chemokine ligand-receptor pair favors metastasis to lymph nodes. These two chemokine ligand-receptor systems are common key mediators of tumor cell metastasis for several malignancies and as such provide key targets for chemotherapy. In this paper, the role of specific chemokines/chemokine receptor interactions in tumor progression, growth and metastasis and the role of chemokine/chemokine receptor interactions in the stromal compartment as related to angiogenesis, metastasis, and immune response to the tumor are reviewed.
Collapse
Affiliation(s)
| | | | - Yee Mon Thu
- Department of Cancer Biology, Nashville, TN 37232, USA
| | - Ann Richmond
- Department of Veterans Affairs, Nashville, TN 37232, USA
- Department of Cancer Biology, Nashville, TN 37232, USA
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Corresponding author Dr. Ann Richmond, Department of Cancer Biology, Vanderbilt University School of Medicine, 432 PRB, 23 Avenue South @ Pierce, Nashville, TN 37232. U.S.A. Tel. +1 615 343 7777 Fax: +1 615 936 2911 e-mail:
| |
Collapse
|
30
|
Ott LW, Resing KA, Sizemore AW, Heyen JW, Cocklin RR, Pedrick NM, Woods HC, Chen JY, Goebl MG, Witzmann FA, Harrington MA. Tumor Necrosis Factor-alpha- and interleukin-1-induced cellular responses: coupling proteomic and genomic information. J Proteome Res 2007; 6:2176-85. [PMID: 17503796 PMCID: PMC2877378 DOI: 10.1021/pr060665l] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pro-inflammatory cytokines, Tumor Necrosis Factor-alpha (TNFalpha) and Interleukin-1 (IL-1) mediate the innate immune response. Dysregulation of the innate immune response contributes to the pathogenesis of cancer, arthritis, and congestive heart failure. TNFalpha- and IL-1-induced changes in gene expression are mediated by similar transcription factors; however, TNFalpha and IL-1 receptor knock-out mice differ in their sensitivities to a known initiator (lipopolysaccharide, LPS) of the innate immune response. The contrasting responses to LPS indicate that TNFalpha and IL-1 regulate different processes. A large-scale proteomic analysis of TNFalpha- and IL-1-induced responses was undertaken to identify processes uniquely regulated by TNFalpha and IL-1. When combined with genomic studies, our results indicate that TNFalpha, but not IL-1, mediates cell cycle arrest.
Collapse
Affiliation(s)
- Lee W Ott
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, MS 4053, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yao PL, Lin YC, Sawhney P, Richburg JH. Transcriptional regulation of FasL expression and participation of sTNF-alpha in response to sertoli cell injury. J Biol Chem 2006; 282:5420-31. [PMID: 17192273 DOI: 10.1074/jbc.m609068200] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Fas/FasL signaling pathway has previously been demonstrated to be critical for triggering germ cell apoptosis in response to mono-(2-ethylhexyl)phthalate (MEHP)-induced Sertoli cell injury. Although Sertoli cells ubiquitously express the FasL protein, MEHP-induced germ cell apoptosis appears to tightly correlate with increased levels of Sertoli cell FasL. Here we characterize the transcriptional regulation of the murine FasL gene in Sertoli cells after MEHP exposure. A serial deletion strategy for 1.5 kb of the 5'-upstream activating sequence of the FasL promoter was used to determine transcriptional activity in response to MEHP. Luciferase activity of the FasL promoter in the rat Sertoli cell line ASC-17D revealed that two regions, -500 to -324 and -1250 to -1000, were necessary to drive the inducible transcription of FasL. Sequence analysis of these two regions revealed two cis-regulatory elements, NF-kappaB and Sp-1. By site-directed mutagenesis, electrophoretic mobility shift and chromatin immunoprecipitation assays, it was confirmed that MEHP-induced FasL expression is enhanced through the transcriptional regulation of both NF-kappaB and Sp-1. Experiments performed both in vitro and in vivo revealed that MEHP exposure results in an increased production of sTNF-alpha and that sTNF-alpha-mediated NF-kappaB activation causes robust increases in FasL levels in both the ASC-17D Sertoli cell line and in primary rat Sertoli cell/germ cell co-cultures. In the seminiferous epithelium, Sertoli cells express TNFR1, whereas germ cells produce TNF-alpha. Therefore, sTNF-alpha released by germ cells after MEHP-induced Sertoli cell injury acts upon Sertoli cell TNFR1 and activates NF-kappaB and Sp-1 that consequently causes a robust induction of FasL expression. These novel findings point to a potential "feed-forward" signaling mechanism by which germ cells prompt Sertoli cells to trigger their apoptotic elimination.
Collapse
Affiliation(s)
- Pei-Li Yao
- Institute for Cellular and Molecular Biology, College of Pharmacy, University of Texas, Austin, Texas 78712-1074, USA
| | | | | | | |
Collapse
|
32
|
Wei CW, Cheng JY, Young TH. Elucidating in vitro cell-cell interaction using a microfluidic coculture system. Biomed Microdevices 2006; 8:65-71. [PMID: 16491333 DOI: 10.1007/s10544-006-6384-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This work presents a novel microfluidic coculture system that improves the accuracy of evaluating the interaction between cocultured cell types. A microfluidic coculture chip, fabricated by CO(2) laser direct-writing on polymethyl methacrylate (PMMA), was designed to separate two cell types using a microchannel, while permitting transfer of cellular media. The system has two up-stream wells and five down-stream wells. As an example, released inflammatory cytokines (e.g., interleukin-1 beta (IL-1 beta) and tumor necrosis factor-alpha (TNF-alpha)), activated in up-stream macrophages, flow through a microfluidic mixing system, generating linear concentration gradients in down-stream wells and inducing down-stream osteoblasts to release prostaglandin E2 (PGE2), a well-known bone resorption marker. Osteoblast viability was assessed by 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay. This novel coculture system can be applied to evaluate cell-cell interaction while physically separating interacting cells.
Collapse
Affiliation(s)
- Cheng-Wey Wei
- Research Center for Applied Sciences, Academia Sinica, Taiwan
| | | | | |
Collapse
|
33
|
Hays AM, Srinivasan D, Witten ML, Carter DE, Lantz RC. Arsenic and cigarette smoke synergistically increase DNA oxidation in the lung. Toxicol Pathol 2006; 34:396-404. [PMID: 16844668 DOI: 10.1080/01926230600824926] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Epidemiological evidence has indicated that arsenic and cigarette smoking exposure act synergistically to increase the incidence of lung cancer. Since oxidative damage of DNA has been linked to cancer, our hypothesis is that aerosolized arsenic and cigarette smoke work synergistically to increase oxidative stress and increase DNA oxidation in the lung. To test this hypothesis male Syrian golden hamsters were exposed to room air (control), aerosolized arsenic compounds (3.2 mg/m3 for 30 minutes), cigarette smoke (5 mg/m3 for 30 minutes), or both smoke and arsenic. Exposures were for 5 days/week for 5 or 28-days. Animals were sacrificed one day after the last exposure. In the 28-day group, glutathione levels and DNA oxidation (8-oxo-2'-deoxyguanosine (8-oxo-dG)) were determined. Our results show that in the 28-day arsenic/smoke group there was a significant decrease in both the reduced and total glutathione levels compared with arsenic or smoke alone. This correlated with a 5-fold increase in DNA oxidation as shown by HPLC. Immunohistochemical localization of 8-oxo-dG showed increase staining in nuclei of airway epithelium and subadjacent interstitial cells. These results show that dual exposure of arsenic and cigarette smoke at environmentally relevant levels can act synergistically to cause DNA damage.
Collapse
Affiliation(s)
- Allison Marie Hays
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona 85724-5044, USA
| | | | | | | | | |
Collapse
|