1
|
Li S, Chen J, Zhou B. The clinical significance of endoplasmic reticulum stress related genes in non-small cell lung cancer and analysis of single nucleotide polymorphism for CAV1. Front Mol Biosci 2024; 11:1414164. [PMID: 39165641 PMCID: PMC11334084 DOI: 10.3389/fmolb.2024.1414164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/09/2024] [Indexed: 08/22/2024] Open
Abstract
In recent years, protein homeostasis imbalance caused by endoplasmic reticulum stress has become a major hallmark of cancer. Studies have shown that endoplasmic reticulum stress is closely related to the occurrence, development, and drug resistance of non-small cell lung cancer, however, the role of various endoplasmic reticulum stress-related genes in non-small cell lung cancer is still unclear. In this study, we established an endoplasmic reticulum stress scores based on the Cancer Genome Atlas for non-small cell lung cancer to reflect patient features and predict prognosis. Survival analysis showed significant differences in overall survival among non-small cell lung cancer patients with different endoplasmic reticulum stress scores. In addition, endoplasmic reticulum stress scores was significantly correlated with the clinical features of non-small cell lung cancer patients, and can be served as an independent prognostic indicator. A nomogram based on endoplasmic reticulum stress scores indicated a certain clinical net benefit, while ssGSEA analysis demonstrated that there was a certain immunosuppressive microenvironment in high endoplasmic reticulum stress scores. Gene Set Enrichment Analysis showed that scores was associated with cancer pathways and metabolism. Finally, weighted gene co-expression network analysis displayed that CAV1 was closely related to the occurrence of non-small cell lung cancer. Therefore, in order to further analyze the role of this gene, Chinese non-smoking females were selected as the research subjects to investigate the relationship between CAV1 rs3779514 and susceptibility and prognosis of non-small cell lung cancer. The results showed that the mutation of rs3779514 significantly reduced the risk of non-small cell lung cancer in Chinese non-smoking females, but no prognostic effect was found. In summary, we proposed an endoplasmic reticulum stress scores, which was an independent prognostic factor and indicated immune characteristics in the microenvironment of non-small cell lung cancer. We also validated the relationship between single nucleotide polymorphism locus of core genes and susceptibility to non-small cell lung cancer.
Collapse
Affiliation(s)
| | | | - Baosen Zhou
- Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
Kalkavan H, Rühl S, Shaw JJP, Green DR. Non-lethal outcomes of engaging regulated cell death pathways in cancer. NATURE CANCER 2023; 4:795-806. [PMID: 37277528 PMCID: PMC10416134 DOI: 10.1038/s43018-023-00571-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/27/2023] [Indexed: 06/07/2023]
Abstract
Regulated cell death (RCD) is essential for successful systemic cancer therapy. Yet, the engagement of RCD pathways does not inevitably result in cell death. Instead, RCD pathways can take part in diverse biological processes if the cells survive. Consequently, these surviving cells, for which we propose the term 'flatliners', harbor important functions. These evolutionarily conserved responses can be exploited by cancer cells to promote their own survival and growth, with challenges and opportunities for cancer therapy.
Collapse
Affiliation(s)
- Halime Kalkavan
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Sebastian Rühl
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- T3 Pharmaceuticals AG, Allschwil, Switzerland
| | - Jeremy J P Shaw
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
3
|
Urzì O, Olofsson Bagge R, Crescitelli R. The dark side of foetal bovine serum in extracellular vesicle studies. J Extracell Vesicles 2022; 11:e12271. [PMID: 36214482 PMCID: PMC9549727 DOI: 10.1002/jev2.12271] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/1912] [Revised: 12/12/1912] [Accepted: 12/12/1912] [Indexed: 11/06/2022] Open
Abstract
Extracellular vesicles (EVs) have been shown to be involved in cell-cell communication and to take part in both physiological and pathological processes. Thanks to their exclusive cargo, which includes proteins, lipids, and nucleic acids from the originating cells, they are gaining interest as potential biomarkers of disease. In recent years, their appealing features have been fascinating researchers from all over the world, thus increasing the number of in vitro studies focused on EV release, content, and biological activities. Cultured cell lines are the most-used source of EVs; however, the EVs released in cell cultures are influenced by the cell culture conditions, such as the use of foetal bovine serum (FBS). FBS is the most common supplement for cell culture media, but it is also a source of contaminants, such as exogenous bovine EVs, RNA, and protein aggregates, that can contaminate the cell-derived EVs and influence their cargo composition. The presence of FBS contaminants in cell-derived EV samples is a well-known issue that limits the clinical applications of EVs, thus increasing the need for standardization. In this review, we will discuss the pros and cons of using FBS in cell cultures as a source of EVs, as well as the protocols used to remove contaminants from FBS.
Collapse
Affiliation(s)
- Ornella Urzì
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational MedicineDepartment of SurgeryInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of BiomedicineNeurosciences and Advanced Diagnostics (Bi.N.D)University of PalermoPalermoItaly
| | - Roger Olofsson Bagge
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational MedicineDepartment of SurgeryInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of SurgerySahlgrenska University HospitalRegion Västra GötalandGothenburgSweden
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research and Wallenberg Centre for Molecular and Translational MedicineDepartment of SurgeryInstitute of Clinical SciencesSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
4
|
Lv W, Fu B, Li M, Kang Y, Bai S, Lu C. Determination of IC 50 values of anticancer drugs on cells by D 2O - single cell Raman spectroscopy. Chem Commun (Camb) 2022; 58:2355-2358. [PMID: 35080537 DOI: 10.1039/d1cc06857a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A simple, sensitive and repeatable D2O-single cell Raman spectroscopy method is developed to quantify the inhibitory activity of anticancer drugs on cancer cell metabolism. The IC50 values obtained from A549 cells incubated with cisplatin and taxol are comparable with results of CCK-8 and ATP luminescent cell viability assays.
Collapse
Affiliation(s)
- Wanxue Lv
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China. .,Center for Advanced Measurement Science (Institute of Life Science Metrology), National Institute of Metrology China, Beijing 100029, China.
| | - Boqiang Fu
- Center for Advanced Measurement Science (Institute of Life Science Metrology), National Institute of Metrology China, Beijing 100029, China.
| | - Manli Li
- Center for Advanced Measurement Science (Institute of Life Science Metrology), National Institute of Metrology China, Beijing 100029, China.
| | - Yu Kang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Shouli Bai
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Chao Lu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
5
|
Ahmadov U, Picard D, Bartl J, Silginer M, Trajkovic-Arsic M, Qin N, Blümel L, Wolter M, Lim JKM, Pauck D, Winkelkotte AM, Melcher M, Langini M, Marquardt V, Sander F, Stefanski A, Steltgens S, Hassiepen C, Kaufhold A, Meyer FD, Seibt A, Kleinesudeik L, Hain A, Münk C, Knobbe-Thomsen CB, Schramm A, Fischer U, Leprivier G, Stühler K, Fulda S, Siveke JT, Distelmaier F, Borkhardt A, Weller M, Roth P, Reifenberger G, Remke M. The long non-coding RNA HOTAIRM1 promotes tumor aggressiveness and radiotherapy resistance in glioblastoma. Cell Death Dis 2021; 12:885. [PMID: 34584066 PMCID: PMC8478910 DOI: 10.1038/s41419-021-04146-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 06/18/2021] [Accepted: 07/22/2021] [Indexed: 11/17/2022]
Abstract
Glioblastoma is the most common malignant primary brain tumor. To date, clinically relevant biomarkers are restricted to isocitrate dehydrogenase (IDH) gene 1 or 2 mutations and O6-methylguanine DNA methyltransferase (MGMT) promoter methylation. Long non-coding RNAs (lncRNAs) have been shown to contribute to glioblastoma pathogenesis and could potentially serve as novel biomarkers. The clinical significance of HOXA Transcript Antisense RNA, Myeloid-Specific 1 (HOTAIRM1) was determined by analyzing HOTAIRM1 in multiple glioblastoma gene expression data sets for associations with prognosis, as well as, IDH mutation and MGMT promoter methylation status. Finally, the role of HOTAIRM1 in glioblastoma biology and radiotherapy resistance was characterized in vitro and in vivo. We identified HOTAIRM1 as a candidate lncRNA whose up-regulation is significantly associated with shorter survival of glioblastoma patients, independent from IDH mutation and MGMT promoter methylation. Glioblastoma cell line models uniformly showed reduced cell viability, decreased invasive growth and diminished colony formation capacity upon HOTAIRM1 down-regulation. Integrated proteogenomic analyses revealed impaired mitochondrial function and determination of reactive oxygen species (ROS) levels confirmed increased ROS levels upon HOTAIRM1 knock-down. HOTAIRM1 knock-down decreased expression of transglutaminase 2 (TGM2), a candidate protein implicated in mitochondrial function, and knock-down of TGM2 mimicked the phenotype of HOTAIRM1 down-regulation in glioblastoma cells. Moreover, HOTAIRM1 modulates radiosensitivity of glioblastoma cells both in vitro and in vivo. Our data support a role for HOTAIRM1 as a driver of biological aggressiveness, radioresistance and poor outcome in glioblastoma. Targeting HOTAIRM1 may be a promising new therapeutic approach.
Collapse
Affiliation(s)
- Ulvi Ahmadov
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Picard
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jasmin Bartl
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Manuela Silginer
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Marija Trajkovic-Arsic
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site Essen, Heidelberg, Germany
| | - Nan Qin
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Lena Blümel
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Marietta Wolter
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Jonathan K M Lim
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - David Pauck
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Alina Marie Winkelkotte
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site Essen, Heidelberg, Germany
| | - Marlen Melcher
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Maike Langini
- Institute for Molecular Medicine I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Molecular Proteomics Laboratory (MPL), Biological-Medical Research Center (BMFZ), Heinrich Heine University, Düsseldorf, Germany
| | - Viktoria Marquardt
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Felix Sander
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Anja Stefanski
- Institute for Molecular Medicine I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Molecular Proteomics Laboratory (MPL), Biological-Medical Research Center (BMFZ), Heinrich Heine University, Düsseldorf, Germany
| | - Sascha Steltgens
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Christina Hassiepen
- Department of Molecular Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Anna Kaufhold
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Frauke-Dorothee Meyer
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Annette Seibt
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Lara Kleinesudeik
- Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anika Hain
- Clinic for Gastroenterology, Hepatology, and Infectiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Carsten Münk
- Clinic for Gastroenterology, Hepatology, and Infectiology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | | | - Alexander Schramm
- Department of Molecular Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Gabriel Leprivier
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Kai Stühler
- Institute for Molecular Medicine I, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
- Molecular Proteomics Laboratory (MPL), Biological-Medical Research Center (BMFZ), Heinrich Heine University, Düsseldorf, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site Essen, Heidelberg, Germany
| | - Felix Distelmaier
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Arndt Borkhardt
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Patrick Roth
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Guido Reifenberger
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Marc Remke
- Division of Pediatric Neuro-Oncogenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Consortium for Translational Cancer Research (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany.
- Department of Neuropathology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
6
|
Tempest R, Guarnerio S, Maani R, Cooper J, Peake N. The Biological and Biomechanical Role of Transglutaminase-2 in the Tumour Microenvironment. Cancers (Basel) 2021; 13:cancers13112788. [PMID: 34205140 PMCID: PMC8199963 DOI: 10.3390/cancers13112788] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
Transglutaminase-2 (TG2) is the most highly and ubiquitously expressed member of the transglutaminase enzyme family and is primarily involved in protein cross-linking. TG2 has been implicated in the development and progression of numerous cancers, with a direct role in multiple cellular processes and pathways linked to apoptosis, chemoresistance, epithelial-mesenchymal transition, and stem cell phenotype. The tumour microenvironment (TME) is critical in the formation, progression, and eventual metastasis of cancer, and increasing evidence points to a role for TG2 in matrix remodelling, modulation of biomechanical properties, cell adhesion, motility, and invasion. There is growing interest in targeting the TME therapeutically in response to advances in the understanding of its critical role in disease progression, and a number of approaches targeting biophysical properties and biomechanical signalling are beginning to show clinical promise. In this review we aim to highlight the wide array of processes in which TG2 influences the TME, focussing on its potential role in the dynamic tissue remodelling and biomechanical events increasingly linked to invasive and aggressive behaviour. Drug development efforts have yielded a range of TG2 inhibitors, and ongoing clinical trials may inform strategies for targeting the biomolecular and biomechanical function of TG2 in the TME.
Collapse
|
7
|
Cingöz A, Ozyerli-Goknar E, Morova T, Seker-Polat F, Esai Selvan M, Gümüş ZH, Bhere D, Shah K, Solaroglu I, Bagci-Onder T. Generation of TRAIL-resistant cell line models reveals distinct adaptive mechanisms for acquired resistance and re-sensitization. Oncogene 2021; 40:3201-3216. [PMID: 33767436 DOI: 10.1038/s41388-021-01697-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 02/01/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces tumor cell-specific apoptosis, making it a prime therapeutic candidate. However, many tumor cells are either innately TRAIL-resistant, or they acquire resistance with adaptive mechanisms that remain poorly understood. In this study, we generated acquired TRAIL resistance models using multiple glioblastoma (GBM) cell lines to assess the molecular alterations in the TRAIL-resistant state. We selected TRAIL-resistant cells through chronic and long-term TRAIL exposure and noted that they showed persistent resistance both in vitro and in vivo. Among known TRAIL-sensitizers, proteosome inhibitor Bortezomib, but not HDAC inhibitor MS-275, was effective in overcoming resistance in all cell models. This was partly achieved through upregulating death receptors and pro-apoptotic proteins, and downregulating major anti-apoptotic members, Bcl-2 and Bcl-xL. We showed that CRISPR/Cas9 mediated silencing of DR5 could block Bortezomib-mediated re-sensitization, demonstrating its critical role. While overexpression of Bcl-2 or Bcl-xL was sufficient to confer resistance to TRAIL-sensitive cells, it failed to override Bortezomib-mediated re-sensitization. With RNA sequencing in multiple paired TRAIL-sensitive and TRAIL-resistant cells, we identified major alterations in inflammatory signaling, particularly in the NF-κB pathway. Inhibiting NF-κB substantially sensitized the most resistant cells to TRAIL, however, the sensitization effect was not as great as what was observed with Bortezomib. Together, our findings provide new models of acquired TRAIL resistance, which will provide essential tools to gain further insight into the heterogeneous therapy responses within GBM tumors. Additionally, these findings emphasize the critical importance of combining proteasome inhibitors and pro-apoptotic ligands to overcome acquired resistance.
Collapse
Affiliation(s)
- Ahmet Cingöz
- Brain Cancer Research and Therapy Laboratory, Koç University Research Center for Translational Medicine, Istanbul, 34450, Turkey
- Koç University School of Medicine, Istanbul, 34450, Turkey
| | - Ezgi Ozyerli-Goknar
- Brain Cancer Research and Therapy Laboratory, Koç University Research Center for Translational Medicine, Istanbul, 34450, Turkey
- Koç University School of Medicine, Istanbul, 34450, Turkey
| | - Tunc Morova
- Koç University School of Medicine, Istanbul, 34450, Turkey
| | - Fidan Seker-Polat
- Brain Cancer Research and Therapy Laboratory, Koç University Research Center for Translational Medicine, Istanbul, 34450, Turkey
- Koç University School of Medicine, Istanbul, 34450, Turkey
| | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Zeynep Hülya Gümüş
- Koç University School of Medicine, Istanbul, 34450, Turkey
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Deepak Bhere
- Center for Stem Cell Therapeutics and Imaging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Khalid Shah
- Center for Stem Cell Therapeutics and Imaging, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ihsan Solaroglu
- Koç University School of Medicine, Istanbul, 34450, Turkey
- Department of Neurosurgery, Koç University School of Medicine, Istanbul, 34010, Turkey
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Laboratory, Koç University Research Center for Translational Medicine, Istanbul, 34450, Turkey.
- Koç University School of Medicine, Istanbul, 34450, Turkey.
| |
Collapse
|
8
|
Platelet-Released Growth Factors and Platelet-Rich Fibrin Induce Expression of Factors Involved in Extracellular Matrix Organization in Human Keratinocytes. Int J Mol Sci 2020; 21:ijms21124404. [PMID: 32575800 PMCID: PMC7378768 DOI: 10.3390/ijms21124404] [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: 05/28/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Platelet-released growth factor (PRGF) is a thrombocyte concentrate lysate which, like its clinically equivalent variations (e.g., Vivostat PRF® (platelet-rich fibrin)), is known to support the healing of chronic and hard-to-heal wounds. However, studies on the effect of PRGF on keratinocytes remain scarce. This study aims to identify genes in keratinocytes that are significantly influenced by PRGF. Therefore, we performed a whole transcriptome and gene ontology (GO) enrichment analysis of PRGF-stimulated human primary keratinocytes. This revealed an increased expression of genes involved in extracellular matrix (ECM) organization. Real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA) analysis confirmed the PRGF-mediated induction of selected ECM-related factors such as transforming growth factor beta-induced protein, fibronectin 1, matrix metalloproteinase-9, transglutaminase 2, fermitin family member 1, collagen type I alpha 1 and collagen type XXII alpha 1. PRGF-induced expression of the above factors was influenced by blockade of the epidermal growth factor receptor (EGFR), a receptor playing a crucial role in wound healing. A differential induction of the investigated factors was also detected in skin explants exposed to PRGF and in experimentally generated in vivo wounds treated with Vivostat PRF®. Together, our study indicates that the induction of ECM-related factors may contribute to the beneficial wound-healing effects of PRGF-based formulations.
Collapse
|
9
|
Chen XE, Chen P, Chen S, Lu J, Ma T, Shi G, Sheng L. Long non-coding RNA FENDRR inhibits migration and invasion of cutaneous malignant melanoma cells. Biosci Rep 2020; 40:BSR20191194. [PMID: 32134466 PMCID: PMC7080643 DOI: 10.1042/bsr20191194] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 01/13/2020] [Accepted: 02/12/2020] [Indexed: 11/17/2022] Open
Abstract
The present study aimed to investigate the effects of lncRNA FENDRR on the migration and invasion of malignant melanoma (MM) cells. The expression levels of FENDRR in MM tissues and MM cell lines were detected using qRT-PCR, followed by construction of FENDRR-knocked down and overexpressed stable cells. Then the effects of FENDRR on cell proliferation, migration and invasion were detected using MTT assay and Transwell assay. The protein expression levels of matrix metallopeptidase 2 (MMP2), MMP9, and related factors in JNK/c-Jun pathway were detected using Western blot. FENDRR was down-regulated in MM tissues and cell lines. Besides, its expression levels in different MM cells were diverse. Knockdown of FENDRR facilitated MM cells proliferation, migration and invasion in A375 cells, while overexpressing FENDRR had reverse results. In addition, MMPs and JNK/c-Jun pathway involved in the FENDRR-mediated regulation of MM cell proliferation, migration and invasion. Our results demonstrated that FENDRR mediated the metastasis phenotype of MM cells by inhibiting the expressions of MMP2 and MMP9 and antagonizing the JNK/c-Jun pathway.
Collapse
Affiliation(s)
- Xu-e Chen
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| | - Pu Chen
- Department of Information, Guizhou Province Hospital of Traditional Chinese Medicine, Guiyang, Guizhou 550001, China
| | - Shanshan Chen
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| | - Jin Lu
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| | - Ting Ma
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| | - Guang Shi
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| | - Liang Sheng
- Department of Dermatology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou 550002, China
| |
Collapse
|
10
|
Zhang C, Wang F, Gao Z, Zhang P, Gao J, Wu X. Regulation of Hippo Signaling by Mechanical Signals and the Cytoskeleton. DNA Cell Biol 2020; 39:159-166. [PMID: 31821009 DOI: 10.1089/dna.2019.5087] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Cong Zhang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, China
- State Education Ministry Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, China
| | - Feng Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zengxin Gao
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Orthopedics, Nanjing Lishui People’s Hospital, Nanjing, China
- Department of Orthopedics, Zhongda Hospital, Lishui Branch, Southeast University, Nanjing, China
| | - Pei Zhang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jiawei Gao
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, China
- State Education Ministry Laboratory of Developmental Genes and Human Diseases, Southeast University, Nanjing, China
| | - Xiaotao Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
11
|
Zhang D, Zhang Y, Cai Z, Tu Y, Hu Z. Dexamethasone and lenvatinib inhibit migration and invasion of non-small cell lung cancer by regulating EKR/AKT and VEGF signal pathways. Exp Ther Med 2019; 19:762-770. [PMID: 31853327 DOI: 10.3892/etm.2019.8225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 02/24/2017] [Indexed: 12/22/2022] Open
Abstract
Migration and invasion is one of the most important features in tumor metastasis and development. Non-small cell lung cancer (NSCLC) is one of the most common types of cancer globally, and has been linked to air contamination. Evidence indicates that cysteine-rich angiogenic inducer 61 (CYR61) is associated with the migration and invasion of NSCLC. Overexpression of CYR61 protein promotes the migration and the transition of tumor-derived vascular endothelial cells in NSCLC. However, the association between CYR61 and NSCLC remains poorly understood. Lenvatinib is an oral multi-target drug that targets various receptors upon tumor angiogenesis. Dexamethasone is widely approved for combination therapy in patients with NSCLC. In the current study, the expression and function of CYR61 in NSCLC was analyzed during the progression of NSCLC. Inhibitory effects on migration and invasion induced by lenvatinib and dexamethasone were determined by migratory and invasion assays. Migratory pathways of extracellular signal-regulated kinases (ERK) and protein kinase B (AKT) were also investigated by targeting vascular endothelial growth factor (VEGF) and CYR61 via synergistic treatment with transforming growth factor-β1 (TGF-β1) and dexamethasone. Therapeutic outcomes of combined treatment with lenvatinib and dexamethasone were assessed in NSCLC-bearing mice. The results of the present study indicate that cooperative treatment of lenvatinib and dexamethasone significantly inhibited TGF-β1-induced cell migration and suppressed tumor growth (P<0.01). Notably, the results demonstrated that dexamethasone eradicated the promotion effects of TGF-β1 on the AKT/epithelial-mesenchymal transition process and lenvatinib extinguished tumor cell metastasis by targeting VEGF. The results of the current study also demonstrate that dexamethasone suppressed the expression of CAG-I and enhanced expression of matrix metalloproteinase-1. Synergistic treatment for NSCLC was demonstrated to be efficacious. In conclusion, dexamethasone inhibited AKT/ERK phosphorylation and lenvatinib antagonism bound VEGF leading to the limitation of migration and invasion of cancer cells in NSCLC.
Collapse
Affiliation(s)
- Daye Zhang
- Department of Pharmacy, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Yongxiang Zhang
- Respiratory and Clinical Care Unit, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Zeyuan Cai
- Department of Cardiovascular Institute, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Ying Tu
- Department of Pharmacy, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| | - Zhansong Hu
- Department of Pharmacy, Tianjin Chest Hospital, Tianjin 300222, P.R. China
| |
Collapse
|
12
|
Saleh R, Taha RZ, Sasidharan Nair V, Alajez NM, Elkord E. PD-L1 Blockade by Atezolizumab Downregulates Signaling Pathways Associated with Tumor Growth, Metastasis, and Hypoxia in Human Triple Negative Breast Cancer. Cancers (Basel) 2019; 11:E1050. [PMID: 31349612 PMCID: PMC6721435 DOI: 10.3390/cancers11081050] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/16/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022] Open
Abstract
Triple negative breast cancer (TNBC) is the most aggressive type of breast cancer, which shows resistance to common breast cancer therapies, as it lacks the expression of the most common breast cancer targets. Therefore, TNBC treatment remains a challenge. Targeting programmed cell death-ligand 1 (PD-L1) by monoclonal antibodies (mAbs), for example, atezolizumab, has revolutionized the treatment for various cancer types. However, the therapeutic efficacy of targeting PD-L1 in TNBC is currently under investigation. In this study, we investigated the molecular mechanisms by which the human TNBC cell line MDA-MB-231, expressing PD-L1, responds to atezolizumab, using RNA-Seq. Transcriptome analysis revealed 388 upregulated and 362 downregulated genes in response to atezolizumab treatment. The expression of selected genes, from RNA-Seq data, was subsequently validated using RT-qPCR in the MDA-MB-231 and MDA-MB-468 TNBC cells following atezolizumab treatment. Bioinformatics analysis revealed that atezolizumab downregulates genes promoting cell migration/invasion and metastasis, epithelial-mesenchymal transition (EMT), cell growth/proliferation/survival, and hypoxia. On the contrary, genes associated with apoptosis and DNA repair were upregulated in response to atezolizumab treatment. Gene set enrichment analyses revealed that a significant number of these genes are related to the NF-kB, PI3K/Akt/mTOR, MAPK, and CD40 signaling pathways. Using functional assays, we confirmed that atezolizumab increases MDA-MB-231 cell apoptosis/necrosis, and reduces their proliferation and viability. Collectively, our findings provide novel insights into the molecular mechanisms/signaling pathways by which atezolizumab exerts inhibitory effects on TNBC, thereby inhibiting EMT/metastasis, tumor growth/survival, and the induction of hypoxia.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Rowaida Z Taha
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha P.O. Box 34110, Qatar.
| |
Collapse
|
13
|
Omar Zaki SS, Kanesan L, Leong MYD, Vidyadaran S. The influence of serum-supplemented culture media in a transwell migration assay. Cell Biol Int 2019; 43:1201-1204. [PMID: 30811086 DOI: 10.1002/cbin.11122] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Our work cautions against the use of serum-supplemented culture media in a transwell migration assay when using chemoattractants other than FBS. At 24 h, a 5% foetal bovine serum (FBS) gradient caused BV2 microglia to migrate toward the lower compartment of the transwell apparatus. Interestingly, FBS-supplemented media in the absence of a gradient also resulted in notable microglia migration. Serum can therefore confound the interpretation of a transwell migration assay when another chemoattractant is used.
Collapse
Affiliation(s)
- Siti Sarah Omar Zaki
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia
| | - Livashini Kanesan
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia
| | - Ming Yeh Danielle Leong
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia.,Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, 126, Jalan Jalil Perkasa 19, 57000, Kuala Lumpur, Malaysia
| | - Sharmili Vidyadaran
- Neuroinflammation Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400, Malaysia
| |
Collapse
|
14
|
Mechanotransduction and Cytoskeleton Remodeling Shaping YAP1 in Gastric Tumorigenesis. Int J Mol Sci 2019; 20:ijms20071576. [PMID: 30934860 PMCID: PMC6480114 DOI: 10.3390/ijms20071576] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/14/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
The essential role of Hippo signaling pathway in cancer development has been elucidated by recent studies. In the gastrointestinal tissues, deregulation of the Hippo pathway is one of the most important driving events for tumorigenesis. It is widely known that Yes-associated protein 1 (YAP1) and WW domain that contain transcription regulator 1 (TAZ), two transcriptional co-activators with a PDZ-binding motif, function as critical effectors negatively regulated by the Hippo pathway. Previous studies indicate the involvement of YAP1/TAZ in mechanotransduction by crosstalking with the extracellular matrix (ECM) and the F-actin cytoskeleton associated signaling network. In gastric cancer (GC), YAP1/TAZ functions as an oncogene and transcriptionally promotes tumor formation by cooperating with TEAD transcription factors. Apart from the classic role of Hippo-YAP1 cascade, in this review, we summarize the current investigations to highlight the prominent role of YAP1/TAZ as a mechanical sensor and responder under mechanical stress and address its potential prognostic and therapeutic value in GC.
Collapse
|
15
|
Eckert RL. Transglutaminase 2 takes center stage as a cancer cell survival factor and therapy target. Mol Carcinog 2019; 58:837-853. [PMID: 30693974 DOI: 10.1002/mc.22986] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/14/2022]
Abstract
Transglutaminase 2 (TG2) has emerged as a key cancer cell survival factor that drives epithelial to mesenchymal transition, angiogenesis, metastasis, inflammation, drug resistance, cancer stem cell survival and stemness, and invasion and migration. TG2 can exist in a GTP-bound signaling-active conformation or in a transamidase-active conformation. The GTP bound conformation of TG2 contributes to cell survival and the transamidase conformation can contribute to cell survival or death. We present evidence suggesting that TG2 has a role in human cancer, summarize what is known about the TG2 mechanism of action in a range of cancer types, and discuss TG2 as a cancer therapy target.
Collapse
Affiliation(s)
- Richard L Eckert
- Department of Biochemistry and Molecular Biology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Extraction of 'Gannanzao' Orange Peel Essential Oil by Response Surface Methodology and its Effect on Cancer Cell Proliferation and Migration. Molecules 2019; 24:molecules24030499. [PMID: 30704118 PMCID: PMC6384855 DOI: 10.3390/molecules24030499] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 11/17/2022] Open
Abstract
The essential oil of ‘Gannanzao’ orange peel was extracted by hydrodistillation, and the extraction conditions were optimized by Box–Behnken response surface methodology. The components of essential oil were analyzed by GC-MS. Thirty-nine different components were detected, accounting for 99.59% of the total oil. Limonene (88.07%) was the prominent component. The optimal extraction conditions were as follows: liquid material ratio of 8.4:1 (mL/g), sodium chloride concentration of 5.3%, and distillation time of 3.5 h. The Cell Counting Kit-8 assay showed that ‘Gannanzao’ orange peel essential oil had good dose-dependent inhibition effect on the proliferation of HepG2 hepatoma cells and HCT116 colorectal cancer cells. When the concentration of the essential oil was 0.6 μL/mL or higher, the viability rate of both cancer cells became lower than 13.0%. The transwell assay indicated the essential oil can inhibit migration of both cancer cells at the concentration of 0.3 μL/mL.
Collapse
|
17
|
Down‐regulation of intracellular anti‐apoptotic proteins, particularly c‐FLIP by therapeutic agents; the novel view to overcome resistance to TRAIL. J Cell Physiol 2018; 233:6470-6485. [PMID: 29741767 DOI: 10.1002/jcp.26585] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/08/2018] [Indexed: 12/24/2022]
|
18
|
Xu Y, Wang Q, Zhang L, Zheng M. 2-Deoxy-d-glucose enhances TRAIL-induced apoptosis in human gastric cancer cells through downregulating JNK-mediated cytoprotective autophagy. Cancer Chemother Pharmacol 2018; 81:555-564. [DOI: 10.1007/s00280-018-3526-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/20/2018] [Indexed: 01/29/2023]
|
19
|
Lee HT, Huang CH, Chen WC, Tsai CS, Chao YL, Liu SH, Chen JH, Wu YY, Lee YJ. Transglutaminase 2 Promotes Migration and Invasion of Lung Cancer Cells. Oncol Res 2018; 26:1175-1182. [PMID: 29301592 PMCID: PMC7844758 DOI: 10.3727/096504018x15149761920868] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Given that the major threat of cancer is metastasis, delineation of the molecular mechanism underlying it would help devise therapeutic strategies. Transglutaminase 2 (TG2), belonging to the transglutaminase superfamily, is a versatile protein with enzymatic and nonenzymatic functions. It mainly localizes inside the cell, but also appears extracellularly. Recent findings have demonstrated the involvement of TG2 in cancer development. Here we examine the role of TG2 in metastasis of lung cancer using a lung cancer cell line CL1-0, which exhibits low invasiveness, and its invasive subline CL1-5. Our results show that CL1-5 cells express a higher amount of TG2 than CL1-0 cells. Overexpression of TG2 in CL1-0 enhances cell migration and invasion, and lowering TG2 expression in CL1-5 cells reduces their ability to do so. The transamidase activity of TG2 is not required since cells expressing the inactive TG2 mutant or treated with a TG2 inhibitor are still able to migrate and invade. TG2-stimulated migration and invasion are, at least in part, mediated by Rac, as inhibition of Rac activity suppresses cell migration and invasion. Lastly, exogenous application of recombinant TG2 protein to CL1-0 cells substantially augments cell migration and invasion, suggesting the significance of extracellular TG2 in promoting these events. Collectively, our results show that TG2 plays a positive role in cell migration and invasion, and this might help metastasis of lung cancer cells.
Collapse
Affiliation(s)
- Hung-Tsung Lee
- Division of Pulmonary Medicine, Antai Tian-Sheng Memorial Hospital, Pingtung, Taiwan, Republic of China
| | - Cheng-Hsieh Huang
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Wuan-Chun Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Chi-Shan Tsai
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Yu-Lin Chao
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Szu-Han Liu
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Jun-Hong Chen
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| | - Yi-Ying Wu
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan, Republic of China
| | - Yi-Ju Lee
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan, Republic of China
| |
Collapse
|
20
|
Shi S, Luo W, Zhang R, Wang C, Zheng Y, Song Y, Wang R, Zhang L, Zhang L, Li W, Luo Z. CRTC2 promotes non-small cell lung cancer A549 migration and invasion in vitro. Thorac Cancer 2018; 9:136-141. [PMID: 29105369 PMCID: PMC5754302 DOI: 10.1111/1759-7714.12550] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND CRTC2 is highly expressed in lung cancer and contributes to lung cancer pathogenesis; however, whether CRTC2 promotes lung cancer metastasis remains unknown. In the present study, we investigated the role of CRTC2 in lung cancer metastasis in vitro. METHODS CRTC2 stable knockdown of lung cancer cell A549 was generated with small hairpin RNA and confirmed by quantitative reverse transcription-PCR and Western blot. Wound healing and invasion transwell assays were performed to explore migration and invasion activity, and Western blot was conducted to detect the expression of related proteins. RESULTS Suppression of CRTC2 significantly inhibited A549 cell migration and invasion in vitro. Mechanistic studies showed that knockdown of CRTC2 greatly downregulated MMP2 and MMP9 expression. CRTC2 silencing remarkably suppressed epithelial-mesenchymal transition by modulating the expression of E-cadherin and vimentin. Furthermore, suppression of CRTC2 expression significantly reduced MAPK/c-Jun N-terminal kinase activity. CONCLUSION CRTC2 may promote A549 migration and invasion by modulation of c-Jun N-terminal kinase-mediated epithelial-mesenchymal transition and matrix metalloproteinase expression.
Collapse
Affiliation(s)
- Shaoqing Shi
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Wei Luo
- Department of Respiratory MedicineThe People's Hospital of LeshanLeshanChina
| | - Rui Zhang
- Department of Thoracic SurgeryThe Seventh People's Hospital of ChengduChengduChina
| | - Chu Wang
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Yuanyuan Zheng
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Yunhua Song
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Rongchun Wang
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Liyan Zhang
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Lihua Zhang
- Department of Geriatric EndocrinologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Weimin Li
- Department of Respiratory MedicineWest China Hospital, Sichuan UniversityChengduChina
| | - Zhuang Luo
- Department of Respiratory MedicineThe First Affiliated Hospital of Kunming Medical UniversityKunmingChina
| |
Collapse
|
21
|
Ishii S, Yamashita K, Harada H, Ushiku H, Tanaka T, Nishizawa N, Yokoi K, Washio M, Ema A, Mieno H, Moriya H, Hosoda K, Waraya M, Katoh H, Watanabe M. The H19-PEG10/IGF2BP3 axis promotes gastric cancer progression in patients with high lymph node ratios. Oncotarget 2017; 8:74567-74581. [PMID: 29088808 PMCID: PMC5650363 DOI: 10.18632/oncotarget.20209] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022] Open
Abstract
We previously demonstrated that the lymph node ratio (LNR) is a prognostic factor associated with EGFR expression, among first priority genes amplified or overexpressed in cancer. Here, we investigated the associations between high LNR and second, third, and fourth priority genes. We performed mRNA expression microarray analysis of tumor tissue from patients with stage III gastric cancer and high or low LNRs. Candidate high LNR-associated genes were further evaluated in 39 patients with stage III gastric cancer. The functional relevance of these genes was evaluated in gastric cancer cell lines. We focused on five genes: H19,PEG10, IGF2BP3, CD177, and PGA3. H19 and PEG10 were confirmed as high LNR-associated genes. H19, PEG10, and IGF2BP3 were found to promote each other’s expression. Knocking down H19 or PEG10 using RNAi decreased cell proliferation, invasion, anchorage-independent growth, and chemoresistance. These genes had a mutual relationship in MKN7 cells. H19 knockdown decreased expression of epithelial-mesenchymal transition-associated genes in MKN74 cells to suppress transformation. Thus, H19 promotes epithelial-mesenchymal transition in gastric cancer and is a potential therapeutic target.
Collapse
Affiliation(s)
- Satoru Ishii
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Keishi Yamashita
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiroki Harada
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hideki Ushiku
- Department of Surgery, Kitasato University Medical Center, Saitama, Japan
| | - Toshimichi Tanaka
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Nobuyuki Nishizawa
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Keigo Yokoi
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Marie Washio
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akira Ema
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiroaki Mieno
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiromitsu Moriya
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kei Hosoda
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Mina Waraya
- Department of Surgery, Sagamino Hospital, Sagamihara, Japan
| | - Hiroshi Katoh
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masahiko Watanabe
- Department of Surgery, Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
22
|
Bagatur Y, Ilter Akulke AZ, Bihorac A, Erdem M, Telci D. Tissue transglutaminase expression is necessary for adhesion, metastatic potential and cancer stemness of renal cell carcinoma. Cell Adh Migr 2017; 12:138-151. [PMID: 28498731 DOI: 10.1080/19336918.2017.1322255] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tissue transglutaminase (TG2) is the ubiquitously expressed member of transglutaminase family and shown to play a critical role in the development and progression of drug resistance malignancies. We have previously showed the association of TG2 upregulation with progression and metastasis of renal cell carcinoma (RCC) and low disease-free survival. In the present study we further investigate the role of TG2 in cell adhesion, migration and invasion of RCC by silencing TG2 expression in Caki-2 and A-498 primary site and Caki-1 and ACHN metastatic site RCC cell lines. Downregulation of TG2 expression led up to a 60% decrease in actin stress fiber formation and adhesion to β 1 integrin (ITGB1) substrates fibronectin, collagen type I and laminin in both primary and metastatic site RCC cell lines. In addition, treatment with siRNAs against TG2 impaired the migration capacity and cellular invasiveness of ITGB1 substrates in all 4 RCC cell lines. Lastly, the knockdown of TG2 in metastatic Caki-1 cells diminished the expression of CD44, CD73-and CD105 cancer stem cell-like markers. We conclude, for the first time, that TG2 expression is critical for cancer cell adhesion, migration, invasiveness and cancer cell-stemness during RCC progression and dissemination. Therefore, combined targeting of TG2 with drugs widely used in the treatment of RCC may be a promising therapeutic strategy for RCC.
Collapse
Affiliation(s)
- Yesim Bagatur
- a Department of Genetics and Bioengineering , Yeditepe University , Istanbul , Turkey
| | | | - Ajna Bihorac
- a Department of Genetics and Bioengineering , Yeditepe University , Istanbul , Turkey
| | - Merve Erdem
- a Department of Genetics and Bioengineering , Yeditepe University , Istanbul , Turkey
| | - Dilek Telci
- a Department of Genetics and Bioengineering , Yeditepe University , Istanbul , Turkey
| |
Collapse
|
23
|
Wei G, Xu Y, Peng T, Yan J, Wang Z, Sun Z. Sanguinarine exhibits antitumor activity via up-regulation of Fas-associated factor 1 in non-small cell lung cancer. J Biochem Mol Toxicol 2017; 31. [PMID: 28296008 DOI: 10.1002/jbt.21914] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/06/2017] [Accepted: 02/09/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Guangxia Wei
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Yahuan Xu
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Tao Peng
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Jie Yan
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Zhengjun Wang
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| | - Zhanwen Sun
- Department of Cardiothoracic Surgery, Huangshi Central Hospital; Affiliated Hospital of Hubei Polytechnic University; Edong Healthcare Group Huangshi 435000 People's Republic of China
| |
Collapse
|
24
|
Lee HJ, Diaz MF, Price KM, Ozuna JA, Zhang S, Sevick-Muraca EM, Hagan JP, Wenzel PL. Fluid shear stress activates YAP1 to promote cancer cell motility. Nat Commun 2017; 8:14122. [PMID: 28098159 PMCID: PMC5253685 DOI: 10.1038/ncomms14122] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 11/04/2016] [Indexed: 12/28/2022] Open
Abstract
Mechanical stress is pervasive in egress routes of malignancy, yet the intrinsic effects of force on tumour cells remain poorly understood. Here, we demonstrate that frictional force characteristic of flow in the lymphatics stimulates YAP1 to drive cancer cell migration; whereas intensities of fluid wall shear stress (WSS) typical of venous or arterial flow inhibit taxis. YAP1, but not TAZ, is strictly required for WSS-enhanced cell movement, as blockade of YAP1, TEAD1-4 or the YAP1–TEAD interaction reduces cellular velocity to levels observed without flow. Silencing of TEAD phenocopies loss of YAP1, implicating transcriptional transactivation function in mediating force-enhanced cell migration. WSS dictates expression of a network of YAP1 effectors with executive roles in invasion, chemotaxis and adhesion downstream of the ROCK–LIMK–cofilin signalling axis. Altogether, these data implicate YAP1 as a fluid mechanosensor that functions to regulate genes that promote metastasis. Fluid frictional forces around cancer cells influence chemokine production and delivery of chemotherapeutic drugs but it is unclear if they directly impact tumour biology through biomechanical effects. Here, the authors show that wall shear stress stimulates cancer cell migration through a ROCK–LIMK–YAP axis.
Collapse
Affiliation(s)
- Hyun Jung Lee
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Miguel F Diaz
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Katherine M Price
- Department of BioSciences, Rice University, Houston, Texas 77030, USA
| | - Joyce A Ozuna
- Department of BioSciences, Rice University, Houston, Texas 77030, USA
| | - Songlin Zhang
- Department of Pathology, The University of Texas Medical School, Houston, Texas 77030, USA
| | - Eva M Sevick-Muraca
- Center for Molecular Imaging, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - John P Hagan
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Pamela L Wenzel
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.,Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| |
Collapse
|
25
|
Zhang H, Chen Z, Miranda RN, Medeiros LJ, McCarty N. TG2 and NF-κB Signaling Coordinates the Survival of Mantle Cell Lymphoma Cells via IL6-Mediated Autophagy. Cancer Res 2016; 76:6410-6423. [PMID: 27488529 DOI: 10.1158/0008-5472.can-16-0595] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/05/2016] [Indexed: 01/01/2023]
Abstract
Expression of the transglutaminase TG2 has been linked to constitutive activation of NF-κB and chemotherapy resistance in mantle cell lymphoma (MCL) cells. TG2 forms complexes with NF-κB components, but mechanistic insights that could be used to leverage therapeutic responses has been lacking. In the current study, we address this issue with the discovery of an unexpected role for TG2 in triggering autophagy in drug-resistant MCL cells through induction of IL6. CRISPR-mediated silencing of TG2 delayed apoptosis while overexpressing TG2 enhanced tumor progression. Under stress, TG2 and IL6 mediate enhanced autophagy formation to promote MCL cell survival. Interestingly, the autophagy product ATG5 involved in autophagosome elongation positively regulated TG2/NF-κB/IL6 signaling, suggesting a positive feedback loop. Our results uncover an interconnected network of TG2/NF-κB and IL6/STAT3 signaling with autophagy regulation in MCL cells, the disruption of which may offer a promising therapeutic strategy. Cancer Res; 76(21); 6410-23. ©2016 AACR.
Collapse
Affiliation(s)
- Han Zhang
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas-Health Science Center at Houston, Houston, Texas
| | - Zheng Chen
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas-Health Science Center at Houston, Houston, Texas
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nami McCarty
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), The University of Texas-Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
26
|
The fibronectin III-1 domain activates a PI3-Kinase/Akt signaling pathway leading to αvβ5 integrin activation and TRAIL resistance in human lung cancer cells. BMC Cancer 2016; 16:574. [PMID: 27484721 PMCID: PMC4970220 DOI: 10.1186/s12885-016-2621-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background Fibronectin is a mechanically sensitive protein which is organized in the extracellular matrix as a network of interacting fibrils. The lung tumor stroma is enriched for fibronectin which is thought to contribute to metastasis and drug resistance. Fibronectin is an elastic, multi-modular protein made up of individually folded domains, some of which can stretch in response to increased mechanical tension. Very little is known about the relationship of fibronectin’s unfolded domains to lung cancer resistance to chemotherapy. In the present study, we evaluated the impact of unfolding the first Type III domain of fibronectin (FnIII-1c) on TNF-related apoptosis inducing ligand (TRAIL) resistance. Methods NCI-H460 non-small cell lung cancer cells were treated with FnIII-1c then assessed for TRAIL-induced apoptosis. Subsequent analysis of FnIII-1c-mediated signaling pathways was also completed. Human non-small cell lung cancer tissue sections were assessed for the expression of vitronectin by immunohistochemistry. Results FnIII-1c inhibited TRAIL-induced activation of caspase 8 and subsequent apoptosis in NCI-H460 lung cancer cells. FnIII-1c treatment was associated with the activation of the phosphatidylinositol-3-kinase/alpha serine/threonine kinase (PI3K/Akt) pathway and the αvβ5 integrin receptor for vitronectin, both of which were required for TRAIL resistance. Immunohistochemical staining of sections from non-small cell lung cancers showed that vitronectin was localized around blood vessels and in the tumor-stroma interface. Conclusions Unfolding of Type III domains within the fibronectin matrix may promote TRAIL resistance through the activation of a PI3K/Akt/αvβ5 signaling axis and point to a novel mechanism by which changes in secondary structure of fibronectin contribute to cancer cell resistance to apoptosis.
Collapse
|
27
|
Pal S, Amin PJ, Sainis KB, Shankar BS. Potential Role of TRAIL in Metastasis of Mutant KRAS Expressing Lung Adenocarcinoma. CANCER MICROENVIRONMENT 2016; 9:77-84. [PMID: 27106232 DOI: 10.1007/s12307-016-0184-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 04/19/2016] [Indexed: 11/26/2022]
Abstract
Apo2L/tumor necrosis factor (TNF)-α-related apoptosis-inducing ligand (TRAIL, TNFSF10) is an important cytokine in the tumor microenvironment and plays a major role in the balance of cell survival/death pathways. Bioinformatic analyses of 839 adenocarcinoma (AC) and 356 squamous cell lung carcinoma patient data (SCC) by cBioPortal (genomic analyses) shows that TRAIL expression leads to differential outcomes of disease free survival in AC and SCC. Oncomine datamining (transcript analyses) reveal that TRAIL is upregulated in 167 SCC as compared to 350 AC patients from six data sets. Genomic analyses using cBioPortal revealed high rates of KRAS mutation in AC accompanied by higher incidence of metastasis and increased amplifications of TRAIL gene in SCC. Bioinformatic analyses of an additional lung cancer patient database also showed that risk of disease progression was significantly increased with high TRAIL expression in AC (461 samples). In vitro studies demonstrated that TRAIL increased phosphorylation of ERK only in adenocarcinoma cell lines with mutant KRAS. This was associated with increased migration that was abrogated by MEK inhibitor PD98059. Effects of increased migration induced by TRAIL persisted even after exposure to ionizing radiation with suppression of DNA damage response. These results help understand the role of TRAIL signaling in metastasis which is essential to develop strategies to revert these signals into pro-apoptotic pathways.
Collapse
Affiliation(s)
- Shyama Pal
- Immunology Section, Radiation Biology & Health Sciences Division, BioScience Group, Bhabha Atomic Research Centre, Modular Laboratories, Mumbai, 400085, India
| | - Prayag J Amin
- Immunology Section, Radiation Biology & Health Sciences Division, BioScience Group, Bhabha Atomic Research Centre, Modular Laboratories, Mumbai, 400085, India
| | - K B Sainis
- Immunology Section, Radiation Biology & Health Sciences Division, BioScience Group, Bhabha Atomic Research Centre, Modular Laboratories, Mumbai, 400085, India
| | - Bhavani S Shankar
- Immunology Section, Radiation Biology & Health Sciences Division, BioScience Group, Bhabha Atomic Research Centre, Modular Laboratories, Mumbai, 400085, India.
| |
Collapse
|
28
|
Singh G, Zhang J, Ma Y, Cerione RA, Antonyak MA. The Different Conformational States of Tissue Transglutaminase Have Opposing Affects on Cell Viability. J Biol Chem 2016; 291:9119-32. [PMID: 26893378 PMCID: PMC4861479 DOI: 10.1074/jbc.m115.699108] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/11/2016] [Indexed: 02/06/2023] Open
Abstract
Tissue transglutaminase (tTG) is an acyltransferase/GTP-binding protein that contributes to the development of various diseases. In human cancer cells, tTG activates signaling pathways that promote cell growth and survival, whereas in other disorders (i.e. neurodegeneration), overexpression of tTG enhances cell death. Therefore, it is important to understand how tTG is differentially regulated and functioning to promote diametrically distinct cellular outcomes. Previous structural studies revealed that tTG adopts either a nucleotide-bound closed conformation or a transamidation-competent open conformation. Here we provide evidence showing that these different conformational states determine whether tTG promotes, or is detrimental to, cell survival, with the open conformation of the protein being responsible for inducing cell death. First, we demonstrate that a nucleotide binding-defective form of tTG, which has previously been shown to induce cell death, assumes an open conformation in solution as assessed by an enhanced sensitivity to trypsin digestion and by small angle x-ray scattering (SAXS) analysis. We next identify two pairs of intramolecular hydrogen bonds that, based on existing x-ray structures, are predicted to form between the most C-terminal β-barrel domain and the catalytic core domain of tTG. By disrupting these hydrogen bonds, we are able to generate forms of tTG that constitutively assume an open conformation and induce apoptosis. These findings provide important insights into how tTG participates in the pathogenesis of neurodegenerative diseases, particularly with regard to the actions of a C-terminal truncated form of tTG (TG-Short) that has been linked to such disorders and induces apoptosis by assuming an open-like conformation.
Collapse
Affiliation(s)
| | | | - Yilun Ma
- From the Department of Molecular Medicine and
| | - Richard A Cerione
- From the Department of Molecular Medicine and the Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853
| | | |
Collapse
|
29
|
Xu J, Xu X, Shi S, Wang Q, Saxton B, He W, Gou X, Jang JH, Nyunoya T, Wang X, Xing C, Zhang L, Lin Y. Autophagy-Mediated Degradation of IAPs and c-FLIP(L) Potentiates Apoptosis Induced by Combination of TRAIL and Chal-24. J Cell Biochem 2015; 117:1136-44. [PMID: 26448608 DOI: 10.1002/jcb.25397] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022]
Abstract
Combination chemotherapy is an effective strategy for increasing anticancer efficacy, reducing side effects and alleviating drug resistance. Here we report that combination of the recently identified novel chalcone derivative, chalcone-24 (Chal-24), and TNF-related apoptosis-inducing ligand (TRAIL) significantly increases cytotoxicity in lung cancer cells. Chal-24 treatment significantly enhanced TRAIL-induced activation of caspase-8 and caspase-3, and the cytotoxicity induced by combination of these agents was effectively suppressed by the pan-caspase inhibitor z-VAD-fmk. Chal-24 and TRAIL combination suppressed expression of cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein large (c-FLIP(L)) and cellular inhibitor of apoptosis proteins (c-IAPs), and ectopic expression of c-FLIP(L) and c-IAPs inhibited the potentiated cytotoxicity. In addition, TRAIL and Chal-24 cooperatively activated autophagy. Suppression of autophagy effectively attenuated cytotoxicity induced by Chal-24 and TRAIL combination, which was associated with attenuation of c-FLIP(L) and c-IAPs degradation. Altogether, these results suggest that Chal-24 potentiates the anticancer activity of TRAIL through autophagy-mediated degradation of c-FLIP(L) and c-IAPs, and that combination of Chal-24 and TRAIL could be an effective approach in improving chemotherapy efficacy.
Collapse
Affiliation(s)
- Jennings Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| | - Xiuling Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| | - Shaoqing Shi
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico.,Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Qiong Wang
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico.,Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Bryanna Saxton
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 510182, P.R. China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 510182, P.R. China
| | - Jun-Ho Jang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China.,Division of Pulmonary and Critical Care Medicine, University of New Mexico and New Mexico VA Health Care System, Albuquerque, New Mexico
| | - Toru Nyunoya
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China.,Division of Pulmonary and Critical Care Medicine, University of New Mexico and New Mexico VA Health Care System, Albuquerque, New Mexico
| | - Xia Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, 55455, Minnesota
| | - Lin Zhang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| |
Collapse
|
30
|
Huang L, Xu AM, Liu W. Transglutaminase 2 in cancer. Am J Cancer Res 2015; 5:2756-2776. [PMID: 26609482 PMCID: PMC4633903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 06/12/2015] [Indexed: 06/05/2023] Open
Abstract
The significant influence of tumor microenvironment on malignant cells has been investigated with enthusiasm in this era of targeted therapy. Transglutaminase 2 (TG2, EC 2.3.2.13), a multi-functional enzyme that catalyzes the formation of intermolecular isopeptide bonds between glutamine and lysine side-chains, has been reported to exert important pathophysiological functions. The aim of this review was to investigate the correlation between TG2 and malignant behaviors, which could provide the rationale for novel approaches in anti-cancer therapy. We performed a systematic and electronic search on Medline, Scopus, and Web of Science for relevant publications from inception to April 2015. The bibliographic references of retrieved articles were further reviewed for additional relevant studies. TG2 exerts important physiological functions and plays vital roles in inflammation mainly through its modulation on the structure and stability of extracellular matrix (ECM). It also regulates EMT of diverse malignant cells through various intracellular and extracellular pathways. TG2 also plays an important role in tumor progression and may serve as a novel prognostic biomarker and therapeutic target in various cancer types. TG2 promotes malignant cell mobility, invasion, and metastasis, and induces chemo-resistance of cancer cells, mainly through its pro-crosslink and signaling transduction mediation propensities. In conclusion, TG2 plays vital roles in malignancy progression, and may have important prognostic and therapeutic significances.
Collapse
Affiliation(s)
- Lei Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical UniversityHefei, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
- Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
- Research Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty of Mannheim, Heidelberg UniversityMannheim, Germany
| | - A-Man Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Anhui Medical UniversityHefei, China
| | - Wei Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
- Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
31
|
Yan D, Ge Y, Deng H, Chen W, An G. Gefitinib upregulates death receptor 5 expression to mediate rmhTRAIL-induced apoptosis in Gefitinib-sensitive NSCLC cell line. Onco Targets Ther 2015; 8:1603-10. [PMID: 26170696 PMCID: PMC4498723 DOI: 10.2147/ott.s73731] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in tumor cells, but when used alone, it is not effective in the treatment of TRAIL-resistant tumors. Some studies have shown that gefitinib interacts with recombinant mutant human TRAIL (rmhTRAIL) to induce high levels of apoptosis in gefitinib-responsive bladder cancer cell lines; however, the molecular mechanisms underlying the anticancer effects are not fully understood. Several reports have shown that the death receptor 5 (DR5) plays an important role in sensitizing cancer cells to apoptosis induced by TRAIL. Therefore, we investigated the effects of the combination of drugs and the expression of the DR5 to analyze the growth of a gefitinib-responsive non-small cell lung cancer cell line PC9, which was treated with rmhTRAIL and gefitinib individually or in combination. Methods Human PC9 non-small cell lung cancer cells harboring an epidermal growth factor receptor mutation were used as a model for the identification of the therapeutic effects of gefitinib alone or in combination with rmhTRAIL, and cytotoxicity was assessed by MTT assays. Cell cycle and apoptosis were investigated using flow cytometry. Moreover, the effects of drugs on DR5, BAX, FLIP, and cleaved-caspase3 proteins expressions were analyzed using Western blot analyses. Finally, quantitative polymerase chain reaction analysis was carried out to assess whether rmhTRAIL and gefitinib modulate the expression of genes related to drug activity. Results Gefitinib and rmhTRAIL synergistically interact to inhibit cell proliferation, and apoptosis assessment demonstrated that associations of drug increased the apoptotic index. rmhTRAIL when used alone downregulated DR5 and upregulated BAX, FLIP, and cleaved-caspase3 proteins expressions. However, results obtained in Western blot analyses demonstrated that the combined treatment-induced cell apoptosis was achieved involving upregulated DR5, cleaved-caspase3, and BAX proteins expression and downregulated FLIP protein expression. Moreover, quantitative polymerase chain reaction showed that gefitinib modulated the expression of targets related to rmhTRAIL activity. Conclusion These results indicate that epidermal growth factor receptor inhibitors enhance rmhTRAIL antitumor activity in the gefitinib-responsive PC9 cell line, and upregulated DR5 expression plays a critical role in activating caspase-signaling apoptotic pathway.
Collapse
Affiliation(s)
- Dong Yan
- Department of Oncology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China ; Translational Molecular pathology, M.D Anderson Cancer Center, Houston, TX, USA
| | - Yang Ge
- Department of Oncology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Haiteng Deng
- School of Sciences, Tsinghua University, Beijing, People's Republic of China
| | - Wenming Chen
- Department of Hematology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Guangyu An
- Department of Oncology, Beijing Chao-yang Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
32
|
Ciprotti M, Tebbutt NC, Lee FT, Lee ST, Gan HK, McKee DC, O'Keefe GJ, Gong SJ, Chong G, Hopkins W, Chappell B, Scott FE, Brechbiel MW, Tse AN, Jansen M, Matsumura M, Kotsuma M, Watanabe R, Venhaus R, Beckman RA, Greenberg J, Scott AM. Phase I Imaging and Pharmacodynamic Trial of CS-1008 in Patients With Metastatic Colorectal Cancer. J Clin Oncol 2015; 33:2609-16. [PMID: 26124477 DOI: 10.1200/jco.2014.60.4256] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE CS-1008 (tigatuzumab) is a humanized, monoclonal immunoglobulin G1 (IgG1) agonistic antibody to human death receptor 5. The purpose of this study was to investigate the impact of CS-1008 dose on the biodistribution, quantitative tumor uptake, and antitumor response in patients with metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with mCRC who had received at least one course of chemotherapy were assigned to one of five dosage cohorts and infused with a weekly dose of CS-1008. Day 1 and day 36 doses were trace-labeled with indium-111 ((111)In), followed by whole-body planar and regional single-photon emission computed tomography (SPECT) imaging at several time points over the course of 10 days. RESULTS Nineteen patients were enrolled. (111)In-CS-1008 uptake in tumor was observed in only 12 patients (63%). (111)In-CS-1008 uptake and pharmacokinetics were not affected by dose or repeated drug administration. (111)In-CS-1008 biodistribution showed gradual blood-pool clearance and no abnormal uptake in normal tissue. No anti-CS-1008 antibody development was detected. One patient achieved partial response (3.7 months duration), eight patients had stable disease, and 10 patients had progressive disease. Clinical benefit rate (stable disease + partial response) in patients with (111)In-CS-1008 uptake in tumor was 58% versus 28% in patients with no uptake. An analysis of individual lesions showed that lesions with antibody uptake were one third as likely to progress as those without antibody uptake (P = .07). Death-receptor-5 expression in archived tumor samples did not correlate with (111)In-CS-1008 uptake (P = .5) or tumor response (P = .6). CONCLUSION Death-receptor-5 imaging with (111)In-CS-1008 reveals interpatient and intrapatient heterogeneity of uptake in tumor, is not dose dependent, and is predictive of clinical benefit in the treatment of patients who have mCRC.
Collapse
Affiliation(s)
- Marika Ciprotti
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Niall C Tebbutt
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Fook-Thean Lee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Sze-Ting Lee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Hui K Gan
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - David C McKee
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Graeme J O'Keefe
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Sylvia J Gong
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Geoffrey Chong
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Wendie Hopkins
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Bridget Chappell
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Fiona E Scott
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Martin W Brechbiel
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Archie N Tse
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Mendel Jansen
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Manabu Matsumura
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Masakatsu Kotsuma
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Rira Watanabe
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Ralph Venhaus
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Robert A Beckman
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Jonathan Greenberg
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY
| | - Andrew M Scott
- Marika Ciprotti, Niall C. Tebbutt, Fook-Thean Lee, Sze-Ting Lee, Hui K. Gan, Wendie Hopkins, Fiona E. Scott, Andrew M. Scott, Ludwig Institute for Cancer Research; David C. McKee, Graeme J. O'Keefe, Sylvia J. Gong, Geoffrey Chong, Bridget Chappell, Andrew M. Scott, Austin Health, Melbourne, Australia; Martin W. Brechbiel, National Cancer Institute, Bethesda, MD; Archie N. Tse, Jonathan Greenberg, Daiichi Sankyo Co., Ltd, Parsippany, NJ; Mendel Jansen, Daiichi Sankyo Development Ltd, Gerrards Cross, Buckinghamshire, United Kingdom; Manabu Matsumura, Masakatsu Kotsuma, Rira Watanabe, Daiichi Sankyo Co., Ltd, Tokyo, Japan; Robert A. Beckman, Georgetown University Medical Center and Ralph Venhaus, Ludwig Institute for Cancer Research, New York, NY.
| |
Collapse
|
33
|
Kim DG, Jung KH, Lee DG, Yoon JH, Choi KS, Kwon SW, Shen HM, Morgan MJ, Hong SS, Kim YS. 20(S)-Ginsenoside Rg3 is a novel inhibitor of autophagy and sensitizes hepatocellular carcinoma to doxorubicin. Oncotarget 2015; 5:4438-51. [PMID: 24970805 PMCID: PMC4147336 DOI: 10.18632/oncotarget.2034] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide. High mortality from HCC is mainly due to widespread prevalence and the lack of effective treatment, since systemic chemotherapy is ineffective, while the targeted agent Sorafenib extends median survival only briefly. The steroidal saponin 20(S)-ginsenoside Rg3 from Panax ginseng C.A. Meyer is proposed to chemosensitize to various therapeutic drugs through an unknown mechanism. Since autophagy often serves as cell survival mechanism in cancer cells exposed to chemotherapeutic agents, we examined the ability of Rg3 to inhibit autophagy and chemosensitize HCC cell lines to doxorubicin in vitro. We show that Rg3 inhibits late stage autophagy, possibly through changes in gene expression. Doxorubicin-induced autophagy plays a protective role in HCC cells, and therefore Rg3 treatment synergizes with doxorubicin to kill HCC cell lines, but the combination is relatively nontoxic in normal liver cells. In addition, Rg3 was well-tolerated in mice and synergized with doxorubicin to inhibit tumor growth in HCC xenografts in vivo. Since novel in vivo inhibitors of autophagy are desirable for clinical use, we propose that Rg3 is such a compound, and that combination therapy with classical chemotherapeutic drugs may represent an effective therapeutic strategy for HCC treatment.
Collapse
Affiliation(s)
- Dong-Gun Kim
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | | | - Da-Gyum Lee
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | - Jung-Ho Yoon
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | - Kyeong Sook Choi
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| | - Sung Won Kwon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Han-Ming Shen
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Michael J Morgan
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado
| | | | - You-Sun Kim
- Department of Biochemistry and Department of Biomedical Sciences, Ajou University School of Medicine, Suwon
| |
Collapse
|
34
|
Abstract
INTRODUCTION Esophageal adenocarcinomas (EAC) are aggressive cancers that are increasing in incidence and associated with a poor prognosis. The identification of highly expressed genes in EAC relative to metaplastic Barrett's esophagus (BE) may provide new targets for novel early cancer detection strategies using endoscopically administered, fluorescently labeled peptides. METHODS Gene expression analysis of BE and EACs were used to identify the cell surface marker transglutaminase 2 (TGM2) as overexpressed in cancer. The expression of two major isoforms of TGM2 was determined by qRT-polymerase chain reaction in an independent cohort of 128 EACs. Protein expression was confirmed by tissue microarrays and immunoblot analysis of EAC cell lines. TGM2 DNA copy number was assessed using single nucleotide polymorphism microarrays and confirmed by qPCR. TGM2 expression in neoadjuvantly treated EACs and following small interfering RNA-mediated knockdown in cisplatin-treated EAC cells was used to determine its possible role in chemoresistance. RESULTS TGM2 is overexpressed in 15 EACs relative to 26 BE samples. Overexpression of both TGM2 isoforms was confirmed in 128 EACs and associated with higher tumor stage, poor differentiation, and increased inflammatory and desmoplastic response. Tissue microarrays and immunohistochemistry confirmed elevated TGM2 protein expression in EAC. Single nucleotide polymorphism and qPCR analysis revealed increased TGM2 gene copy number as one mechanism underlying elevated TGM2 expression. TGM2 was highly expressed in resistant EAC after patient treatment with neoadjuvant chemotherapy/radiation suggesting a role for TGM2 in chemoresistance. CONCLUSION TGM2 may be a useful cell surface biomarker for early detection of EAC.
Collapse
|
35
|
Nong Y, Wu D, Lin Y, Zhang Y, Bai L, Tang H. Tenascin-C expression is associated with poor prognosis in hepatocellular carcinoma (HCC) patients and the inflammatory cytokine TNF-α-induced TNC expression promotes migration in HCC cells. Am J Cancer Res 2015; 5:782-791. [PMID: 25973315 PMCID: PMC4396033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023] Open
Abstract
Although tenascin-c (TNC) in inflammatory microenvironment contributes to progression in some tumors, its role in hepatocellular carcinoma (HCC) in metastasis and the mechanism by which TNC expression is regulated in HCC cells are elusive. In this study, we examined TNC expression in 100 HCC tissue samples by immunohistochemistry and compared which between the groups with or without metastasis. TNC expression was higher in metastatic HCC tissues than that in the non-metastatic HCC tissues, which was associated with the Knodell inflammation scores. Importantly, high level of TNC expression was associated with lower survival rate and shorter survival time in the HCC patients. We then investigated the mechanism by which TNC expression is regulated in HCC cells with an in vitro cell culture system. The recombinant TNF-α and conditioned medium from macrophages induced TNC expression at both mRNA and protein levels in HepG2 cells. The induction of TNC expression by conditioned medium from macrophages was suppressed by a TNF-α neutralizing antibody. TNF-α-promoted cell migration was inhibited by a TNC siRNA. In addition, TNF-α-induced TNC expression was blocked by a NF-κB pathway inhibitor. These results suggest that TNF-α in the tumor microenvironment induces TNC expression in HCC cells through the NF-κB pathway, which in turn, promotes HCC cell migration. Thus, TNC may play an important role in promoting HCC metastasis and TNC expression could be a predictive factor for poor prognosis in HCC patients.
Collapse
Affiliation(s)
- Yunhong Nong
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute2425 Ridgecrest Dr. SE, Albuquerque, NM 87108, USA
| | - Yongqiang Zhang
- Biorepository, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| |
Collapse
|
36
|
Aragona P, Aguennouz M, Rania L, Postorino E, Sommario MS, Roszkowska AM, De Pasquale MG, Pisani A, Puzzolo D. Matrix metalloproteinase 9 and transglutaminase 2 expression at the ocular surface in patients with different forms of dry eye disease. Ophthalmology 2014; 122:62-71. [PMID: 25240629 DOI: 10.1016/j.ophtha.2014.07.048] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/17/2014] [Accepted: 07/25/2014] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To evaluate the expression of matrix metalloproteinase 9 (MMP9) and transglutaminase 2 (TG2) in different forms of dry eye. DESIGN Case control study. PARTICIPANTS Seventy-five female subjects divided into 3 groups: group 1, 15 healthy controls; group 2, 30 subjects with Sjögren syndrome (SS); and group 3, 30 subjects with Meibomian gland dysfunction (MGD). METHODS A clinical assessment was carried out and impression cytologic specimens were processed for immunoperoxidase staining for MMP9 and TG2 and real-time polymerase chain reaction analyses were carried out for MMP9, TG2, interleukin-6, interferon-γ, B-cell lymphoma 2, and caspase 3. To study MMP9 and TG2 expression after anti-inflammatory treatment, patients were divided into 2 subgroups, one treated with saline and the other treated with saline plus topical corticosteroid eye drops (0.5% loteprednol etabonate) 4 times daily for 15 days. For statistical analysis, Student t test, Mann-Whitney U test, and Spearman's correlation coefficient were used as appropriate. MAIN OUTCOME MEASURES Conjunctival expression of MMP9 and TG2. RESULTS MMP9 and TG2 expression were higher in both patient groups than in controls (P < 0.0001). Group 2 patients showed higher expression than group 3 (P < 0.0001). The Spearman's correlation coefficient showed in group 2 a positive correlation between MMP9 and TG2 expression (ρ = 0.437; P = 0.01), but no correlation in group 3 (ρ = 0.143; P = 0.45). Corticosteroid treatment significantly reduced MMP9 and TG2 expression in both groups, ameliorating symptoms and signs. A much higher percentage reduction was observed in SS. CONCLUSIONS The pathogenic mechanisms of the 2 forms of dry eye give an account for the different MMP9 and TG2 expressions in the 2 groups of patients. The higher expression in SS is determined by the direct autoimmune insult to the ocular surface epithelia, whereas in MGD patients, with an epithelial damage due to an unbalanced tear secretion, the molecules expression is significantly lower, although higher than in controls. The corticosteroid treatment induced a reduction of both molecules, although higher in SS than in MGD, because of its direct inhibitory effect on inflammation.
Collapse
Affiliation(s)
- Pasquale Aragona
- Department of Experimental Medical-Surgical Specialties, Regional Referral Center for Ocular Surface Diseases, University of Messina, Messina, Italy.
| | | | - Laura Rania
- Department of Experimental Medical-Surgical Specialties, Regional Referral Center for Ocular Surface Diseases, University of Messina, Messina, Italy
| | - Elisa Postorino
- Department of Experimental Medical-Surgical Specialties, Regional Referral Center for Ocular Surface Diseases, University of Messina, Messina, Italy
| | - Margherita Serena Sommario
- Department of Experimental Medical-Surgical Specialties, Regional Referral Center for Ocular Surface Diseases, University of Messina, Messina, Italy
| | - Anna Maria Roszkowska
- Department of Experimental Medical-Surgical Specialties, Regional Referral Center for Ocular Surface Diseases, University of Messina, Messina, Italy
| | | | - Antonina Pisani
- Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Domenico Puzzolo
- Department of Biomedical Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
37
|
Xu X, Wells A, Padilla MT, Kato K, Kim KC, Lin Y. A signaling pathway consisting of miR-551b, catalase and MUC1 contributes to acquired apoptosis resistance and chemoresistance. Carcinogenesis 2014; 35:2457-66. [PMID: 25085901 DOI: 10.1093/carcin/bgu159] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Acquired chemoresistance is a major challenge in cancer therapy. While the oncoprotein Mucin-1 (MUC1) performs multiple roles in the development of diverse human tumors, whether MUC1 is involved in acquired chemoresistance has not been determined. Using an acquired chemoresistance lung cancer cell model, we show that MUC1 expression was substantially increased in cells with acquired apoptosis resistance (AR). Knockdown of MUC1 expression effectively increased the sensitivity of these cells to the apoptotic cytotoxicity of anticancer therapeutics, suggesting that MUC1 contributes to acquired chemoresistance. Decreased catalase expression and increased cellular reactive oxygen species (ROS) accumulation were found to be associated with MUC1 overexpression. Scavenging ROS with butylated hydroxyanisole or supplying exogenous catalase dramatically suppressed MUC1 expression through destabilizing MUC1 protein, suggesting that reduced catalase expression mediated ROS accumulation is accounted for MUC1 overexpression. Further, we found that increased miR-551b expression in the AR cells inhibited the expression of catalase and potentiated ROS accumulation and MUC1 expression. Finally, by manipulating MUC1 expression, we found that MUC1 promotes EGFR-mediated activation of the cell survival cascade involving Akt/c-FLIP/COX-2 in order to protect cancer cells from responding to anticancer agents. Thus, our results establish a pathway consisting of miR-551b/catalase/ROS that results in MUC1 overexpression, and intervention against this pathway could be exploited to overcome acquired chemoresistance.
Collapse
Affiliation(s)
- Xiuling Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive, Albuquerque, NM 87108, USA and Department of Physiology & Lung Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| | - Alexandria Wells
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive, Albuquerque, NM 87108, USA and Department of Physiology & Lung Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| | - Mabel T Padilla
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive, Albuquerque, NM 87108, USA and Department of Physiology & Lung Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| | - Kosuke Kato
- Department of Physiology & Lung Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| | - Kwang Chul Kim
- Department of Physiology & Lung Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest Drive, Albuquerque, NM 87108, USA and Department of Physiology & Lung Center, Temple University School of Medicine, 3420 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
38
|
Eckert RL, Kaartinen MT, Nurminskaya M, Belkin AM, Colak G, Johnson GVW, Mehta K. Transglutaminase regulation of cell function. Physiol Rev 2014; 94:383-417. [PMID: 24692352 DOI: 10.1152/physrev.00019.2013] [Citation(s) in RCA: 330] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transglutaminases (TGs) are multifunctional proteins having enzymatic and scaffolding functions that participate in regulation of cell fate in a wide range of cellular systems and are implicated to have roles in development of disease. This review highlights the mechanism of action of these proteins with respect to their structure, impact on cell differentiation and survival, role in cancer development and progression, and function in signal transduction. We also discuss the mechanisms whereby TG level is controlled and how TGs control downstream targets. The studies described herein begin to clarify the physiological roles of TGs in both normal biology and disease states.
Collapse
|
39
|
Cheng P, Dai W, Wang F, Lu J, Shen M, Chen K, Li J, Zhang Y, Wang C, Yang J, Zhu R, Zhang H, Zheng Y, Guo CY, Xu L. Ethyl pyruvate inhibits proliferation and induces apoptosis of hepatocellular carcinoma via regulation of the HMGB1-RAGE and AKT pathways. Biochem Biophys Res Commun 2014; 443:1162-1168. [PMID: 24361892 DOI: 10.1016/j.bbrc.2013.12.064] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 12/11/2013] [Indexed: 01/10/2023]
Abstract
Ethyl pyruvate (EP) was recently identified as a stable lipophilic derivative of pyruvic acid with significant antineoplastic activities. The high mobility group box-B1 (HMGB1)-receptor for advanced glycation end-products (RAGE) and the protein kinase B (Akt) pathways play a crucial role in tumorigenesis and development of many malignant tumors. We tried to observe the effects of ethyl pyruvate on liver cancer growth and explored its effects in hepatocellular carcinoma model. In this study, three hepatocellular carcinoma cell lines were treated with ethyl pyruvate. An MTT colorimetric assay was used to assess the effects of EP on cell proliferation. Flow cytometry and TUNEL assays were used to analyze apoptosis. Real-time PCR, Western blotting and immunofluorescence demonstrated ethyl pyruvate reduced the HMGB1-RAGE and AKT pathways. The results of hepatoma orthotopic tumor model verified the antitumor effects of ethyl pyruvate in vivo. EP could induce apoptosis and slow the growth of liver cancer. Moreover, EP decreased the expression of HMGB1, RAGE, p-AKT and matrix metallopeptidase-9 (MMP9) and increased the Bax/Bcl-2 ratio. In conclusion, this study demonstrates that ethyl pyruvate induces apoptosis and cell-cycle arrest in G phase in hepatocellular carcinoma cells, plays a critical role in the treatment of cancer.
Collapse
Affiliation(s)
- Ping Cheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Miao Shen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Chengfen Wang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Jing Yang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Rong Zhu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Huawei Zhang
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Yuanyuan Zheng
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China
| | - Chuan-Yong Guo
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China.
| | - Ling Xu
- Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
40
|
Freitas DP, Teixeira CA, Santos-Silva F, Vasconcelos MH, Almeida GM. Therapy-induced enrichment of putative lung cancer stem-like cells. Int J Cancer 2013; 134:1270-8. [PMID: 24105655 DOI: 10.1002/ijc.28478] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 08/29/2013] [Indexed: 12/18/2022]
Abstract
Tumour drug resistance is a major issue in the management of lung cancer patients as almost all lung tumours are either intrinsically resistant or quickly develop acquired resistance to chemotherapeutic drugs. Cancer drug resistance has recently been linked, at least in part, to the existence of cancer stem-like cells (CSLCs), a small sub-population of cells within the tumour that possess stem-like properties. CSLCs are often isolated by fluorescence activated cell sorting (FACS) according to the expression of certain stem-like cell membrane markers. Conflicting results regarding the specificity of particular stem cell surface markers for isolating CSLCs have, however, been recently reported. Therefore, alternative strategies enabling the identification and study of CSLCs should be considered, particularly in tumour types where appropriate stem cell markers are not well established and validated, like in lung cancer. In this article, we review data indicating therapy-selection as a valid approach for putative lung CSLCs enrichment. We believe that this strategy would be determinant for correctly assessing and characterising the sub-populations of CSLCs that are able to survive chemo or radiotherapy regimens and, at the same time, also have the ability to recapitulate and sustain tumour growth. Using therapy-induced enrichment of CSLCs may, therefore, prove to be an extremely useful method for studying CSLCs and provide new clues regarding potential therapeutic targets for their efficient elimination, which will undoubtedly play a decisive role in improving lung cancer patients' survival.
Collapse
Affiliation(s)
- Daniela P Freitas
- Cancer Drug Resistance Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Expression Regulation in Cancer Group, Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | | | | | | | | |
Collapse
|
41
|
Abstract
Cell-matrix adhesion is a fundamental biological process that governs survival, migration, and proliferation of living eukaryotic cells. Paxillin is an important central player in a network of adhesome proteins that form focal adhesion complexes. Phosphorylation of tyrosine and serine residues in paxillin is critical for the coordinated sequential recruitment of other adaptor and kinase proteins to adhesion complexes. Recently, the phosphorylation of serine178 in paxillin has been shown to be vital for epithelial cell adhesion and migration. In vivo and in vitro evidence have shown that transglutaminase (TG)-2 positively regulates this phosphorylation. Here, we propose three possible mechanisms that may explain these observations. First, TG-2 itself may be an adhesome member directly interacting with paxillin in a non-covalent way. Second, TG-2 may cross link a mitogen-activated protein kinase kinase kinase (MAP3K), which eventually activates c-Jun N-terminal kinase (JNK), and the latter phosphorylates paxillin. Lastly, TG-2 may have intrinsic kinase activity that phosphorylates paxillin. Future studies investigating these hypotheses on TG-2-paxillin relationships are necessary in order to address this fundamental process in cell matrix adhesion signaling.
Collapse
Affiliation(s)
- Evelyn Png
- Ocular Surface Research Group; Singapore Eye Research Institute; Singapore
| | - Louis Tong
- Ocular Surface Research Group; Singapore Eye Research Institute; Singapore; Department of Cornea and External Eye Disease; Singapore National Eye Center; Singapore; Office of Clinical Science; Duke-NUS Graduate Medical School; Singapore; Department of Ophthalmology; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| |
Collapse
|
42
|
GONZALEZ-VILLASANA VIANEY, GUTIÉRREZ-PUENTE YOLANDA, TARI ANAM. Cyclooxygenase-2 utilizes Jun N-terminal kinases to induce invasion, but not tamoxifen resistance, in MCF-7 breast cancer cells. Oncol Rep 2013; 30:1506-10. [DOI: 10.3892/or.2013.2549] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/29/2013] [Indexed: 11/05/2022] Open
|
43
|
Flusberg DA, Sorger PK. Modulating cell-to-cell variability and sensitivity to death ligands by co-drugging. Phys Biol 2013; 10:035002. [PMID: 23735516 DOI: 10.1088/1478-3975/10/3/035002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) holds promise as an anti-cancer therapeutic but efficiently induces apoptosis in only a subset of tumor cell lines. Moreover, even in clonal populations of responsive lines, only a fraction of cells dies in response to TRAIL and individual cells exhibit cell-to-cell variability in the timing of cell death. Fractional killing in these cell populations appears to arise not from genetic differences among cells but rather from differences in gene expression states, fluctuations in protein levels and the extent to which TRAIL-induced death or survival pathways become activated. In this study, we ask how cell-to-cell variability manifests in cell types with different sensitivities to TRAIL, as well as how it changes when cells are exposed to combinations of drugs. We show that individual cells that survive treatment with TRAIL can regenerate the sensitivity and death-time distribution of the parental population, demonstrating that fractional killing is a stable property of cell populations. We also show that cell-to-cell variability in the timing and probability of apoptosis in response to treatment can be tuned using combinations of drugs that together increase apoptotic sensitivity compared to treatment with one drug alone. In the case of TRAIL, modulation of cell-to-cell variability by co-drugging appears to involve a reduction in the threshold for mitochondrial outer membrane permeabilization.
Collapse
Affiliation(s)
- Deborah A Flusberg
- Center for Cell Decision Processes, Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | | |
Collapse
|
44
|
Flusberg DA, Roux J, Spencer SL, Sorger PK. Cells surviving fractional killing by TRAIL exhibit transient but sustainable resistance and inflammatory phenotypes. Mol Biol Cell 2013; 24:2186-200. [PMID: 23699397 PMCID: PMC3708725 DOI: 10.1091/mbc.e12-10-0737] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Cells that survive fractional killing by TRAIL or FasR agonists enter a state of resistance accompanied by inflammatory phenotypes. This state is transient, decaying over the course of several days, but can be sustained by periodic TRAIL treatments. This finding has implications for optimal dosing strategies of extrinsic cell death agents. When clonal populations of human cells are exposed to apoptosis-inducing agents, some cells die and others survive. This fractional killing arises not from mutation but from preexisting, stochastic differences in the levels and activities of proteins regulating apoptosis. Here we examine the properties of cells that survive treatment with agonists of two distinct death receptors, tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) and anti-FasR antibodies. We find that “survivor” cells are highly resistant to a second ligand dose applied 1 d later. Resistance is reversible, resetting after several days of culture in the absence of death ligand. “Reset” cells appear identical to drug-naive cells with respect to death ligand sensitivity and gene expression profiles. TRAIL survivors are cross-resistant to activators of FasR and vice versa and exhibit an NF-κB–dependent inflammatory phenotype. Remarkably, reversible resistance is induced in the absence of cell death when caspase inhibitors are present and can be sustained for 1 wk or more, also without cell death, by periodic ligand exposure. Thus stochastic differences in cell state can have sustained consequences for sensitivity to prodeath ligands and acquisition of proinflammatory phenotypes. The important role played by periodicity in TRAIL exposure for induction of opposing apoptosis and survival mechanisms has implications for the design of optimal therapeutic agents and protocols.
Collapse
Affiliation(s)
- Deborah A Flusberg
- Center for Cell Decision Processes, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
45
|
Wang Q, Chen W, Xu X, Li B, He W, Padilla MT, Jang JH, Nyunoya T, Amin S, Wang X, Lin Y. RIP1 potentiates BPDE-induced transformation in human bronchial epithelial cells through catalase-mediated suppression of excessive reactive oxygen species. Carcinogenesis 2013; 34:2119-28. [PMID: 23633517 DOI: 10.1093/carcin/bgt143] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cell survival signaling is important for the malignant phenotypes of cancer cells. Although the role of receptor-interacting protein 1 (RIP1) in cell survival signaling is well documented, whether RIP1 is directly involved in cancer development has never been studied. In this report, we found that RIP1 expression is substantially increased in human non-small cell lung cancer and mouse lung tumor tissues. RIP1 expression was remarkably increased in cigarette smoke-exposed mouse lung. In human bronchial epithelial cells (HBECs), RIP1 was significantly induced by cigarette smoke extract or benzo[a]pyrene diol epoxide (BPDE), the active form of the tobacco-specific carcinogen benzo(a)pyrene. In RIP1 knockdown HBECs, BPDE-induced cytotoxicity was significantly increased, which was associated with induction of cellular reactive oxygen species (ROS) and activation of mitogen-activated protein kinases (MAPKs), including c-jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK) and p38. Scavenging ROS suppressed BPDE-induced MAPK activation and inhibiting ROS or MAPKs substantially blocked BPDE-induced cytotoxicity, suggesting ROS-mediated MAPK activation is involved in BPDE-induced cell death. The ROS-reducing enzyme catalase is destabilized in an ERK- and JNK-dependent manner in RIP1 knockdown HBECs and application of catalase effectively blocked BPDE-induced ROS accumulation and cytotoxicity. Importantly, BPDE-induced transformation of HBECs was significantly reduced when RIP1 expression was suppressed. Altogether, these results strongly suggest an oncogenic role for RIP1, which promotes malignant transformation through protecting DNA-damaged cells against carcinogen-induced cytotoxicity associated with excessive ROS production.
Collapse
Affiliation(s)
- Qiong Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rao-Bindal K, Rao CK, Yu L, Kleinerman ES. Expression of c-FLIP in pulmonary metastases in osteosarcoma patients and human xenografts. Pediatr Blood Cancer 2013; 60:575-9. [PMID: 23255321 PMCID: PMC3883385 DOI: 10.1002/pbc.24412] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 11/01/2012] [Indexed: 01/09/2023]
Abstract
OBJECTIVE(S) We have previously shown that Fas expression inversely correlates with the metastatic potential of osteosarcoma (OS) to the lung. FasL is constitutively expressed in the lung microenvironment and eliminates Fas(+) OS cells leaving Fas(-) cells to form metastases. Absence of FasL in the lung epithelium or blocking the Fas-signaling pathway interfered with this clearance mechanism allowing Fas(+) cells to remain and form lung metastases. We also demonstrated that while the majority of patient OS lung metastases were Fas(-), 10-20% of the lesions contained Fas(+) cells, suggesting that these cells were not sensitive to FasL-induced apoptosis. The expression of c-FLIP, an inhibitor of the Fas pathway, has been associated with tumor development, progression, and resistance to chemotherapy. We therefore evaluated the expression of c-FLIP in OS patient tumor specimens and human xenograft lung metastases. METHODS OS patient tissues, which included both primary and metastatic lesions, were evaluated for the expression of c-FLIP. In addition, tumors from human OS xenografts were examined for c-FLIP expression. RESULTS c-FLIP expression was significantly higher in the lung metastases than in the primary tumors. CONCLUSION(S) c-FLIP may play an important role in the metastatic potential of OS to the lung. Inhibition of c-FLIP may be a future therapeutic target.
Collapse
Affiliation(s)
- Krithi Rao-Bindal
- Division of Pediatrics, The University of Texas, MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
47
|
He W, Wang Q, Xu J, Xu X, Padilla MT, Ren G, Gou X, Lin Y. Attenuation of TNFSF10/TRAIL-induced apoptosis by an autophagic survival pathway involving TRAF2- and RIPK1/RIP1-mediated MAPK8/JNK activation. Autophagy 2012; 8:1811-21. [PMID: 23051914 PMCID: PMC3541290 DOI: 10.4161/auto.22145] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although it is known that tumor necrosis factor-related apoptosis-inducing ligand (TNFSF10/TRAIL) induces autophagy, the mechanism by which autophagy is activated by TNFSF10 is still elusive. In this report, we show evidence that TRAF2- and RIPK1-mediated MAPK8/JNK activation is required for TNFSF10-induced cytoprotective autophagy. TNFSF10 activated autophagy rapidly in cancer cell lines derived from lung, bladder and prostate tumors. Blocking autophagy with either pharmacological inhibitors or siRNAs targeting the key autophagy factors BECN1/Beclin 1 or ATG7 effectively increased TNFSF10-induced apoptotic cytotoxicity, substantiating a cytoprotective role for TNFSF10-induced autophagy. Blocking MAPK8 but not NFκB effectively blocked autophagy, suggesting that MAPK8 is the main pathway for TNFSF10-induced autophagy. In addition, blocking MAPK8 effectively inhibited degradation of BCL2L1/Bcl-xL and reduction of the autophagy-suppressing BCL2L1-BECN1complex. Knockdown of TRAF2 or RIPK1 effectively suppressed TNFSF10-induced MAPK8 activation and autophagy. Furthermore, suppressing autophagy inhibited expression of antiapoptosis factors BIRC2/cIAP1, BIRC3/cIAP2, XIAP and CFLAR/c-FLIP and increased the formation of TNFSF10-induced death-inducing signaling complex (DISC). These results reveal a critical role for the MAPK8 activation pathway through TRAF2 and RIPK1 for TNFSF10-induced autophagy that blunts apoptosis in cancer cells. Thus, suppression of MAPK8-mediated autophagy could be utilized for sensitizing cancer cells to therapy with TNFSF10.
Collapse
Affiliation(s)
- Weiyang He
- Department of Urology; The First Affiliated Hospital of Chongqing Medical University; Chongqing, China
- Molecular Oncology and Epigenetics Laboratory; The First Affiliated Hospital of Chongqing Medical University; Chongqing, China
- Molecular Biology and Lung Cancer Program; Lovelace Respiratory Research Institute; Albuquerque, NM USA
| | - Qiong Wang
- Molecular Biology and Lung Cancer Program; Lovelace Respiratory Research Institute; Albuquerque, NM USA
| | - Jennings Xu
- Molecular Biology and Lung Cancer Program; Lovelace Respiratory Research Institute; Albuquerque, NM USA
| | - Xiuling Xu
- Molecular Biology and Lung Cancer Program; Lovelace Respiratory Research Institute; Albuquerque, NM USA
| | - Mabel T. Padilla
- Molecular Biology and Lung Cancer Program; Lovelace Respiratory Research Institute; Albuquerque, NM USA
| | - Guosheng Ren
- Molecular Oncology and Epigenetics Laboratory; The First Affiliated Hospital of Chongqing Medical University; Chongqing, China
| | - Xin Gou
- Department of Urology; The First Affiliated Hospital of Chongqing Medical University; Chongqing, China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program; Lovelace Respiratory Research Institute; Albuquerque, NM USA
| |
Collapse
|
48
|
Liu YP, Yang CJ, Huang MS, Yeh CT, Wu ATH, Lee YC, Lai TC, Lee CH, Hsiao YW, Lu J, Shen CN, Lu PJ, Hsiao M. Cisplatin selects for multidrug-resistant CD133+ cells in lung adenocarcinoma by activating Notch signaling. Cancer Res 2012; 73:406-16. [PMID: 23135908 DOI: 10.1158/0008-5472.can-12-1733] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Platinum-based chemotherapy is the first-line treatment for non-small cell lung cancer, but recurrence occurs in most patients. Recent evidence suggests that CD133(+) cells are the cause of drug resistance and tumor recurrence. However, the correlation between chemotherapy and regulation of CD133(+) cells has not been investigated methodically. In this study, we revealed that CD133(+) lung cancer cells labeled by a human CD133 promoter-driven GFP reporter exhibited drug resistance and stem cell characteristics. Treatment of H460 and H661 cell lines with low-dose cisplatin (IC(20)) was sufficient to enrich CD133(+) cells, to induce DNA damage responses, and to upregulate ABCG2 and ABCB1 expression, which therefore increased the cross-resistance to doxorubicin and paclitaxel. This cisplatin-induced enrichment of CD133(+) cells was mediated through Notch signaling as judged by increased levels of cleaved Notch1 (NICD1). Pretreatment with the γ-secretase inhibitor, N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butyl ester (DAPT), or Notch1 short hairpin RNAs (shRNA) remarkably reduced the cisplatin-induced enrichment of CD133(+) cells and increased the sensitivity to doxorubicin and paclitaxel. Ectopic expression of NICD1 reversed the action of DAPT on drug sensitivity. Immunohistochemistry showed that CD133(+) cells were significantly increased in the relapsed tumors in three of six patients with lung cancer who have received cisplatin treatment. A similar effect was observed in animal experiments as cisplatin treatment increased Notch1 cleavage and the ratio of CD133(+) cells in engrafted tumors. Intratumoral injection of DAPT with cisplatin treatment significantly reduced CD133(+) cell number. Together, our results showed that cisplatin induces the enrichment of CD133(+) cells, leading to multidrug resistance by the activation of Notch signaling.
Collapse
Affiliation(s)
- Yu-Peng Liu
- Institute of Clinical Medicine, National Cheng Kung University, Medical College, Tainan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jia LT, Chen SY, Yang AG. Cancer gene therapy targeting cellular apoptosis machinery. Cancer Treat Rev 2012; 38:868-876. [PMID: 22800735 DOI: 10.1016/j.ctrv.2012.06.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/21/2012] [Accepted: 06/22/2012] [Indexed: 01/14/2023]
Abstract
The unraveling of cellular apoptosis machinery provides novel targets for cancer treatment, and gene therapy targeting this suicidal system has been corroborated to cause inflammation-free autonomous elimination of neoplastic cells. The apoptotic machinery can be targeted by introduction of a gene encoding an inducer, mediator or executioner of apoptotic cell death or by inhibition of anti-apoptotic gene expression. Strategies targeting cancer cells, which are achieved by selective gene delivery, specific gene expression or secretion of target proteins via genetic modification of autologous cells, dictate the outcome of apoptosis-based cancer gene therapy. Despite so far limited clinical success, gene therapy targeting the apoptotic machinery has great potential to benefit patients with threatening malignancies provided the availability of efficient and specific gene delivery and administration systems.
Collapse
Affiliation(s)
- Lin-Tao Jia
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, China.
| | | | | |
Collapse
|
50
|
Yang L, Xu L. GPR56 in cancer progression: current status and future perspective. Future Oncol 2012; 8:431-40. [PMID: 22515446 DOI: 10.2217/fon.12.27] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell adhesion is a critical process during cancer progression and is mediated by transmembrane receptors. Recently, GPR56, a member of the adhesion family of G protein-coupled receptors, was established as a new type of adhesion receptor that binds to extracellular matrix proteins and shown to play inhibitory roles in melanoma progression. Further studies revealed that the extracellular portion and the seven transmembrane domains of GPR56 function antagonistically to regulate VEGF production and angiogenesis via a signaling pathway mediated by PKCα. Tissue transglutaminase was identified as the first extracellular matrix protein that binds to GPR56. It is a crosslinking enzyme in the extracellular matrix but is also expressed in the cytosol. Tissue transglutaminase plays pleiotropic roles in cancer progression. Whether and how it might mediate GPR56-regulated cancer progression awaits further investigation.
Collapse
Affiliation(s)
- Liquan Yang
- Department of Biomedical Genetics, Department of Dermatology, James P Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | |
Collapse
|