1
|
Ries A, Slany A, Pirker C, Mader JC, Mejri D, Mohr T, Schelch K, Flehberger D, Maach N, Hashim M, Hoda MA, Dome B, Krupitza G, Berger W, Gerner C, Holzmann K, Grusch M. Primary and hTERT-Transduced Mesothelioma-Associated Fibroblasts but Not Primary or hTERT-Transduced Mesothelial Cells Stimulate Growth of Human Mesothelioma Cells. Cells 2023; 12:2006. [PMID: 37566084 PMCID: PMC10417280 DOI: 10.3390/cells12152006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/12/2023] Open
Abstract
Pleural mesothelioma (PM) is an aggressive malignancy that develops in a unique tumor microenvironment (TME). However, cell models for studying the TME in PM are still limited. Here, we have generated and characterized novel human telomerase reverse transcriptase (hTERT)-transduced mesothelial cell and mesothelioma-associated fibroblast (Meso-CAF) models and investigated their impact on PM cell growth. Pleural mesothelial cells and Meso-CAFs were isolated from tissue of pneumothorax and PM patients, respectively. Stable expression of hTERT was induced by retroviral transduction. Primary and hTERT-transduced cells were compared with respect to doubling times, hTERT expression and activity levels, telomere lengths, proteomes, and the impact of conditioned media (CM) on PM cell growth. All transduced derivatives exhibited elevated hTERT expression and activity, and increased mean telomere lengths. Cell morphology remained unchanged, and the proteomes were similar to the corresponding primary cells. Of note, the CM of primary and hTERT-transduced Meso-CAFs stimulated PM cell growth to the same extent, while CM derived from mesothelial cells had no stimulating effect, irrespective of hTERT expression. In conclusion, all new hTERT-transduced cell models closely resemble their primary counterparts and, hence, represent valuable tools to investigate cellular interactions within the TME of PM.
Collapse
Affiliation(s)
- Alexander Ries
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Astrid Slany
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
| | - Christine Pirker
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Johanna C. Mader
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
| | - Doris Mejri
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Thomas Mohr
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Waehringer Guertel 38, 1090 Vienna, Austria
- ScienceConsult—DI Thomas Mohr KG, Enzianweg 10a, 2353 Guntramsdorf, Austria
| | - Karin Schelch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.A.H.); (B.D.)
| | - Daniela Flehberger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Nadine Maach
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Muhammad Hashim
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.A.H.); (B.D.)
| | - Balazs Dome
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.A.H.); (B.D.)
- National Korányi Institute of Pulmonology, Korányi Frigyes u. 1, 1122 Budapest, Hungary
- Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Rath Gyorgy u. 7-9, 1122 Budapest, Hungary
- Department of Translational Medicine, Lund University, Sölvegatan 19, 22184 Lund, Sweden
| | - Georg Krupitza
- Department of Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090 Vienna, Austria; (A.S.); (J.C.M.); (C.G.)
| | - Klaus Holzmann
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| | - Michael Grusch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria; (A.R.); (C.P.); (D.M.); (T.M.); (K.S.); (D.F.); (N.M.); (M.H.); (W.B.); (K.H.)
| |
Collapse
|
2
|
Synthetic Secoisolariciresinol Diglucoside (LGM2605) Prevents Asbestos-Induced Inflammation and Genotoxic Cell Damage in Human Mesothelial Cells. Int J Mol Sci 2022; 23:ijms231710085. [PMID: 36077483 PMCID: PMC9456329 DOI: 10.3390/ijms231710085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/25/2022] Open
Abstract
Although alveolar macrophages play a critical role in malignant transformation of mesothelial cells following asbestos exposure, inflammatory and oxidative processes continue to occur in the mesothelial cells lining the pleura that may contribute to the carcinogenic process. Malignant transformation of mesothelial cells following asbestos exposure occurs over several decades; however, amelioration of DNA damage, inflammation, and cell injury may impede the carcinogenic process. We have shown in an in vitro model of asbestos-induced macrophage activation that synthetic secoisolariciresinol diglucoside (LGM2605), given preventively, reduced inflammatory cascades and oxidative/nitrosative cell damage. Therefore, it was hypothesized that LGM2605 could also be effective in reducing asbestos-induced activation and the damage of pleural mesothelial cells. LGM2605 treatment (50 µM) of huma n pleural mesothelial cells was initiated 4 h prior to exposure to asbestos (crocidolite, 20 µg/cm2). Supernatant and cells were evaluated at 0, 2, 4, and 8 h post asbestos exposure for reactive oxygen species (ROS) generation, DNA damage (oxidized guanine), inflammasome activation (caspase-1 activity) and associated pro-inflammatory cytokine release (IL-1β, IL-18, IL-6, TNFα, and HMGB1), and markers of oxidative stress (malondialdehyde (MDA) and 8-iso-prostaglandin F2a (8-iso-PGF2α). Asbestos induced a time-dependent ROS increase that was significantly (p < 0.0001) reduced (29.4%) by LGM2605 treatment. LGM2605 pretreatment also reduced levels of asbestos-induced DNA damage by 73.6% ± 1.0%. Although levels of inflammasome-activated cytokines, IL-1β and IL-18, reached 29.2 pg/mL ± 0.7 pg/mL and 43.9 pg/mL ± 0.8 pg/mL, respectively, LGM2605 treatment significantly (p < 0.0001) reduced cytokine levels comparable to baseline (non-asbestos exposed) values (3.8 pg/mL ± 0.2 pg/mL and 5.4 pg/mL ± 0.2 pg/mL, respectively). Furthermore, levels of IL-6 and TNFα in asbestos-exposed mesothelial cells were high (289.1 pg/mL ± 2.9 pg/mL and 511.3 pg/mL ± 10.2 pg/mL, respectively), while remaining undetectable with LGM2605 pretreatment. HMGB1 (a key inflammatory mediator and initiator of malignant transformation) release was reduced 75.3% ± 0.4% by LGM2605. Levels of MDA and 8-iso-PGF2α, markers of oxidative cell injury, were significantly (p < 0.001) reduced by 80.5% ± 0.1% and 76.6% ± 0.3%, respectively. LGM2605, given preventively, reduced ROS generation, DNA damage, and inflammasome-activated cytokine release and key inflammatory mediators implicated in asbestos-induced malignant transformation of normal mesothelial cells.
Collapse
|
3
|
Hajj GNM, Cavarson CH, Pinto CAL, Venturi G, Navarro JR, Lima VCCD. Malignant pleural mesothelioma: an update. J Bras Pneumol 2021; 47:e20210129. [PMID: 34909922 PMCID: PMC8836658 DOI: 10.36416/1806-3756/e20210129] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022] Open
Abstract
Malignant mesotheliomas are rare types of cancers that affect the mesothelial surfaces, usually the pleura and peritoneum. They are associated with asbestos exposure, but due to a latency period of more than 30 years and difficult diagnosis, most cases are not detected until they reach advanced stages. Treatment options for this tumor type are very limited and survival ranges from 12 to 36 months. This review discusses the molecular physiopathology, current diagnosis, and latest therapeutic options for this disease.
Collapse
Affiliation(s)
- Glaucia N M Hajj
- Instituto International de Pesquisa, A.C. Camargo Cancer Center, São Paulo (SP), Brasil.,Instituto Nacional de Oncogenômica e Inovação Terapêutica, São Paulo (SP), Brasil
| | - Carolina H Cavarson
- Instituto International de Pesquisa, A.C. Camargo Cancer Center, São Paulo (SP), Brasil.,Instituto Nacional de Oncogenômica e Inovação Terapêutica, São Paulo (SP), Brasil
| | | | - Gabriela Venturi
- Instituto International de Pesquisa, A.C. Camargo Cancer Center, São Paulo (SP), Brasil.,BP Mirante, São Paulo (SP), Brasil
| | | | - Vladmir C Cordeiro de Lima
- Instituto Nacional de Oncogenômica e Inovação Terapêutica, São Paulo (SP), Brasil.,Rede D'Or, São Paulo (SP), Brasil
| |
Collapse
|
4
|
Cai C, Nishimura T, Hwang J, Hu XM, Kuroda A. Asbestos Detection with Fluorescence Microscopy Images and Deep Learning. SENSORS (BASEL, SWITZERLAND) 2021; 21:4582. [PMID: 34283157 PMCID: PMC8272007 DOI: 10.3390/s21134582] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 11/23/2022]
Abstract
Fluorescent probes can be used to detect various types of asbestos (serpentine and amphibole groups); however, the fiber counting using our previously developed software was not accurate for samples with low fiber concentration. Machine learning-based techniques (e.g., deep learning) for image analysis, particularly Convolutional Neural Networks (CNN), have been widely applied to many areas. The objectives of this study were to (1) create a database of a wide-range asbestos concentration (0-50 fibers/liter) fluorescence microscopy (FM) images in the laboratory; and (2) determine the applicability of the state-of-the-art object detection CNN model, YOLOv4, to accurately detect asbestos. We captured the fluorescence microscopy images containing asbestos and labeled the individual asbestos in the images. We trained the YOLOv4 model with the labeled images using one GTX 1660 Ti Graphics Processing Unit (GPU). Our results demonstrated the exceptional capacity of the YOLOv4 model to learn the fluorescent asbestos morphologies. The mean average precision at a threshold of 0.5 (mAP@0.5) was 96.1% ± 0.4%, using the National Institute for Occupational Safety and Health (NIOSH) fiber counting Method 7400 as a reference method. Compared to our previous counting software (Intec/HU), the YOLOv4 achieved higher accuracy (0.997 vs. 0.979), particularly much higher precision (0.898 vs. 0.418), recall (0.898 vs. 0.780) and F-1 score (0.898 vs. 0.544). In addition, the YOLOv4 performed much better for low fiber concentration samples (<15 fibers/liter) compared to Intec/HU. Therefore, the FM method coupled with YOLOv4 is remarkable in detecting asbestos fibers and differentiating them from other non-asbestos particles.
Collapse
Affiliation(s)
- Changjie Cai
- Department of Occupational and Environmental Health, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, OK 73069, USA;
| | - Tomoki Nishimura
- Unit of Biotechnology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8530, Japan;
| | - Jooyeon Hwang
- Department of Occupational and Environmental Health, University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, OK 73069, USA;
| | - Xiao-Ming Hu
- Center for Analysis and Prediction of Storms, University of Oklahoma, Norman, OK 73072, USA;
| | - Akio Kuroda
- Unit of Biotechnology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8530, Japan;
| |
Collapse
|
5
|
Gillezeau CN, van Gerwen M, Ramos J, Liu B, Flores R, Taioli E. Biomarkers for malignant pleural mesothelioma: a meta-analysis. Carcinogenesis 2019; 40:1320-1331. [DOI: 10.1093/carcin/bgz103] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/13/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023] Open
Abstract
Abstract
Malignant pleural mesothelioma (MPM) is a rare but aggressive cancer, and early detection is associated with better survival. Mesothelin, fibulin-3 and osteopontin have been suggested as screening biomarkers. The study conducted a meta-analysis of the mean differences of mesothelin, osteopontin and fibulin-3 in blood and pleural samples. PubMed searches were conducted for studies that measured levels of mesothelin, osteopontin and fibulin-3 in participants with MPM compared with malignancy, benign lung disease or healthy participants. Thirty-two studies with mesothelin levels, 12 studies with osteopontin levels and 9 studies with fibulin-3 levels were included in the meta-analysis. Statistically significant mean differences were seen between MPM patients and all other comparison groups for mesothelin blood and pleural levels. Statistically significant differences in blood osteopontin levels were seen between participants with benign lung disease and healthy participants compared with participants with MPM, but not when comparing participants with cancer with MPM participants. There were not enough studies that reported osteopontin levels in pleural fluid to complete a meta-analysis. Statistically significant differences were seen in both blood and pleural levels of fibulin-3 in MPM patients compared with all other groups. On the basis of these results, mesothelin and fibulin-3 levels appear to be significantly lower in all control groups compared with those with MPM, making them good candidates for screening biomarkers. Osteopontin may be a useful biomarker for screening healthy individuals or those with benign lung disease but would not be useful for screening patients with malignancies.
Collapse
Affiliation(s)
- Christina N Gillezeau
- Institute for Translational Epidemiology and Department of Population Health Science and Policy, New York, NY, USA
| | - Maaike van Gerwen
- Institute for Translational Epidemiology and Department of Population Health Science and Policy, New York, NY, USA
| | - Julio Ramos
- Institute for Translational Epidemiology and Department of Population Health Science and Policy, New York, NY, USA
| | - Bian Liu
- Institute for Translational Epidemiology and Department of Population Health Science and Policy, New York, NY, USA
| | - Raja Flores
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emanuela Taioli
- Institute for Translational Epidemiology and Department of Population Health Science and Policy, New York, NY, USA
| |
Collapse
|
6
|
Ishida T, Fujihara N, Nishimura T, Funabashi H, Hirota R, Ikeda T, Kuroda A. Live-cell imaging of macrophage phagocytosis of asbestos fibers under fluorescence microscopy. Genes Environ 2019; 41:14. [PMID: 31178942 PMCID: PMC6549298 DOI: 10.1186/s41021-019-0129-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/05/2019] [Indexed: 11/10/2022] Open
Abstract
Background Frustrated phagocytosis occurs when an asbestos fiber > 10 μm in length is engulfed imperfectly by a macrophage, and it is believed to be associated with chromosomal instability. Few studies have focused on dynamic cellular imaging to assess the toxicity of hazardous inorganic materials such as asbestos. One reason for this is the relative lack of fluorescent probes available to facilitate experimental visualization of inorganic materials. We recently developed asbestos-specific fluorescent probes based on asbestos-binding proteins, and achieved efficient fluorescent labeling of asbestos. Results Live-cell imaging with fluorescent asbestos probes was successfully utilized to dynamically analyze asbestos phagocytosis. The fluorescently labeled asbestos fibers were phagocytosed by RAW 264.7 macrophages. Internalized fibers of < 5 μm in length were visualized clearly via overlaid phase contrast and fluorescence microscopy images, but they were not clearly depicted using phase contrast images alone. Approximately 60% of the cells had phagocytosed asbestos fibers after 2 h, but over 96% of cells remained alive even 24 h after the addition of asbestos fibers. Immediate cell death was only observed when an asbestos fiber was physically pulled from a cell by an external force. Notably, at 24 h after the addition of asbestos fibers an approximately 4-fold increase in the number of binucleated cells was observed. Monitoring of individual cell divisions of cells that had phagocytosed asbestos suggested that binucleated cells were formed via the inhibition of cell separation, by asbestos fibers of > 10 μm in length that were localized in the proximity of the intercellular bridge. Conclusions Fluorescently labeled asbestos facilitated visualization of the dynamic biological processes that occur during and after the internalization of asbestos fibers, and indicated that (i) frustrated phagocytosis itself does not lead to immediate cell death unless the asbestos fiber is physically pulled from the cell by an external force, and (ii) macrophages that have phagocytosed asbestos can divide but sometimes the resulting daughter cells fuse, leading to the formation of a binucleated cell. This fusion only seemed to occur when a comparatively long asbestos fiber (> 10 μm) was shared by two daughter cells.
Collapse
Affiliation(s)
- Takenori Ishida
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| | - Nobutoshi Fujihara
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| | - Tomoki Nishimura
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| | - Hisakage Funabashi
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| | - Ryuichi Hirota
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| | - Takeshi Ikeda
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| | - Akio Kuroda
- Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi Hiroshima, Hiroshima, 739-8530 Japan
| |
Collapse
|
7
|
The Future of Mesothelioma Research: Basic Science Research. CARING FOR PATIENTS WITH MESOTHELIOMA: PRINCIPLES AND GUIDELINES 2019. [PMCID: PMC7119960 DOI: 10.1007/978-3-319-96244-3_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Our current understanding of mesothelioma in terms of disease induction, development, and treatment is underpinned by decades of basic laboratory science. In this chapter, we discuss the tools that have been developed to aid our understanding of mesothelioma such as cell lines and animal models. We then go on to detail the current use and understanding of conventional therapies for mesothelioma, e.g. chemotherapy, surgery, and radiotherapy, plus their mechanisms of action, and why they may be ineffective. Finally, we discuss a range of newer treatments that are either undergoing clinical trials or are still in the earlier stages of preclinical investigation. These include a growing number of immunotherapies (e.g. checkpoint inhibitors), plus targeted therapies, the search for clinical biomarkers to predict whether patients with mesothelioma might respond to particular treatments, and combined therapies where conventional treatments may be added to newer drugs. The strategy of repositioning existing drugs, approved for other diseases, to treat mesothelioma is also discussed.
Collapse
|
8
|
The Synthetic Lignan Secoisolariciresinol Diglucoside Prevents Asbestos-Induced NLRP3 Inflammasome Activation in Murine Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7395238. [PMID: 29075366 PMCID: PMC5615985 DOI: 10.1155/2017/7395238] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The interaction of asbestos with macrophages drives two key processes that are linked to malignancy: (1) the generation of reactive oxygen species (ROS)/reactive nitrogen species (RNS) and (2) the activation of an inflammation cascade that drives acute and chronic inflammation, with the NLRP3 inflammasome playing a key role. Synthetic secoisolariciresinol diglucoside (SDG), LGM2605, is a nontoxic lignan with anti-inflammatory and antioxidant properties and was evaluated for protection from asbestos in murine peritoneal macrophages (MF). METHODS MFs were exposed to crocidolite asbestos ± LGM2605 given 4 hours prior to exposure and evaluated at various times for NLRP3 expression, secretion of inflammasome-activated cytokines (IL-1β and IL-18), proinflammatory cytokines (IL-6, TNFα, and HMGB1), NF-κB activation, and levels of total nitrates/nitrites. RESULTS Asbestos induces a significant (p < 0.0001) increase in the NLRP3 subunit, release of proinflammatory cytokines, NLRP3-activated cytokines, NF-κB, and levels of nitrates/nitrites. LGM2605 significantly reduced NLRP3 ranging from 40 to 81%, IL-1β by 89-96%, and TNFα by 67-78%, as well as activated NF-κB by 48-49% while decreasing levels of nitrates/nitrites by 85-93%. CONCLUSIONS LGM2605 reduced asbestos-induced NLRP3 expression, proinflammatory cytokine release, NF-κB activation, and nitrosative stress in MFs supporting its possible use in preventing the asbestos-induced inflammatory cascade leading to malignancy.
Collapse
|
9
|
Keshava S, Rao LVM, Pendurthi UR. Intrapleural Adenoviral-mediated Endothelial Cell Protein C Receptor Gene Transfer Suppresses the Progression of Malignant Pleural Mesothelioma in a Mouse Model. Sci Rep 2016; 6:36829. [PMID: 27833109 PMCID: PMC5104979 DOI: 10.1038/srep36829] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/21/2016] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive thoracic cancer with a high mortality rate as it responds poorly to standard therapeutic interventions. Our recent studies showed that expression of endothelial cell protein C receptor (EPCR) in MPM cells suppresses tumorigenicity. The present study was aimed to investigate the mechanism by which EPCR suppresses MPM tumor growth and evaluate whether EPCR gene therapy could suppress the progression of MPM in a mouse model of MPM. Measurement of cytokines from the pleural lavage showed that mice implanted with MPM cells expressing EPCR had elevated levels of IFNγ and TNFα compared to mice implanted with MPM cells lacking EPCR. In vitro studies demonstrated that EPCR expression renders MPM cells highly susceptible to IFNγ + TNFα-induced apoptosis. Intrapleural injection of Ad.EPCR into mice with an established MPM originating from MPM cells lacking EPCR reduced the progression of tumor growth. Ad.EPCR treatment elicited recruitment of macrophages and NK cells into the tumor microenvironment and increased IFNγ and TNFα levels in the pleural space. Ad.EPCR treatment resulted in a marked increase in tumor cell apoptosis. In summary, our data show that EPCR expression in MPM cells promotes tumor cell apoptosis, and intrapleural EPCR gene therapy suppresses MPM progression.
Collapse
Affiliation(s)
- Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - L Vijaya Mohan Rao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| | - Usha R Pendurthi
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Texas, USA
| |
Collapse
|
10
|
Kadariya Y, Menges CW, Talarchek J, Cai KQ, Klein-Szanto AJ, Pietrofesa RA, Christofidou-Solomidou M, Cheung M, Mossman BT, Shukla A, Testa JR. Inflammation-Related IL1β/IL1R Signaling Promotes the Development of Asbestos-Induced Malignant Mesothelioma. Cancer Prev Res (Phila) 2016; 9:406-414. [PMID: 26935421 DOI: 10.1158/1940-6207.capr-15-0347] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 02/25/2016] [Indexed: 11/16/2022]
Abstract
Exposure to asbestos is causally associated with the development of malignant mesothelioma, a cancer of cells lining the internal body cavities. Malignant mesothelioma is an aggressive cancer resistant to all current therapies. Once inhaled or ingested, asbestos causes inflammation in and around tissues that come in contact with these carcinogenic fibers. Recent studies suggest that inflammation is a major contributing factor in the development of many types of cancer, including malignant mesothelioma. The NALP3/NLRP3 inflammasome, including the component ASC, is thought to be an important mediator of inflammation in cells that sense extracellular insults, such as asbestos, and activate a signaling cascade resulting in release of mature IL1β and recruitment of inflammatory cells. To determine if inflammasome-mediated inflammation contributes to asbestos-induced malignant mesothelioma, we chronically exposed Asc-deficient mice and wild-type littermates to asbestos and evaluated differences in tumor incidence and latency. The Asc-deficient mice showed significantly delayed tumor onset and reduced malignant mesothelioma incidence compared with wild-type animals. We also tested whether inflammation-related release of IL1β contributes to tumor development in an accelerated mouse model of asbestos-induced malignant mesothelioma. Nf2(+/-);Cdkn2a(+/-) mice exposed to asbestos in the presence of anakinra, an IL1 receptor (IL1R) antagonist, showed a marked delay in the median time of malignant mesothelioma onset compared with similarly exposed mice given vehicle control (33.1 weeks vs. 22.6 weeks, respectively). Collectively, these studies provide evidence for a link between inflammation-related IL1β/IL1R signaling and the development of asbestos-induced malignant mesothelioma. Furthermore, these findings provide rationale for chemoprevention strategies targeting IL1β/IL1R signaling in high-risk, asbestos-exposed populations. Cancer Prev Res; 9(5); 406-14. ©2016 AACR.
Collapse
Affiliation(s)
- Yuwaraj Kadariya
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Craig W Menges
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | | | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Andres J Klein-Szanto
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111.,Histopathology Facility, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Ralph A Pietrofesa
- Department of Medicine, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Melpo Christofidou-Solomidou
- Department of Medicine, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Mitchell Cheung
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Brooke T Mossman
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, 05405-0068
| | - Arti Shukla
- Department of Pathology, University of Vermont College of Medicine, Burlington, VT, 05405-0068
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| |
Collapse
|
11
|
Asbestos Induces Oxidative Stress and Activation of Nrf2 Signaling in Murine Macrophages: Chemopreventive Role of the Synthetic Lignan Secoisolariciresinol Diglucoside (LGM2605). Int J Mol Sci 2016; 17:322. [PMID: 26938529 PMCID: PMC4813184 DOI: 10.3390/ijms17030322] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/15/2016] [Accepted: 02/23/2016] [Indexed: 11/29/2022] Open
Abstract
The interaction of asbestos fibers with macrophages generates harmful reactive oxygen species (ROS) and subsequent oxidative cell damage that are key processes linked to malignancy. Secoisolariciresinol diglucoside (SDG) is a non-toxic, flaxseed-derived pluripotent compound that has antioxidant properties and may thus function as a chemopreventive agent for asbestos-induced mesothelioma. We thus evaluated synthetic SDG (LGM2605) in asbestos-exposed, elicited murine peritoneal macrophages as an in vitro model of tissue phagocytic response to the presence of asbestos in the pleural space. Murine peritoneal macrophages (MFs) were exposed to crocidolite asbestos fibers (20 µg/cm2) and evaluated at various times post exposure for cytotoxicity, ROS generation, malondialdehyde (MDA), and levels of 8-iso Prostaglandin F2α (8-isoP). We then evaluated the ability of LGM2605 to mitigate asbestos-induced oxidative stress by administering LGM2605 (50 µM) 4-h prior to asbestos exposure. We observed a significant (p < 0.0001), time-dependent increase in asbestos-induced cytotoxicity, ROS generation, and the release of MDA and 8-iso Prostaglandin F2α, markers of lipid peroxidation, which increased linearly over time. LGM2605 treatment significantly (p < 0.0001) reduced asbestos-induced cytotoxicity and ROS generation, while decreasing levels of MDA and 8-isoP by 71%–88% and 41%–73%, respectively. Importantly, exposure to asbestos fibers induced cell protective defenses, such as cellular Nrf2 activation and the expression of phase II antioxidant enzymes, HO-1 and Nqo1 that were further enhanced by LGM2605 treatment. LGM2605 boosted antioxidant defenses, as well as reduced asbestos-induced ROS generation and markers of oxidative stress in murine peritoneal macrophages, supporting its possible use as a chemoprevention agent in the development of asbestos-induced malignant mesothelioma.
Collapse
|
12
|
Bioanalytical techniques for detecting biomarkers of response to human asbestos exposure. Bioanalysis 2016; 7:1157-73. [PMID: 26039812 DOI: 10.4155/bio.15.53] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Asbestos exposure is known to cause lung cancer and mesothelioma and its health and economic impacts have been well documented. The exceptionally long latency periods of most asbestos-related diseases have hampered preventative and precautionary steps thus far. We aimed to summarize the state of knowledge on biomarkers of response to asbestos exposure. Asbestos is not present in human biological fluids; rather it is inhaled and trapped in lung tissue. Biomarkers of response, which reflect a change in biologic function in response to asbestos exposure, are analyzed. Several classes of molecules have been studied and evaluated for their potential utility as biomarkers of asbestos exposure. These studies range from small molecule oxidative stress biomarkers to proteins involved in immune responses.
Collapse
|
13
|
Pietrofesa RA, Velalopoulou A, Arguiri E, Menges CW, Testa JR, Hwang WT, Albelda SM, Christofidou-Solomidou M. Flaxseed lignans enriched in secoisolariciresinol diglucoside prevent acute asbestos-induced peritoneal inflammation in mice. Carcinogenesis 2015; 37:177-87. [PMID: 26678224 PMCID: PMC4745643 DOI: 10.1093/carcin/bgv174] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/11/2015] [Indexed: 12/18/2022] Open
Abstract
Following acute exposure to crocidolite asbestos fibers, flaxseed lignans, enriched in secoisolariciresinol diglucoside (SDG), significantly reduced peritoneal inflammation, proinflammatory/profibrogenic cytokine release and oxidative/nitrosative stress in mice. Our findings support the potential role of SDG, which is safe and well-tolerated, in the chemoprevention of malignant mesothelioma. Malignant mesothelioma (MM), linked to asbestos exposure, is a highly lethal form of thoracic cancer with a long latency period, high mortality and poor treatment options. Chronic inflammation and oxidative tissue damage caused by asbestos fibers are linked to MM development. Flaxseed lignans, enriched in secoisolariciresinol diglucoside (SDG), have antioxidant, anti-inflammatory and cancer chemopreventive properties. As a prelude to chronic chemoprevention studies for MM development, we tested the ability of flaxseed lignan component (FLC) to prevent acute asbestos-induced inflammation in MM-prone Nf2+/mu mice. Mice (n = 16–17 per group) were placed on control (CTL) or FLC-supplemented diets initiated 7 days prior to a single intraperitoneal bolus of 400 µg of crocidolite asbestos. Three days post asbestos exposure, mice were evaluated for abdominal inflammation, proinflammatory/profibrogenic cytokine release, WBC gene expression changes and oxidative and nitrosative stress in peritoneal lavage fluid (PLF). Asbestos-exposed mice fed CTL diet developed acute inflammation, with significant (P < 0.0001) elevations in WBCs and proinflammatory/profibrogenic cytokines (IL-1ß, IL-6, TNFα, HMGB1 and active TGFß1) relative to baseline (BL) levels. Alternatively, asbestos-exposed FLC-fed mice had a significant (P < 0.0001) decrease in PLF WBCs and proinflammatory/profibrogenic cytokine levels relative to CTL-fed mice. Importantly, PLF WBC gene expression of cytokines (IL-1ß, IL-6, TNFα, HMGB1 and TGFß1) and cytokine receptors (TNFαR1 and TGFßR1) were also downregulated by FLC. FLC also significantly (P < 0.0001) blunted asbestos-induced nitrosative and oxidative stress. FLC reduces acute asbestos-induced peritoneal inflammation, nitrosative and oxidative stress and may thus prove to be a promising agent in the chemoprevention of MM.
Collapse
Affiliation(s)
| | | | | | - Craig W Menges
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA and
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA and
| | - Wei-Ting Hwang
- Department of Biostatistics and Epidemiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
14
|
Inhibition of hedgehog signaling reduces the side population in human malignant mesothelioma cell lines. Cancer Gene Ther 2015. [PMID: 26206198 PMCID: PMC4541142 DOI: 10.1038/cgt.2015.31] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Deregulation of crucial embryonic pathways, including hedgehog signaling, has been frequently implicated in a variety of human cancers and is emerging as an important target for anticancer therapy. This study evaluated the potential anticancer effects of cyclopamine, a chemical inhibitor of hedgehog signaling, in human malignant mesothelioma (HMM) cell lines. Cyclopamine treatment significantly decreased the proliferation of HMM cells by promoting apoptosis and shifting the cell cycle toward dormant phase. The clonogenicity and mobility of HMM cells were significantly decreased by cyclopamine treatment. Treatment of HMM cells with cyclopamine significantly reduced the abundance of side population cells, which were measured using an assay composed of Hoechst 33342 dye staining and subsequent flow cytometry. Furthermore, the expression levels of stemness-related genes were significantly affected by cyclopamine treatment. Taken together, the present study showed that targeting hedgehog signaling could reduce a more aggressive subpopulation of the cancer cells, suggesting an alternative approach for HMM therapy.
Collapse
|
15
|
Hakiri S, Osada H, Ishiguro F, Murakami H, Murakami-Tonami Y, Yokoi K, Sekido Y. Functional differences between wild-type and mutant-type BRCA1-associated protein 1 tumor suppressor against malignant mesothelioma cells. Cancer Sci 2015; 106:990-9. [PMID: 26011428 PMCID: PMC4556387 DOI: 10.1111/cas.12698] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/29/2022] Open
Abstract
Malignant mesothelioma (MM) shows inactivation of the BRCA1-associated protein 1 (BAP1) gene. In this study, we found BAP1 mutations in 5 (26%) of the 19 cell lines that we established from Japanese MM patients, and examined functional differences between the WT and mutant BAP1. First, we studied the subcellular localization of BAP1, demonstrating that the WT primarily resides in the nucleus and that the mutant BAP1 is found in the cytoplasm of the cells. Transduction of the WT BAP1 vector into MM cells with homozygous deletion at the BAP1 3′ side resulted in both inhibition of cell proliferation and anchorage-independent cell growth, whereas BAP1 mutants of a missense or C-terminal truncated form showed impaired growth inhibitory effects. Next, we studied how BAP1 is involved in MM cell survival after irradiation (IR), which causes DNA damage. After IR, we found that both WT and mutant BAP1 were similarly phosphorylated and phospho-BAP1 localized mainly in the nucleus. Interestingly, BRCA1 proteins were decreased in the MM cells with BAP1 deletion, and transduction of the mutants as well as WT BAP1 increased BRCA1 proteins, suggesting that BAP1 may promote DNA repair partly through stabilizing BRCA1. Furthermore, using the MM cells with BAP1 deletion, we found that WT BAP1, and even a missense mutant, conferred a higher survival rate after IR compared to the control vector. Our results suggested that, whereas WT BAP1 suppresses MM cell proliferation and restores cell survival after IR damage, some mutant BAP1 may also moderately retain these functions.
Collapse
Affiliation(s)
- Shuhei Hakiri
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hirotaka Osada
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Genetics, Program in Function Construction Medicine, Nagoya, Japan
| | - Futoshi Ishiguro
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideki Murakami
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Pathology, Aichi Medical University, Nagakute, Japan
| | - Yuko Murakami-Tonami
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kohei Yokoi
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshitaka Sekido
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Genetics, Program in Function Construction Medicine, Nagoya, Japan
| |
Collapse
|
16
|
Acencio MMP, Soares B, Marchi E, Silva CSR, Teixeira LR, Broaddus VC. Inflammatory Cytokines Contribute to Asbestos-Induced Injury of Mesothelial Cells. Lung 2015; 193:831-7. [PMID: 26059286 DOI: 10.1007/s00408-015-9744-4] [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: 01/07/2015] [Accepted: 05/13/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Several diseases have been related to asbestos exposure, including the pleural tumor mesothelioma. The mechanism of pleural injury by asbestos fibers is not yet fully understood. The inflammatory response with release of mediators leading to a dysregulation of apoptosis may play a pivotal role in the pathophysiology of asbestos-induced pleural disease. OBJECTIVE To determine whether pro-inflammatory cytokines produced by asbestos-exposed pleural mesothelial cells modify the injury induced by the asbestos. METHODS Mouse pleural mesothelial cells (PMC) were exposed to crocidolite or chrysotile asbestos fibers (3.0 μg/cm(2)) for 4, 24, or 48 h and assessed for viability, necrosis and apoptosis, and the production of cytokines IL-1β, IL-6 and macrophage inflammatory protein-2 (MIP-2). Cells exposed to fibers were also treated with antibodies anti-IL-1β, anti-IL-6, anti- IL-1β+anti-IL-6 or anti-MIP-2 or their irrelevant isotypes, and assessed for apoptosis and necrosis. Non-exposed cells and cells treated with wollastonite, an inert particle, were used as controls. RESULTS Mesothelial cells exposed to either crocidolite or chrysotile underwent both apoptosis and necrosis and released cytokines IL-1β, IL-6 and MIP-2. In the crocidolite group, apoptosis and the levels of all cytokines were higher than in the chrysotile group, at comparable concentrations. Neutralization of IL-1β andIL-6, but not MIP-2, inhibited apoptosis and necrosis, especially in the cells exposed to crocidolite fibers. CONCLUSIONS Both crocidolite and chrysotile asbestos fibers induced apoptosis and produced an acute inflammatory response characterized by elevated levels of IL-1β, IL-6 and MIP-2 in cultured mouse PMC. IL-1β and IL-6, but not MIP-2, were shown to contribute to asbestos-induced injury, especially in the crocidolite group.
Collapse
Affiliation(s)
- Milena Marques Pagliarelli Acencio
- Pleura Laboratory, Pulmonary Division - Heart Institute (InCor), University of São Paulo Medical School, Rua Dr. Eneas Carvalho Aguiar 44, 10 andar, Cerqueira Cesar, São Paulo, 05403-000, Brazil.
| | - Barbara Soares
- Pleura Laboratory, Pulmonary Division - Heart Institute (InCor), University of São Paulo Medical School, Rua Dr. Eneas Carvalho Aguiar 44, 10 andar, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Evaldo Marchi
- Pleura Laboratory, Pulmonary Division - Heart Institute (InCor), University of São Paulo Medical School, Rua Dr. Eneas Carvalho Aguiar 44, 10 andar, Cerqueira Cesar, São Paulo, 05403-000, Brazil.,Medical College of Jundiai, São Paulo, Brazil
| | - Carlos Sergio Rocha Silva
- Pleura Laboratory, Pulmonary Division - Heart Institute (InCor), University of São Paulo Medical School, Rua Dr. Eneas Carvalho Aguiar 44, 10 andar, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Lisete Ribeiro Teixeira
- Pleura Laboratory, Pulmonary Division - Heart Institute (InCor), University of São Paulo Medical School, Rua Dr. Eneas Carvalho Aguiar 44, 10 andar, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - V C Broaddus
- Lung Biology Center, UCSF, San Francisco, CA, USA
| |
Collapse
|
17
|
Gaudino G, Yang H, Carbone M. HGF/Met Signaling Is a Key Player in Malignant Mesothelioma Carcinogenesis. Biomedicines 2014; 2:327-344. [PMID: 28548074 PMCID: PMC5344271 DOI: 10.3390/biomedicines2040327] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 12/25/2022] Open
Abstract
Malignant mesothelioma (MM) is a highly aggressive cancer related to asbestos or erionite exposure and resistant to current therapies. Hepatocyte Growth Factor (HGF) and its tyrosine kinase receptor Met regulate cell growth, survival, motility/migration, and invasion. HGF and Met are expressed in MM cells, suggesting that the HGF/Met signaling plays a role in development and progression of this tumor, by autocrine and/or paracrine mechanisms. Upregulation and ligand-independent activation of Met, which is under suppressive control of miR-34 family members, correlate with enhanced invasion, migration and metastatic potential in several cancers, including MM. Moreover, Simian Virus 40 (SV40) Tag expression also induces a HGF autocrine circuit in an Rb-dependent manner in human mesothelial cells (HM) and possibly other cell types, enhancing cell adhesion, invasion and angiogenesis. The resulting activation of Met causes HM transformation and cell cycle progression, and contributes to virus particle assembling and infection of adjacent cells. The constitutive activation of Met, frequently occurring in MM, has been successfully targeted in preclinical models of MM. In conclusion, Met expression, activation state, subcellular localization and also HGF co-receptors expression, such as CD44, have clinical relevance for novel targeted therapies in a cancer for which no effective treatment is currently available.
Collapse
Affiliation(s)
- Giovanni Gaudino
- University of Hawai'i Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA.
| | - Haining Yang
- University of Hawai'i Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA.
| | - Michele Carbone
- University of Hawai'i Cancer Center, 701 Ilalo Street, Honolulu, HI 96813, USA.
| |
Collapse
|
18
|
Arzt L, Kothmaier H, Halbwedl I, Quehenberger F, Popper HH. Signal transducer and activator of transcription 1 (STAT1) acts like an oncogene in malignant pleural mesothelioma. Virchows Arch 2014; 465:79-88. [DOI: 10.1007/s00428-014-1584-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 03/17/2014] [Accepted: 04/22/2014] [Indexed: 11/28/2022]
|
19
|
Kim MC, Cui FJ, Kim Y. Hydrogen peroxide promotes epithelial to mesenchymal transition and stemness in human malignant mesothelioma cells. Asian Pac J Cancer Prev 2014; 14:3625-30. [PMID: 23886156 DOI: 10.7314/apjcp.2013.14.6.3625] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Reactive oxygen species (ROS) are known to promote mesothelial carcinogenesis that is closely associated with asbestos fibers and inflammation. Epithelial to mesenchymal cell transition (EMT) is an important process involved in the progression of tumors, providing cancer cells with aggressiveness. The present study was performed to determine if EMT is induced by H2O2 in human malignant mesothelioma (HMM) cells. Cultured HMM cells were treated with H2O2, followed by measuring expression levels of EMT-related genes and proteins. Immunohistochemically, TWIST1 expression was confined to sarcomatous cells in HMM tissues, but not in epithelioid cells. Treatment of HMM cells with H2O2 promoted EMT, as indicated by increased expression levels of vimentin, SLUG and TWIST1, and decreased E-cadherin expression. Expression of stemness genes such as OCT4, SOX2 and NANOG was also significantly increased by treatment of HMM cells with H2O2. Alteration of these genes was mediated via activation of hypoxia inducible factor 1 alpha (HIF-1α) and transforming growth factor beta 1 (TGF-β1). Considering that treatment with H2O2 results in excess ROS, the present study suggests that oxidative stress may play a critical role in HMM carcinogenesis by promoting EMT processes and enhancing the expression of stemness genes.
Collapse
Affiliation(s)
- Myung-Chul Kim
- Laboratory of Clinical Pathology, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, South Korea
| | | | | |
Collapse
|
20
|
Integrated high-resolution array CGH and SKY analysis of homozygous deletions and other genomic alterations present in malignant mesothelioma cell lines. Cancer Genet 2013; 206:191-205. [PMID: 23830731 DOI: 10.1016/j.cancergen.2013.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 04/23/2013] [Accepted: 04/30/2013] [Indexed: 02/04/2023]
Abstract
High-resolution oligonucleotide array comparative genomic hybridization (aCGH) and spectral karyotyping (SKY) were applied to a panel of malignant mesothelioma (MMt) cell lines. SKY has not been applied to MMt before, and complete karyotypes are reported based on the integration of SKY and aCGH results. A whole genome search for homozygous deletions (HDs) produced the largest set of recurrent and non-recurrent HDs for MMt (52 recurrent HDs in 10 genomic regions; 36 non-recurrent HDs). For the first time, LINGO2, RBFOX1/A2BP1, RPL29, DUSP7, and CCSER1/FAM190A were found to be homozygously deleted in MMt, and some of these genes could be new tumor suppressor genes for MMt. Integration of SKY and aCGH data allowed reconstruction of chromosomal rearrangements that led to the formation of HDs. Our data imply that only with acquisition of structural and/or numerical karyotypic instability can MMt cells attain a complete loss of tumor suppressor genes located in 9p21.3, which is the most frequently homozygously deleted region. Tetraploidization is a late event in the karyotypic progression of MMt cells, after HDs in the 9p21.3 region have already been acquired.
Collapse
|
21
|
Ramos-Nino ME. The role of chronic inflammation in obesity-associated cancers. ISRN ONCOLOGY 2013; 2013:697521. [PMID: 23819063 PMCID: PMC3683483 DOI: 10.1155/2013/697521] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 05/12/2013] [Indexed: 12/20/2022]
Abstract
There is a strong relationship between metabolism and immunity, which can become deleterious under conditions of metabolic stress. Obesity, considered a chronic inflammatory disease, is one example of this link. Chronic inflammation is increasingly being recognized as an etiology in several cancers, particularly those of epithelial origin, and therefore a potential link between obesity and cancer. In this review, the connection between the different factors that can lead to the chronic inflammatory state in the obese individual, as well as their effect in tumorigenesis, is addressed. Furthermore, the association between obesity, inflammation, and esophageal, liver, colon, postmenopausal breast, and endometrial cancers is discussed.
Collapse
Affiliation(s)
- Maria E. Ramos-Nino
- Department of Pathology and Department of Medical Laboratory Sciences, University of Vermont, Burlington, VT, USA
| |
Collapse
|
22
|
Activated leukocyte cell-adhesion molecule (ALCAM) promotes malignant phenotypes of malignant mesothelioma. J Thorac Oncol 2012; 7:890-9. [PMID: 22722789 DOI: 10.1097/jto.0b013e31824af2db] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Cell-adhesion molecules play important roles involving the malignant phenotypes of human cancer cells. However, detailed characteristics of aberrant expression status of cell-adhesion molecules in malignant mesothelioma (MM) cells and their possible biological roles for MM malignancy remain poorly understood. METHODS DNA microarray analysis was employed to identify aberrantly expressing genes using 20 MM cell lines. Activated leukocyte cell-adhesion molecule (ALCAM) expression in MM cell lines was analyzed with quantitative reverse transcription-polymerase chain reaction and Western blot analyses in 47 primary MM specimens with immunohistochemistry. ALCAM knockdown in MM cell lines was performed with lentivirus-mediated short hairpin RNA (shRNA) transduction. Purified soluble ALCAM (sALCAM) protein was used for in vitro experiments, whereas MM cell lines infected with the sALCAM-expressing lentivirus were tested for tumorigenicity in vivo. RESULTS ALCAM, a member of the immunoglobulin superfamily, was detected as one of the most highly upregulated genes among 103 cell-adhesion molecules with microarray analysis. Elevated expression levels of ALCAM messenger RNA and protein were detected in all 20 cell lines. Positive staining of ALCAM was detected in 26 of 47 MM specimens (55%) with immunohistochemistry. ALCAM knockdown with shRNA suppressed cell migration and invasion of MM cell lines. Purified sALCAM protein impaired the migration and invasion of MM cells in vitro, and the infection of sALCAM-expressing virus into MM cells significantly prolonged survival periods of MM-transplanted nude mice in vivo. CONCLUSION Our study suggests that overexpression of ALCAM contributes to tumor progression in MM and that ALCAM might be a potential therapeutic target of MM.
Collapse
|
23
|
Cornelissen R, Lievense LA, Heuvers ME, Maat AP, Hendriks RW, Hoogsteden HC, Hegmans JP, Aerts JG. Dendritic cell-based immunotherapy in mesothelioma. Immunotherapy 2012; 4:1011-22. [DOI: 10.2217/imt.12.108] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesothelioma is a rare thoracic malignancy with a dismal prognosis. Current treatment options are scarce and clinical outcomes are rather disappointing. Due to the immunogenic nature of mesothelioma, several studies have investigated immunotherapeutic strategies to improve the prognosis of patients with mesothelioma. In the last decade, progress in knowledge of the modulation of the immune system to attack the tumor has been remarkable, but the optimal strategy for immunotherapy has yet to be unraveled. Because of their potent antigen-presenting capacity, dendritic cells are acknowledged as a promising agent in immunotherapeutic approaches in a number of malignancies. This review gives an update and provides a future perspective in which immunotherapy may improve the outcome of mesothelioma therapy.
Collapse
Affiliation(s)
- Robin Cornelissen
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Lysanne A Lievense
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Marlies E Heuvers
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Alexander P Maat
- Department of Thoracic Surgery, Erasmus Medical Center – Daniel den Hoed Cancer Center, University Medical Center, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Henk C Hoogsteden
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Joost P Hegmans
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Joachim G Aerts
- Department of Pulmonary Medicine, Erasmus MC, SV-125, PO-Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
24
|
Menges CW, Sementino E, Talarchek J, Xu J, Chernoff J, Peterson JR, Testa JR. Group I p21-activated kinases (PAKs) promote tumor cell proliferation and survival through the AKT1 and Raf-MAPK pathways. Mol Cancer Res 2012; 10:1178-88. [PMID: 22798428 DOI: 10.1158/1541-7786.mcr-12-0082] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Group I p21-activated kinases (PAK) are important effectors of the small GTPases Rac and Cdc42, which regulate cell motility/migration, survival, proliferation, and gene transcription. Hyperactivation of these kinases have been reported in many tumor types, making PAKs attractive targets for therapeutic intervention. PAKs are activated by growth factor-mediated signaling and are negatively regulated by the tumor suppressor neurofibromatosis type 2 (NF2)/Merlin. Thus, tumors characterized by NF2 inactivation would be expected to show hyperactivated PAK signaling. On the basis of this rationale, we evaluated the status of PAK signaling in malignant mesothelioma, an aggressive neoplasm that is resistant to current therapies and shows frequent inactivation of NF2. We show that group I PAKs are activated in most mesotheliomas and mesothelioma cell lines and that genetic or pharmacologic inhibition of PAKs is sufficient to inhibit mesothelioma cell proliferation and survival. We also identify downstream effectors and signaling pathways that may contribute mechanistically to PAK-related tumorigenesis. Specifically, we show that inhibition of PAK results in attenuation of AKT and Raf-MAPK signaling and decreased tumor cell viability. Collectively, these data suggest that pharmacologic inhibition of group I PAKs may have therapeutic efficacy in tumors characterized by PAK activation.
Collapse
Affiliation(s)
- Craig W Menges
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Shi Y, Moura U, Opitz I, Soltermann A, Rehrauer H, Thies S, Weder W, Stahel RA, Felley-Bosco E. Role of hedgehog signaling in malignant pleural mesothelioma. Clin Cancer Res 2012; 18:4646-56. [PMID: 22733539 DOI: 10.1158/1078-0432.ccr-12-0599] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of this study was to assess the activity of hedgehog signaling pathway in malignant pleural mesothelioma (MPM). EXPERIMENTAL DESIGN The expression of hedgehog signaling components was assessed by quantitative PCR and in situ hybridization in 45 clinical samples. Primary MPM cultures were developed in serum-free condition in 3% oxygen and were used to investigate the effects of smoothened (SMO) inhibitors or GLI1 silencing on cell growth and hedgehog signaling. In vivo effects of SMO antagonists were determined in an MPM xenograft growing in nude mice. RESULTS A significant increase in GLI1, sonic hedgehog, and human hedgehog interacting protein gene expression was observed in MPM tumors compared with nontumoral pleural tissue. SMO antagonists inhibited GLI1 expression and cell growth in sensitive primary cultures. This effect was mimicked by GLI1 silencing. Reduced survivin and YAP protein levels were also observed. Survivin protein levels were rescued by overexpression of GLI1 or constitutively active YAP1. Treatment of tumor-bearing mice with the SMO inhibitor HhAntag led to a significant inhibition of tumor growth in vivo accompanied by decreased Ki-67 and nuclear YAP immunostaining and a significant difference in selected gene expression profile in tumors. CONCLUSIONS An aberrant hedgehog signaling is present in MPM, and inhibition of hedgehog signaling decreases tumor growth indicating potential new therapeutic approach.
Collapse
Affiliation(s)
- Yandong Shi
- Laboratory of Molecular Oncology, Clinic and Policlinic of Oncology, Division of Thoracic Surgery, Institute of Surgical Pathology, University Hospital Zürich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Astoul P, Roca E, Galateau-Salle F, Scherpereel A. Malignant Pleural Mesothelioma: From the Bench to the Bedside. Respiration 2012; 83:481-93. [DOI: 10.1159/000339259] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
27
|
Kadota K, Suzuki K, Colovos C, Sima CS, Rusch VW, Travis WD, Adusumilli PS. A nuclear grading system is a strong predictor of survival in epitheloid diffuse malignant pleural mesothelioma. Mod Pathol 2012; 25:260-71. [PMID: 21983936 PMCID: PMC4080411 DOI: 10.1038/modpathol.2011.146] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Epithelioid mesothelioma is the most prevalent subtype of diffuse malignant pleural mesothelioma in which only staging is prognostic for survival. In this study of epithelioid diffuse malignant pleural mesothelioma, we investigate the prognostic utility of nuclear features. The slides of 232 epithelioid diffuse malignant pleural mesothelioma patients (14 stage I, 54 stage II, 130 stage III, and 34 stage IV) from a single institution were reviewed for the following seven nuclear features: nuclear atypia, nuclear/cytoplasmic ratio, chromatin pattern, intranuclear inclusions, prominence of nucleoli, mitotic count, and atypical mitoses. MIB-1 immunohistochemistry was performed using tissue microarray, and MIB-1 labeling index was recorded as the percentage of positive tumor cells. Median overall survival of all patients was 16 months and correlated with nuclear atypia (P<0.001), chromatin pattern (P=0.031), prominence of nucleoli (P<0.001), mitotic count (P<0.001), and atypical mitoses (P<0.001) by univariate analysis. Multivariate analysis revealed nuclear atypia (P=0.012) and mitotic count (P<0.001) as independent prognostic factors, and these two factors were utilized to create a three-tier nuclear grade score. The resulting nuclear grade stratified patients into three distinct prognostic groups: grade I (n=107, median overall survival=28 months), grade II (n=91, 14 months), and grade III (n=34, 5 months). Not only was nuclear grade an independent predictor of overall survival (P<0.001), but it was also a stronger discriminator of survival than all currently available factors. Furthermore, nuclear grade was associated with time to recurrence (P=0.004) in patients who underwent complete surgical resection (n=159). MIB-1 labeling index correlated with mitotic count (P<0.001) and nuclear atypia (P=0.037) and stratified overall survival (P<0.001) and time to recurrence (P=0.048), confirming the prognostic value of the nuclear grade. Nuclear grading in epithelioid mesothelioma provides a simple, practical, and cost-effective prognostic tool that better stratifies clinical outcome and time to recurrence than currently available clinicopathologic factors.
Collapse
Affiliation(s)
- Kyuichi Kadota
- Division of Thoracic Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA,Department of Diagnostic Pathology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Kei Suzuki
- Division of Thoracic Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Christos Colovos
- Division of Thoracic Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Camelia S Sima
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Valerie W Rusch
- Division of Thoracic Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - William D Travis
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Prasad S Adusumilli
- Division of Thoracic Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA,Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
28
|
Qi F, Carbone M, Yang H, Gaudino G. Simian virus 40 transformation, malignant mesothelioma and brain tumors. Expert Rev Respir Med 2012; 5:683-97. [PMID: 21955238 DOI: 10.1586/ers.11.51] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Simian virus 40 (SV40) is a DNA virus isolated in 1960 from contaminated polio vaccines, that induces mesotheliomas, lymphomas, brain and bone tumors, and sarcomas, including osteosarcomas, in hamsters. These same tumor types have been found to contain SV40 DNA and proteins in humans. Mesotheliomas and brain tumors are the two tumor types that have been most consistently associated with SV40, and the range of positivity has varied about from 6 to 60%, although a few reported 100% of positivity and a few reported 0%. It appears unlikely that SV40 infection alone is sufficient to cause human malignancy, as we did not observe an epidemic of cancers following the administration of SV40-contaminated vaccines. However, it seems possible that SV40 may act as a cofactor in the pathogenesis of some tumors. In vitro and animal experiments showing cocarcinogenicity between SV40 and asbestos support this hypothesis.
Collapse
Affiliation(s)
- Fang Qi
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | | | | |
Collapse
|
29
|
Affiliation(s)
- W Weder
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Chemotherapy and targeted therapies for unresectable malignant mesothelioma. Lung Cancer 2011; 73:256-63. [PMID: 21620512 DOI: 10.1016/j.lungcan.2011.04.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Revised: 04/11/2011] [Accepted: 04/25/2011] [Indexed: 12/29/2022]
Abstract
The global burden of mesothelioma is expected to increase in the coming decades. As a result the development of more effective therapies with an emphasis on personalized treatments based on validated prognostic and predictive biomarkers is an essential requirement. Progress has been made in the last decade with the development of newer generation anti-folates leading to the current standard of care of pemetrexed and cisplatin in patients with unresectable disease. However, the median overall survival of patients with this combination treatment is only 12 months. There is no consensus regarding second line therapy for patients who have progressed or not responded to pemetrexed based therapies although gemcitabine in combination with a platinum compound or single agent vinorelbine is a reasonable option. The development of effective targeted agents that are active in mesothelioma has to date been disappointing. Strategies involving the addition of bevacizumab to pemetrexed and cisplatin in the frontline setting, the histone deacetylase inhibitor vorinostat as second line therapy and studies evaluating the utility of maintenance therapy in mesothelioma are all ongoing and appear promising. In addition clinical trials investigating immunotherapy and gene therapy in combination with chemotherapy could potentially improve the prognosis of patients with mesothelioma.
Collapse
|
31
|
Jakobsen JN, Sørensen JB. Review on clinical trials of targeted treatments in malignant mesothelioma. Cancer Chemother Pharmacol 2011; 68:1-15. [PMID: 21553148 DOI: 10.1007/s00280-011-1655-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/14/2011] [Indexed: 12/21/2022]
Abstract
PURPOSE Malignant mesothelioma (MM) is an aggressive tumor of the serosal surfaces with a poor prognosis. Advances in the understanding of tumor biology have led to the development of several targeted treatments, which have been evaluated in clinical trials. This article is a comprehensive review of all clinical trials evaluating the effect of targeted treatments in MM. METHODS An extensive literature search was performed in January 2011 using pubmed and medline. No constraints on publication date were applied. RESULTS Thirty-two trials exploring 17 different targeted agents in MM were found. Treatment in first- and second-line targeted agents induced response rates ranging from 0-14% and 0-16%, respectively. The tyrosine kinase inhibitor sunitinib induced partial response in 10% and stable disease in 66% of MPM patients as second-line treatment. A preliminary analysis of a phase II/III trial suggests that addition of bevacizumab to pemetrexed and cisplatin first-line treatment significantly improves disease control (CR + PR + SD) in the bevacizumab arm (73.5%) compared with treatment with pemetrexed and cisplatin without bevacizumab (43.2%) (P = 0.010). Another phase II trial did not observe any significant clinical benefit of adding of bevacizumab to gemcitabine and cisplatin. CONCLUSIONS Disease stabilization is reported in some patients with several targeted treatments and might be beneficial in subgroups of patients or in combination with classic chemotherapy. None of the hitherto explored targeted treatments can currently be recommended as standard treatment in MM.
Collapse
|
32
|
Murakami H, Mizuno T, Taniguchi T, Fujii M, Ishiguro F, Fukui T, Akatsuka S, Horio Y, Hida T, Kondo Y, Toyokuni S, Osada H, Sekido Y. LATS2 is a tumor suppressor gene of malignant mesothelioma. Cancer Res 2011; 71:873-83. [PMID: 21245096 DOI: 10.1158/0008-5472.can-10-2164] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Malignant mesothelioma (MM) is an aggressive neoplasm associated with asbestos exposure. We carried out genome-wide array-based comparative genomic hybridization analysis with 14 MM cell lines. Three cell lines showed overlapping homozygous deletion at chromosome 13q12, which harbored the LATS2 (large tumor suppressor homolog 2) gene. With 6 other MM cell lines and 25 MM tumors, we found 10 inactivating homozygous deletions or mutations of LATS2 among 45 MMs. LATS2 encodes a serine/threonine kinase, a component of the Hippo tumor-suppressive signaling pathway, and we transduced LATS2 in MM cells with its mutation. Transduction of LATS2 inactivated oncoprotein YAP, a transcriptional coactivator, via phosphorylation, and inhibited MM cell growth. We also analyzed LATS2 immunohistochemically and found that 13 of 45 MM tumors had low expression of LATS2. Because NF2 is genetically mutated in 40% to 50% of MM, our data indicate that Hippo pathway dysregulation is frequent in MM cells with inactivation of LATS2 or an upstream regulator of this pathway, Merlin, which is encoded by NF2. Thus, our results suggest that the inactivation of LATS2 is one of the key mechanisms for constitutive activation of YAP, which induces deregulation of MM cell proliferation.
Collapse
Affiliation(s)
- Hideki Murakami
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Synthetic double-stranded RNA stimulates the expression of interferon-inducible protein 10 in human mesothelial cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 18:176-9. [PMID: 21106778 DOI: 10.1128/cvi.00007-10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interferon-inducible protein 10 (IP-10) is a chemokine playing an important role in the restriction of viral spread. A time- and dose-dependent increase in IP-10 is found upon activation of viral receptors expressed on mesothelial cells, which provides novel evidence for a link between viral infections and inflammation of serous membranes.
Collapse
|
34
|
Wheatley-Price P, Yang B, Patsios D, Patel D, Ma C, Xu W, Leighl N, Feld R, Cho BCJ, O'Sullivan B, Roberts H, Tsao MS, Tammemagi M, Anraku M, Chen Z, de Perrot M, Liu G. Soluble mesothelin-related Peptide and osteopontin as markers of response in malignant mesothelioma. J Clin Oncol 2010; 28:3316-22. [PMID: 20498407 DOI: 10.1200/jco.2009.26.9944] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE In malignant mesothelioma (MM), radiologic assessment of disease status is difficult. Both soluble mesothelin-related peptide (SMRP) and osteopontin (OP) have utility in distinguishing MM from benign pleural disease. We evaluated whether SMRP and OP also correlated with the disease course of MM. PATIENTS AND METHODS Serial plasma samples from patients with MM were prospectively collected, and SMRP and OP levels were measured. Radiologic tests across time periods showing disease progression, stability, or shrinkage were compared with corresponding changes in SMRP/OP levels. RESULTS From 41 patients, 165 samples were collected (range, 2 to 10; median 4). At study entry, 37 of 41 patients had measurable disease, of whom 92% (34 of 37) had elevated baseline SMRP levels; four of 41 patients had no evidence of recurrence and each had normal baseline SMRP levels. In 21 patients receiving systemic therapy, percentage change in SMRP more than 10% correlated with the radiologic assessment by a trained thoracic radiologist (P < .001), by formal Response Evaluation Criteria in Solid Tumors (RECIST; P = .008), or by modified RECIST (P < .001). All seven patients who underwent surgical resection with negative margins had elevated preoperative SMRP levels that fell to normal postoperatively. Rising SMRP was observed in all patients with radiologic disease progression. No associations were found with OP. CONCLUSION Percentage changes in SMRP levels, but not changes in OP levels, are a potentially useful marker of disease course. These findings should be validated prospectively for a role as an objective adjunctive measure of disease course in both clinical trials and clinical practice.
Collapse
|
35
|
Combined treatment with cisplatin and sirolimus to enhance cell death in human mesothelioma. J Thorac Cardiovasc Surg 2010; 139:1233-40. [DOI: 10.1016/j.jtcvs.2009.06.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Accepted: 06/21/2009] [Indexed: 11/23/2022]
|
36
|
Wörnle M, Sauter M, Kastenmüller K, Ribeiro A, Mussack T, Ladurner R, Sitter T. Role of toll-like receptor 3, RIG-I, and MDA5 in the expression of mesothelial IL-8 induced by viral RNA. Appl Biochem Biotechnol 2010; 160:1179-87. [PMID: 19472082 DOI: 10.1007/s12010-009-8643-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Accepted: 04/06/2009] [Indexed: 11/30/2022]
Abstract
Interleukin-8 (IL-8) is a chemokine that has been shown to be a potent chemoattractant for polymorphonuclear neutrophils from the vascular compartment into the pleural space during infectious pleural effusions. Mesothelial cells express the viral receptors Toll-like receptor 3 (TLR3), RIG-I, and MDA5. Activation of these receptors by viral RNA exemplified by poly (I:C) RNA leads to a time- and dose-dependent increase of mesothelial IL-8 synthesis. To show the specific effect of viral receptors, knockdown experiments with short interfering RNA specific for TLR3, RIG-I and MDA5 were performed. This novel finding of functional expression of these viral sensors on human mesothelial cells may indicate a novel link between viral infections and mesothelial inflammation and indicates a pathophysiologic role of viral receptors in these processes.
Collapse
Affiliation(s)
- Markus Wörnle
- Medical Policlinic, Ludwig-Maximilians-University, Pettenkoferstrasse 8a, 80336 Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
37
|
Deniz H, Kibar Y, Güldür ME, Bakır K. Is D2-40 a useful marker for distinguishing malignant mesothelioma from pulmonary adenocarcinoma and benign mesothelial proliferations? Pathol Res Pract 2009; 205:749-52. [DOI: 10.1016/j.prp.2009.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2008] [Revised: 05/26/2009] [Accepted: 06/05/2009] [Indexed: 10/20/2022]
|
38
|
Kobayashi H, Yamada Y, Kanayama S, Furukawa N, Noguchi T, Haruta S, Yoshida S, Sakata M, Sado T, Oi H. The role of hepatocyte nuclear factor-1beta in the pathogenesis of clear cell carcinoma of the ovary. Int J Gynecol Cancer 2009; 19:471-9. [PMID: 19407577 DOI: 10.1111/igc.0b013e3181a19eca] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PROBLEM Clear cell carcinoma (CCC) of the ovary has a number of features distinguishing it from other epithelial ovarian carcinomas (EOC) because of its characteristic histology and biology, frequent concurrence with endometriotic lesion, and highly chemoresistant nature resulting in an extremely poor prognosis. The incidence of CCC has been steadily increasing in Japan. They comprise approximately 20% of all EOC. Understanding the mechanisms of CCC development and elucidating pathogenesis and pathophysiology are intrinsic to prevention and effective therapies for CCC. METHOD OF STUDY This article reviews the English language literature for biology, pathogenesis, and pathophysiological studies on endometriosis-associated EOC. Several data are discussed in the context of endometriosis and CCC biology. RESULTS Recent studies based on genome-wide expression analysis technology have noted specific expression of hepatocyte nuclear factor-1beta (HNF-1beta) in endometriosis and CCC, suggesting that early differentiation into the clear cell lineage takes place in the endometriosis. The HNF-1beta-dependent pathway of CCC will be discussed, which are providing new insights into regulation of apoptosis and glycogen synthesis and resistance of CCC to anticancer agents. CONCLUSIONS This review summarizes recent advances in the HNF-1beta and its target genes; the potential challenges to the understanding of carcinogenesis, pathogenesis, and pathophysiology of CCC; and a possible novel model is proposed.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Prat O, Bérenguer F, Steinmetz G, Ruat S, Sage N, Quéméneur E. Alterations in gene expression in cultured human cells after acute exposure to uranium salt: Involvement of a mineralization regulator. Toxicol In Vitro 2009; 24:160-8. [PMID: 19654044 DOI: 10.1016/j.tiv.2009.07.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 07/08/2009] [Accepted: 07/30/2009] [Indexed: 10/20/2022]
Abstract
The risk of exposure of workers or populations to materials, such as uranium, of nuclear fuel process origins is a major concern worldwide. Our goal is to improve the knowledge of mechanisms ruling its chemical toxicity, and to search for proteins as potential indicator of effect. Such a marker of internal damage remains to be discovered in the case of uranium. This study, based on DNA microarrays, reports a comparative gene expression analysis following acute uranium exposure of several human cell lines taken from kidneys or lungs as representative targets. Among uranium altered genes, no common gene was found between cells originating from lungs and kidney. In contrast, a set of 24 altered genes was common to two kidney cell lines. Transcriptional levels of a subset of renal genes were assessed with qRT-PCR. Furthermore, we highlighted a gene (SPP1) coding for a secreted protein (osteopontin) linked to ectopic mineralization. Immunoblotting assays showed that uranyl ions affect the excretion of osteopontin in a time- and dose-dependent manner. We consider that osteopontin, described as associated with bone resorbtion and kidney mineral stones, is a worthwhile candidate to be tested in vivo as a potential indicator of uranyl mineralization effects.
Collapse
|
40
|
Tucker TA, Dean C, Komissarov AA, Koenig K, Mazar AP, Pendurthi U, Allen T, Idell S. The urokinase receptor supports tumorigenesis of human malignant pleural mesothelioma cells. Am J Respir Cell Mol Biol 2009; 42:685-96. [PMID: 19635932 DOI: 10.1165/rcmb.2008-0433oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a lethal neoplasm for which current therapy is unsatisfactory. The urokinase plasminogen activator receptor (uPAR) is associated with increased virulence of many solid neoplasms, but its role in the pathogenesis of MPM is currently unclear. We found that REN human pleural MPM cells expressed 4- to 10-fold more uPAR than MS-1 or M9K MPM cells or MeT5A human pleural mesothelial cells. In a new orthotopic murine model of MPM, we found that the kinetics of REN cell tumorigenesis is accelerated versus MS-1 or M9K cells, and that REN instillates generated larger tumors expressing increased uPAR, were more invasive, and caused earlier mortality. While REN, MS-1, and M9K tumors were all associated with prominent extravascular fibrin deposition, excised REN tumor homogenates were characterized by markedly increased uPAR at both the mRNA and protein levels. REN cells exhibited increased thymidine incorporation, which was attenuated in uPAR-silenced cells (P < 0.01). REN cells traversed three-dimensional fibrin gels while MS-1, M9K, and MeT5A cells did not. uPAR siRNA or uPAR blocking antibodies decreased REN cell migration and invasion, while uPA and fetal bovine serum augmented the effects. Transfection of relatively low uPAR expressing MS-1 cells with uPAR cDNA increased proliferation and migration in vitro and tumor formation in vivo. These observations link overexpression of uPAR to the pathogenesis of MPM, demonstrate that this receptor contributes to accelerated tumor growth in part through interactions with uPA, and suggest that uPAR may be a promising target for therapeutic intervention.
Collapse
Affiliation(s)
- Torry A Tucker
- The Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US HWY 271, Biomedical Research Building, Lab C-5, Tyler, TX 75708, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Stahel RA, Felley-Bosco E, Opitz I, Weder W. Malignant pleural mesothelioma. Future Oncol 2009; 5:391-402. [PMID: 19374545 DOI: 10.2217/fon.09.7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Malignant pleural mesothelioma continues to be a challenge. The diagnosis and treatment of patients with malignant pleural mesothelioma requires a multidisciplinary approach. The diagnosis is best made by thoracoscopic biopsy and the aid of immunohistochemistry. Molecular studies identified inactivation of the neurofibromatosis-2 gene and INK4alpha/ARF to be key events in tumorigenesis. Based on the results of a Phase III trial, the combination of cisplatin with pemetrexed has become the preferred choice for chemotherapy, although there is suggestive evidence for the activity of other platin combinations based on Phase II studies. The optimal second-line chemotherapy remains to be defined. Surgical interventions ranging from pleurectomy/decortication to extrapleural pneumonectomy are increasingly offered in specialized centers, and the results of multimodality approaches with neoadjuvant or adjuvant chemotherapy and extrapleural pneumonectomy are encouraging. Ongoing investigations are defining the role of postoperative radiotherapy and the clinical activity of tyrosine kinase inhibitors targeting VEGFR2, histone deacetylase inhibitors and proteosome inhibitors.
Collapse
Affiliation(s)
- Rolf A Stahel
- Laboratory of Molecular Oncology, Clinic and Policlinic of Oncology, University Hosptial, CH-8091 Zürich, Switzerland.
| | | | | | | |
Collapse
|
42
|
Henzi T, Blum WV, Pfefferli M, Kawecki TJ, Salicio V, Schwaller B. SV40-induced expression of calretinin protects mesothelial cells from asbestos cytotoxicity and may be a key factor contributing to mesothelioma pathogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:2324-36. [PMID: 19435792 DOI: 10.2353/ajpath.2009.080352] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The calcium-binding protein calretinin has emerged as a useful marker for the identification of mesotheliomas of the epithelioid and mixed types, but its putative role in tumor development has not been addressed previously. Although exposure to asbestos fibers is considered the main cause of mesothelioma, undoubtedly, not all mesothelioma patients have a history of asbestos exposure. The question as to whether the SV40 virus is involved as a possible co-factor is still highly debated. Here we show that increased expression of SV40 early gene products in the mesothelial cell line MeT-5A induces the expression of calretinin and that elevated calretinin levels strongly correlate with increased resistance to asbestos cytotoxicity. Calretinin alone mediates a significant part of this protective effect because cells stably transfected with calretinin cDNA were clearly more resistant to the toxic effects of crocidolite than mock-transfected control cells. Down-regulation of calretinin by antisense methods restored the sensitivity to asbestos toxicity to a large degree. The protective effect observed in clones with higher calretinin expression levels could be eliminated by phosphatidylinositol 3-kinase (PI3K) inhibitors, implying an important role for the PI3K/AKT signaling (survival) pathway in mediating the protective effect. Up-regulation of calretinin, resulting from either asbestos exposure or SV40 oncoproteins, may be a common denominator that leads to increased resistance to asbestos cytotoxicity and thereby contributes to mesothelioma carcinogenesis.
Collapse
Affiliation(s)
- Thomas Henzi
- Unit of Anatomy, Department of Medicine, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland
| | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Wörnle M, Sauter M, Kastenmüller K, Ribeiro A, Roeder M, Schmid H, Krötz F, Mussack T, Ladurner R, Sitter T. Novel role of toll-like receptor 3, RIG-I and MDA5 in poly (I:C) RNA-induced mesothelial inflammation. Mol Cell Biochem 2009; 322:193-206. [PMID: 19005739 DOI: 10.1007/s11010-008-9957-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 10/28/2008] [Indexed: 11/26/2022]
Abstract
Viral inflammation and infection of mesothelial cells (MC) are a major problem in several organ systems including pleura, pericardium and peritoneum. Toll-like receptors (TLRs) are an essential part of the innate immune system for early recognition of pathogen-associated molecular patterns. TLRs recognise molecular patterns associated with microbial pathogens and induce an immune response. TLR3 recognises dsRNA of viral origin as exemplified by poly (I:C) RNA, a synthetic analogue of viral dsRNA. The helicases RIG-I and MDA5 may also act as sensors of viral infections. MC exhibit an expression of TLR3, RIG-I and MDA5. Poly (I:C) RNA stimulation resulted in an up-regulation of proinflammatory cytokines and chemokines as well as type I interferons. This novel finding of functional expression of viral sensors on human MC may indicate a novel link between viral infections and mesothelial inflammation and indicates a pathophysiologic role of viral receptors in these processes.
Collapse
Affiliation(s)
- Markus Wörnle
- Medical Policlinic, Ludwig-Maximilians-University, Pettenkoferstrasse 8a, 80336, Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Pulmonary pathology includes a large spectrum of both neoplastic and non-neoplastic diseases that affect the lung. Many of these are a result of the unusual relationship of the lung with the outside world. Every breath that a human takes brings the outside world into the body in the form of infectious agents, organic and inorganic particles, and noxious agents of all types. Although the lung has many defense mechanisms to protect itself from these insults, these are not infallible; therefore, lung pathology arises. Damage to the lung is particularly important given the role of the lung in the survival of the organism. Any impairment of lung function has widespread effects throughout the body, since all organs depend on the lungs for the oxygen they need. Pulmonary pathology catalogs the changes in the lung tissues and the mechanisms through which these occur. This chapter presents a review of lung pathology and the current state of knowledge about the pathogenesis of each disease. It suggests that a clear understanding of both morphology and mechanism is required for the development of new therapies and preventive measures.
Collapse
|
46
|
Comparison of semiquantitative fluorescence imaging and PET tracer uptake in mesothelioma models as a monitoring system for growth and therapeutic effects. Nucl Med Biol 2008; 35:851-60. [DOI: 10.1016/j.nucmedbio.2008.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Revised: 08/10/2008] [Accepted: 08/26/2008] [Indexed: 11/22/2022]
|
47
|
Pacurari M, Yin XJ, Zhao J, Ding M, Leonard SS, Schwegler-Berry D, Ducatman BS, Sbarra D, Hoover MD, Castranova V, Vallyathan V. Raw single-wall carbon nanotubes induce oxidative stress and activate MAPKs, AP-1, NF-kappaB, and Akt in normal and malignant human mesothelial cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:1211-7. [PMID: 18795165 PMCID: PMC2535624 DOI: 10.1289/ehp.10924] [Citation(s) in RCA: 192] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2007] [Accepted: 05/14/2008] [Indexed: 05/22/2023]
Abstract
BACKGROUND Single-wall carbon nanotubes (SWCNTs), with their unique physicochemical and mechanical properties, have many potential new applications in medicine and industry. There has been great concern subsequent to preliminary investigations of the toxicity, biopersistence, pathogenicity, and ability of SWCNTs to translocate to subpleural areas. These results compel studies of potential interactions of SWCNTs with mesothelial cells. OBJECTIVE Exposure to asbestos is the primary cause of malignant mesothelioma in 80-90% of individuals who develop the disease. Because the mesothelial cells are the primary target cells of asbestos-induced molecular changes mediated through an oxidant-linked mechanism, we used normal mesothelial and malignant mesothelial cells to investigate alterations in molecular signaling in response to a commercially manufactured SWCNT. METHODS In the present study, we exposed mesothelial cells to SWCNTs and investigated reactive oxygen species (ROS) generation, cell viability, DNA damage, histone H2AX phosphorylation, activation of poly(ADP-ribose) polymerase 1 (PARP-1), stimulation of extracellular signal-regulated kinase (ERKs), Jun N-terminal kinases (JNKs), protein p38, and activation of activator protein-1 (AP-1), nuclear factor kappaB (NF-kappaB), and protein serine-threonine kinase (Akt). RESULTS Exposure to SWCNTs induced ROS generation, increased cell death, enhanced DNA damage and H2AX phosphorylation, and activated PARP, AP-1, NF-kappaB, p38, and Akt in a dose-dependent manner. These events recapitulate some of the key molecular events involved in mesothelioma development associated with asbestos exposure. CONCLUSIONS The cellular and molecular findings reported here do suggest that SWCNTs can cause potentially adverse cellular responses in mesothelial cells through activation of molecular signaling associated with oxidative stress, which is of sufficient significance to warrant in vivo animal exposure studies.
Collapse
Affiliation(s)
- Maricica Pacurari
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Xuejun J. Yin
- Department of Pathology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Jinshun Zhao
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Ming Ding
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Steve S. Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Diane Schwegler-Berry
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Barbara S. Ducatman
- Department of Pathology, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Deborah Sbarra
- Division of Respiratory Disease Studies, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Mark D. Hoover
- Division of Respiratory Disease Studies, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Vincent Castranova
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Val Vallyathan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
- Address correspondence to V. Vallyathan, NIOSH/CDC, 1095 Willowdale Rd., Morgantown, WV 26505 USA. Telephone: (304) 285-5770. Fax: (304) 285-5938. E-mail:
| |
Collapse
|
48
|
Yokoyama T, Osada H, Murakami H, Tatematsu Y, Taniguchi T, Kondo Y, Yatabe Y, Hasegawa Y, Shimokata K, Horio Y, Hida T, Sekido Y. YAP1 is involved in mesothelioma development and negatively regulated by Merlin through phosphorylation. Carcinogenesis 2008; 29:2139-46. [PMID: 18725387 DOI: 10.1093/carcin/bgn200] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
We previously reported the results of bacterial artificial chromosome array comprehensive genomic hybridization of malignant pleural mesotheliomas (MPMs), including two cases with high-level amplification in the 11q22 locus. In this study, we found that the YAP1 gene encoding a transcriptional coactivator was localized in this amplified region and overexpressed in both cases, suggesting it as a candidate oncogene in this region. We analyzed the involvement of YAP1 in MPM proliferation, as well as its functional and physical interaction with Merlin encoded by the neurofibromatosis type 2 (NF2) tumor suppressor gene, which is frequently mutated in MPMs. YAP1-RNA interference suppressed growth of a mesothelioma cell line NCI-H290 with NF2 homozygous deletion, probably through cell-cycle arrest and apoptosis induction, whereas YAP1 transfection promoted the growth of MeT-5A, an immortalized mesothelial cell line. We also found that the introduction of NF2 into NCI-H290 induced phosphorylation at serine 127 of YAP1, which was accompanied by reduction of nuclear localization of YAP1, whereas nuclear localization of a YAP1 S 127A mutant was not affected. Furthermore, results of immunoprecipitation and in vitro pull-down assays indicated a physical interaction between Merlin and YAP1. These results suggest that YAP1 is involved in mesothelial cell growth and that the transcriptional coactivator activity of YAP1 is functionally inhibited by Merlin through the induction of phosphorylation and cytoplasmic retention of YAP1. This is the first report of negative regulatory signaling from Merlin to YAP1 in mammalian cells. Future studies of transcriptional targets of YAP1 in MPMs may shed light on the molecular mechanisms of MPM development and lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Toshihiko Yokoyama
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT, Tschopp J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008; 320:674-7. [PMID: 18403674 DOI: 10.1126/science.1156995] [Citation(s) in RCA: 2028] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The inhalation of airborne pollutants, such as asbestos or silica, is linked to inflammation of the lung, fibrosis, and lung cancer. How the presence of pathogenic dust is recognized and how chronic inflammatory diseases are triggered are poorly understood. Here, we show that asbestos and silica are sensed by the Nalp3 inflammasome, whose subsequent activation leads to interleukin-1beta secretion. Inflammasome activation is triggered by reactive oxygen species, which are generated by a NADPH oxidase upon particle phagocytosis. (NADPH is the reduced form of nicotinamide adenine dinucleotide phosphate.) In a model of asbestos inhalation, Nalp3-/- mice showed diminished recruitment of inflammatory cells to the lungs, paralleled by lower cytokine production. Our findings implicate the Nalp3 inflammasome in particulate matter-related pulmonary diseases and support its role as a major proinflammatory "danger" receptor.
Collapse
Affiliation(s)
- Catherine Dostert
- Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland
| | | | | | | | | | | |
Collapse
|
50
|
Tsou JA, Galler JS, Wali A, Ye W, Siegmund KD, Groshen S, Laird PW, Turla S, Koss MN, Pass HI, Laird-Offringa IA. DNA methylation profile of 28 potential marker loci in malignant mesothelioma. Lung Cancer 2007; 58:220-30. [PMID: 17659810 PMCID: PMC2752414 DOI: 10.1016/j.lungcan.2007.06.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 05/28/2007] [Accepted: 06/19/2007] [Indexed: 11/24/2022]
Abstract
Patients with malignant mesothelioma (MM), an aggressive cancer associated with asbestos exposure, usually present clinically with advanced disease and this greatly reduces the likelihood of curative treatment. MM is difficult to diagnose without invasive techniques; the development of non-invasively detectable molecular markers would therefore be highly beneficial. DNA methylation changes in cancer cells provide powerful markers that are potentially detectable non-invasively in DNA shed into bodily fluids. Here we examined the methylation status of 28 loci in 52 MM tumors to investigate their potential as molecular markers for MM. To exclude candidate MM markers that might be positive in biopsies/pleural fluid due to contaminating surrounding non-tumor lung tissue/DNA, we also examined the methylation of these markers in lung samples (age- or environmentally induced hypermethylation is frequently observed in non-cancerous lung). Statistically significantly increased methylation in MM versus non-tumor lung samples was found for estrogen receptor 1 (ESR1; p = 0.0002), solute carrier family 6 member 20 (SLC6A20; p = 0.0022) and spleen tyrosine kinase (SYK; p=0.0003). Examination of associations between methylation levels of the 28 loci and clinical parameters suggest associations of the methylation status of metallothionein genes with gender, histology, asbestos exposure, and lymph node involvement, and the methylation status of leucine zipper tumor suppressor 1 (LZTS1) and SLC6A20 with survival.
Collapse
Affiliation(s)
- Jeffrey A. Tsou
- Norris Cancer Center and Departments of Surgery and of Biochemistry and Molecular Biology, Room NOR6420, 1441 Eastlake Ave., Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9176, USA
| | - Janice S. Galler
- Norris Cancer Center and Departments of Surgery and of Biochemistry and Molecular Biology, Room NOR6420, 1441 Eastlake Ave., Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9176, USA
| | - Anil Wali
- Department of Thoracic Surgery, Karamanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Wei Ye
- Biostatistics Core, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9175, USA
| | - Kimberly D. Siegmund
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9011, USA
| | - Susan Groshen
- Biostatistics Core, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9175, USA
| | - Peter W. Laird
- Norris Cancer Center and Departments of Surgery and of Biochemistry and Molecular Biology, Room NOR6420, 1441 Eastlake Ave., Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9176, USA
| | - Sally Turla
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9092, USA
| | - Michael N. Koss
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9092, USA
| | - Harvey I. Pass
- Department of Thoracic Surgery, Karamanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Ite A. Laird-Offringa
- Norris Cancer Center and Departments of Surgery and of Biochemistry and Molecular Biology, Room NOR6420, 1441 Eastlake Ave., Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9176, USA
- Corresponding author: Tel.: +1 323 865 0655; fax: +1 323 865 0158. E-mail address: (I.A. Laird-Offringa)
| |
Collapse
|