1
|
Philipp LM, Yesilyurt UU, Surrow A, Künstner A, Mehdorn AS, Hauser C, Gundlach JP, Will O, Hoffmann P, Stahmer L, Franzenburg S, Knaack H, Schumacher U, Busch H, Sebens S. Epithelial and Mesenchymal-like Pancreatic Cancer Cells Exhibit Different Stem Cell Phenotypes Associated with Different Metastatic Propensities. Cancers (Basel) 2024; 16:686. [PMID: 38398077 PMCID: PMC10886860 DOI: 10.3390/cancers16040686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is mostly diagnosed at advanced or even metastasized stages, limiting the prognoses of patients. Metastasis requires high tumor cell plasticity, implying phenotypic switching in response to changing environments. Here, epithelial-mesenchymal transition (EMT), being associated with an increase in cancer stem cell (CSC) properties, and its reversion are important. Since it is poorly understood whether different CSC phenotypes exist along the EMT axis and how these impact malignancy-associated properties, we aimed to characterize CSC populations of epithelial and mesenchymal-like PDAC cells. Single-cell cloning revealed CSC (Holoclone) and non-CSC (Paraclone) clones from the PDAC cell lines Panc1 and Panc89. The Panc1 Holoclone cells showed a mesenchymal-like phenotype, dominated by a high expression of the stemness marker Nestin, while the Panc89 Holoclone cells exhibited a SOX2-dominated epithelial phenotype. The Panc89 Holoclone cells showed enhanced cell growth and a self-renewal capacity but slow cluster-like invasion. Contrarily, the Panc1 Holoclone cells showed slower cell growth and self-renewal ability but were highly invasive. Moreover, cell variants differentially responded to chemotherapy. In vivo, the Panc1 and Panc89 cell variants significantly differed regarding the number and size of metastases, as well as organ manifestation, leading to different survival outcomes. Overall, these data support the existence of different CSC phenotypes along the EMT axis in PDAC, manifesting different metastatic propensities.
Collapse
Affiliation(s)
- Lisa-Marie Philipp
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Umut-Ulas Yesilyurt
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Arne Surrow
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Anne-Sophie Mehdorn
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Charlotte Hauser
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Jan-Paul Gundlach
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Olga Will
- Molecular Imaging North Competence Center, Clinic of Radiology and Neuroradiology, Kiel University, UKSH, Campus Kiel, 24118 Kiel, Germany
| | - Patrick Hoffmann
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Lea Stahmer
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Kiel University, 24118 Kiel, Germany
| | - Hendrike Knaack
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
- Academic Affairs Office, Hannover Medical School, 30625 Hannover, Germany
| | - Udo Schumacher
- Department of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| |
Collapse
|
2
|
Liu Q, Ma Z, Cao Q, Zhao H, Guo Y, Liu T, Li J. Perineural invasion-associated biomarkers for tumor development. Biomed Pharmacother 2022; 155:113691. [PMID: 36095958 DOI: 10.1016/j.biopha.2022.113691] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Perineural invasion (PNI) is the process of neoplastic invasion of peripheral nerves and is considered to be the fifth mode of cancer metastasis. PNI has been detected in head and neck tumors and pancreatic, prostate, bile duct, gastric, and colorectal cancers. It leads to poor prognostic outcomes and high local recurrence rates. Despite the increasing number of studies on PNI, targeted therapeutic modalities have not been proposed. The identification of PNI-related biomarkers would facilitate the non-invasive and early diagnosis of cancers, the establishment of prognostic panels, and the development of targeted therapeutic approaches. In this review, we compile information on the molecular mediators involved in PNI-associated cancers. The expression and prognostic significance of molecular mediators and their receptors in PNI-associated cancers are analyzed, and the possible mechanisms of action of these mediators in PNI are explored, as well as the association of cells in the microenvironment where PNI occurs.
Collapse
Affiliation(s)
- Qi Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhiming Ma
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Qian Cao
- Department of Education, The Second Hospital of Jilin University, Changchun 130041, China
| | - Hongyu Zhao
- Gastroenterology and Center of Digestive Endoscopy, The Second Hospital of Jilin University, Changchun 130041, China
| | - Yu Guo
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Jiannan Li
- Department of General Surgery, The Second Hospital of Jilin University, Changchun 130041, China.
| |
Collapse
|
3
|
Wang JW, Wang HL, Liu Q, Hu K, Yuan Q, Huang SK, Wan JH. L1CAM expression in either metastatic brain lesion or peripheral blood is correlated with peripheral platelet count in patients with brain metastases from lung cancer. Front Oncol 2022; 12:990762. [DOI: 10.3389/fonc.2022.990762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundSystemic immune-inflammation states across the heterogeneous population of brain metastases from lung cancer are very important, especially in the context of complex brain-immune bidirectional communication. Previous studies from our team and others have shown that the L1 cell adhesion molecule (L1CAM) is deeply involved in the aggressive phenotype, immunosuppressive tumor microenvironment (TME), and metastasis during multiple malignancies, which may lead to an unfavorable outcome. However, little is known about the relationship between the L1CAM expression and the systemic immune-inflammation macroenvironment beyond the TME in brain metastases from lung cancer.MethodsTwo cohorts of patients with brain metastases from lung cancer admitted to the National Cancer Center, Cancer Hospital of Chinese Academy of Medical Sciences, were studied in the present research. The L1CAM expression in cranial metastatic lesions by immunohistochemistry was explored in patients treated with neurosurgical resection, whereas the L1CAM expression in peripheral blood by ELISA was tested in patients treated with non-surgical antitumor management. Furthermore, based on peripheral blood cell counts in the CBC test, six systemic immune-inflammation biomarkers [neutrophil count, lymphocyte count, platelet count, systemic immune-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio] were calculated. Then, the relationship between the L1CAM expression and these systemic immune-inflammation biomarkers was analyzed. In addition, these systemic immune-inflammation biomarkers were also used to compare the systemic immune-inflammation states in two cohorts of patients with brain metastases from lung cancer.ResultsPositive L1CAM expressions in the metastatic brain lesions were accompanied with significantly increased peripheral platelet counts in patients treated with neurosurgical tumor resection (P < 0.05). Similarly, in patients treated with non-surgical antitumor management, L1CAM expressions in the peripheral blood were positively correlated with peripheral platelet counts (P < 0.05). In addition, patients prepared for neurosurgical tumor resection were presented with poorer systemic immune-inflammation states in comparison with the one with non-surgical antitumor management, which was characterized by a significant increase in peripheral neutrophil counts (P < 0.01), SII (P < 0.05), and NLR (P < 0.05) levels.ConclusionThe L1CAM expression in either the metastatic brain lesion or peripheral blood is positively correlated with the peripheral platelet count in patients with brain metastases from lung cancer. In addition, brain metastases that are prepared for neurosurgical tumor resection show poor systemic immune-inflammation states.
Collapse
|
4
|
Zhang J, Guo Z, Xie Q, Zhong C, Gao X, Yang Q. Tryptophan hydroxylase 1 drives glioma progression by modulating the serotonin/L1CAM/NF-κB signaling pathway. BMC Cancer 2022; 22:457. [PMID: 35473609 PMCID: PMC9044587 DOI: 10.1186/s12885-022-09569-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/13/2022] [Indexed: 12/31/2022] Open
Abstract
Background Glioma is one of the main causes of cancer-related mortality worldwide and is associated with high heterogeneity. However, the key players determining the fate of glioma remain obscure. In the present study, we shed light on tumor metabolism and aimed to investigate the role of tryptophan hydroxylase 1 (TPH-1) in the advancement of glioma. Method Herein, the levels of TPH-1 expression in glioma tissues were evaluated using The Cancer Genome Atlas (TCGA) database. Further, the proliferative characteristics and migration ability of TPH-1 overexpressing LN229/T98G cells were evaluated. Additionally, we performed a cytotoxicity analysis using temozolomide (TMZ) in these cells. We also examined the tumor growth and survival time in a mouse model of glioma treated with chemotherapeutic agents and a TPH-1 inhibitor. Results The results of both clinical and experimental data showed that excess TPH-1 expression resulted in sustained glioma progression and a dismal overall survival in these patients. Mechanistically, TPH-1 increased the production of serotonin in glioma cells. The elevated serotonin levels then augmented the NF-κB signaling pathway through the upregulation of the L1-cell adhesion molecule (L1CAM), thereby contributing to cellular proliferation, invasive migration, and drug resistance. In vivo experiments demonstrated potent antitumor effects, which benefited further from the synergistic combination of TMZ and LX-1031. Conclusion Taken together, these data suggested that TPH-1 facilitated cellular proliferation, migration, and chemoresistance in glioma through the serotonin/L1CAM/NF-κB pathway. By demonstrating the link of amino acid metabolic enzymes with tumor development, our findings may provide a potentially viable target for therapeutic manipulation aimed at eradicating glioma. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09569-2.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Neurosurgery, Ya' an people's Hospital, Ya' an, 625000, People's Republic of China
| | - Zhangchao Guo
- Department of Neurosurgery, Ya' an people's Hospital, Ya' an, 625000, People's Republic of China. .,Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China.
| | - Qiangli Xie
- Department of Cardiology, Chengdu Qingbaijiang District People's Hospital, Chengdu, People's Republic of China
| | - Chuanhong Zhong
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Xiangyu Gao
- Pediatrics of Ya' an people's Hospital, Ya' an, People's Republic of China
| | - Qiumei Yang
- Department of Geriatrics, Luzhou people's Hospital, Luzhou, People's Republic of China
| |
Collapse
|
5
|
Hu J, Lin SL, Schachner M. A fragment of cell adhesion molecule L1 reduces amyloid-β plaques in a mouse model of Alzheimer's disease. Cell Death Dis 2022; 13:48. [PMID: 35013124 PMCID: PMC8748658 DOI: 10.1038/s41419-021-04348-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 10/02/2021] [Accepted: 10/13/2021] [Indexed: 02/05/2023]
Abstract
Deposition of amyloid-β (Aβ) in the brain is one of the important histopathological features of Alzheimer's disease (AD). Previously, we reported a correlation between cell adhesion molecule L1 (L1) expression and the occurrence of AD, but its relationship was unclear. Here, we report that the expression of L1 and a 70 kDa cleavage product of L1 (L1-70) was reduced in the hippocampus of AD (APPswe) mice. Interestingly, upregulation of L1-70 expression in the hippocampus of 18-month-old APPswe mice, by parabiosis involving the joining of the circulatory system of an 18-month-old APPswe mouse with a 2-month-old wild-type C57BL/6 mouse, reduced amyloid plaque deposition. Furthermore, the reduction was accompanied by the appearance of a high number of activated microglia. Mechanistically, we observed that L1-70 could combine with topoisomerase 1 (Top1) to form a complex, L1-70/Top1, that was able to regulate expression of macrophage migration inhibitory factor (MIF), resulting in the activation of microglia and reduction of Aβ plaques. Also, transforming growth factor β1 (TGFβ-1) transferred from the blood of young wild-type C57BL/6 mice to the aged AD mice, was identified as a circulating factor that induces full-length L1 and L1-70 expression. All together, these findings suggest that L1-70 contributes to the clearance of Aβ in AD, thereby adding a novel perspective in understanding AD pathogenesis.
Collapse
Affiliation(s)
- Junkai Hu
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
| | - Stanley Li Lin
- Deaprtment of Cell Biology, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, Shantou, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| |
Collapse
|
6
|
Arnold S, Kortland J, Maltseva DV, Nersisyan SA, Samatov TR, Lezius S, Tonevitsky AG, Milde-Langosch K, Wicklein D, Schumacher U, Stürken C. Fra-2 overexpression upregulates pro-metastatic cell-adhesion molecules, promotes pulmonary metastasis, and reduces survival in a spontaneous xenograft model of human breast cancer. J Cancer Res Clin Oncol 2021; 148:1525-1542. [PMID: 34693476 PMCID: PMC9114065 DOI: 10.1007/s00432-021-03812-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/22/2021] [Indexed: 11/28/2022]
Abstract
Purpose The transcription factor Fra-2 affects the invasive potential of breast cancer cells by dysregulating adhesion molecules in vitro. Previous results suggested that it upregulates the expression of E- and P-selectin ligands. Such selectin ligands are important members of the leukocyte adhesion cascade, which govern the adhesion and transmigration of cancer cells into the stroma of the host organ of metastasis. As so far, no in vivo data are available, this study was designed to elucidate the role of Fra-2 expression in a spontaneous breast cancer metastasis xenograft model. Methods The effect of Fra-2 overexpression in two stable Fra-2 overexpressing clones of the human breast cancer cell line MDA MB231 on survival and metastatic load was studied after subcutaneous injection into scid and E- and P-selectin-deficient scid mice. Results Fra-2 overexpression leads to a significantly shorter overall survival and a higher amount of spontaneous lung metastases not only in scid mice, but also in E- and P-deficient mice, indicating that it regulates not only selectin ligands, but also selectin-independent adhesion processes. Conclusion Thus, Fra-2 expression influences the metastatic potential of breast cancer cells by changing the expression of adhesion molecules, resulting in increased adherence to endothelial cells in a breast cancer xenograft model. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03812-2.
Collapse
Affiliation(s)
- Sabrina Arnold
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Jan Kortland
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Diana V Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya Str. 13/4, 117997, Moscow, Russia
| | - Stepan A Nersisyan
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya Str. 13/4, 117997, Moscow, Russia
| | - Timur R Samatov
- Evotec International GmbH, Marie-Curie-Str. 7, 37079, Göttingen, Germany
| | - Susanne Lezius
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Alexander G Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya Str. 13/4, 117997, Moscow, Russia.,Scientific Research Center Bioclinicum, Ugreshskaya Str. 2/85, 115088, Moscow, Russia
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Christine Stürken
- Institute of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| |
Collapse
|
7
|
Wandmacher AM, Mehdorn AS, Sebens S. The Heterogeneity of the Tumor Microenvironment as Essential Determinant of Development, Progression and Therapy Response of Pancreatic Cancer. Cancers (Basel) 2021; 13:4932. [PMID: 34638420 PMCID: PMC8508450 DOI: 10.3390/cancers13194932] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is commonly diagnosed at advanced stages and most anti-cancer therapies have failed to substantially improve prognosis of PDAC patients. As a result, PDAC is still one of the deadliest tumors. Tumor heterogeneity, manifesting at multiple levels, provides a conclusive explanation for divergent survival times and therapy responses of PDAC patients. Besides tumor cell heterogeneity, PDAC is characterized by a pronounced inflammatory stroma comprising various non-neoplastic cells such as myofibroblasts, endothelial cells and different leukocyte populations which enrich in the tumor microenvironment (TME) during pancreatic tumorigenesis. Thus, the stromal compartment also displays a high temporal and spatial heterogeneity accounting for diverse effects on the development, progression and therapy responses of PDAC. Adding to this heterogeneity and the impact of the TME, the microbiome of PDAC patients is considerably altered. Understanding this multi-level heterogeneity and considering it for the development of novel therapeutic concepts might finally improve the dismal situation of PDAC patients. Here, we outline the current knowledge on PDAC cell heterogeneity focusing on different stromal cell populations and outline their impact on PDAC progression and therapy resistance. Based on this information, we propose some novel concepts for treatment of PDAC patients.
Collapse
Affiliation(s)
| | - Anna Maxi Wandmacher
- Department of Internal Medicine II, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3, 24105 Kiel, Germany;
| | - Anne-Sophie Mehdorn
- Department of General, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany;
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Str. 3, Building U30 Entrance 1, 24105 Kiel, Germany
| |
Collapse
|
8
|
Zhao X, Liu S, Chen X, Zhao J, Li F, Zhao Q, Xie T, Huang L, Zhang Z, Qi Y, Yang Y, Zhao S, Zhang Y. L1CAM overexpression promotes tumor progression through recruitment of regulatory T cells in esophageal carcinoma. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0182. [PMID: 33710805 PMCID: PMC8185865 DOI: 10.20892/j.issn.2095-3941.2020.0182] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/20/2020] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE L1 cell adhesion molecule (L1CAM) exhibits oncogenic activity in tumors. However, the link between L1CAM and the tumor microenvironment remains poorly understood in patients with esophageal squamous cell carcinoma (ESCC). In this study, we investigated how L1CAM expression in ESCC affects the oncogenic characteristics of tumor cells and the tumor microenvironment. METHODS Human ESCC samples were collected, and the mRNA and protein levels of L1CAM were examined by real-time PCR and immunohistochemistry. Overexpression and knockdown gene expression assays were used for mechanistic studies. The cell proliferation and cell cycle were measured with CCK-8 assays and flow cytometry. Cell migration and invasion ability were measured with Transwell assays. Multiplex bead-based assays were performed to identity the factors downstream of L1CAM. Xenograft studies were performed in nude mice to evaluate the effects of L1CAM on tumor growth and regulatory T cell (Treg) recruitment. RESULTS L1CAM expression was significantly elevated in ESCC tissues (P < 0.001) and correlated with poorer prognosis (P < 0.05). Ablation of L1CAM in ESCC cells inhibited tumor growth and migration, and increased tumor cell apoptosis (P < 0.05). In the tumor microenvironment, L1CAM expression correlated with Treg infiltration in ESCC by affecting CCL22 secretion. Mechanistically, L1CAM facilitated CCL22 expression by activating the PI3K/Akt/NF-κB signaling pathway. Furthermore, CCL22 promoted Treg recruitment to the tumor site; the Tregs then secreted TGF-β, which in turn promoted L1CAM expression via Smad2/3 in a positive feedback loop. CONCLUSIONS Our findings provide new insight into the mechanism of immune evasion mediated by L1CAM, suggesting that targeting L1CAM-CCL22-TGF-β crosstalk between tumor cells and Tregs may offer a unique means to improve treatment of patients with ESCC.
Collapse
Affiliation(s)
- Xuan Zhao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Shasha Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jianyi Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qitai Zhao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tan Xie
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lan Huang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yu Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yang Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Song Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou 450052, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou 450052, China
| |
Collapse
|
9
|
Liu A, Zhou J, Bi X, Hou G, Li SS, Chen Q, Xu H, Cao X. Aptamer-SH2 superbinder-based targeted therapy for pancreatic ductal adenocarcinoma. Clin Transl Med 2021; 11:e337. [PMID: 33783993 PMCID: PMC7908048 DOI: 10.1002/ctm2.337] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) exhibits the poorest prognosis of all solid tumors with a 5-year survival rate of less than 10% and a median survival of 6 months after diagnosis. Numerous targeted agents have been developed and evaluated to improve the survival benefit in patients with PDAC. Unfortunately, most agents have been proven futile mainly owing to the dense stroma and the sophisticated signaling pathways of PDAC. Here, we show the potent effectiveness of Aptamer-SH2 superbinder-(Arg)9 conjugate on the treatment of PDAC. In this conjugate, DNA aptamer selected against PDAC cell line confers the function of specifically recognizing and binding to the PDAC cells and activated pancreatic stellate cells (PSCs) in stroma; cell penetrating peptide (Arg)9 facilitates the intracellular delivery of fused proteins; SH2 superbinder conducts the drastic blockade of multiple phosphotyrosines (pY)-based signaling pathways in tumor cells. METHODS PDAC-associated pY were reanalyzed by bioinformatics screen. XQ-2d and SH2 superbinder-(Arg)9 were crosslinked with BMH to form XQ-2d-SH2 CM-(Arg)9 conjugate. Immunofluorescence was utilized to assess the potency of the conjugate entering cells. MTT and wound healing assays were performed to evaluate the proliferation or migration of PANC-1 and BxPC-3 cells, respectively. Western blot and Pulldown assays revealed that conjugate influenced several pY-based signaling pathways. Tumor-bearing mice were used to validate XQ-2d-SH2 CM-(Arg)9, which restrained the growth and metastasis of cancer cells. RESULTS XQ-2d-His-SH2 CM-(Arg)9 conjugate restrained proliferation, invasion, and metastasis of PDAC cells with potent efficacy via blocking the activity of several pY-related signaling cascades. XQ-2d-His-SH2 CM-(Arg)9 could eliminate the dense stroma of PDAC and then arrive at tumor tissues. CONCLUSIONS XQ-2d-SH2 CM-(Arg)9 conjugate may efficiently destroy the pancreatic stroma and show potent antitumor efficacy with minimal toxic effect by regulating tumor cell proliferation and metastasis in vitro and in vivo, which makes it to be a promising targeted therapy of PDAC.
Collapse
Affiliation(s)
- An‐Dong Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Xiao‐Yang Bi
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Guo‐Qing Hou
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Shawn Shun‐Cheng Li
- Department of Biochemistry, Schulich School of Medicine and DentistryWestern UniversityLondonOntarioCanada
| | - Qing Chen
- Department of Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Hui Xu
- Ultrastructural Pathology Laboratory, Department of Pathology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| | - Xuan Cao
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanP. R. China
| |
Collapse
|
10
|
Altevogt P, Ben-Ze'ev A, Gavert N, Schumacher U, Schäfer H, Sebens S. Recent insights into the role of L1CAM in cancer initiation and progression. Int J Cancer 2020; 147:3292-3296. [PMID: 32588424 DOI: 10.1002/ijc.33177] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/08/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022]
Abstract
First described as a neuronal cell adhesion molecule, L1CAM was later identified to be present at increased levels in primary tumors and metastases of various types of cancer. Here, we describe the multifaceted roles of L1CAM that are involved in diverse fundamental steps during tumor initiation and progression, as well as in chemoresistance. Recently, Ganesh et al reported that L1CAM identifies metastasis-initiating cells in colorectal carcinoma exhibiting stem-like cell features, increased tumorigenic potential and enhanced chemoresistance. In this review, we highlight recent advances in L1CAM research with particular emphasis on its role in de-differentiation processes and cancer cell stemness supporting the view that L1CAM is a powerful prognostic factor and a suitable target for improved therapy of metastatic and drug-resistant tumors.
Collapse
Affiliation(s)
- Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg, Germany
| | - Avri Ben-Ze'ev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nancy Gavert
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Heiner Schäfer
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University and University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| |
Collapse
|
11
|
Giordano M, Cavallaro U. Different Shades of L1CAM in the Pathophysiology of Cancer Stem Cells. J Clin Med 2020; 9:E1502. [PMID: 32429448 PMCID: PMC7291284 DOI: 10.3390/jcm9051502] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
L1 cell adhesion molecule (L1CAM) is aberrantly expressed in several tumor types where it is causally linked to malignancy and therapy resistance, acting also as a poor prognosis factor. Accordingly, several approaches have been developed to interfere with L1CAM function or to deliver cytotoxic agents to L1CAM-expressing tumors. Metastatic dissemination, tumor relapse and drug resistance can be fueled by a subpopulation of neoplastic cells endowed with peculiar biological properties that include self-renewal, efficient DNA repair, drug efflux machineries, quiescence, and immune evasion. These cells, known as cancer stem cells (CSC) or tumor-initiating cells, represent, therefore, an ideal target for tumor eradication. However, the molecular and functional traits of CSC have been unveiled only to a limited extent. In this context, it appears that L1CAM is expressed in the CSC compartment of certain tumors, where it plays a causal role in stemness itself and/or in biological processes intimately associated with CSC (e.g., epithelial-mesenchymal transition (EMT) and chemoresistance). This review summarizes the role of L1CAM in cancer focusing on its functional contribution to CSC pathophysiology. We also discuss the clinical usefulness of therapeutic strategies aimed at targeting L1CAM in the context of anti-CSC treatments.
Collapse
Affiliation(s)
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, 20128 Milan, Italy;
| |
Collapse
|
12
|
Cave DD, Di Guida M, Costa V, Sevillano M, Ferrante L, Heeschen C, Corona M, Cucciardi A, Lonardo E. TGF-β1 secreted by pancreatic stellate cells promotes stemness and tumourigenicity in pancreatic cancer cells through L1CAM downregulation. Oncogene 2020; 39:4271-4285. [PMID: 32291413 PMCID: PMC7239770 DOI: 10.1038/s41388-020-1289-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 02/08/2023]
Abstract
Pancreatic stellate cells (PSCs) secrete high levels of transforming growth factor-β1 (TGF-β1) that contributes to the development of pancreatic ductal adenocarcinoma (PDAC). TGF-β1 modulates the expression of L1 cell adhesion molecule (L1CAM), but its role in tumour progression still remains controversial. To clarify L1 function in PDAC and cellular phenotypes, we performed L1CAM cell sorting, silencing and overexpression in several primary pancreatic cancer cells. PSCs silenced for TGF-β1 were used for crosstalk experiments. We found that TGF-β1 secreted by PSCs negatively regulates L1CAM expression, through canonical TGF-β-Smad2/3 signalling, leading to a more aggressive PDAC phenotype. Cells with reduced expression of L1CAM harboured enhanced stemness potential and tumourigenicity. Inactivation of TGF-β1 signalling in PSCs strongly reduced the aggressiveness of PDAC cells. Our data provide functional proof and mechanistic insights for the tumour-suppressive function of L1CAM via reducing stemness. Rescuing L1CAM expression in cancer cells through targeting of TGF-β1 reverses stemness and bears the potential to improve the still miserable prognosis of PDAC patients.
Collapse
Affiliation(s)
- Donatella Delle Cave
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Martina Di Guida
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Marta Sevillano
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luigi Ferrante
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy
| | | | - Marco Corona
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Antonio Cucciardi
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy
| | - Enza Lonardo
- Institute of Genetics and Biophysics 'Adriano Buzzati-Traverso' (IGB), CNR, Via Pietro Castellino 111, 80131, Naples, Italy. .,Spanish National Cancer Research Centre, CNIO, Madrid, Spain.
| |
Collapse
|
13
|
Zinn R, Otterbein H, Lehnert H, Ungefroren H. RAC1B: A Guardian of the Epithelial Phenotype and Protector Against Epithelial-Mesenchymal Transition. Cells 2019; 8:cells8121569. [PMID: 31817229 PMCID: PMC6952788 DOI: 10.3390/cells8121569] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 12/27/2022] Open
Abstract
The small GTPase Ras-related C3 botulinum toxin substrate 1B (RAC1B) has been shown to potently inhibit transforming growth factor (TGF)-β1-induced cell migration and epithelial-mesenchymal transition (EMT) in pancreatic and breast epithelial cells, but the underlying mechanism has remained obscure. Using a panel of pancreatic ductal adenocarcinoma (PDAC)-derived cell lines of different differentiation stages, we show that RAC1B is more abundantly expressed in well differentiated as opposed to poorly differentiated cells. Interestingly, RNA interference-mediated knockdown of RAC1B decreased expression of the epithelial marker protein E-cadherin, encoded by CDH1, and enhanced its TGF-β1-induced downregulation, whereas ectopic overexpression of RAC1B upregulated CDH1 expression and largely prevented its TGF-β1-induced silencing of CDH1. Conversely, knockdown of RAC1B, or deletion of the RAC1B-specific exon 3b by CRISPR/Cas-mediated genomic editing, enhanced basal and TGF-β1-induced upregulation of mesenchymal markers like Vimentin, and EMT-associated transcription factors such as SNAIL and SLUG. Moreover, we demonstrate that knockout of RAC1B enhanced the cells’ migratory activity and derepressed TGF-β1-induced activation of the mitogen-activated protein kinase ERK2. Pharmacological inhibition of ERK1/2 activation in RAC1B-depleted cells rescued cells from the RAC1B knockdown-induced enhancement of cell migration, TGF-β1-induced downregulation of CDH1, and upregulation of SNAI1. We conclude that RAC1B promotes epithelial gene expression and suppresses mesenchymal gene expression by interfering with TGF-β1-induced MEK-ERK signaling, thereby protecting cells from undergoing EMT and EMT-associated responses like acquisition of cell motility.
Collapse
Affiliation(s)
- Rabea Zinn
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hannah Otterbein
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hendrik Lehnert
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
| | - Hendrik Ungefroren
- First Department of Medicine, UKSH, Campus Lübeck, 23552 Lübeck, Germany; (R.Z.); (H.O.); (H.L.)
- Department of General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Correspondence: ; Tel.: +49-451-3101-7866
| |
Collapse
|
14
|
Clinical significance and biological role of L1 cell adhesion molecule in gastric cancer. Br J Cancer 2019; 121:1058-1068. [PMID: 31754264 PMCID: PMC6964673 DOI: 10.1038/s41416-019-0646-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 10/09/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022] Open
Abstract
Background L1 cell adhesion molecule (L1CAM) is highly expressed in malignant tumours and might play a pivotal role in tumour progression. Methods We analysed by immunohistochemistry L1CAM protein expression in formalin-fixed, paraffin-embedded specimens from 309 GC patients. We performed propensity score matching (PSM) analysis to clarify the prognostic impact of L1CAM in GC patients. We evaluated L1CAM gene expression in fresh frozen specimens from another group of 131 GC patients to establish its clinical relevance. The effects of changes in L1CAM were investigated in vitro and in vivo. Results L1CAM was mainly expressed in tumour cells of GC tissues. Elevated L1CAM expression was an independent prognostic factor for overall and disease-free survival, and an independent risk factor for distant metastasis in GC patients. PSM analysis showed that high L1CAM expression was significantly associated with poor prognosis. L1CAM gene expression using fresh frozen specimens successfully validated all of these findings in an independent cohort. Inhibition of L1CAM suppressed cell proliferation, cycle progress, invasion, migration and anoikis resistance in GC cells. Furthermore, L1CAM inhibition suppressed the growth of peritoneal metastasis. Conclusion L1CAM may serve as a feasible biomarker for identification of patients who have a high risk of recurrence of GC.
Collapse
|
15
|
Mesmar F, Dai B, Ibrahim A, Hases L, Jafferali MH, Jose Augustine J, DiLorenzo S, Kang Y, Zhao Y, Wang J, Kim M, Lin CY, Berkenstam A, Fleming J, Williams C. Clinical candidate and genistein analogue AXP107-11 has chemoenhancing functions in pancreatic adenocarcinoma through G protein-coupled estrogen receptor signaling. Cancer Med 2019; 8:7705-7719. [PMID: 31568691 PMCID: PMC6912054 DOI: 10.1002/cam4.2581] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/13/2019] [Accepted: 09/13/2019] [Indexed: 12/17/2022] Open
Abstract
Despite advances in cancer therapeutics, pancreatic cancer remains difficult to treat and often develops resistance to chemotherapies. We have evaluated a bioavailable genistein analogue, AXP107-11 which has completed phase Ib clinical trial, as an approach to sensitize tumor cells to chemotherapy. Using organotypic cultures of 14 patient-derived xenografts (PDX) of pancreatic ductal adenocarcinoma, we found that addition of AXP107-11 indeed sensitized 57% of cases to gemcitabine treatment. Results were validated using PDX models in vivo. Further, RNA-Seq from responsive and unresponsive tumors proposed a 41-gene treatment-predictive signature. Functional and molecular assays were performed in cell lines and demonstrated that the effect was synergistic. Transcriptome analysis indicated activation of G-protein-coupled estrogen receptor (GPER1) as the main underlying mechanism of action, which was corroborated using GPER1-selective agonists and antagonists. GPER1 expression in pancreatic tumors was indicative of survival, and our study proposes that activation of GPER1 may constitute a new avenue for pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Fahmi Mesmar
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.,Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Bingbing Dai
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed Ibrahim
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Linnea Hases
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden.,Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Mohammed Hakim Jafferali
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| | - Jithesh Jose Augustine
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sebastian DiLorenzo
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.,National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Ya'an Kang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Zhao
- Department of Bioinformatics and Computing Science, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computing Science, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Kim
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chin-Yo Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | | | - Jason Fleming
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cecilia Williams
- Department of Protein Science, KTH Royal Institute of Technology, Science for Life Laboratory, Solna, Sweden
| |
Collapse
|
16
|
Mrazkova B, Dzijak R, Imrichova T, Kyjacova L, Barath P, Dzubak P, Holub D, Hajduch M, Nahacka Z, Andera L, Holicek P, Vasicova P, Sapega O, Bartek J, Hodny Z. Induction, regulation and roles of neural adhesion molecule L1CAM in cellular senescence. Aging (Albany NY) 2019; 10:434-462. [PMID: 29615539 PMCID: PMC5892697 DOI: 10.18632/aging.101404] [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: 12/20/2017] [Accepted: 03/22/2018] [Indexed: 12/12/2022]
Abstract
Aging involves tissue accumulation of senescent cells (SC) whose elimination through senolytic approaches may evoke organismal rejuvenation. SC also contribute to aging-associated pathologies including cancer, hence it is imperative to better identify and target SC. Here, we aimed to identify new cell-surface proteins differentially expressed on human SC. Besides previously reported proteins enriched on SC, we identified 78 proteins enriched and 73 proteins underrepresented in replicatively senescent BJ fibroblasts, including L1CAM, whose expression is normally restricted to the neural system and kidneys. L1CAM was: 1) induced in premature forms of cellular senescence triggered chemically and by gamma-radiation, but not in Ras-induced senescence; 2) induced upon inhibition of cyclin-dependent kinases by p16INK4a; 3) induced by TGFbeta and suppressed by RAS/MAPK(Erk) signaling (the latter explaining the lack of L1CAM induction in RAS-induced senescence); and 4) induced upon downregulation of growth-associated gene ANT2, growth in low-glucose medium or inhibition of the mevalonate pathway. These data indicate that L1CAM is controlled by a number of cell growth- and metabolism-related pathways during SC development. Functionally, SC with enhanced surface L1CAM showed increased adhesion to extracellular matrix and migrated faster. Our results provide mechanistic insights into senescence of human cells, with implications for future senolytic strategies.
Collapse
Affiliation(s)
- Blanka Mrazkova
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| | - Rastislav Dzijak
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| | - Terezie Imrichova
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| | - Lenka Kyjacova
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| | - Peter Barath
- Institute of Chemistry, Slovak Academy of Sciences, Bratislava 84538, Slovakia
| | - Petr Dzubak
- Institute of Molecular and Translational Medicine, Palacky University, Olomouc 77147, Czech Republic
| | - Dusan Holub
- Institute of Molecular and Translational Medicine, Palacky University, Olomouc 77147, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Palacky University, Olomouc 77147, Czech Republic
| | - Zuzana Nahacka
- Laboratory of Molecular Therapy, Institute of Biotechnology of the ASCR, Prague 14220, Czech Republic
| | - Ladislav Andera
- Laboratory of Molecular Therapy, Institute of Biotechnology of the ASCR, Prague 14220, Czech Republic
| | - Petr Holicek
- Laboratory of Molecular Therapy, Institute of Biotechnology of the ASCR, Prague 14220, Czech Republic
| | - Pavla Vasicova
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| | - Olena Sapega
- Laboratory of Immunological and Tumour Models, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| | - Jiri Bartek
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic.,Danish Cancer Society Research Center, Copenhagen DK-2100, Denmark.,Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Zdenek Hodny
- Department of Genome Integrity, Institute of Molecular Genetics of the ASCR, Prague 14220, Czech Republic
| |
Collapse
|
17
|
Wang B, Li J, Li X, Ou Y. Identifying prognosis and metastasis-associated genes associated with Ewing sarcoma by weighted gene co-expression network analysis. Oncol Lett 2019; 18:3527-3536. [PMID: 31516570 PMCID: PMC6732955 DOI: 10.3892/ol.2019.10681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/25/2019] [Indexed: 02/06/2023] Open
Abstract
Ewing sarcoma (ES) is a highly malignant pediatric tumor with a low survival rate and a high rate of metastasis. However, there have been limited reports on the exploration of new biomarkers of ES. Therefore, the aim of the present study was to identify the potential hub genes associated with overall vital survival (OVS) and metastasis in ES. Traditional methods for identifying differentially expressed genes lack the in-depth information of mechanistic studies. In this study, a weighted co-expression network for ES was constructed through weighted gene co-expression network analysis to identify co-expression modules associated with clinical phenotypes. The hub genes in the metastasis- and OVS-related co-expression modules were extracted, and the association between the hub genes and patient OVS was verified in another independent Gene Expression Omnibus dataset. Functional annotations and protein-protein interaction analysis of co-expression modules were also used to understand the potential regulatory mechanisms. The results of the functional enrichment analysis revealed that the OVS-associated module was mainly enriched in the cell cycle and immune response, and the metastasis-associated module was enriched in metabolism. A total of four genes (proteasome subunit α4, L1 cell adhesion molecule, serine/threonine kinase receptor-associated protein and cytotoxic T-lymphocyte-associated protein 4) in the OVS-related module and two genes (calcium voltage-gated channel auxiliary subunit γ2 and γ-aminobutyric acid type B receptor subunit 2) in the metastasis-related module were selected as hub genes. Further research on the hub genes identified in the present study may contribute to the understanding of the mechanism of ES metastasis and progression.
Collapse
Affiliation(s)
- Ben Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Jie Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Xin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| |
Collapse
|
18
|
Guha Majumdar A, Subramanian M. Hydroxychavicol from Piper betle induces apoptosis, cell cycle arrest, and inhibits epithelial-mesenchymal transition in pancreatic cancer cells. Biochem Pharmacol 2019; 166:274-291. [PMID: 31154000 DOI: 10.1016/j.bcp.2019.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022]
Abstract
Pancreatic cancer is a major cause of cancer-related mortality around the world. Currently, options for diagnosis and treatment are extremely limited, which culminates in a very high mortality rate. Intensive research spanning more than four decades has met several roadblocks in terms of improvement in overall survival. In this study, we have evaluated the effect of Hydroxychavicol (HC), a naturally occurring and abundantly isolatable allylarene from Piper betle leaves on pancreatic cancer cells. Our investigation reveals that HC inhibits proliferation and epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. HC induces DNA damage, as evidenced by γ-H2AX, 53BP1 induction and comet assay, which further results in mitotic catastrophe and apoptosis. The apoptosis induced by HC is JNK pathway-dependent and caspase-mediated. HC also inhibits migration and invasion of pancreatic cancer cells via a generalized repression of genes involved in EMT. A quantitative real time PCR-based array revealed at least 14 different genes to be differentially expressed upon HC treatment in pancreatic cancer cells. These results show significant potential of HC as an anticancer agent against pancreatic cancer.
Collapse
Affiliation(s)
- Ananda Guha Majumdar
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | - Mahesh Subramanian
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
19
|
Liu X, Min S, Wu N, Liu H, Wang T, Li W, Shen Y, Zhao C, Wang H, Qian Z, Xu H, Chen Y, Wang X. miR-193a-3p inhibition of the Slug activator PAK4 suppresses non-small cell lung cancer aggressiveness via the p53/Slug/L1CAM pathway. Cancer Lett 2019; 447:56-65. [PMID: 30685413 DOI: 10.1016/j.canlet.2019.01.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/12/2018] [Accepted: 01/21/2019] [Indexed: 01/10/2023]
Abstract
L1 cell adhesion molecule (L1CAM) promotes invasiveness and metastasis in non-small cell lung cancer (NSCLC) cells and is upregulated by the p53-regulated transcription factor Slug. p21-activated kinase 4 (PAK4) directly phosphorylates Slug, resulting in pro-malignant Slug stabilization. We hypothesized that microRNA-based negative regulation of PAK4 would reduce L1CAM-induced NSCLC aggressiveness via destabilizing Slug. We found that elevated L1CAM expression was tightly correlated with p53 loss-of-function and reduced NSCLC patient survival. L1CAM suppression reduced NSCLC cell migration and invasiveness in vitro as well as tumor formation and distal metastasis in vivo. Mechanistically, p53 restricts L1CAM expression through the β-catenin/Slug pathway, with levels of β-catenin and Slug positively correlating with L1CAM expression in NSCLC tumors. The microRNA miR-193a-3p directly targets PAK4 and suppresses downstream p-Slug and L1CAM expression. Silencing PAK4, Slug, and L1CAM mirrored miR-193a-3p's effects upon the migration and invasiveness of NSCLC cells in vitro. Decreased miR-193a-3p levels correlated with elevated PAK4, p-Slug, and L1CAM levels in NSCLC tumors. Our findings support a model of miR-193a-3p as a suppressor of metastatic disease progression in NSCLC via modulation of the p53/Slug/L1CAM pathway.
Collapse
Affiliation(s)
- Xincheng Liu
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Shengping Min
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Nan Wu
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Hongli Liu
- Department of Gynecological Oncology, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Tao Wang
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Wei Li
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Yuanbing Shen
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Chengling Zhao
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Hongtao Wang
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Zhongqing Qian
- Department of Immunology, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Huanbai Xu
- Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated First People's Hospital, Shanghai, China
| | - Yuqing Chen
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China.
| | - Xiaojing Wang
- Anhui Clinical and Preclinical Key Laboratory of Respiratory Disease, Department of Respiration, First Affiliated Hospital, Bengbu Medical College, Bengbu, Anhui Province, China.
| |
Collapse
|
20
|
Novel Immunotherapeutic Approaches for Neuroblastoma and Malignant Melanoma. J Immunol Res 2018; 2018:8097398. [PMID: 30510968 PMCID: PMC6232800 DOI: 10.1155/2018/8097398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/15/2018] [Indexed: 01/24/2023] Open
Abstract
Neuroblastoma (NB) and malignant melanoma (MM), tumors of pediatric age and adulthood, respectively, share a common origin, both of them deriving from the neural crest cells. Although NB and MM have a different behavior, in respect to age of onset, primary tissue involvement and metastatic spread, the prognosis for high stage-affected patients is still poor, in spite of aggressive treatment strategies and the huge amount of new discovered biological knowledge. For these reasons researchers are continuously attempting to find out new treatment options, which in a near future could be translated to the clinical practice. In the last two decades, a strong effort has been spent in the field of translational research of immunotherapy which led to satisfactory results. Indeed, several immunotherapeutic clinical trials have been performed and some of them also resulted beneficial. Here, we summarize preclinical studies based on immunotherapeutic approaches applied in models of both NB and MM.
Collapse
|
21
|
Kha ML, Hesse L, Deisinger F, Sipos B, Röcken C, Arlt A, Sebens S, Helm O, Schäfer H. The antioxidant transcription factor Nrf2 modulates the stress response and phenotype of malignant as well as premalignant pancreatic ductal epithelial cells by inducing expression of the ATF3 splicing variant ΔZip2. Oncogene 2018; 38:1461-1476. [PMID: 30302023 DOI: 10.1038/s41388-018-0518-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 08/27/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits one of the worst survival rates of all cancers. While death rates show declining trends in the majority of cancers, PDAC registers rising rates. Based on the recently described crosstalk between TGF-β1 and Nrf2 in the PDAC development, the involvement of ATF3 and its splice variant ΔZip2 in TGF-β1- and Nrf2-driven pancreatic tumorigenesis was investigated. As demonstrated here, PDAC (Panc1, T3M4) cells or premalignant H6c7 pancreatic ductal epithelial cells differentially express ΔZip2- and ATF3, relating to stronger Nrf2 activity seen in Panc1 cells and TGF-ß1 activity in T3M4 or H6c7 cells, respectively. Treatment with the electrophile/oxidative stress inducer tBHQ or the cytostatic drug gemcitabine strongly elevated ΔZip2 expression in a Nrf2-dependent fashion. The differential expression of ATF3 and ΔZip2 in response to Nrf2 and TGF-ß1 relates to differential ATF3-gene promoter usage, giving rise of distinct splice variants. Nrf2-dependent ΔZip2 expression confers resistance against gemcitabine-induced apoptosis, only partially relating to interference with ATF3 and its proapoptotic activity, e.g., through CHOP-expression. In fact, ΔZip2 autonomously activates expression of cIAP anti-apoptotic proteins. Moreover, ΔZip2 favors and ATF3 suppresses growth and clonal expansion of PDAC cells, again partially independent of each other. Using a Panc1 tumor xenograft model in SCID-beige mice, the opposite activities of ATF3 and ΔZip2 on tumor-growth and chemoresistance were verified in vivo. Immunohistochemical analyses confirmed ΔZip2 and Nrf2 coexpression in cancerous and PanIN structures of human PDAC and chronic pancreatitis tissues, respectively, which to some extent was reciprocal to ATF3 expression. It is concluded that depending on selective ATF3-gene promoter usage by Nrf2, the ΔZip2 expression is induced in response to electrophile/oxidative (here through tBHQ) and xenobiotic (here through gemcitabine) stress, providing apoptosis protection and growth advantages to pancreatic ductal epithelial cells. This condition may substantially add to pancreatic carcinogenesis driven by chronic inflammation.
Collapse
Affiliation(s)
- My-Lan Kha
- Laboratory of Molecular Gastroenterology & Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Lisa Hesse
- Laboratory of Molecular Gastroenterology & Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Florian Deisinger
- Laboratory of Molecular Gastroenterology & Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Bence Sipos
- Department of Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstr. 8, 72076, Tübingen, Germany
| | - Christoph Röcken
- Institute of Pathology, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 14, Arnold-Heller-Straße 3, 24105, Kiel, Germany.,Biomaterial Bank of the Comprehensive Cancer Center Kiel, UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Alexander Arlt
- Laboratory of Gastrointestinal Signal Transduction, Department of Internal Medicine I, UKSH Campus Kiel, Bldg. 6, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Susanne Sebens
- Biomaterial Bank of the Comprehensive Cancer Center Kiel, UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany.,Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Ole Helm
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University & UKSH Campus Kiel, Bldg. 17, Arnold-Heller-Straße 3, 24105, Kiel, Germany.
| |
Collapse
|
22
|
L1CAM induces perineural invasion of pancreas cancer cells by upregulation of metalloproteinase expression. Oncogene 2018; 38:596-608. [PMID: 30171263 DOI: 10.1038/s41388-018-0458-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 07/11/2018] [Accepted: 07/14/2018] [Indexed: 11/08/2022]
Abstract
Pancreas cancer cells have a tendency to invade along nerves. Such cancerous nerve invasion (CNI) is associated with poor outcome; however, the exact mechanism that drives cancer cells to disseminate along nerves is unknown. Immunohistochemical analysis of human pancreatic ductal adenocarcinoma (PDAC) specimens showed overexpression of the L1 cell adhesion molecule (L1CAM) in cancer cells and in adjacent Schwann cells (SC) in invaded nerves. By modeling the neural microenvironment, we found that L1CAM secreted from SCs acts as a strong chemoattractant to cancer cells, through activation of MAP kinase signaling. L1CAM also upregulated expression of metalloproteinase-2 (MMP-2) and MMP-9 by PDAC cells, through STAT3 activation. Using a transgenic Pdx-1-Cre/KrasG12D /p53R172H (KPC) mouse model, we show that treatment with anti-L1CAM Ab significantly reduces CNI in vivo. We provide evidence of a paracrine response between SCs and cancer cells in the neural niche, which promotes cancer invasion via L1CAM secretion.
Collapse
|
23
|
Knaack H, Lenk L, Philipp LM, Miarka L, Rahn S, Viol F, Hauser C, Egberts JH, Gundlach JP, Will O, Tiwari S, Mikulits W, Schumacher U, Hengstler JG, Sebens S. Liver metastasis of pancreatic cancer: the hepatic microenvironment impacts differentiation and self-renewal capacity of pancreatic ductal epithelial cells. Oncotarget 2018; 9:31771-31786. [PMID: 30167093 PMCID: PMC6114965 DOI: 10.18632/oncotarget.25884] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/21/2018] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at advanced stages with the liver as the main site of metastases. The hepatic microenvironment has been shown to determine outgrowth of liver metastases. Cancer stem cells (CSCs) are essential for initiation and maintenance of tumors and acquisition of CSC-properties has been linked to Epithelial-Mesenchymal-Transition. Thus, this study aimed at elucidating whether and how the hepatic microenvironment impacts stemness and differentiation of disseminated pancreatic ductal epithelial cells (PDECs). Culture of premalignant H6c7-kras and malignant Panc1 PDECs together with hepatocytes and hepatic stellate cells (HSC) promoted self-renewal capacity of both PDEC lines. This was indicated by higher colony formation compared to cells cocultured with hepatocytes and hepatic myofibroblasts. Different Panc1 colony types derived from an HSC-enriched coculture were expanded and characterized revealing that holoclones exhibited an enhanced colony formation ability, elevated and exclusive expression of the CSC-marker Nestin and a more pronounced mesenchymal phenotype compared to paraclones. Moreover, Panc1 holoclone cells showed an increased tumorigenic potential in vivo leading to formation of undifferentiated tumors in 7/10 animals, while inoculation of paraclone cells only led to formation of tumors in 2/10 animals being smaller in number and size. Holoclone tumors were characterized by elevated expression of mesenchymal markers, complete loss of E-cadherin expression and high expression of Nestin. Finally, Etanercept-mediated TNF-α blocking partly reversed the mesenchymal CSC-phenotype of Panc1 holoclone cells. Overall, these data provide evidence that the hepatic microenvironment determines stemness and differentiation of PDECs, thereby substantially contributing to liver metastases of PDAC.
Collapse
Affiliation(s)
- Hendrike Knaack
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Lennart Lenk
- Department of Pediatrics, UKSH Campus Kiel, Kiel, Germany
| | - Lisa-Marie Philipp
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Lauritz Miarka
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Sascha Rahn
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Fabrice Viol
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Charlotte Hauser
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH Campus Kiel, Kiel, Germany
| | - Jan-Hendrik Egberts
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH Campus Kiel, Kiel, Germany
| | - Jan-Paul Gundlach
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH Campus Kiel, Kiel, Germany
| | - Olga Will
- Molecular Imaging North Competence Center, Clinic of Radiology and Neuroradiology, CAU and UKSH Campus Kiel, Kiel, Germany
| | - Sanjay Tiwari
- Molecular Imaging North Competence Center, Clinic of Radiology and Neuroradiology, CAU and UKSH Campus Kiel, Kiel, Germany
| | - Wolfgang Mikulits
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Udo Schumacher
- Centre of Experimental Medicine, Department of Anatomy and Experimental Morphology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University Dortmund, Dortmund, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| |
Collapse
|
24
|
Added Value of Estrogen Receptor, Progesterone Receptor, and L1 Cell Adhesion Molecule Expression to Histology-Based Endometrial Carcinoma Recurrence Prediction Models: An ENITEC Collaboration Study. Int J Gynecol Cancer 2018; 28:514-523. [DOI: 10.1097/igc.0000000000001187] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
ObjectivesEndometrial carcinoma mortality is mainly caused by recurrent disease, and various immunohistochemical markers to predict recurrences have been studied. Loss of the estrogen receptor (ER) and progesterone receptor (PR) and the presence of the L1 cell adhesion molecule (L1CAM) are promising markers, but their combined value has not been studied.Materials and MethodsExpression of ER, PR, and L1CAM was immunohistochemically determined in 293 endometrial carcinomas from 11 collaborating European Network for Individualized Treatment of Endometrial Cancer centers. Estrogen receptor, PR, or L1CAM staining was considered positive or negative when expressed by greater than or equal to 10% or less than 10% of the tumor cells, respectively. The association between these markers and clinicopathological markers, and their combined value in predicting survival were calculated, both in the entire cohort and in a selected groups of stage I endometrioid and low-risk stage I endometrioid carcinomas.ResultsEstrogen receptor and PR were negative in 19% and 28% of the cases, respectively, and L1CAM was positive in 18%. All 3 were associated with advanced stage, high-grade, nonendometrioid histology, lymphovascular space invasion (LVSI), and reduced disease-free survival. Only advanced stage, loss of PR, and LVSI were associated with reduced disease-free survival in multivariate analysis. A prognostic model including these 3 markers was superior to 1 including only the 3 immunohistochemical markers, which was superior to the traditional model. In both the stage I endometrioid and the low-risk stage I endometrioid groups, only loss of PR was associated with reduced disease-free survival.ConclusionsLoss of ER and PR, and the presence of L1CAM are associated with high risk characteristics, and loss of PR is the strongest predictor of recurrent disease. Although a combination of these 3 markers is slightly superior to the traditional histological markers, a prognostic model including stage, PR expression, and LVSI is the most promising model in the identification of high risk carcinomas. In the stage I endometrioid carcinomas, PR immunohistochemistry appears to be of additional value in predicting recurrences.
Collapse
|
25
|
Prognostic value and clinicopathologic characteristics of L1 cell adhesion molecule (L1CAM) in a large series of vulvar squamous cell carcinomas. Oncotarget 2018; 7:26192-205. [PMID: 27028855 PMCID: PMC5041974 DOI: 10.18632/oncotarget.8353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 03/04/2016] [Indexed: 12/21/2022] Open
Abstract
Background Vulvar cancer treatment is mostly curative, but also has high morbidity rates. In a search for markers that can identify patients at risk of metastases, we investigated the prognostic value of L1-cell adhesion molecule (L1CAM) in large series of vulvar squamous cell carcinomas (VSCCs). L1CAM promotes cell motility and is an emerging prognostic factor for metastasis in many cancer subtypes. Results L1CAM expression was observed at the invasive front or in spray-patterned parts of 17% of the tumours. L1CAM-positive tumours expressed vimentin more often, but L1CAM expression was not associated with TP53 or CTNNB1 mutations. Five-year survival was worse for patients with L1CAM expression (overall survival 46.1% vs 63.6%, P=.014, disease specific survival 63.8% vs 80.0%, P=.018). Multivariate analysis indicates L1CAM expression as an independent prognostic marker (HR 2.9, 95% CI 1.10–7.68). An in vitro spheroid invasion assay showed decreased invasion of L1CAM-expressing VSCC spindle cells after treatment with L1CAM-neutralising antibodies. Materials and Methods Paraffin-embedded tumour tissue from two cohorts (N=103 and 245) of primary VSCCs were stained for L1CAM, vimentin and E-cadherin. Patients of the first cohort were tested for human papilloma virus infection and sequenced for TP53 and CTNNB1 (β-catenin) mutations. The expression of L1CAM was correlated to clinical characteristics and patient survival. Conclusion This is the first study to show high L1CAM-expression at the infiltrating margin of VSCC's. L1CAM-expressing VSCCs had a significantly worse prognosis compared to L1CAM-negative tumours. The highest expression was observed in spindle-shaped cells, where it might be correlated to their invasive capacity.
Collapse
|
26
|
Howley BV, Howe PH. TGF-beta signaling in cancer: post-transcriptional regulation of EMT via hnRNP E1. Cytokine 2018; 118:19-26. [PMID: 29396052 DOI: 10.1016/j.cyto.2017.12.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022]
Abstract
The TGFβ signaling pathway is a critical regulator of cancer progression in part through induction of the epithelial to mesenchymal transition (EMT). This process is aberrantly activated in cancer cells, facilitating invasion of the basement membrane, survival in the circulatory system, and dissemination to distant organs. The mechanisms through which epithelial cells transition to a mesenchymal state involve coordinated transcriptional and post-transcriptional control of gene expression. One such mechanism of control is through the RNA binding protein hnRNP E1, which regulates splicing and translation of a cohort of EMT and stemness-associated transcripts. A growing body of evidence indicates a major role for hnRNP E1 in the control of epithelial cell plasticity, especially in the context of carcinoma progression. Here, we review the multiple mechanisms through which hnRNP E1 functions to control EMT and metastatic progression.
Collapse
Affiliation(s)
- Breege V Howley
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Philip H Howe
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
27
|
Rahn S, Zimmermann V, Viol F, Knaack H, Stemmer K, Peters L, Lenk L, Ungefroren H, Saur D, Schäfer H, Helm O, Sebens S. Diabetes as risk factor for pancreatic cancer: Hyperglycemia promotes epithelial-mesenchymal-transition and stem cell properties in pancreatic ductal epithelial cells. Cancer Lett 2017; 415:129-150. [PMID: 29222037 DOI: 10.1016/j.canlet.2017.12.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/15/2017] [Accepted: 12/01/2017] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with hyperglycemia and a risk to develop pancreatic ductal adenocarcinoma (PDAC), one of the most fatal malignancies. Cancer stem cells (CSC) are essential for initiation and maintenance of tumors, and acquisition of CSC-features is linked to epithelial-mesenchymal-transition (EMT). The present study investigated whether hyperglycemia promotes EMT and CSC-features in premalignant and malignant pancreatic ductal epithelial cells (PDEC). Under normoglycemia (5 mM d-glucose), Panc1 PDAC cells but not premalignant H6c7-kras cells exhibited a mesenchymal phenotype along with pronounced colony formation. While hyperglycemia (25 mM d-glucose) did not impact the mesenchymal phenotype of Panc1 cells, CSC-properties were aggravated exemplified by increased Nanog expression and Nanog-dependent formation of holo- and meroclones. In H6c7-kras cells, high glucose increased secretion of Transforming-Growth-Factor-beta1 (TGF-β1) as well as TGF-β1 signaling, and in a TGF-β1-dependent manner reduced E-cadherin expression, increased Nestin expression and number of meroclones. Finally, reduced E-cadherin expression was detected in pancreatic ducts of hyperglycemic but not normoglycemic mice. These data suggest that hyperglycemia promotes the acquisition of mesenchymal and CSC-properties in PDEC by activating TGF-β signaling and might explain how T2DM facilitates pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Sascha Rahn
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Vivien Zimmermann
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Fabrice Viol
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hendrike Knaack
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Kerstin Stemmer
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Lena Peters
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Lennart Lenk
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Hendrik Ungefroren
- Department of General Surgery and Thoracic Surgery, UKSH Campus Kiel, Germany; First Department of Medicine, UKSH Campus Lübeck, Lübeck, Germany
| | - Dieter Saur
- II. Medizinische Klinik und Poliklinik, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Heiner Schäfer
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Ole Helm
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel (CAU) and University Medical Center Schleswig-Holstein (UKSH) Campus Kiel, Kiel, Germany.
| |
Collapse
|
28
|
Ungefroren H, Witte D, Mihara K, Rauch BH, Henklein P, Jöhren O, Bonni S, Settmacher U, Lehnert H, Hollenberg MD, Kaufmann R, Gieseler F. Transforming Growth Factor- β1/Activin Receptor-like Kinase 5-Mediated Cell Migration is Dependent on the Protein Proteinase-Activated Receptor 2 but not on Proteinase-Activated Receptor 2-Stimulated G q-Calcium Signaling. Mol Pharmacol 2017; 92:519-532. [PMID: 28842394 DOI: 10.1124/mol.117.109017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/07/2017] [Indexed: 02/14/2025] Open
Abstract
Transforming growth factor-β (TGF-β), serine proteinases such as trypsin, and proteinase-activated receptor 2 (PAR2) promote tumor development by stimulating invasion and metastasis. Previously, we found that in cancer cells derived from pancreatic ductal adenocarcinoma (PDAC) PAR2 protein is necessary for TGF-β1-dependent cell motility. Here, we show in the same cells that, conversely, the type I TGF-β receptor activin receptor-like kinase 5 is dispensable for trypsin and PAR2 activating peptide (PAR2-AP)-induced migration. To reveal whether Gq-calcium signaling is a prerequisite for PAR2 to enhance TGF-β signaling, we investigated the effects of PAR2-APs, PAR2 mutation and PAR2 inhibitors on TGF-β1-induced migration, reporter gene activity, and Smad activation. Stimulation of cells with PAR2-AP alone failed to enhance basal or TGF-β1-induced C-terminal phosphorylation of Smad3, Smad-dependent activity of a luciferase reporter gene, and cell migration. Consistently, in complementary loss of function studies, abrogation of the PAR2-Gq-calcium signaling arm failed to suppress TGF-β1-induced cell migration, reporter gene activity, and Smad3 activation. Together, our findings suggest that the calcium-regulating motif is not required for PAR2 to synergize with TGF-β1 to promote cell motility. Additional experiments in PDAC cells revealed that PAR2 and TGF-β1 synergy may involve TGF-β1 induction of enzymes that cause autocrine cleavage/activation of PAR2, possibly through a biased signaling function. Our results suggest that although reducing PAR2 protein expression may potentially block TGF-β's prooncogenic function, inhibiting PAR2-Gq-calcium signaling alone would not be sufficient to achieve this effect.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - David Witte
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Koichiro Mihara
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Bernhard H Rauch
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Petra Henklein
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Olaf Jöhren
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Shirin Bonni
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Utz Settmacher
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Hendrik Lehnert
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Morley D Hollenberg
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Roland Kaufmann
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| | - Frank Gieseler
- First Department of Medicine, UKSH, and University of Lübeck, Lübeck, Germany (H.U., D.W., H.L., F.G.); Department of Physiology and Pharmacology and Department of Medicine, Inflammation Research Network-Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (K.M., M.D.H.); Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, Greifswald, Germany (B.H.R.); Charité - University Medicine Berlin, Institute of Biochemistry, CharitéCrossOver, Berlin, Germany (P.H.); Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany (O.J.); Arnie Charbonneau Cancer Institute and Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (S.B.); and Department of General, Visceral and Vascular Surgery, Jena University Hospital, Jena, Germany (U.S., R.K.)
| |
Collapse
|
29
|
Schrevel M, Corver WE, Vegter ME, Ter Haar NT, Dreef EJ, Beltman JJ, Kenter G, Bosse T, de Kroon CD, Jordanova ES. L1 cell adhesion molecule (L1CAM) is a strong predictor for locoregional recurrences in cervical cancer. Oncotarget 2017; 8:87568-87581. [PMID: 29152102 PMCID: PMC5675654 DOI: 10.18632/oncotarget.20976] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 08/21/2017] [Indexed: 12/19/2022] Open
Abstract
Background L1 cell adhesion molecule (L1CAM) has been shown to be a prognostic marker in various cancer types, and has been suggested to play a role in epithelial mesenchymal transition (EMT). Here, we determined the prognostic significance of L1CAM in cervical cancer and its association with vimentin expression on tumor cells, indicative of EMT. Methods Formalin-fixed, paraffin-embedded primary tumor samples from 372 cervical cancer patients were collected for immunohistochemical analysis of L1CAM expression. In 109 FFPE specimens, the percentage of vimentin expressing tumor cells was determined by flow cytometry. Results Positive L1CAM expression (≥10% of tumor cells) was associated with disease-free survival, validated using RNAseq TCGA data. L1CAM expression was independently associated with locoregional recurrence-free survival (hazard ratio 2.62, 95% CI 1.33 – 5.17, P = 0.006), and strongly associated with percentage of vimentin expressing tumor cells (P = 0.003). Expression of both L1CAM and vimentin indicated a subgroup with the highest risk of recurrence (hazard ratio 3.15, 95% CI 1.25 – 7.92, P = 0.015). Conclusion L1CAM might be a promising new prognostic marker for locoregional recurrences in cervical cancer, and its association with vimentin expression suggests that L1CAM might affect tumor aggressiveness, possibly through EMT.
Collapse
Affiliation(s)
- Marlies Schrevel
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Willem E Corver
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Margit E Vegter
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Natalja T Ter Haar
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Enno J Dreef
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jogchum J Beltman
- Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gemma Kenter
- Department of Gynecology, VUmc, Centre for Gynecologic Oncology, Amsterdam, The Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis D de Kroon
- Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ekaterina S Jordanova
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Gynecology, VUmc, Centre for Gynecologic Oncology, Amsterdam, The Netherlands
| |
Collapse
|
30
|
TGF-β1 affects cell-cell adhesion in the heart in an NCAM1-dependent mechanism. J Mol Cell Cardiol 2017; 112:49-57. [PMID: 28870505 DOI: 10.1016/j.yjmcc.2017.08.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/23/2017] [Accepted: 08/31/2017] [Indexed: 12/22/2022]
Abstract
The contractile property of the myocardium is maintained by cell-cell junctions enabling cardiomyocytes to work as a syncytium. Alterations in cell-cell junctions are observed in heart failure, a disease characterized by the activation of Transforming Growth Factor beta 1 (TGFβ1). While TGFβ1 has been implicated in diverse biologic responses, its molecular function in controlling cell-cell adhesion in the heart has never been investigated. Cardiac-specific transgenic mice expressing active TGFβ1 were generated to model the observed increase in activity in the failing heart. Activation of TGFβ1 in the heart was sufficient to drive ventricular dysfunction. To begin to understand the function of this important molecule we undertook an extensive structural analysis of the myocardium by electron microscopy and immunostaining. This approach revealed that TGFβ1 alters intercalated disc structures and cell-cell adhesion in ventricular myocytes. Mechanistically, we found that TGFβ1 induces the expression of neural adhesion molecule 1 (NCAM1) in cardiomyocytes in a p38-dependent pathway, and that selective targeting of NCAM1 was sufficient to rescue the cell adhesion defect observed when cardiomyocytes were treated with TGFβ1. Importantly, NCAM1 was upregulated in human heart samples from ischemic and non-ischemic cardiomyopathy patients and NCAM1 protein levels correlated with the degree of TGFβ1 activity in the human cardiac ventricle. Overall, we found that TGFβ1 is deleterious to the heart by regulating the adhesion properties of cardiomyocytes in an NCAM1-dependent mechanism. Our results suggest that inhibiting NCAM1 would be cardioprotective, counteract the pathological action of TGFβ1 and reduce heart failure severity.
Collapse
|
31
|
Geismann C, Grohmann F, Dreher A, Häsler R, Rosenstiel P, Legler K, Hauser C, Egberts JH, Sipos B, Schreiber S, Linkermann A, Hassan Z, Schneider G, Schäfer H, Arlt A. Role of CCL20 mediated immune cell recruitment in NF-κB mediated TRAIL resistance of pancreatic cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:782-796. [PMID: 28188806 DOI: 10.1016/j.bbamcr.2017.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 01/25/2017] [Accepted: 02/06/2017] [Indexed: 01/11/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest cancers. From a clinical view, the transcription factor NF-κB is of particular importance, since this pathway confers apoptosis resistance and limits drug efficacy. Whereas the role of the most abundant NF-κB subunit p65/RelA in therapeutic resistance is well documented, only little knowledge of the RelA downstream targets and their functional relevance in TRAIL mediated apoptosis in PDAC is available. In the present study TRAIL resistant and sensitive PDAC cell lines were analyzed for differentially expressed RelA target genes, to define RelA downstream targets mediating TRAIL resistance. The most upregulated target gene was then further functionally characterized. Unbiased genome-wide expression analysis demonstrated that the chemokine CCL20 represents the strongest TRAIL inducible direct RelA target gene in resistant PDAC cells. Unexpectedly, targeting CCL20 by siRNA, blocking antibodies or by downregulation of the sole CCL20 receptor CCR6 had no effect on PDAC cell death or cancer cell migration, arguing against an autocrine role of CCL20 in PDAC. However, by using an ex vivo indirect co-culture system we were able to show that CCL20 acts paracrine to recruit immune cells. Importantly, CCL20-recruited immune cells further increase TRAIL resistance of CCL20-producing PDAC cells. In conclusion, our data show a functional role of a RelA-CCL20 pathway in PDAC TRAIL resistance. We demonstrate how the therapy-induced cross-talk of cancer cells with immune cells affects treatment responses, knowledge needed to tailor novel bi-specific treatments, which target tumor cell as well as immune cells.
Collapse
Affiliation(s)
- Claudia Geismann
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH-Campus Kiel, Kiel, Germany
| | - Frauke Grohmann
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH-Campus Kiel, Kiel, Germany
| | - Anita Dreher
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH-Campus Kiel, Kiel, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, UKSH Campus Kiel, Germany
| | | | - Karen Legler
- Division of Molecular Oncology, Institute for Experimental Cancer Research, UKSH Campus Kiel, Kiel, Germany
| | | | | | - Bence Sipos
- Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Stefan Schreiber
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH-Campus Kiel, Kiel, Germany; Institute of Clinical Molecular Biology, UKSH Campus Kiel, Germany
| | - Andreas Linkermann
- Clinic for Nephrology and Hypertension, Christian-Albrechts-University, Kiel, Germany
| | - Zonera Hassan
- Technische Universität München, Klinikum rechts der Isar, II. Medizinische Klinik, Munich, Germany
| | - Günter Schneider
- Technische Universität München, Klinikum rechts der Isar, II. Medizinische Klinik, Munich, Germany
| | - Heiner Schäfer
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH-Campus Kiel, Kiel, Germany; Institute of Experimental Cancer Research, UKSH Campus Kiel, Germany
| | - Alexander Arlt
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH-Campus Kiel, Kiel, Germany.
| |
Collapse
|
32
|
Bergmann F. [Pancreatic acinar neoplasms : Comparative molecular characterization]. DER PATHOLOGE 2016; 37:191-195. [PMID: 27807633 DOI: 10.1007/s00292-016-0235-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Pancreatic acinar cell carcinomas are biologically aggressive neoplasms for which treatment options are very limited. The molecular mechanisms of tumor initiation and progression are largely not understood and precursor lesions have not yet been identified. In this study, pancreatic acinar cell carcinomas were cytogenetically characterized as well as by molecular and immunohistochemical analyses. Corresponding investigations were carried out on pancreatic ductal adenocarcinomas and pancreatic neuroendocrine neoplasms augmented by functional analyses. We show that pancreatic acinar cell carcinomas display a microsatellite stable, chromosomal unstable genotype, characterized by recurrent chromosomal imbalances that clearly discriminate them from pancreatic ductal adenocarcinomas and neuroendocrine neoplasms. Based on findings obtained from comparative genomic hybridization, candidate genes could be identified, such as deleted in colorectal cancer (DCC) and c-MYC. Furthermore, several therapeutic targets were identified in acinar cell carcinomas and other pancreatic neoplasms, including epidermal growth factor receptor (EGFR), L1 cell adhesion molecule (L1CAM) and heat shock protein 90 (HSP90). Moreover, L1CAM was shown to play a significant role in the tumorigenesis of pancreatic ductal adenocarcinoma. Functional analyses in cell lines derived from pancreatic neuroendocrine neoplasms revealed promising anti-tumorigenic effects using EGFR and HSP90 inhibitors affecting the cell cycle and in the case of HSP90, regulating several other oncogenes. Finally, based on mutational analyses of mitochondrial DNA, molecular evidence is provided that acinar cell cystadenomas (or better cystic acinar transformation) represent non-clonal lesions, suggesting an inflammatory reactive non-neoplastic nature.
Collapse
Affiliation(s)
- F Bergmann
- Pathologisches Institut, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland.
| |
Collapse
|
33
|
van der Putten LJ, Visser NC, van de Vijver K, Santacana M, Bronsert P, Bulten J, Hirschfeld M, Colas E, Gil-Moreno A, Garcia A, Mancebo G, Alameda F, Trovik J, Kopperud RK, Huvila J, Schrauwen S, Koskas M, Walker F, Weinberger V, Minar L, Jandakova E, Snijders MP, van den Berg-van Erp S, Matias-Guiu X, Salvesen HB, Amant F, Massuger LF, Pijnenborg JM. L1CAM expression in endometrial carcinomas: an ENITEC collaboration study. Br J Cancer 2016; 115:716-24. [PMID: 27505134 PMCID: PMC5023774 DOI: 10.1038/bjc.2016.235] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 01/07/2023] Open
Abstract
Background: Identification of aggressive endometrioid endometrial carcinomas (EECs) and non-endometrioid carcinomas (NEECs) is essential to improve outcome. L1 cell adhesion molecule (L1CAM) expression is a strong prognostic marker in stage I EECs, but less is known about L1CAM expression in advanced-stage EECs and NEECs. This study analyses L1CAM expression in a clinically representative cohort of endometrial carcinomas. Methods: The expression of L1CAM was immunohistochemically determined in 1199 endometrial carcinomas, treated at one of the European Network for Individualized Treatment of Endometrial Cancer (ENITEC) centres. Staining was considered positive when >10% of the tumour cells expressed L1CAM. The association between L1CAM expression and several clincopathological characteristics and disease outcome was calculated. Results: In all, L1CAM was expressed in 10% of the 935 stage I EECs, 18% of the 160 advanced stage EECs, and 75% of the 104 NEECs. The expression of L1CAM was associated with advanced stage, nodal involvement, high tumour grade, non-endometrioid histology, lymphovascular space invasion, and distant recurrences in all cases, and with reduced survival in the EECs, but not in the NEECs. Conclusions: The expression of L1CAM is a strong predictor of poor outcome in EECs, but not NEECs. It is strongly associated with non-endometrioid histology and distant spread, and could improve the postoperative selection of high-risk endometrial carcinomas. The value of L1CAM expression in the preoperative selection of high-risk endometrial carcinomas should be studied.
Collapse
Affiliation(s)
- Louis Jm van der Putten
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein 10, Nijmegen 6525GA, The Netherlands
| | - Nicole Cm Visser
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein 10, Nijmegen 6525GA, The Netherlands
| | - Koen van de Vijver
- Department of Pathology, Anthoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam 1066CX, The Netherlands
| | - Maria Santacana
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Avenida Alcalde Rovira Roure, 80, Lleida 25198, Spain
| | - Peter Bronsert
- Institute of Pathology, University Medical Center Freiburg, Hugstetter Strasse 55, Freiburg 79106, Germany
| | - Johan Bulten
- Department of Pathology, Radboud University Medical Center, Geert Grooteplein 10, Nijmegen 6525GA, The Netherlands
| | - Marc Hirschfeld
- Department of Obstetrics and Gynecology, University Medical Center Freiburg, Hugstetter Strasse 55, Freiburg 79106, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg D-69120, Germany
| | - Eva Colas
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain
| | - Antonio Gil-Moreno
- Biomedical Research Group in Gynecology, Vall Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain.,Gynecological Department, Vall Hebron University Hospital, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain
| | - Angel Garcia
- Department of Pathology, Vall Hebron University Hospital, Passeig de la Vall d'Hebron, 119-129, Barcelona 08035, Spain
| | - Gemma Mancebo
- Department of Obstetrics and Gynecology, Hospital del Mar, Passeig Marítim, 25-29, Barcelona 08003, Spain
| | - Fransesc Alameda
- Department of Pathology, Hospital del Mar, Passeig Marítim, 25-29, Barcelona 08003, Spain
| | - Jone Trovik
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Reidun K Kopperud
- Department of Clinical Science, University of Bergen, PO Box 7800, Bergen 5020, Norway.,Center for Cancer Biomarkers (CCBIO), University of Bergen, PO Box 7800, Bergen 5020, Norway
| | - Jutta Huvila
- Department of Pathology, University of Turku, PO Box 7245, Laskut, Turku 01051, Finland
| | - Stefanie Schrauwen
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospital Gasthuisberg, Herestraat 49, Leuven 3000, Belgium
| | - Martin Koskas
- Department of Obstetrics and Gynecology, Bichat-Claude Bernard Hospital, 46 Rue Henri Huchard, Paris 75018, France
| | - Francine Walker
- Department of Pathology, Bichat-Claude Bernard Hospital, 46 Rue Henri Huchard, Paris 75018, France
| | - Vit Weinberger
- Department of Gynecology and Obstetrics, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Lubos Minar
- Department of Gynecology and Obstetrics, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Eva Jandakova
- Institute of Pathology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Marc Plm Snijders
- Department of Obstetrics and Gynaecology, Canisius-Wilhelmina Hospital, Weg door Jonkerbos 100, Nijmegen 6532SZ, The Netherlands
| | - Saskia van den Berg-van Erp
- Department of Pathology, Canisius-Wilhelmina Hospital, Weg door Jonkerbos 100, Nijmegen 6532SZ, The Netherlands
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLLEIDA, Avenida Alcalde Rovira Roure, 80, Lleida 25198, Spain
| | - Helga B Salvesen
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Jonas Lies vei 65, Bergen 5021, Norway
| | - Frederic Amant
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University Hospital Gasthuisberg, Herestraat 49, Leuven 3000, Belgium
| | - Leon Fag Massuger
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Geert Grooteplein 10, Nijmegen 6525GA, The Netherlands
| | - Johanna Ma Pijnenborg
- Department of Obstetrics and Gynaecology, TweeSteden Hospital, Doctor Deelenlaan 5, Tilburg 5042AD, The Netherlands
| |
Collapse
|
34
|
Samatov TR, Wicklein D, Tonevitsky AG. L1CAM: Cell adhesion and more. ACTA ACUST UNITED AC 2016; 51:25-32. [DOI: 10.1016/j.proghi.2016.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/20/2016] [Indexed: 12/17/2022]
|
35
|
Genrich G, Kruppa M, Lenk L, Helm O, Broich A, Freitag-Wolf S, Röcken C, Sipos B, Schäfer H, Sebens S. The anti-oxidative transcription factor Nuclear factor E2 related factor-2 (Nrf2) counteracts TGF-β1 mediated growth inhibition of pancreatic ductal epithelial cells -Nrf2 as determinant of pro-tumorigenic functions of TGF-β1. BMC Cancer 2016; 16:155. [PMID: 26915435 PMCID: PMC4766703 DOI: 10.1186/s12885-016-2191-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/17/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nuclear factor E2 related factor-2 (Nrf2) is an oxidative stress inducible transcription factor being essential in regulating cell homeostasis. Thus, acute induction of Nrf2 in epithelial cells exposed to inflammation confers protection from oxidative cell damage and mutagenesis supporting an anti-tumorigenic role for Nrf2. However, pancreatic ductal adenocarcinoma (PDAC) is characterized by persistent Nrf2 activity conferring therapy resistance which points to a pro-tumorigenic role of Nrf2. A similar dichotomous role in tumorigenesis is described for the Transforming Growth Factor-beta 1 (TGF-β1). The present study therefore aimed at elucidating whether the switch of Nrf2 function towards a tumor promoting one relates to the modulation of TGF-β1 induced cell responses and whether this might occur early in PDAC development. METHODS In situ analysis comprised immunohistochemical stainings of activated (phosphorylated) Nrf2 and Ki67 in pancreatic tissues containing normal ducts and pancreatic intraepithelial neoplasia (PanINs). In vitro, Nrf2 levels in benign (H6c7-pBp), premalignant (H6c7-kras) and malignant (Colo357) pancreatic ductal epithelial cells were modulated by Nrf2 specific siRNA or Nrf2 overexpression. Then, the effect of Nrf2 alone and in combination with TGF-β1 on cell growth and survival was investigated by cell counting, Ki67 staining and apoptosis assays. The underlying cell signaling was investigated by western blotting. Statistical analysis was performed by Shapiro-Wilk test for normal distribution. Parametric data were analyzed by one-way ANOVA, while non-parametric data were analyzed by Kruskal-Wallis one-way ANOVA on ranks. RESULTS Significantly elevated expression of activated Nrf2 and Ki67 could be detected in PanINs but not in normal pancreatic ductal epithelium. While the effect of Nrf2 on basal cell growth of H6c7-pBp, H6c7-kras and Colo357 cells was minor, it clearly attenuated the growth inhibiting effects of TGF-β1 in all cell lines. This enhanced Nrf2-mediated cell survival was predominantly based on an enhanced proliferative activity. Accordingly, expression of p21 expression along with expression of phospho-p38 and phospho-Smad3 was diminished whereas Erk-phosphorylation was enhanced under these conditions. CONCLUSIONS Overall, our data demonstrate that Nrf2 being elevated in early precursor lesions counteracts the growth inhibiting function of TGF-β1 already in benign and premalignant pancreatic ductal epithelial cells. This could represent one fundamental mechanism underlying the functional switch of both- TGF-β1 and Nrf2 - which may manifest already in early stages of PDAC development.
Collapse
Affiliation(s)
- Geeske Genrich
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Marcus Kruppa
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Lennart Lenk
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Ole Helm
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Anna Broich
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, UKSH Campus Kiel, Brunswiker Str. 10, 24105, Kiel, Germany.
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 14, 24105, Kiel, Germany.
| | - Bence Sipos
- Department of Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstr. 8, 72076, Tübingen, Germany.
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Hepatology, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 6, 24105, Kiel, Germany.
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis, Institute for Experimental Cancer Research, Christian-Albrechts-University Kiel, Arnold-Heller-Str. 3, Building 17, 24105, Kiel, Germany.
| |
Collapse
|
36
|
Yu X, Yang F, Fu DL, Jin C. L1 cell adhesion molecule as a therapeutic target in cancer. Expert Rev Anticancer Ther 2016; 16:359-71. [PMID: 26781307 DOI: 10.1586/14737140.2016.1143363] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
L1 cell adhesion molecule (L1CAM) is the prototype member of the L1-family of closely related neural adhesion molecules. L1CAM is differentially expressed in the normal nervous system as well as pathological tissues and displays a wide range of biological activities. In human malignancies, L1CAM plays a vital role in tumor growth, invasion and metastasis. Recently, increasing evidence has suggested that L1CAM exerts a variety of functions at different steps of tumor progression through a series of signaling pathways. In addition, L1CAM has been identified as a promising target for cancer therapy by using synthetic and natural inhibitors. In this review, we provide an up-to-date overview of the role of L1CAM involved in cancers and the rationale for L1CAM as a novel molecular target for cancer therapy.
Collapse
Affiliation(s)
- Xinzhe Yu
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - Feng Yang
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - De-Liang Fu
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - Chen Jin
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| |
Collapse
|
37
|
Van Gool IC, Stelloo E, Nout RA, Nijman HW, Edmondson RJ, Church DN, MacKay HJ, Leary A, Powell ME, Mileshkin L, Creutzberg CL, Smit VTHBM, Bosse T. Prognostic significance of L1CAM expression and its association with mutant p53 expression in high-risk endometrial cancer. Mod Pathol 2016; 29:174-81. [PMID: 26743472 DOI: 10.1038/modpathol.2015.147] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/12/2015] [Indexed: 02/07/2023]
Abstract
Studies in early-stage, predominantly low- and intermediate-risk endometrial cancer have demonstrated that L1 cell adhesion molecule (L1CAM) overexpression identifies patients at increased risk of recurrence, yet its prognostic significance in high-risk endometrial cancer is unclear. To evaluate this, its frequency, and the relationship of L1CAM with the established endometrial cancer biomarker p53, we analyzed the expression of both markers by immunohistochemistry in a pilot series of 116 endometrial cancers (86 endometrioid, 30 non-endometrioid subtype) with high-risk features (such as high tumor grade and deep myometrial invasion) and correlated results with clinical outcome. We used The Cancer Genome Atlas (TCGA) endometrial cancer series to validate our findings. Using the previously reported cutoff of 10% positive staining, 51/116 (44%) tumors were classified as L1CAM-positive, with no significant association between L1CAM positivity and the rate of distant metastasis (P=0.195). However, increasing the threshold for L1CAM positivity to 50% resulted in a reduction of the frequency of L1CAM-positive tumors to 24% (28/116), and a significant association with the rate of distant metastasis (P=0.018). L1CAM expression was strongly associated with mutant p53 in the high-risk and TCGA series (P<0.001), although a substantial fraction (36% of endometrioid, 10% of non-endometrioid morphology) of p53-mutant endometrial cancers displayed <10% L1CAM positivity. Moreover, 30% of p53-wild-type non-endometrioid endometrial cancers demonstrated diffuse L1CAM staining, suggesting p53-independent mechanisms of L1CAM overexpression. In conclusion, the previously proposed threshold for L1CAM positivity of >10% does not predict prognosis in high-risk endometrial cancer, whereas an alternative threshold (>50%) does. L1CAM expression is strongly, but not universally, associated with mutant p53, and may be strong enough for clinical implementation as prognostic marker in combination with p53. The high frequency of L1CAM expression in high-risk endometrial cancers suggests that it may also be a promising therapeutic target in this tumor subset.
Collapse
Affiliation(s)
- Inge C Van Gool
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellen Stelloo
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Remi A Nout
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans W Nijman
- Department of Gynecology, University Medical Center Groningen, Groningen, The Netherlands
| | - Richard J Edmondson
- Institute of Cancer Sciences, University of Manchester, St Marys Hospital, Manchester, UK
| | - David N Church
- Molecular and Population Genetics Laboratory, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.,Oxford Cancer Centre, Churchill Hospital, Oxford, UK
| | - Helen J MacKay
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Princess Margaret Hospital/University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Leary
- Department of Medicine, Gynecology Unit, Gustave Roussy, Villejuif, France
| | - Melanie E Powell
- Department of Clinical Oncology, Barts Health NHS Trust, London, UK
| | - Linda Mileshkin
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
38
|
Zhang Y, Bi L, Qiu Y, Wang Y, Ding J, Zhuang Y, Tian Y, Cai H. Elevated sL1-CAM levels in BALF and serum of IPF patients. Respirology 2015; 21:143-8. [PMID: 26610737 DOI: 10.1111/resp.12659] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/21/2015] [Accepted: 07/24/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Yingwei Zhang
- Department of Respiratory Diseases; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| | - Lintao Bi
- Department of Oncology and Hematology; China-Japan Union Hospital Affiliated to Jilin University; Changchun China
| | - Yuying Qiu
- Department of Respiratory Diseases; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| | - Yang Wang
- Department of Radiology; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| | - Jingjing Ding
- Department of Respiratory Diseases; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| | - Yi Zhuang
- Department of Respiratory Diseases; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| | - Yaqiong Tian
- Department of Respiratory Diseases; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| | - Hourong Cai
- Department of Respiratory Diseases; Nanjing Drum Tower Hospital affiliated to Medical School of Nanjing University; Nanjing China
| |
Collapse
|
39
|
Altevogt P, Doberstein K, Fogel M. L1CAM in human cancer. Int J Cancer 2015; 138:1565-76. [DOI: 10.1002/ijc.29658] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/19/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany and Department of Dermatology, Venereology and Allergology; University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg; Mannheim Germany
| | - Kai Doberstein
- Ovarian Cancer Research Center, Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA
| | - Mina Fogel
- Central Laboratories; Kaplan Medical Center; Rehovot Israel
| |
Collapse
|
40
|
Arfmann-Knübel S, Struck B, Genrich G, Helm O, Sipos B, Sebens S, Schäfer H. The Crosstalk between Nrf2 and TGF-β1 in the Epithelial-Mesenchymal Transition of Pancreatic Duct Epithelial Cells. PLoS One 2015. [PMID: 26226105 PMCID: PMC4520686 DOI: 10.1371/journal.pone.0132978] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nrf2 and TGF-β1 both affect tumorigenesis in a dual fashion, either by preventing carcinogen induced carcinogenesis and suppressing tumor growth, respectively, or by conferring cytoprotection and invasiveness to tumor cells during malignant transformation. Given the involvement of Nrf2 and TGF-β1 in the adaptation of epithelial cells to persistent inflammatory stress, e.g. of the pancreatic duct epithelium during chronic pancreatitis, a crosstalk between Nrf2 and TGF-β1 can be envisaged. By using premalignant human pancreatic duct cells (HPDE) and the pancreatic ductal adenocarcinoma cell line Colo357, we could show that Nrf2 and TGF-β1 independently but additively conferred an invasive phenotype to HPDE cells, whereas acting synergistically in Colo357 cells. This was accompanied by differential regulation of EMT markers like vimentin, Slug, L1CAM and E-cadherin. Nrf2 activation suppressed E-cadherin expression through an as yet unidentified ARE related site in the E-cadherin promoter, attenuated TGF-β1 induced Smad2/3-activity and enhanced JNK-signaling. In Colo357 cells, TGF-β1 itself was capable of inducing Nrf2 whereas in HPDE cells TGF-β1 per-se did not affect Nrf2 activity, but enhanced Nrf2 induction by tBHQ. In Colo357, but not in HPDE cells, the effects of TGF-β1 on invasion were sensitive to Nrf2 knock-down. In both cell lines, E-cadherin re-expression inhibited the proinvasive effect of Nrf2. Thus, the increased invasion of both cell lines relates to the Nrf2-dependent downregulation of E-cadherin expression. In line, immunohistochemistry analysis of human pancreatic intraepithelial neoplasias in pancreatic tissues from chronic pancreatitis patients revealed strong Nrf2 activity already in premalignant epithelial duct cells, accompanied by partial loss of E-cadherin expression. Our findings indicate that Nrf2 and TGF-β1 both contribute to malignant transformation through distinct EMT related mechanisms accounting for an invasive phenotype. Provided a crosstalk between both pathways, Nrf2 and TGF-β1 mutually promote their tumorigenic potential, a condition manifesting already at an early stage during inflammation induced carcinogenesis of the pancreas.
Collapse
Affiliation(s)
- Sarah Arfmann-Knübel
- Laboratory of Molecular Gastroenterology, Dept. of Internal Medicine I, UKSH Campus Kiel, Arnold-Heller-Str. 3, Bldg. 6, 24105, Kiel, Germany
| | - Birte Struck
- Laboratory of Molecular Gastroenterology, Dept. of Internal Medicine I, UKSH Campus Kiel, Arnold-Heller-Str. 3, Bldg. 6, 24105, Kiel, Germany
| | - Geeske Genrich
- Group Inflammatory Carcinogenesis, Institute of Experimental Medicine, CAU Kiel, Arnold-Heller-Str. 3, Bldg. 17, 24105, Kiel, Germany
| | - Ole Helm
- Group Inflammatory Carcinogenesis, Institute of Experimental Medicine, CAU Kiel, Arnold-Heller-Str. 3, Bldg. 17, 24105, Kiel, Germany
| | - Bence Sipos
- Department of Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstraße 8, 72076, Tübingen, Germany
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis, Institute of Experimental Medicine, CAU Kiel, Arnold-Heller-Str. 3, Bldg. 17, 24105, Kiel, Germany
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology, Dept. of Internal Medicine I, UKSH Campus Kiel, Arnold-Heller-Str. 3, Bldg. 6, 24105, Kiel, Germany
- * E-mail:
| |
Collapse
|
41
|
Slug-dependent upregulation of L1CAM is responsible for the increased invasion potential of pancreatic cancer cells following long-term 5-FU treatment. PLoS One 2015; 10:e0123684. [PMID: 25860483 PMCID: PMC4393253 DOI: 10.1371/journal.pone.0123684] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 01/05/2023] Open
Abstract
Background Pancreatic adenocarcinoma is a lethal disease with 5-year survival of less than 5%. 5-fluorouracil (5-FU) is a principal first-line therapy, but treatment only extends survival modestly and is seldom curative. Drug resistance and disease recurrence is typical and there is a pressing need to overcome this. To investigate acquired 5-FU resistance in pancreatic adenocarcinoma, we established chemoresistant monoclonal cell lines from the Panc 03.27 cell line by long-term exposure to increasing doses of 5-FU. Results 5-FU-resistant cell lines exhibited increased expression of markers associated with multidrug resistance explaining their reduced sensitivity to 5-FU. In addition, 5-FU-resistant cell lines showed alterations typical for an epithelial-to-mesenchymal transition (EMT), including upregulation of mesenchymal markers and increased invasiveness. Microarray analysis revealed the L1CAM pathway as one of the most upregulated pathways in the chemoresistant clones, and a significant upregulation of L1CAM was seen on the RNA and protein level. In pancreatic cancer, expression of L1CAM is associated with a chemoresistant and migratory phenotype. Using esiRNA targeting L1CAM, or by blocking the extracellular part of L1CAM with antibodies, we show that the increased invasiveness observed in the chemoresistant cells functionally depends on L1CAM. Using esiRNA targeting β-catenin and/or Slug, we demonstrate that in the chemoresistant cell lines, L1CAM expression depends on Slug rather than β-catenin. Conclusion Our findings establish Slug-induced L1CAM expression as a mediator of a chemoresistant and migratory phenotype in pancreatic adenocarcinoma cells.
Collapse
|
42
|
Goebel L, Grage-Griebenow E, Gorys A, Helm O, Genrich G, Lenk L, Wesch D, Ungefroren H, Freitag-Wolf S, Sipos B, Röcken C, Schäfer H, Sebens S. CD4 + T cells potently induce epithelial-mesenchymal-transition in premalignant and malignant pancreatic ductal epithelial cells-novel implications of CD4 + T cells in pancreatic cancer development. Oncoimmunology 2015; 4:e1000083. [PMID: 26137395 DOI: 10.1080/2162402x.2014.1000083] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/06/2023] Open
Abstract
Chronic pancreatitis (CP) is a risk factor of pancreatic ductal adenocarcinoma (PDAC) and characterized by a pronounced desmoplastic reaction with CD4+ T cells accounting for the majority of the stromal T cell infiltrate. Epithelial-mesenchymal-transition (EMT) is a critical process for metastasis by which epithelial/carcinoma cells become enabled to disseminate probably prior to tumor formation. To investigate whether CD4+ T cells induce EMT in human pancreatic ductal epithelial cells, premalignant H6c7 cells were mono- or co-cultured with human CD4+CD25+CD127-CD49d- regulatory T cells (T-regs) or CD4+CD25- T-effector cells (T-effs) being isolated by negative magnetic bead separation from blood of healthy donors. Particularly in the presence of activated T-effs, H6c7 cells acquired a spindle-shaped morphology, reduced E-cadherin expression, and elevated expression of the mesenchymal proteins vimentin, L1CAM, and ZEB-1. This was accompanied by an increased invasive behavior. Moreover, activated T-effs exerted similar effects in the PDAC cell line T3M4. Blocking of TNF-α and IL-6 being released at greater amounts into supernatants during co-cultures with activated T-effs attenuated the EMT-associated alterations in H6c7 cells. Supporting these findings, EMT-associated alterations (exemplified by reduced E-cadherin expression and enhanced expression of vimentin and L1CAM) were predominantly detected in ductal epithelium of CP tissues surrounded by a dense stroma enriched with CD4+ T cells. Overall this study points to a novel role of CD4+ T cells beyond their immune function in pancreatic tumorigenesis and underscores the view that EMT induction in pancreatic ductal epithelial cells represents an early event in PDAC development being essentially promoted by inflammatory processes.
Collapse
Affiliation(s)
- Lisa Goebel
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Evelin Grage-Griebenow
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Artur Gorys
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Ole Helm
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Geeske Genrich
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Lennart Lenk
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| | - Daniela Wesch
- Institute of Immunology; Christian-Albrechts-University and UKSH Campus Kiel ; Kiel, Germany
| | | | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics; UKSH Campus Kiel ; Kiel, Germany
| | - Bence Sipos
- Department of Pathology and Neuropathology; University Hospital Tübingen ; Tübingen, Germany
| | | | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Hepatology; Department of Internal Medicine I; UKSH Campus Kiel ; Kiel, Germany
| | - Susanne Sebens
- Group Inflammatory Carcinogenesis; Institute for Experimental Medicine; Christian-Albrechts-University Kiel and University Hospital Schleswig-Holstein (UKSH) Campus Kiel ; Kiel, Germany
| |
Collapse
|
43
|
Rac1b negatively regulates TGF-β1-induced cell motility in pancreatic ductal epithelial cells by suppressing Smad signalling. Oncotarget 2014; 5:277-90. [PMID: 24378395 PMCID: PMC3960208 DOI: 10.18632/oncotarget.1696] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor (TGF)-β1 promotes progression of pancreatic ductal adenocarcinoma (PDAC) by enhancing epithelial-mesenchymal transition, cell migration/invasion, and metastasis, in part by cooperating with the small GTPase Rac1. Prompted by the observation of higher expression of Rac1b, an alternatively spliced Rac1 isoform, in pancreatic ductal epithelial cells and in patients with chronic pancreatitis vs. PDAC, as well as in long-time vs. short-time survivors among PDAC patients, we asked whether Rac1b might negatively affect TGF-β1 prometastatic function. Interestingly, the non-malignant pancreatic ductal epithelial cell line H6c7 exhibited a higher ratio of active Rac1b to total Rac1b than the TGF-β1-responsive PDAC cell lines Panc-1 and Colo357. Notably, siRNA-mediated silencing of Rac1b increased TGF-β1/Smad-dependent migratory activities in H6c7, Colo357, and Panc-1 cells, while ectopic overexpression of Rac1b in Panc-1 cells attenuated TGF-β1-induced cell motility. Depletion of Rac1b in Panc-1 cells enhanced TGF-β1/Smad-dependent expression of promoter-reporter genes and of the endogenous Slug gene. Moreover, Rac1b depletion resulted in a higher and more sustained C-terminal phosphorylation of Smad3 and Smad2, suggesting that Rac1b is involved in Smad2/3 dephosphorylation/inactivation. Since pharmacologic or siRNA-mediated inhibition of Smad3 but not Smad2 was able to alleviate the Rac1b siRNA effect on TGF-β1-induced cell migration, our results suggests that Rac1b inhibits TGF-β1-induced cell motility in pancreatic ductal epithelial cells by blocking the function of Smad3. Moreover, Rac1b may act as an endogenous inhibitor of Rac1 in TGF-β1-mediated migration and possibly metastasis. Hence, it could be exploited for diagnostic/prognostic purposes or even therapeutically in late-stage PDAC as an antimetastatic agent.
Collapse
|
44
|
Suh DH, Kim MA, Kim HS, Chung HH, Park NH, Song YS, Kang SB. L1 cell adhesion molecule expression is associated with pelvic lymph node metastasis and advanced stage in diabetic patients with endometrial cancer: a matched case control study. J Cancer Prev 2014; 19:231-9. [PMID: 25337593 PMCID: PMC4189503 DOI: 10.15430/jcp.2014.19.3.231] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 08/31/2014] [Accepted: 09/01/2014] [Indexed: 11/26/2022] Open
Abstract
Background: Diabetic patients with endometrial cancer had more lymph node metastasis than non-diabetic patients with endometrial cancer. L1 cell adhesion molecule (L1CAM) could be possibly associated with lymph node metastasis in diabetic patients with endometrial cancer via epithelial-mesenchymal transition. We aimed to investigate the association between L1CAM expression and lymph node metastasis in diabetic patients with endometrial cancer. Methods: We conducted a matched case control study of 68 endometrial cancer patients who comprise each 34 diabetic and non-diabetic patients. L1CAM expression was evaluated by immunohistochemistry using fresh formalin-fixed paraffin-embedded tissue block of the patients. The association between L1CAM expression and pelvic lymph node metastasis was assessed according to the presence of diabetes. Results: Of the 68 patients, 13 (19.1%) were positive for L1CAM immunostaining. Positive rate of L1CAM expression in diabetic endometrial cancer patients was similar to that in non-diabetic endometrial cancer patients (14.7% vs. 23.5%, P = 0.355). Tumor recurred more frequently in patients with positive L1CAM expression than those with negative L1CAM expression (33.3% vs. 1.6%, P = 0.019). However, we failed to find any significant association between L1CAM expression and lymph node metastasis. Only for the diabetic patients (n = 34), patients with pelvic lymph node metastasis had more L1CAM expression than those without lymph node metastasis (50.0% vs. 3.6%, P = 0.035). Advanced stage was the only risk factor for recurrence that showed a significant association with L1CAM expression for the diabetic endometrial cancer patients (P = 0.006), as well as all the enrolled patients (P = 0.014). Conclusion: L1CAM expression is associated with pelvic lymph node metastasis and advanced stage in diabetic patients with endometrial cancer.
Collapse
Affiliation(s)
- Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Min A Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Hoon Chung
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Noh Hyun Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea ; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea ; Major in Biomodulation, World Class University, Seoul National University, Seoul, Korea
| | - Soon-Beom Kang
- Women's Gynecologic Oncology Center, Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
45
|
Doberstein K, Harter PN, Haberkorn U, Bretz NP, Arnold B, Carretero R, Moldenhauer G, Mittelbronn M, Altevogt P. Antibody therapy to human L1CAM in a transgenic mouse model blocks local tumor growth but induces EMT. Int J Cancer 2014; 136:E326-39. [DOI: 10.1002/ijc.29222] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 08/18/2014] [Accepted: 09/09/2014] [Indexed: 11/07/2022]
Affiliation(s)
- Kai Doberstein
- Translational Immunology, D015, Tumor Immunology Programme German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Patrick N. Harter
- Edinger Institute (Neurological Institute), Goethe University Frankfurt; Frankfurt Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine; University Hospital Heidelberg; Heidelberg Germany
| | - Niko P. Bretz
- Translational Immunology, D015, Tumor Immunology Programme German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Bernd Arnold
- Molecular Immunology, D050, Tumor Immunology Programme German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Rafael Carretero
- Molecular Immunology, D050, Tumor Immunology Programme German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Gerhard Moldenhauer
- Translational Immunology, D015, Tumor Immunology Programme German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Michel Mittelbronn
- Edinger Institute (Neurological Institute), Goethe University Frankfurt; Frankfurt Germany
| | - Peter Altevogt
- Translational Immunology, D015, Tumor Immunology Programme German Cancer Research Center (DKFZ); Heidelberg Germany
| |
Collapse
|
46
|
Pancreatic stellate cells and CX3CR1: occurrence in normal pancreas and acute and chronic pancreatitis and effect of their activation by a CX3CR1 agonist. Pancreas 2014; 43:708-19. [PMID: 24681877 PMCID: PMC4315317 DOI: 10.1097/mpa.0000000000000109] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Numerous studies suggest important roles of the chemokine, fractalkine (CX3CL1), in acute/chronic pancreatitis; however, the possible mechanisms of the effects are unclear. Pancreatic stellate cells (PSCs) can play important roles in pancreatitis, secreting inflammatory cytokines/chemokines, as well as proliferation. Therefore, we investigated CX3CL1 receptor (CX3CR1) occurrence in normal pancreas and pancreatitis (acute/chronic) tissues and the effects of CX3CL1 on activated PSCs. METHODS CX3CR1 expression/localization in normal pancreas and pancreatitis (acute/chronic) tissues was evaluated with immunohistochemical analysis. CX3CR1 expression and effects of CX3CL1 on activated PSCs were examined with real-time polymerase chain reaction, BrdU (5-bromo-2-deoxyuridine) assays, and Western blotting. RESULTS In normal pancreas, acinar cells expressed CX3CR1 within granule-like formations in the cytoplasm, whereas in acute/chronic pancreatitis, acinar, ductal, and activated PSCs expressed CX3CR1 on cell membranes. With activation of normal PSCs, CX3CR1 is increased. CX3CL1 activated multiple signaling cascades in PSCs. CX3CL1 did not induce inflammatory genes expression in activated PSCs, but induced proliferation. CONCLUSIONS CX3CR1s are expressed in normal pancreas. Expression is increased in acute/chronic pancreatitis, and the CX3CR1s are activated. CX3CL1 induces proliferation of activated PSCs without increasing release of inflammatory mediators. These results suggest that CX3CR1 activation of PSCs could be important in their effects in pancreatitis, especially to PSC proliferation in pancreatitis where CX3CL1 levels are elevated.
Collapse
|
47
|
Grage-Griebenow E, Schäfer H, Sebens S. The fatal alliance of cancer and T cells: How pancreatic tumor cells gather immunosuppressive T cells. Oncoimmunology 2014; 3:e29382. [PMID: 25114835 PMCID: PMC4126073 DOI: 10.4161/onci.29382] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 05/27/2014] [Indexed: 11/19/2022] Open
Abstract
Immune evasion is a hallmark of cancer. We recently identified the adhesion molecule L1CAM as biomarker of pancreatic ductal adenocarcinoma (PDAC) associated with poor prognosis. During inflammation-associated carcinogenesis, L1CAM drives the enrichment of highly immunosuppressive CD4+CD25-CD69+ T cells. Thus, L1CAM may serve as a target in immunomodulatory therapy for PDAC.
Collapse
Affiliation(s)
- Evelin Grage-Griebenow
- Group of Inflammatory Carcinogenesis; Institute for Experimental Medicine; Kiel, Germany
| | - Heiner Schäfer
- Laboratory of Molecular Gastroenterology & Hepatology; Department of Internal Medicine I; Kiel, Germany
| | - Susanne Sebens
- Group of Inflammatory Carcinogenesis; Institute for Experimental Medicine; Kiel, Germany
| |
Collapse
|
48
|
A standardized staining protocol for L1CAM on formalin-fixed, paraffin-embedded tissues using automated platforms. Int J Biol Markers 2014; 29:e180-3. [PMID: 24242293 DOI: 10.5301/jbm.5000055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2013] [Indexed: 11/20/2022]
Abstract
The L1 cell adhesion molecule (L1CAM) is overexpressed in many human cancers and can serve as a biomarker for prognosis in most of these cancers (including type I endometrial carcinomas). Here we provide an optimized immunohistochemical staining procedure for a widely used automated platform (VENTANA™), which has recourse to commercially available primary antibody and detection reagents. In parallel, we optimized the staining on a semi-automated BioGenix (i6000)
immunostainer. These protocols yield good stainings and should represent the basis for a reliable and standardized immunohistochemical detection of L1CAM in a variety of malignancies in different laboratories.
Collapse
|
49
|
Helm O, Mennrich R, Petrick D, Goebel L, Freitag-Wolf S, Röder C, Kalthoff H, Röcken C, Sipos B, Kabelitz D, Schäfer H, Oberg HH, Wesch D, Sebens S. Comparative characterization of stroma cells and ductal epithelium in chronic pancreatitis and pancreatic ductal adenocarcinoma. PLoS One 2014; 9:e94357. [PMID: 24797069 PMCID: PMC4010424 DOI: 10.1371/journal.pone.0094357] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 03/13/2014] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by an extensive stroma being also present in chronic pancreatitis (CP). Using immunohistochemistry, the stroma of CP and PDAC was comprehensively analyzed and correlated with epithelial/carcinoma-related alterations and clinicopathological patient characteristics. While there were no significant differences between CP and PDAC regarding the distribution of CD3+ T cells and α-SMA+ fibroblasts, proportions of CD4+ and CD8+ T cells were significantly lower and numbers of CD25+(CD4+) and FoxP3+(CD4+) regulatory T cells were greater in PDAC compared with CP. Macrophages were more prevalent in CP, but localized more closely to carcinoma cells in PDAC, as were γδ-T cells. Duct-related FoxP3 and L1CAM expression increased from CP to PDAC, while vimentin expression was similarly abundant in both diseases. Moreover, stromal and epithelial compartments of well-differentiated tumors and CPs shared considerable similarities, while moderately and poorly differentiated tumors significantly differed from CP tissues. Analysis of 27 parameters within each pancreatic disease revealed a significant correlation of i) CD4+ and FoxP3+CD4+ T cells with FoxP3 expression in PDAC cells, ii) α-SMA+ fibroblasts with L1CAM expression and proliferation in PDAC cells, iii) CD3 and CD8 expression with γδ-TCR expression in both pancreatic diseases and iv) CD68+ and CD163+ macrophages with vimentin expression in PDAC cells. High expression of FoxP3, vimentin and L1CAM in PDAC cells as well as a tumor-related localization of macrophages each tended to correlate with higher tumor grade. Multivariate survival analysis revealed a younger age at time of surgery as a positive prognostic marker for PDAC patients with the most frequently operated disease stage T3N1M0. Overall this study identified several interrelationships between stroma and epithelial/carcinoma cells in PDACs but also in CP, which in light of previous experimental data strongly support the view that the inflammatory stroma contributes to malignancy-associated alterations already in precursor cells during CP.
Collapse
Affiliation(s)
- Ole Helm
- Institute for Experimental Medicine, Group Inflammatory Carcinogenesis, UKSH Campus Kiel, Kiel, Germany
| | - Ruben Mennrich
- Institute for Experimental Medicine, Group Inflammatory Carcinogenesis, UKSH Campus Kiel, Kiel, Germany
| | | | - Lisa Goebel
- Institute for Experimental Medicine, Group Inflammatory Carcinogenesis, UKSH Campus Kiel, Kiel, Germany
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, UKSH Campus Kiel, Kiel, Germany
| | - Christian Röder
- Institute of Experimental Cancer Research, UKSH Campus Kiel, Kiel, Germany
| | - Holger Kalthoff
- Institute of Experimental Cancer Research, UKSH Campus Kiel, Kiel, Germany
| | | | - Bence Sipos
- Institute of Pathology; University Tübingen, Tübingen, Germany
| | | | - Heiner Schäfer
- Department of Internal Medicine I, Laboratory of Molecular Gastroenterology & Hepatology, UKSH Campus Kiel, Kiel, Germany
| | | | - Daniela Wesch
- Institute of Immunology, UKSH Campus Kiel, Kiel, Germany
| | - Susanne Sebens
- Institute for Experimental Medicine, Group Inflammatory Carcinogenesis, UKSH Campus Kiel, Kiel, Germany
- * E-mail:
| |
Collapse
|
50
|
MiRNA 17 family regulates cisplatin-resistant and metastasis by targeting TGFbetaR2 in NSCLC. PLoS One 2014; 9:e94639. [PMID: 24722426 PMCID: PMC3983236 DOI: 10.1371/journal.pone.0094639] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 03/18/2014] [Indexed: 11/30/2022] Open
Abstract
MicroRNAs (miRNAs) have been proven to play crucial roles in cancer, including tumor chemotherapy resistance and metastasis of non-small-cell lung cancer (NSCLC). TGFβ signal pathway abnormality is widely found in cancer and correlates with tumor proliferation, apoptosis and metastasis. Here, miR-17, 20a, 20b were detected down-regulated in A549/DDP cells (cisplatin resistance) compared with A549 cells (cisplatin sensitive). Over-expression of miR-17, 20a, 20b can not only decrease cisplatin-resistant but also reduce migration by inhibiting epithelial-to-mesenchymal transition (EMT) in A549/DDP cells. These functions of miR-17, 20a, 20b may be caused at least in part via inhibition of TGFβ signal pathway, as miR-17, 20a, 20b are shown to directly target and repress TGF-beta receptor 2 (TGFβR2) which is an important component of TGFβ signal pathway. Consequently, our study suggests that miRNA 17 family (including miR-17, 20a, 20b) can act as TGFβR2 suppressor for reversing cisplatin-resistant and suppressing metastasis in NSCLC.
Collapse
|