1
|
Yuan H, Zhang Y, Liu F, Wu Y, Huang X, Liu X, Jiang L, Xiao B, Zhu Y, Chen Q, Wu P, Jiang K. Exploring the biological mechanism and clinical value of perineural invasion in pancreatic cancer. Cancer Lett 2025; 613:217515. [PMID: 39892698 DOI: 10.1016/j.canlet.2025.217515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Pancreatic cancer (PC) is an extremely aggressive malignancy, with a 5-year survival rate of only 13 %. Perineural invasion (PNI) is a hallmark pathological feature of PC and is observed in almost all cases. Accordingly, PC ranks highly among solid tumors in terms of PNI incidence. The interaction between PC and the nervous system plays a pivotal role in tumor growth and metastasis. In PC, PNI is a key driver of local tumor progression, distant metastasis, and poor prognosis. Clarification of tumor-nerve crosstalk and the underlying molecular mechanisms is needed to facilitate the development of new therapeutic strategies to slow PC progression and alleviate PNI-associated symptoms. In this review, we present a comprehensive overview of the manifestations and characteristics of PNI in PC, summarize the molecular networks that regulate PNI, examine the relationship between PNI and the tumor microenvironment, and discuss the current research challenges and future directions in this critical area.
Collapse
Affiliation(s)
- Hao Yuan
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yufeng Zhang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Fengyuan Liu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yang Wu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xumin Huang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xinjian Liu
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Luyang Jiang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Bin Xiao
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yi Zhu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China; Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China.
| | - Qun Chen
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Pengfei Wu
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Kuirong Jiang
- Pancreas Centre, First Affiliated Hospital, Nanjing Medical University, Nanjing, China; Pancreas Institute, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Hernández-Hatibi S, Borau C, Martínez-Bosch N, Navarro P, García-Aznar JM, Guerrero PE. Quantitative characterization of the 3D self-organization of PDAC tumor spheroids reveals cell type and matrix dependence through advanced microscopy analysis. APL Bioeng 2025; 9:016116. [PMID: 40161492 PMCID: PMC11952832 DOI: 10.1063/5.0242490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by an abundant tumor-associated stroma composed from pancreatic stellate cells, which play a critical role in tumor progression. Developing accurate in vitro models requires understanding the complex interactions between tumor cells and their microenvironment. In this study, we present a quantitative imaging-based characterization of the three dimensional (3D) self-organization of PDAC tumour spheroids using a microfluidic platform that mimics key aspects of the tumor microenvironment. Our model incorporates collagen type I hydrogels to recreate the extracellular matrix, activated human pancreatic stellate cells (HPSCs), and various tumor cell types. Advanced imaging techniques, including Lattice Lightsheet Microscopy, allowed us to analyze the 3D growth and spatial organization of the spheroids, revealing intricate biomechanical interactions. Our results indicate that alterations in matrix properties-such as stiffness, pore size, and hydraulic permeability-due to variations in collagen concentration significantly influence the growth patterns and organization of PDAC spheroids, depending on tumor subtype and epithelial-mesenchymal phenotype. Higher collagen concentrations promoted larger spheroids in epithelial-like cell lines, while mesenchymal-type cells required increased collagen for self-organization into smaller spheroids. Furthermore, coculture with HPSCs affected spheroid formation distinctly based on each PDAC cell line's genetic and phenotypic traits. HPSCs had opposing effects on epithelial-like cell lines: one cell line exhibited enhanced spheroid growth, while another showed inhibited formation, whereas mesenchymal-like spheroids showed minimal impact. These results provide insights into tumor-stroma interactions, emphasizing the importance of the cell-specific and matrix-dependent factors for advancing our understanding of PDAC progression and informing future therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Research Institute (HMRI), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain
| | | | | | | |
Collapse
|
3
|
Hartwig C, Müller J, Klett H, Kouhestani D, Mittelstädt A, Anthuber A, David P, Brunner M, Jacobsen A, Glanz K, Swierzy I, Roßdeutsch L, Klösch B, Grützmann R, Wittenberger T, Sohn K, Weber GF. Discrimination of pancreato-biliary cancer and pancreatitis patients by non-invasive liquid biopsy. Mol Cancer 2024; 23:28. [PMID: 38308296 PMCID: PMC10836044 DOI: 10.1186/s12943-024-01943-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Current diagnostics for the detection of pancreato-biliary cancers (PBCs) need to be optimized. We therefore propose that methylated cell-free DNA (cfDNA) derived from non-invasive liquid biopsies serves as a novel biomarker with the ability to discriminate pancreato-biliary cancers from non-cancer pancreatitis patients. METHODS Differentially methylated regions (DMRs) from plasma cfDNA between PBCs, pancreatitis and clinical control samples conditions were identified by next-generation sequencing after enrichment using methyl-binding domains and database searches to generate a discriminatory panel for a hybridization and capture assay with subsequent targeted high throughput sequencing. RESULTS The hybridization and capture panel, covering around 74 kb in total, was applied to sequence a cohort of 25 PBCs, 25 pancreatitis patients, 25 clinical controls, and seven cases of Intraductal Papillary Mucinous Neoplasia (IPMN). An unbiased machine learning approach identified the 50 most discriminatory methylation markers for the discrimination of PBC from pancreatitis and controls resulting in an AUROC of 0.85 and 0.88 for a training (n = 45) and a validation (n = 37) data set, respectively. The panel was also able to distinguish high grade from low grade IPMN samples. CONCLUSIONS We present a proof of concept for a methylation biomarker panel with better performance and improved discriminatory power than the current clinical marker CA19-9 for the discrimination of pancreato-biliary cancers from non-cancerous pancreatitis patients and clinical controls. This workflow might be used in future diagnostics for the detection of precancerous lesions, e.g. the identification of high grade IPMNs vs. low grade IPMNs.
Collapse
Affiliation(s)
- Christina Hartwig
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Institute for Interfacial Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Jan Müller
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- Center for Integrative Bioinformatics Vienna (CIBIV), Max Perutz Labs, University of Vienna and Medical University of Vienna, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | | | - Dina Kouhestani
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anke Mittelstädt
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anna Anthuber
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Paul David
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Maximilian Brunner
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Anne Jacobsen
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Karolina Glanz
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Izabela Swierzy
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lotta Roßdeutsch
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Bettina Klösch
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | | | - Kai Sohn
- Innovation Field In-vitro Diagnostics, Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany.
| | - Georg F Weber
- Department of Surgery, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.
- Comprehensive Cancer Center, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| |
Collapse
|
4
|
Fernandes AS, Oliveira C, Reis RL, Martins A, Silva TH. Marine-Inspired Drugs and Biomaterials in the Perspective of Pancreatic Cancer Therapies. Mar Drugs 2022; 20:689. [PMID: 36355012 PMCID: PMC9698933 DOI: 10.3390/md20110689] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 05/12/2024] Open
Abstract
Despite its low prevalence, pancreatic cancer (PC) is one of the deadliest, typically characterised as silent in early stages and with a dramatically poor prognosis when in its advanced stages, commonly associated with a high degree of metastasis. Many efforts have been made in pursuing innovative therapeutical approaches, from the search for new cytotoxic drugs and other bioactive compounds, to the development of more targeted approaches, including improved drug delivery devices. Marine biotechnology has been contributing to this quest by providing new chemical leads and materials originating from different organisms. In this review, marine biodiscovery for PC is addressed, particularly regarding marine invertebrates (namely sponges, molluscs, and bryozoans), seaweeds, fungi, and bacteria. In addition, the development of biomaterials based on marine-originating compounds, particularly chitosan, fucoidan, and alginate, for the production of advanced cancer therapies, is also discussed. The key role that drug delivery can play in new cancer treatments is highlighted, as therapeutical outcomes need to be improved to give further hope to patients.
Collapse
Affiliation(s)
- Andreia S. Fernandes
- 3B’s Research Group, I3Bs–Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Catarina Oliveira
- 3B’s Research Group, I3Bs–Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs–Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Albino Martins
- 3B’s Research Group, I3Bs–Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Tiago H. Silva
- 3B’s Research Group, I3Bs–Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| |
Collapse
|
5
|
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
|
6
|
Bizzozero L, Pergolizzi M, Pascal D, Maldi E, Villari G, Erriquez J, Volante M, Serini G, Marchiò C, Bussolino F, Arese M. Tumoral Neuroligin 1 Promotes Cancer-Nerve Interactions and Synergizes with the Glial Cell Line-Derived Neurotrophic Factor. Cells 2022; 11:280. [PMID: 35053395 PMCID: PMC8774081 DOI: 10.3390/cells11020280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Many nervous proteins are expressed in cancer cells. In this report, we asked whether the synaptic protein neuroligin 1 (NLGN1) was expressed by prostatic and pancreatic carcinomas; in addition, given the tendency of these tumors to interact with nerves, we asked whether NLGN1 played a role in this process. Through immunohistochemistry on human tissue microarrays, we showed that NLGN1 is expressed by prostatic and pancreatic cancer tissues in discrete stages and tumor districts. Next, we performed in vitro and in vivo assays, demonstrating that NLGN1 promotes cancer cell invasion and migration along nerves. Because of the established role of the neurotrophic factor glial cell line-derived neurotrophic factor (GDNF) in tumor-nerve interactions, we assessed a potential NLGN1-GDNF cooperation. We found that blocking GDNF activity with a specific antibody completely inhibited NLGN1-induced in vitro cancer cell invasion of nerves. Finally, we demonstrated that, in the presence of NLGN1, GDNF markedly activates cofilin, a cytoskeletal regulatory protein, altering filopodia dynamics. In conclusion, our data further prove the existence of a molecular and functional cross-talk between the nervous system and cancer cells. NLGN1 was shown here to function along one of the most represented neurotrophic factors in the nerve microenvironment, possibly opening new therapeutic avenues.
Collapse
Affiliation(s)
- Laura Bizzozero
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Margherita Pergolizzi
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Davide Pascal
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Elena Maldi
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (E.M.); (C.M.)
| | - Giulia Villari
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | | | - Marco Volante
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
| | - Guido Serini
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Caterina Marchiò
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy; (E.M.); (C.M.)
- Department of Medical Sciences, University of Turin, 10124 Turin, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| | - Marco Arese
- Department of Oncology, University of Torino, 10060 Candiolo, Italy; (L.B.); (M.P.); (D.P.); (G.V.); (M.V.); (G.S.); (F.B.)
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, Italy;
| |
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
|
Marine bacterial exopolysaccharide EPS11 inhibits migration and invasion of liver cancer cells by directly targeting collagen I. J Biol Chem 2021; 297:101133. [PMID: 34461092 PMCID: PMC8449266 DOI: 10.1016/j.jbc.2021.101133] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/20/2021] [Accepted: 08/26/2021] [Indexed: 02/02/2023] Open
Abstract
Many natural polysaccharides have significant anticancer activity with low toxicity, but the complex chemical structures make in-depth studies of the involved mechanisms extremely difficult. The purpose of this study was to investigate the effect of the marine bacterial exopolysaccharide (exopolysaccharide 11 [EPS11]) on liver cancer metastasis to explore the underlying target protein and molecular mechanism. We found that EPS11 significantly suppressed cell adhesion, migration, and invasion in liver cancer cells. Proteomic analysis showed that EPS11 induced downregulation of proteins related to the extracellular matrix–receptor interaction signaling pathway. In addition, the direct pharmacological target of EPS11 was identified as collagen I using cellular thermal shift assays. Surface plasmon resonance and pull-down assays further confirmed the specific binding of EPS11 to collagen I. Moreover, EPS11 was shown to inhibit tumor metastasis by directly modulating collagen I activity via the β1-integrin–mediated signaling pathway. Collectively, our study demonstrated for the first time that collagen I could be a direct pharmacological target of polysaccharide drugs. Moreover, directly targeting collagen I may be a promising strategy for finding novel carbohydrate-based drugs.
Collapse
|
9
|
Kamionka EM, Qian B, Gross W, Bergmann F, Hackert T, Beretta CA, Dross N, Ryschich E. Collagen Organization Does Not Influence T-Cell Distribution in Stroma of Human Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13153648. [PMID: 34359549 PMCID: PMC8344977 DOI: 10.3390/cancers13153648] [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: 05/20/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The excessive desmoplasia is the hallmark of human pancreatic cancer that influences the local T-cell-based immune response. In the present work, the stromal collagen organization in normal and malignant pancreatic tissues as well as its relationsship to T-cell distribution in pancreatic cancer were studied. It was found that differences in collagen organization do not change the spatial orientation of T-cell migration and do not influence the availability of tumor cells for T-cells. The results of the study do not support the concept of use of stroma collagen organization for improvement of spatial T-cell distribution in the tumor. Abstract The dominant intrastromal T-cell infiltration in pancreatic cancer is mainly caused by the contact guidance through the excessive desmoplastic reaction and could represent one of the obstacles to an effective immune response in this tumor type. This study analyzed the collagen organization in normal and malignant pancreatic tissues as well as its influence on T-cell distribution in pancreatic cancer. Human pancreatic tissue was analyzed using immunofluorescence staining and multiphoton and SHG microscopy supported by multistep image processing. The influence of collagen alignment on activated T-cells was studied using 3D matrices and time-lapse microscopy. It was found that the stroma of malignant and normal pancreatic tissues was characterized by complex individual organization. T-cells were heterogeneously distributed in pancreatic cancer and there was no relationship between T-cell distribution and collagen organization. There was a difference in the angular orientation of collagen alignment in the peritumoral and tumor-cell-distant stroma regions in the pancreatic ductal adenocarcinoma tissue, but there was no correlation in the T-cell densities between these regions. The grade of collagen alignment did not influence the directionality of T-cell migration in the 3D collagen matrix. It can be concluded that differences in collagen organization do not change the spatial orientation of T-cell migration or influence stromal T-cell distribution in human pancreatic cancer. The results of the present study do not support the rationale of remodeling of stroma collagen organization for improvement of T-cell–tumor cell contact in pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Eva-Maria Kamionka
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Baifeng Qian
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Wolfgang Gross
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Frank Bergmann
- Department of Pathology, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany;
| | - Thilo Hackert
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
| | - Carlo A. Beretta
- CellNetworks Math-Clinic, University of Heidelberg, Bioquant BQ001, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany;
- Institute for Anatomy and Cell Biology, University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Nicolas Dross
- Nikon Imaging Center, University of Heidelberg, 69120 Heidelberg, Germany;
| | - Eduard Ryschich
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Im Neuenheimer Feld 365/420, 69120 Heidelberg, Germany; (E.-M.K.); (B.Q.); (W.G.); (T.H.)
- Correspondence: ; Tel.: +49-6221-56-6110; Fax: +49-6221-56-5199
| |
Collapse
|
10
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
11
|
Morphological Heterogeneity in Pancreatic Cancer Reflects Structural and Functional Divergence. Cancers (Basel) 2021; 13:cancers13040895. [PMID: 33672734 PMCID: PMC7924365 DOI: 10.3390/cancers13040895] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Pancreatic cancer has a poor prognosis, which is largely due to resistance to treatment. Tumor heterogeneity is a known cause for treatment failure and has been studied at the molecular level. Morphological heterogeneity is common but has not been investigated, despite the fact that pathology examination is an integral part of clinical diagnostics. This study assessed whether morphological heterogeneity reflects structural and functional diversity in key cancer biological processes. Using archival tissues from resected pancreatic cancer, we selected four common and distinct morphological phenotypes and demonstrated that these differed significantly for a panel of 26 structural and functional features of the cancer-cell and stromal compartments. The strong link between these features and morphological phenotypes allowed prediction of the latter based on the results for the panel of features. The findings of this study indicate that morphological heterogeneity reflects biological diversity and that its assessment may potentially provide clinically relevant information. Abstract Inter- and intratumor heterogeneity is an important cause of treatment failure. In human pancreatic cancer (PC), heterogeneity has been investigated almost exclusively at the genomic and transcriptional level. Morphological heterogeneity, though prominent and potentially easily assessable in clinical practice, remains unexplored. This proof-of-concept study aims at demonstrating that morphological heterogeneity reflects structural and functional divergence. From the wide morphological spectrum of conventional PC, four common and distinctive patterns were investigated in 233 foci from 39 surgical specimens. Twenty-six features involved in key biological processes in PC were analyzed (immuno-)histochemically and morphometrically: cancer cell proliferation (Ki67) and migration (collagen fiber alignment, MMP14), cancer stem cells (CD44, CD133, ALDH1), amount, composition and spatial arrangement of extracellular matrix (epithelial proximity, total collagen, collagen I and III, fibronectin, hyaluronan), cancer-associated fibroblasts (density, αSMA), and cancer-stroma interactions (integrins α2, α5, α1; caveolin-1). All features differed significantly between at least two of the patterns. Stromal and cancer-cell-related features co-varied with morphology and allowed prediction of the morphological pattern. In conclusion, morphological heterogeneity in the cancer-cell and stromal compartments of PC correlates with structural and functional diversity. As such, histopathology has the potential to inform on the operationality of key biological processes in individual tumors.
Collapse
|
12
|
Sala M, Ros M, Saltel F. A Complex and Evolutive Character: Two Face Aspects of ECM in Tumor Progression. Front Oncol 2020; 10:1620. [PMID: 32984031 PMCID: PMC7485352 DOI: 10.3389/fonc.2020.01620] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor microenvironment, including extracellular matrix (ECM) and stromal cells, is a key player during tumor development, from initiation, growth and progression to metastasis. During all of these steps, remodeling of matrix components occurs, changing its biochemical and physical properties. The global and basic cancer ECM model is that tumors are surrounded by activated stromal cells, that remodel physiological ECM to evolve into a stiffer and more crosslinked ECM than in normal conditions, thereby increasing invasive capacities of cancer cells. In this review, we show that this too simple model does not consider the complexity, specificity and heterogeneity of each organ and tumor. First, we describe the general ECM in context of cancer. Then, we go through five invasive and most frequent cancers from different origins (breast, liver, pancreas, colon, and skin), and show that each cancer has its own specific matrix, with different stromal cells, ECM components, biochemical properties and activated signaling pathways. Furthermore, in these five cancers, we describe the dual role of tumor ECM: as a protective barrier against tumor cell proliferation and invasion, and as a major player in tumor progression. Indeed, crosstalk between tumor and stromal cells induce changes in matrix organization by remodeling ECM through invadosome formation in order to degrade it, promoting tumor progression and cell invasion. To sum up, in this review, we highlight the specificities of matrix composition in five cancers and the necessity not to consider the ECM as one general and simple entity, but one complex, dynamic and specific entity for each cancer type and subtype.
Collapse
|
13
|
Jiang HY, Najmeh S, Martel G, MacFadden-Murphy E, Farias R, Savage P, Leone A, Roussel L, Cools-Lartigue J, Gowing S, Berube J, Giannias B, Bourdeau F, Chan CHF, Spicer JD, McClure R, Park M, Rousseau S, Ferri LE. Activation of the pattern recognition receptor NOD1 augments colon cancer metastasis. Protein Cell 2020; 11:187-201. [PMID: 31956962 PMCID: PMC7026222 DOI: 10.1007/s13238-019-00687-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
While emerging data suggest nucleotide oligomerization domain receptor 1 (NOD1), a cytoplasmic pattern recognition receptor, may play an important and complementary role in the immune response to bacterial infection, its role in cancer metastasis is entirely unknown. Hence, we sought to determine the effects of NOD1 on metastasis. NOD1 expression in paired human primary colon cancer, human and murine colon cancer cells were determined using immunohistochemistry and immunoblotting (WB). Clinical significance of NOD1 was assessed using TCGA survival data. A series of in vitro and in vivo functional assays, including adhesion, migration, and metastasis, was conducted to assess the effect of NOD1. C12-iE-DAP, a highly selective NOD1 ligand derived from gram-negative bacteria, was used to activate NOD1. ML130, a specific NOD1 inhibitor, was used to block C12-iE-DAP stimulation. Stable knockdown (KD) of NOD1 in human colon cancer cells (HT29) was constructed with shRNA lentiviral transduction and the functional assays were thus repeated. Lastly, the predominant signaling pathway of NOD1-activation was identified using WB and functional assays in the presence of specific kinase inhibitors. Our data demonstrate that NOD1 is highly expressed in human colorectal cancer (CRC) and human and murine CRC cell lines. Clinically, we demonstrate that this increased NOD1 expression negatively impacts survival in patients with CRC. Subsequently, we identify NOD1 activation by C12-iE-DAP augments CRC cell adhesion, migration and metastasis. These effects are predominantly mediated via the p38 mitogen activated protein kinase (MAPK) pathway. This is the first study implicating NOD1 in cancer metastasis, and thus identifying this receptor as a putative therapeutic target.
Collapse
Affiliation(s)
- Henry Y Jiang
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Sara Najmeh
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Guy Martel
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Elyse MacFadden-Murphy
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Raquel Farias
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Paul Savage
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, QC, H3A 1A3, Canada
| | - Arielle Leone
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Lucie Roussel
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Jonathan Cools-Lartigue
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Stephen Gowing
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Julie Berube
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Betty Giannias
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada
| | - France Bourdeau
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada
| | - Carlos H F Chan
- Department of Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Jonathan D Spicer
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Rebecca McClure
- Department of Pathology, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, Canada
| | - Morag Park
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, QC, H3A 1A3, Canada
| | - Simon Rousseau
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Lorenzo E Ferri
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada. .,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
| |
Collapse
|
14
|
Ma F, Tremmel DM, Li Z, Lietz CB, Sackett SD, Odorico JS, Li L. In Depth Quantification of Extracellular Matrix Proteins from Human Pancreas. J Proteome Res 2019; 18:3156-3165. [PMID: 31200599 DOI: 10.1021/acs.jproteome.9b00241] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Extracellular matrix (ECM) is an important component of the pancreatic microenvironment which regulates β cell proliferation, differentiation, and insulin secretion. Protocols have recently been developed for the decellularization of the human pancreas to generate functional scaffolds and hydrogels. In this work, we characterized human pancreatic ECM composition before and after decellularization using isobaric dimethylated leucine (DiLeu) labeling for relative quantification of ECM proteins. A novel correction factor was employed in the study to eliminate the bias introduced during sample preparation. In comparison to the commonly employed sample preparation methods (urea and FASP) for proteomic analysis, a recently developed surfactant and chaotropic agent assisted sequential extraction/on pellet digestion (SCAD) protocol has provided an improved strategy for ECM protein extraction of human pancreatic ECM matrix. The quantitative proteomic results revealed the preservation of matrisome proteins while most of the cellular proteins were removed. This method was compared with a well-established label-free quantification (LFQ) approach which rendered similar expressions of different categories of proteins (collagens, ECM glycoproteins, proteoglycans, etc.). The distinct expression of ECM proteins was quantified comparing adult and fetal pancreas ECM, shedding light on the correlation between matrix composition and postnatal β cell maturation. Despite the distinct profiles of different subcategories in the native pancreas, the distribution of matrisome proteins exhibited similar trends after the decellularization process. Our method generated a large data set of matrisome proteins from a single tissue type. These results provide valuable insight into the possibilities of constructing a bioengineered pancreas. It may also facilitate better understanding of the potential roles that matrisome proteins play in postnatal β cell maturation.
Collapse
Affiliation(s)
- Fengfei Ma
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Daniel M Tremmel
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Zihui Li
- Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | - Christopher B Lietz
- Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| | - Sara Dutton Sackett
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Jon S Odorico
- Department of Surgery, Division of Transplantation, School of Medicine and Public Health , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States
| | - Lingjun Li
- School of Pharmacy , University of Wisconsin-Madison , Madison , Wisconsin 53705 , United States.,Department of Chemistry , University of Wisconsin-Madison , Madison , Wisconsin 53706 , United States
| |
Collapse
|
15
|
Casadonte R, Kriegsmann M, Perren A, Baretton G, Deininger S, Kriegsmann K, Welsch T, Pilarsky C, Kriegsmann J. Development of a Class Prediction Model to Discriminate Pancreatic Ductal Adenocarcinoma from Pancreatic Neuroendocrine Tumor by MALDI Mass Spectrometry Imaging. Proteomics Clin Appl 2018; 13:e1800046. [DOI: 10.1002/prca.201800046] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 11/05/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Mark Kriegsmann
- Institute of PathologyUniversity of Heidelberg Heidelberg 69120 Germany
| | - Aurel Perren
- Institute of PathologyUniversity of Bern Bern 3012 Switzerland
| | - Gustavo Baretton
- Institute of PathologyUniversity Hospital Carl Gustav Carus at the Technical University of Dresden Dresden 01307 Germany
| | | | - Katharina Kriegsmann
- Department of HematologyOncology and RheumatologyUniversity of Heidelberg Heidelberg 69120 Germany
| | - Thilo Welsch
- Institute of PathologyUniversity Hospital Carl Gustav Carus at the Technical University of Dresden Dresden 01307 Germany
| | - Christian Pilarsky
- Institute of PathologyUniversity Hospital Carl Gustav Carus at the Technical University of Dresden Dresden 01307 Germany
| | - Jörg Kriegsmann
- Proteopath GmbH Trier 54296 Germany
- MVZ for HistologyCytology and Molecular Diagnostics Trier 54296 Germany
| |
Collapse
|
16
|
Hadavi E, Leijten J, Brinkmann J, Jonkheijm P, Karperien M, van Apeldoorn A. Fibronectin and Collagen IV Microcontact Printing Improves Insulin Secretion by INS1E Cells. Tissue Eng Part C Methods 2018; 24:628-636. [PMID: 30306836 DOI: 10.1089/ten.tec.2018.0151] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
IMPACT STATEMENT This research deals with finding a proper bioengineering strategy for the creation of improved β-cell replacement therapy in type 1 diabetes. It specifically deals with the microenvironment of β-cells and its relationship to their endocrine function.
Collapse
Affiliation(s)
- Elahe Hadavi
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Jeroen Leijten
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Jenny Brinkmann
- 2 MESA+ Institute for Nanotechnology, Molecular Nanofabrication Group, University of Twente , Enschede, The Netherlands
| | - Pascal Jonkheijm
- 2 MESA+ Institute for Nanotechnology, Molecular Nanofabrication Group, University of Twente , Enschede, The Netherlands
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands
| | - Aart van Apeldoorn
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente , Enschede, The Netherlands .,3 Complex Tissue Regeneration Department, MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University , Maastricht, The Netherlands
| |
Collapse
|
17
|
Ma L, Zhang B, Zhou C, Li Y, Li B, Yu M, Luo Y, Gao L, Zhang D, Xue Q, Qiu Q, Lin B, Zou J, Yang H. The comparison genomics analysis with glioblastoma multiforme (GBM) cells under 3D and 2D cell culture conditions. Colloids Surf B Biointerfaces 2018; 172:665-673. [PMID: 30243220 DOI: 10.1016/j.colsurfb.2018.09.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/27/2018] [Accepted: 09/14/2018] [Indexed: 12/16/2022]
Abstract
GBM, the most common and aggressive malignant primary brain tumors which needs new research approach to reveal the underline molecular mechanism of tumor progression. The 3D in vitro tumor model can be a simple and effective way to study tumor characteristics with ability to replicate of the tumor milieu. In the current study, we adopted the DNA microarray to analyze the gene expression of GBM tumor cells cultured under 2D cell culture flasks and 3D PLA porous scaffolds for 4,7 and 14 days. For 14 day old cultures, 8117 and 3060 genes expression were upregulated and downregulated respectively. Further KEGG pathway analysis revealed, the upregulated genes were mainly enriched/involved in PPAR and PI3K-Akt signaling pathways whereas the downregulated genes were mainly contributed in metabolism, ECM related and TGF-beta pathways. Thus, our approach of establishing 3D in vitro tumor model provides realistic results and proves itself a powerful tool for understanding the inner nature of GBM and can be considered as potential platform for drug screening.
Collapse
Affiliation(s)
- Liang Ma
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| | - Bin Zhang
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| | - Changchun Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Yuting Li
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Binjie Li
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, People's Republic of China
| | - Yichen Luo
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Lei Gao
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Trumpington Street, Cambridge, CB2 1PZ, United Kingdom
| | - Qian Xue
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Qingchong Qiu
- Zhejiang California International NanoSystems Institute, Zhejiang University, Hangzhou, 310029, People's Republic of China
| | - Biaoyang Lin
- Zhejiang California International NanoSystems Institute, Zhejiang University, Hangzhou, 310029, People's Republic of China
| | - Jun Zou
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Huayong Yang
- State Key Lab of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, People's Republic of China; School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, People's Republic of China
| |
Collapse
|
18
|
The Extracellular Matrix and Pancreatic Cancer: A Complex Relationship. Cancers (Basel) 2018; 10:cancers10090316. [PMID: 30200666 PMCID: PMC6162452 DOI: 10.3390/cancers10090316] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/31/2018] [Accepted: 09/02/2018] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extraordinarily dense fibrotic stroma that impedes tumor perfusion and delivery of anticancer drugs. Since the extracellular matrix (ECM) comprises the bulk of the stroma, it is primarily responsible for the increased interstitial tissue pressure and stiff mechanical properties of the stroma. Besides its mechanical influence, the ECM provides important biochemical and physical cues that promote survival, proliferation, and metastasis. By serving as a nutritional source, the ECM also enables PDAC cells to survive under the nutrient-poor conditions. While therapeutic strategies using stroma-depleting drugs have yielded disappointing results, an increasing body of research indicates the ECM may offer a variety of potential therapeutic targets. As preclinical studies of ECM-targeted drugs have shown promising effects, a number of clinical trials are currently investigating agents with the potential to advance the future treatment of PDAC. Thus, the present review seeks to give an overview of the complex relationship between the ECM and PDAC.
Collapse
|
19
|
Ansari D, Friess H, Bauden M, Samnegård J, Andersson R. Pancreatic cancer: disease dynamics, tumor biology and the role of the microenvironment. Oncotarget 2018; 9:6644-6651. [PMID: 29464100 PMCID: PMC5814240 DOI: 10.18632/oncotarget.24019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is known for its propensity to metastasize. Recent studies have challenged the commonly held belief that pancreatic cancer is a stepwise process, where tumor cells disseminate late in primary tumor development. Instead it has been suggested that pancreatic tumor cells may disseminate early and develop independently and in parallel to the primary tumor. Circulating tumor cells can be found in most patients with pancreatic cancer, even in those with localized stage. Also, recent phylogenetic analyses have revealed evidence for a branched evolution where metastatic lineages can develop early in tumor development. In this Review, we discuss current models of pancreatic cancer progression and the importance of the tumor microenvironment, in order to better understand the recalcitrant nature of this disease.
Collapse
Affiliation(s)
- Daniel Ansari
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Helmut Friess
- Department of Surgery, Technical University of Munich, Munich, Germany
| | - Monika Bauden
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Johan Samnegård
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Roland Andersson
- Department of Surgery, Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
20
|
Duchalais E, Guilluy C, Nedellec S, Touvron M, Bessard A, Touchefeu Y, Bossard C, Boudin H, Louarn G, Neunlist M, Van Landeghem L. Colorectal Cancer Cells Adhere to and Migrate Along the Neurons of the Enteric Nervous System. Cell Mol Gastroenterol Hepatol 2017; 5:31-49. [PMID: 29188232 PMCID: PMC5696385 DOI: 10.1016/j.jcmgh.2017.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/02/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND & AIMS In several types of cancers, tumor cells invade adjacent tissues by migrating along the resident nerves of the tumor microenvironment. This process, called perineural invasion, typically occurs along extrinsic nerves, with Schwann cells providing physical guidance for the tumor cells. However, in the colorectal cancer microenvironment, the most abundant nervous structures belong to the nonmyelinated intrinsic enteric nervous system (ENS). In this study, we investigated whether colon cancer cells interact with the ENS. METHODS Tumor epithelial cells (TECs) from human primary colon adenocarcinomas and cell lines were cocultured with primary cultures of ENS and cultures of human ENS plexus explants. By combining confocal and atomic force microscopy, as well as video microscopy, we assessed tumor cell adhesion and migration on the ENS. We identified the adhesion proteins involved using a proteomics approach based on biotin/streptavidin interaction, and their implication was confirmed further using selective blocking antibodies. RESULTS TEC adhered preferentially and with stronger adhesion forces to enteric nervous structures than to mesenchymal cells. TEC adhesion to ENS involved direct interactions with enteric neurons. Enteric neuron removal from ENS cultures led to a significant decrease in tumor cell adhesion. TECs migrated significantly longer and further when adherent on ENS compared with on mesenchymal cells, and their trajectory faithfully followed ENS structures. Blocking N-cadherin and L1CAM decreased TEC migration along ENS structures. CONCLUSIONS Our data show that the enteric neuronal network guides tumor cell migration, partly via L1CAM and N-cadherin. These results open a new avenue of research on the underlying mechanisms and consequences of perineural invasion in colorectal cancer.
Collapse
Key Words
- AFM, atomic force microscope
- Adhesion
- Colorectal Cancer
- DMEM, Dulbecco's modified Eagle medium
- ENS, enteric nervous system
- Enteric Neurons
- GFP, green fluorescent protein
- MCS, multiple cloning site
- Migration
- PBS, phosphate-buffered saline
- TEC, tumor epithelial cell
- Tuj, tubulin III
- pcENS, primary culture enteric nervous system
- α-SMA, α–smooth muscle actin
Collapse
Affiliation(s)
- Emilie Duchalais
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Université de Nantes, Nantes, France
- Clinique de Chirurgie Digestive et Endocrinienne, Centre Hospitalier Universitaire de Nantes, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Correspondence Address correspondence to: Emilie Duchalais, MD, Inserm U1235, 1 Rue Gaston Veil, 44000 Nantes, France. fax: +33 2 40 41 11 10.Inserm U12351 Rue Gaston VeilNantes44000France
| | | | - Steven Nedellec
- Université de Nantes, Nantes, France
- Micropicell, Nantes, France
| | - Melissa Touvron
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - Anne Bessard
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Université de Nantes, Nantes, France
| | - Yann Touchefeu
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Université de Nantes, Nantes, France
| | - Céline Bossard
- Université de Nantes, Nantes, France
- Service d’Anatomie et Cytologie Pathologiques, Centre Hospitalier Universitaire de Nantes, France
| | - Hélène Boudin
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Université de Nantes, Nantes, France
| | - Guy Louarn
- Université de Nantes, Nantes, France
- Institut des Matériaux Jean Rouxel, Centre National de la Recherche Scientifique, Nantes, France
| | - Michel Neunlist
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Université de Nantes, Nantes, France
| | - Laurianne Van Landeghem
- Inserm U1235, Institut des Maladies de l'Appareil Digestif, Nantes, France
- Université de Nantes, Nantes, France
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
21
|
Drifka CR, Loeffler AG, Esquibel CR, Weber SM, Eliceiri KW, Kao WJ. Human pancreatic stellate cells modulate 3D collagen alignment to promote the migration of pancreatic ductal adenocarcinoma cells. Biomed Microdevices 2017; 18:105. [PMID: 27819128 DOI: 10.1007/s10544-016-0128-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the ability for cancer cells to aggressively infiltrate and navigate through a dense stroma during the metastatic process. Key features of the PDAC stroma include an abundant population of activated pancreatic stellate cells (PSCs) and highly aligned collagen fibers; however, important questions remain regarding how collagen becomes aligned and what the biological manifestations are. To better understand how PSCs, aligned collagen, and PDAC cells might cooperate during the transition to invasion, we utilized a microchannel-based in vitro tumor model and advanced imaging technologies to recreate and examine in vivo-like heterotypic interactions. We found that PSCs participate in a collaborative process with cancer cells by orchestrating the alignment of collagen fibers that, in turn, are permissive to enhanced cell migration. Additionally, direct contact between PSCs, collagen, and PDAC cells is critical to invasion and co-migration of both cell types. This suggests PSCs may accompany and assist in navigating PDAC cells through the stromal terrain. Together, our data provides a new role for PSCs in stimulating the metastatic process and underscores the importance of collagen alignment in cancer progression.
Collapse
Affiliation(s)
- Cole R Drifka
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin, Madison, WI, USA.,Morgridge Institute for Research, Madison, WI, USA
| | - Agnes G Loeffler
- Department of Surgical Pathology, University of Wisconsin, Madison, WI, USA.,University of Wisconsin Comprehensive Carbone Cancer Center, Madison, WI, USA
| | - Corinne R Esquibel
- Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin, Madison, WI, USA
| | - Sharon M Weber
- University of Wisconsin Comprehensive Carbone Cancer Center, Madison, WI, USA.,Department of Surgery, University of Wisconsin, Madison, WI, USA
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,Laboratory for Optical and Computational Instrumentation (LOCI), University of Wisconsin, Madison, WI, USA.,Morgridge Institute for Research, Madison, WI, USA.,University of Wisconsin Comprehensive Carbone Cancer Center, Madison, WI, USA
| | - W John Kao
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. .,University of Wisconsin Comprehensive Carbone Cancer Center, Madison, WI, USA. .,Department of Surgery, University of Wisconsin, Madison, WI, USA. .,Faculties of Medicine and Engineering, University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong.
| |
Collapse
|
22
|
Carpenter PM, Sivadas P, Hua SS, Xiao C, Gutierrez AB, Ngo T, Gershon PD. Migration of breast cancer cell lines in response to pulmonary laminin 332. Cancer Med 2017; 6:220-234. [PMID: 27878981 PMCID: PMC5269569 DOI: 10.1002/cam4.957] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 10/10/2016] [Accepted: 10/14/2016] [Indexed: 02/06/2023] Open
Abstract
Because tumor cell motility is a requirement for metastasis, we hypothesized that lung tissue harbors substances that induce tumor cell migration. MCF-7 breast carcinoma cells exposed to small airway epithelial cells and conditioned medium exhibited dose-dependent tumor cell migration. Among the extracellular matrix proteins in the conditioned medium identified by mass spectrometry, laminin 332 (LM332) had the greatest contribution to the migration of MCF-7 cells. Immunoblotting and immunohistochemistry for LM332-specific chains identified LM332 in the lung and in pulmonary epithelial cells. Antibodies to either LM332 or its integrin receptor inhibited MCF-7 motility, and knockdown of LM332 chains also reduced its migration-inducing activity. Taken together, these findings implicate LM332 as a component of lung tissue that can induce motility in breast carcinoma cells that have been transported to lung during metastasis. Earlier studies on LM332 in tumor progression have examined LM332 expression in tumor cells. This investigation, in comparison, provides evidence that the tumor promoting potential of LM332 may originate in the lung microenvironment rather than in tumor cells alone. Furthermore, this study provides evidence that the motility-inducing properties of the microenvironment can reside in epithelial cells. The findings raise the possibility that LM332 plays a role in the pulmonary metastases of breast carcinoma and may provide a target for antimetastasis therapy.
Collapse
Affiliation(s)
- Philip M. Carpenter
- Department of PathologyKeck School of Medicine, the University of Southern CaliforniaLos AngelesCalifornia
- Department of PathologyUniversity of CaliforniaIrvineCalifornia
| | - Priyanka Sivadas
- Department of PathologyKeck School of Medicine, the University of Southern CaliforniaLos AngelesCalifornia
| | - Spencer S. Hua
- Department of PathologyUniversity of CaliforniaIrvineCalifornia
| | - Cally Xiao
- Department of Pharmacology and Experimental TherapeuticsUniversity Hospital of CologneCologneGermany
| | | | - Tuan Ngo
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCalifornia
| | - Paul D. Gershon
- Department of Molecular Biology and BiochemistryUniversity of CaliforniaIrvineCalifornia
| |
Collapse
|
23
|
Drifka CR, Loeffler AG, Mathewson K, Keikhosravi A, Eickhoff JC, Liu Y, Weber SM, Kao WJ, Eliceiri KW. Highly aligned stromal collagen is a negative prognostic factor following pancreatic ductal adenocarcinoma resection. Oncotarget 2016; 7:76197-76213. [PMID: 27776346 PMCID: PMC5342807 DOI: 10.18632/oncotarget.12772] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/03/2016] [Indexed: 01/18/2023] Open
Abstract
Risk factors for pancreatic ductal adenocarcinoma (PDAC) progression after surgery are unclear, and additional prognostic factors are needed to inform treatment regimens and therapeutic targets. PDAC is characterized by advanced sclerosis of the extracellular matrix, and interactions between cancer cells, fibrillar collagen, and other stromal components play an integral role in progression. Changes in stromal collagen alignment have been shown to modulate cancer cell behavior and have important clinical value in other cancer types, but little is known about its role in PDAC and prognostic value. We hypothesized that the alignment of collagen is associated with PDAC patient survival. To address this, pathology-confirmed tissues from 114 PDAC patients that underwent curative-intent surgery were retrospectively imaged with Second Harmonic Generation (SHG) microscopy, quantified with fiber segmentation algorithms, and correlated to patient survival. The same tissue regions were analyzed for epithelial-to-mesenchymal (EMT), α-SMA, and syndecan-1 using complimentary immunohistostaining and visualization techniques. Significant inter-tumoral variation in collagen alignment was found, and notably high collagen alignment was observed in 12% of the patient cohort. Stratification of patients according to collagen alignment revealed that high alignment is an independent negative factor following PDAC resection (p = 0.0153, multivariate). We also found that epithelial expression of EMT and the stromal expression of α-SMA and syndecan-1 were positively correlated with collagen alignment. In summary, stromal collagen alignment may provide additional, clinically-relevant information about PDAC tumors and underscores the importance of stroma-cancer interactions.
Collapse
Affiliation(s)
- Cole R. Drifka
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Agnes G. Loeffler
- Department of Surgical Pathology, University of Wisconsin, Madison, WI, USA
| | - Kara Mathewson
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
| | - Adib Keikhosravi
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
| | - Jens C. Eickhoff
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, WI, USA
| | - Yuming Liu
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
| | - Sharon M. Weber
- Department of Surgery, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - W. John Kao
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Department of Surgery, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
24
|
Abstract
OBJECTIVES Differentiation of pancreatic cancer (PCA) from chronic pancreatitis (CP) is challenging. We searched for peptide markers in urine to develop a diagnostic peptide marker model. METHODS Capillary electrophoresis-mass spectrometry was used to search for peptides in urine of patients with PCA (n = 39) or CP (n = 41). Statistical different peptides were included in a peptide multimarker model. Peptide markers were sequence identified and validated by immunoassay and immunohistochemistry (IHC). RESULTS Applied to a validation cohort of 54 patients with PCA and 52 patients with CP, the peptide model correctly classified 47 patients with PCA and 44 patients with CP (area under the curve, 0.93; 87% sensitivity; 85% specificity). All 5 patients with PCA with concomitant CP were classified positive. Urine proteome analysis outperformed carbohydrate antigen 19-9 (area under the curve, 0.84) by a 15% increase in sensitivity at the same specificity. From 99 healthy subjects, only four were misclassified. Fetuin-A was the most prominent peptide marker source for PCA as verified by immunoassay and IHC. In silico protease mapping of the peptide markers' terminal sequences pointed to increased meprin-A activity in PCA, which in IHC was associated with neoangiogenesis. CONCLUSIONS Urinary proteome analysis differentiates PCA from CP and may serve as PCA screening tool.
Collapse
|
25
|
Influence of Immune Myeloid Cells on the Extracellular Matrix During Cancer Metastasis. CANCER MICROENVIRONMENT 2016; 9:45-61. [PMID: 26956475 PMCID: PMC4842183 DOI: 10.1007/s12307-016-0181-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/12/2016] [Indexed: 01/04/2023]
Abstract
The extracellular matrix (ECM) is one of the most important components within the tumor microenvironment that supports cancer development and metastasis. Under normal physiological conditions, the ECM is a tightly regulated network providing structural and biochemical support. However, the ECM becomes highly disorganized during neoplastic progression and consequently, stimulates cancer cell transformation, growth and spread. Cancer development and progression is also known to greatly benefit from the support of immune myeloid cells, which have multiple pro-tumorigenic functions including promoting tumor growth, migration and invasion, stimulating angiogenesis and suppressing anti-tumor responses. An increasing number of studies have shown that myeloid cells alter the ECM to support metastatic cancer progression and in turn, the ECM can influence the function of infiltrating myeloid cells. However, the exact nature of this relationship, such as the mechanisms employed and their molecular targets remains unclear. This review discusses evidence for the reciprocal dependence of myeloid cells and the tumor ECM for efficient tumor development and explores potential mechanisms involved in these interactions. A better understanding of this relationship has exciting implications for the development of new therapeutic treatments for metastatic cancer.
Collapse
|
26
|
Jolly LA, Novitskiy S, Owens P, Massoll N, Cheng N, Fang W, Moses HL, Franco AT. Fibroblast-Mediated Collagen Remodeling Within the Tumor Microenvironment Facilitates Progression of Thyroid Cancers Driven by BrafV600E and Pten Loss. Cancer Res 2016; 76:1804-13. [PMID: 26818109 DOI: 10.1158/0008-5472.can-15-2351] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/14/2016] [Indexed: 01/01/2023]
Abstract
Contributions of the tumor microenvironment (TME) to progression in thyroid cancer are largely unexplored and may illuminate a basis for understanding rarer aggressive cases of this disease. In this study, we investigated the relationship between the TME and thyroid cancer progression in a mouse model where thyroid-specific expression of oncogenic BRAF and loss of Pten (Braf(V600E)/Pten(-/-)/TPO-Cre) leads to papillary thyroid cancers (PTC) that rapidly progress to poorly differentiated thyroid cancer (PDTC). We found that fibroblasts were recruited to the TME of Braf(V600E)/Pten(-/-)/TPO-Cre thyroid tumors. Conditioned media from cell lines established from these tumors, but not tumors driven by mutant H-ras, induced fibroblast migration and proliferation in vitro Notably, the extracellular matrix of Braf(V600E)/Pten(-/-)/TPO-Cre tumors was enriched with stromal-derived fibrillar collagen, compared with wild-type or Hras-driven tumors. Further, type I collagen enhanced the motility of Braf(V600E)/Pten(-/-)/TPO-Cre tumor cells in vitro In clinical specimens, we found COL1A1 and LOX to be upregulated in PTC and expressed at highest levels in PDTC and anaplastic thyroid cancer. Additionally, increased expression levels of COL1A1 and LOX were associated with decreased survival in thyroid cancer patients. Overall, our results identified fibroblast recruitment and remodeling of the extracellular matrix as pivotal features of the TME in promoting thyroid cancer progression, illuminating candidate therapeutic targets and biomarkers in advanced forms of this malignancy. Cancer Res; 76(7); 1804-13. ©2016 AACR.
Collapse
Affiliation(s)
- Lee Ann Jolly
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sergey Novitskiy
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Phillip Owens
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Nicole Massoll
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nikki Cheng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Wei Fang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Harold L Moses
- Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Aime T Franco
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas.
| |
Collapse
|
27
|
Drifka CR, Tod J, Loeffler AG, Liu Y, Thomas GJ, Eliceiri KW, Kao WJ. Periductal stromal collagen topology of pancreatic ductal adenocarcinoma differs from that of normal and chronic pancreatitis. Mod Pathol 2015; 28:1470-80. [PMID: 26336888 DOI: 10.1038/modpathol.2015.97] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 07/16/2015] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma continues to be one of the most difficult diseases to manage with one of the highest cancer mortality rates. This is due to several factors including nonspecific symptomatology and subsequent diagnosis at an advanced stage, aggressive metastatic behavior that is incompletely understood, and limited response to current therapeutic regimens. As in other cancers, there is great interest in studying the role of the tumor microenvironment in pancreatic ductal adenocarcinoma and whether components of this environment could serve as research and therapeutic targets. In particular, attention has turned toward the desmoplastic collagen-rich pancreatic ductal adenocarcinoma stroma for both biological and clinical insight. In this study, we used quantitative second harmonic generation microscopy to investigate stromal collagen organization and structure in human pancreatic ductal adenocarcinoma pathology tissues compared with non-neoplastic tissues. Collagen topology was characterized in whole-tissue microarray cores and at specific pathology-annotated epithelial-stroma interfaces representing 241 and 117 patients, respectively. We quantitatively demonstrate that a unique collagen topology exists in the periductal pancreatic ductal adenocarcinoma stroma. Specifically, collagen around malignant ducts shows increased alignment, length, and width compared with normal ducts and benign ducts in a chronic pancreatitis background. These findings indicate that second harmonic generation imaging can provide quantitative information about fibrosis that complements traditional histopathologic insights and can serve as a rich field for investigation into pathogenic and clinical implications of reorganized collagen as a pancreatic ductal adenocarcinoma disease marker.
Collapse
Affiliation(s)
- Cole R Drifka
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
| | - Jo Tod
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Agnes G Loeffler
- Department of Pathology, University of Wisconsin, Madison, WI, USA.,Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Yuming Liu
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
| | - Gareth J Thomas
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA.,Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA
| | - W John Kao
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA.,Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA.,Department of Surgery, University of Wisconsin, Madison, WI, USA.,School of Pharmacy, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
28
|
Qiu F, Sun R, Deng N, Guo T, Cao Y, Yu Y, Wang X, Zou B, Zhang S, Jing T, Ling T, Xie J, Zhang Q. miR-29a/b enhances cell migration and invasion in nasopharyngeal carcinoma progression by regulating SPARC and COL3A1 gene expression. PLoS One 2015; 10:e0120969. [PMID: 25786138 PMCID: PMC4364736 DOI: 10.1371/journal.pone.0120969] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/27/2015] [Indexed: 12/15/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor associated with a genetic predisposition, Epstein-Barr virus infection and chromosomal abnormalities. Recently, several miRNAs have been shown to target specific mRNAs to regulate NPC development and progression. However, the involvement of miRNAs in processes leading to NPC migration and invasion remains to be elucidated. We predicted that miR-29a/b are associated with dysregulated genes controlling NPC through an integrated interaction network of miRNAs and genes. miR-29a/b over-expression in NPC cell lines had no significant effect on proliferation, whereas miR-29b mildly increased the percentage of cells in the G1 phase with a concomitant decrease in the percentage of cells in S phase. Furthermore, we demonstrated that miR-29a/b might be responsible for increasing S18 cell migration and invasion, and only COL3A1 was identified as a direct target of miR-29b despite the fact that both SPARC and COL3A1 were inhibited by miR-29a/b over-expression. Meanwhile, SPARC proteins were increased in metastatic NPC tissue and are involved in NPC progression. Unexpectedly, we identified that miRNA-29b expression was elevated in the serum of NPC patients with a high risk of metastasis. The 5-year actuarial overall survival rates in NPC patients with high serum miR-29b expression was significantly shorter than those with low serum miR-29b expression; therefore, serum miR-29b expression could be a promising prognostic marker.
Collapse
Affiliation(s)
- Feifei Qiu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rui Sun
- State Key Laboratory of Oncology in Southern China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ning Deng
- Key Laboratory of Molecular Immunology and Antibody Engineering of Guangdong Province, Antibody Engineering Center in Jinan University, Guangzhou, China
| | - Tianyu Guo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yange Cao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ying Yu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xuejun Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Bingcheng Zou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Songmei Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Tao Jing
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Tao Ling
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun Xie
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture, Pearl River Fisheries Research Institute of CAFS, Guangzhou, Guangdong, China
- * E-mail: (QZ); (JX)
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- * E-mail: (QZ); (JX)
| |
Collapse
|
29
|
Zhu W, Li W, Yang G, Fu C, Jiang G, Hu Q. Vitronectin silencing inhibits hepatocellular carcinoma in vitro and in vivo. Future Oncol 2014; 11:251-8. [PMID: 25179307 DOI: 10.2217/fon.14.202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To explore if inhibition of vitronectin can be used for the treatment of hepatocellular carcinoma. MATERIALS & METHODS RNAi technology was used to silence the expression of VTN in HepG2 and SMMC 7721 cells. Change of growth characteristics in these cells was evaluated. RESULTS VTN silencing does not affect growth characteristics of cancer cells in monolayer cell culture, but could suppress the colonized growth of cells in soft agar. VTN-siRNA suppresses colony formation more than 80% compared with that of control in SMMC7721cells and leads to the inhibition of colony formation of over 70% in HepG2 cells. In addition, VTN silencing decreases the size of tumor xenografts in nude mice, particularly in male mice, with an inhibition rate of 46.6%. CONCLUSION VTN plays a significant role in the malignant growth of tumor. Inhibition of VTN could potentially be applied for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Toxicology, Guangzhou Center for Disease Control & Prevention, Guangzhou, Guangdong 510080, PR China
| | | | | | | | | | | |
Collapse
|
30
|
Flate E, Stalvey JRD. Motility of select ovarian cancer cell lines: effect of extra-cellular matrix proteins and the involvement of PAK2. Int J Oncol 2014; 45:1401-11. [PMID: 25050916 PMCID: PMC4151804 DOI: 10.3892/ijo.2014.2553] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/11/2014] [Indexed: 12/18/2022] Open
Abstract
The interaction between tumor cells and extracellular matrix (ECM) proteins influences cell migration and the invasive behavior of cancer cells. In this study, we provide experimental evidence that collagen I and fibronectin affect ovarian cancer cell migration. In vitro wound healing assays and transwell migration assays were used to measure both total wound healing and directionality of individually migrating OV2008 and C13 ovarian cancer cells on glass, collagen I and fibronectin. Involvement of p21-activated kinase 2 (Pak2) in the motility of these cell lines was investigated using a chemical inhibitor as well as siRNA transfection. Culturing ovarian cancer cells on collagen type I (COLL) increased the migratory ability of OV2008 and C13 cells by increasing the directional migration of cells. In contrast, fibronectin (FN) decreased the migratory ability of OV2008 cells by reducing their ability to migrate directionally. When both cell lines are cultured on COLL and treated with increasing concentrations of a PAK inhibitor (IPA-3), there is a dose-dependent response such that there is a decrease in migration with an increase in inhibitor concentration. Further experiments utilizing PAK2 knockdown via siRNA transfection demonstrated significantly reduced migration of OV2008 cells on COLL as compared to those receiving control siRNA. In conclusion, our results indicate that FN and COLL affect the motility of the selected ovarian cancer cells lines and the effect of COLL is likely mediated, at least in part, by PAK2. A better understanding of the molecular events that contribute to tumor invasion and metastasis is crucial for developing novel treatment strategies to improve the long-term survival of women with ovarian cancer. As PAK2 is involved in motility, it should be further explored as a pro-metastatic gene in ovarian cancer.
Collapse
|
31
|
Stromnes IM, DelGiorno KE, Greenberg PD, Hingorani SR. Stromal reengineering to treat pancreas cancer. Carcinogenesis 2014; 35:1451-60. [PMID: 24908682 DOI: 10.1093/carcin/bgu115] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma co-opts multiple cellular and extracellular mechanisms to create a complex cancer organ with an unusual proclivity for metastasis and resistance to therapy. Cell-autonomous events are essential for the initiation and maintenance of pancreatic ductal adenocarcinoma, but recent studies have implicated critical non-cell autonomous processes within the robust desmoplastic stroma that promote disease pathogenesis and resistance. Thus, non-malignant cells and associated factors are culprits in tumor growth, immunosuppression and invasion. However, even this increasing awareness of non-cell autonomous contributions to disease progression is tempered by the conflicting roles stromal elements can play. A greater understanding of stromal complexity and complicity has been aided in part by studies in highly faithful genetically engineered mouse models of pancreatic ductal adenocarcinoma. Insights gleaned from such studies are spurring the development of therapies designed to reengineer the pancreas cancer stroma and render it permissive to agents targeting cell-autonomous events or to reinstate immunosurveillance. Integrating conventional and immunological treatments in the context of stromal targeting may provide the key to a durable clinical impact on this formidable disease.
Collapse
Affiliation(s)
- Ingunn M Stromnes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA, Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Kathleen E DelGiorno
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Philip D Greenberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA, Department of Immunology, University of Washington, Seattle, WA 98195, USA, Department of Medicine, Division of Medical Oncology, University of Washington School of Medicine, Seattle, WA 98195, USA and
| | - Sunil R Hingorani
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA, Department of Medicine, Division of Medical Oncology, University of Washington School of Medicine, Seattle, WA 98195, USA and Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| |
Collapse
|
32
|
Hartmann N, Giese NA, Giese T, Poschke I, Offringa R, Werner J, Ryschich E. Prevailing role of contact guidance in intrastromal T-cell trapping in human pancreatic cancer. Clin Cancer Res 2014; 20:3422-33. [PMID: 24763614 DOI: 10.1158/1078-0432.ccr-13-2972] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is characterized by extensive collagen-rich stroma. T cells that infiltrate pancreatic cancers frequently become trapped in the stroma and do not contact tumor cells. Here, we aimed to analyze how chemokines and extracellular matrix (ECM) collagen interact in mediating T-cell infiltration in PDAC. EXPERIMENTAL DESIGN T-cell distribution and ECM structure within tumors were analyzed. Chemokine concentrations in human PDAC were compared with the levels of immune cell infiltration. We assessed the influences of selected chemokines and collagen on directed and random T-cell movement using in vitro migration systems. RESULTS PDAC overproduced several T-cell-active chemokines, but their levels were not correlated with intratumoral T-cell infiltration. In the absence of collagen, directed migration of activated T cells was induced by chemokines. Interestingly, collagen itself promoted high migratory activity of T cells, but completely abolished chemokine-guided movement. This effect was not altered by a β1-integrin blocking antibody. Activated T cells actively migrated in low-density collagen matrices, but migration was inhibited in dense collagen. Accordingly, T cells were heterogeneously distributed in the pancreatic cancer stroma, with the majority residing in areas of low-density collagen far from tumor clusters. CONCLUSION The excessive desmoplasia in PDAC promotes T-cell migration by contact guidance, which abrogates tumor cell-directed movement. Furthermore, dense collagen networks represent a physical barrier, additionally rearranging T-cell distribution to favor tumor stroma. These mechanisms are mainly responsible for intrastromal T-cell trapping in pancreatic cancer and may hinder the development of T-cell-based immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Isabel Poschke
- Department of Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany
| | - Rienk Offringa
- Department of Translational Cancer Research, German Cancer Research Center, Heidelberg, Germany
| | - Jens Werner
- Authors' Affiliations: Departments of General Surgery and
| | | |
Collapse
|
33
|
Ajdžanović VZ, Medigović IM, Pantelić JB, Milošević VL. Soy isoflavones and cellular mechanics. J Bioenerg Biomembr 2014; 46:99-107. [PMID: 24346760 DOI: 10.1007/s10863-013-9536-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 12/05/2013] [Indexed: 01/30/2023]
Abstract
Soy isoflavones are diphenolic compounds that are frequently used for alternative treatment of ageing symptoms in both genders. They operate at principally two hierarchical levels of functional organization - cellular and molecular, while these 'types' of action appear to have indefinite borders. Soy isoflavone action at the cellular level involves inter alia the effects on cell mechanics. This epigenetic and modular determinant of cell function and fate is defined by: the anchorage to extracellular matrix (ECM) and neighboring cells, cytoskeleton organization, membrane tension and vesicle trafficking. Soy isoflavones have been reported to: (i) generally fashion an inert cell phenotype in some cancers and enhance the cell anchorage in connective tissues, via the effects on ECM proteins, focal adhesion kinases-mediated events and matrix metalloproteinases inhibition; (ii) affect cytoskeleton integrity, the effects being related to Ca(2+) ions fluxes and involving cell retraction or differentiation/proliferation-related variations in mechanical status; (iii) increase, remain "silent" or decrease membrane tension/fluidity, which depends on polarity and a number and arrangement of functional groups in applied isoflavone; (iv) provoke inhibitory effects on vesicle trafficking and exo-/endocytosis, which are usually followed by changed cell morphology. Here we present and discuss the abundance of effects arising from cells' "encounter" with soy isoflavones, focusing on different morphofunctional definers of cell mechanics.
Collapse
Affiliation(s)
- Vladimir Z Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11060, Belgrade, Serbia,
| | | | | | | |
Collapse
|
34
|
Chen H, Nalbantoglu J. Ring cell migration assay identifies distinct effects of extracellular matrix proteins on cancer cell migration. BMC Res Notes 2014; 7:183. [PMID: 24674422 PMCID: PMC3986826 DOI: 10.1186/1756-0500-7-183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 03/17/2014] [Indexed: 11/15/2022] Open
Abstract
Background Alterations in cell migration are a hallmark of cancer cell invasion and metastasis. In vitro assays commonly used to study cell migration, including the scratch wound healing assay, Boyden chamber assay, and newly developed advanced systems with microfluidics, each have several disadvantages. Findings Here we describe an easy and cost-effective in vitro assay for cell migration employing cloning rings to create gaps in the cell monolayer (“ring cell migration assay”). The assay was used to quantitate innate differences in cell motility and the effect of various extracellular matrix proteins on migration of five cancer cell lines: U87 and U251N glioma cells, MDA-MB-231and MCF-7 breast cancer cells, and HeLa cervical cancer cells. Interestingly, collagen was a general promoter of cell migration for all five cancer cell lines, without affecting cell proliferation. Conclusions Taken together, the ring cell migration assay is an easy, convenient and cost-effective assay to study cell migration in vitro.
Collapse
Affiliation(s)
| | - Josephine Nalbantoglu
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal Quebec, Canada.
| |
Collapse
|
35
|
Salvatori M, Katari R, Patel T, Peloso A, Mugweru J, Owusu K, Orlando G. Extracellular Matrix Scaffold Technology for Bioartificial Pancreas Engineering: State of the Art and Future Challenges. J Diabetes Sci Technol 2014; 8:159-169. [PMID: 24876552 PMCID: PMC4454093 DOI: 10.1177/1932296813519558] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Emergent technologies in regenerative medicine may soon overcome the limitations of conventional diabetes therapies. Collaborative efforts across the subfields of stem cell technology, islet encapsulation, and biomaterial carriers seek to produce a bioengineered pancreas capable of restoring endocrine function in patients with insulin-dependent diabetes. These technologies rely on a robust understanding of the extracellular matrix (ECM), the supportive 3-dimensional network of proteins necessary for cellular attachment, proliferation, and differentiation. Although these functions can be partially approximated by biosynthetic carriers, novel decellularization protocols have allowed researchers to discover the advantages afforded by the native pancreatic ECM. The native ECM has proven to be an optimal platform for recellularization and whole-organ pancreas bioengineering, an exciting new field with the potential to resolve the dire shortage of transplantable organs. This review seeks to contextualize recent findings, discuss current research goals, and identify future challenges of regenerative medicine as it applies to diabetes management.
Collapse
Affiliation(s)
| | - Ravi Katari
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Timil Patel
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Andrea Peloso
- Wake Forest School of Medicine, Winston-Salem, NC, USA Department of Surgery, School of Medicine, University of Pavia, Pavia, Italy
| | - Jon Mugweru
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kofi Owusu
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
36
|
Pancreatic stellate cells promote hapto-migration of cancer cells through collagen I-mediated signalling pathway. Br J Cancer 2013; 110:409-20. [PMID: 24201748 PMCID: PMC3899756 DOI: 10.1038/bjc.2013.706] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/27/2013] [Accepted: 10/15/2013] [Indexed: 01/05/2023] Open
Abstract
Background: Pancreatic stellate cells (PSCs) promote metastasis as well as local growth of pancreatic cancer. However, the factors mediating the effect of PSCs on pancreatic cancer cells have not been clearly identified. Methods: We used a modified Boyden chamber assay as an in vitro model to investigate the role of PSCs in migration of Panc1 and UlaPaCa cells and to identify the underlying mechanisms. Results: PSC supernatant (PSC-SN) dose-dependently induced the trans-migration of Panc1 and UlaPaCa cells, mainly via haptokinesis and haptotaxis, respectively. In contrast to poly-L-lysine or fibronectin, collagen I resembled PSC-SN with respect to its effect on cancer cell behaviours, including polarised morphology, facilitated adhesion, accelerated motility and stimulated trans-migration. Blocking antibodies against integrin α2/β1 subunits significantly attenuated PSC-SN- or collagen I-promoted cell trans-migration and adhesion. Moreover, both PSC-SN and collagen I induced the formation of F-actin and focal adhesions in cells, which was consistent with the constantly enhanced phosphorylation of focal adhesion kinase (FAK, Tyr397). Inhibition of FAK function by an inhibitor or small interference RNAs significantly diminished the effect of PSC-SN or collagen I on haptotaxis/haptokinesis of pancreatic cancer cells. Conclusion: Collagen I is the major mediator for PSC-SN-induced haptokinesis of Panc1 and haptotaxis of UlaPaCa by activating FAK signalling via binding to integrin α2β1.
Collapse
|
37
|
Yao J, Hu XF, Feng XS, Gao SG. Pleiotrophin promotes perineural invasion in pancreatic cancer. World J Gastroenterol 2013; 19:6555-6558. [PMID: 24151381 PMCID: PMC3801368 DOI: 10.3748/wjg.v19.i39.6555] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/19/2013] [Accepted: 08/15/2013] [Indexed: 02/06/2023] Open
Abstract
Perineural invasion (PNI) in pancreatic cancer is an important cause of local recurrence, but little is known about its mechanism. Pleiotrophin (PTN) is an important neurotrophic factor. It is of interest that our recent experimental data showed its involvement in PNI of pancreatic cancer. PTN strongly presents in the cytoplasm of pancreatic cancer cells, and high expression of PTN and its receptor may contribute to the high PNI of pancreatic cancer. Correspondingly, PNI is prone to happen in PTN-positive tumors. We thus hypothesize that, as a neurite growth-promoting factor, PTN may promote PNI in pancreatic cancer. PTN is released at the time of tumor cell necrosis, and binds with its high-affinity receptor, N-syndecan on pancreatic nerves, to promote neural growth in pancreatic cancer. Furthermore, neural destruction leads to a distorted neural homeostasis. Neurons and Schwann cells produce more N-syndecan in an effort to repair the pancreatic nerves. However, the abundance of N-syndecan attracts further PTN-positive cancer cells to the site of injury, creating a vicious cycle. Ultimately, increased PTN and N-syndecan levels, due to the continuous nerve injury, may promote cancer invasion and propagation along the neural structures. Therefore, it is meaningful to discuss the relationship between PTN/N-syndecan signaling and PNI in pancreatic cancer, which may lead to a better understanding of the mechanism of PNI in pancreatic cancer.
Collapse
|
38
|
Drifka CR, Eliceiri KW, Weber SM, Kao WJ. A bioengineered heterotypic stroma-cancer microenvironment model to study pancreatic ductal adenocarcinoma. LAB ON A CHIP 2013; 13:3965-75. [PMID: 23959166 PMCID: PMC3834588 DOI: 10.1039/c3lc50487e] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Interactions between neoplastic epithelial cells and components of a reactive stroma in pancreatic ductal adenocarcinoma (PDAC) are of key significance behind the disease's dismal prognosis. Despite extensive published research in the importance of stroma-cancer interactions in other cancers and experimental evidence supporting the importance of the microenvironment in PDAC progression, a reproducible three-dimensional (3D) in vitro model for exploring stroma-cancer interplay and evaluating therapeutics in a physiologically relevant context has been lacking. We introduce a humanized microfluidic model of the PDAC microenvironment incorporating multicellularity, extracellular matrix (ECM) components, and a spatially defined 3D microarchitecture. Pancreatic stellate cells (PSCs) isolated from clinically-evaluated human tissue specimens were co-cultured with pancreatic ductal adenocarcinoma cells as an accessible 3D construct that maintained important tissue features and disease behavior. Multiphoton excitation (MPE) and Second Harmonic Generation (SHG) imaging techniques were utilized to image the intrinsic signal of stromal collagen in human pancreatic tissues and live cell-collagen interactions within the optically-accessible microfluidic tissue model. We further evaluated the dose-response of the model with the anticancer agent paclitaxel. This bioengineered model of the PDAC stroma-cancer microenvironment provides a complementary platform to elucidate the complex stroma-cancer interrelationship and to evaluate the efficacy of potential therapeutics in a humanized system that closely recapitulates key PDAC microenvironment characteristics.
Collapse
Affiliation(s)
- Cole R. Drifka
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
- Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Sharon M. Weber
- Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Surgery, University of Wisconsin, Madison, WI, USA
| | - W. John Kao
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI, USA
- Paul P. Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI, USA
- Department of Surgery, University of Wisconsin, Madison, WI, USA
- School of Pharmacy, University of Wisconsin, Madison, WI, USA
- UW Institute for Clinical and Translational Research, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
39
|
Cabarcas SM, Sun L, Mathews L, Thomas S, Zhang X, Farrar WL. The differentiation of pancreatic tumor-initiating cells by vitronectin can be blocked by cilengitide. Pancreas 2013; 42:861-70. [PMID: 23462327 PMCID: PMC3676482 DOI: 10.1097/mpa.0b013e318279d568] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Pancreatic cancer is a leading cancer type and its molecular pathology is poorly understood. The only potentially curative therapeutic option available is complete surgical resection; however, this is inadequate as most of the patients are diagnosed at an advanced or metastatic stage. Tumor-initiating cells (TICs) constitute a subpopulation of cells within a solid tumor that sustain tumor growth, metastasis, and chemo/radioresistance. Within pancreatic cancer, TICs have been identified based on the expression of specific cell surface markers. METHODS We use a sphere formation assay to enrich putative TICs and use human serum as a driver of differentiation. We demonstrate by using specific blocking reagents that we can inhibit the differentiation process and maintain TIC-associated markers and genes. RESULTS We can induce differentiation of pancreatospheres with the addition of human serum, and we identified vitronectin as an inducer of differentiation. We inhibit differentiation by human serum using an arginine-glycine-aspartate-specific peptide, which is Cilengitide; hence, demonstrating this differentiation is mediated via specific integrin receptors. CONCLUSIONS Overall, our studies further the definition of pancreatic TICs and provide further insight into both the maintenance and differentiation of this lethal population.
Collapse
Affiliation(s)
- Stephanie M Cabarcas
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Zhu J, Nie S, Wu J, Lubman DM. Target proteomic profiling of frozen pancreatic CD24+ adenocarcinoma tissues by immuno-laser capture microdissection and nano-LC-MS/MS. J Proteome Res 2013; 12:2791-804. [PMID: 23679566 DOI: 10.1021/pr400139c] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cellular heterogeneity of solid tumors represents a common problem in mass spectrometry (MS)-based analysis of tissue specimens. Combining immuno-laser capture microdissection (iLCM) and mass spectrometry (MS) provides a means to study proteins that are specific for pure cell subpopulations in complex tissues. CD24, as a cell surface marker for detecting pancreatic cancer stem cells (CSCs), is directly correlated with the development and metastasis of pancreatic cancer. Herein, we describe an in-depth proteomic profiling of frozen pancreatic CD24(+) adenocarcinoma cells from early stage tumors using iLCM and LC-MS/MS and a comparison with CD24(-) cells dissected from patient-matched adjacent normal tissues. Approximately 40 nL of tissue was procured from each specimen and subjected to tandem MS analysis in triplicate. A total of 2665 proteins were identified, with 375 proteins in common that were significantly differentially expressed in CD24(+) versus CD24(-) cells by at least a 2-fold change. The major groups of the differentially overexpressed proteins are involved in promoting tumor cell migration and invasion, immune escape, and tumor progression. Three selected candidates relevant to mediating immune escape, CD59, CD70, and CD74, and a tumor promoter, TGFBI, were further validated by immunohistochemistry analysis on tissue microarrays. These proteins showed significantly increased expression in a large group of clinical pancreatic adenocarcinomas but were negative in all normal pancreas samples. The significant coexpression of these proteins with CD24 suggests that they may play important roles in the progression of pancreatic cancer and could serve as promising prognosis markers and novel therapeutic targets for this deadly disease.
Collapse
Affiliation(s)
- Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, Michigan 48109, United States
| | | | | | | |
Collapse
|
41
|
Shin E, Yoon Y, Ahn J, Park S. TC-VGC: a tumor classification system using variations in genes' correlation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2011; 104:e87-e101. [PMID: 21531474 DOI: 10.1016/j.cmpb.2011.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 01/11/2011] [Accepted: 03/07/2011] [Indexed: 05/30/2023]
Abstract
Classification analysis of microarray data is widely used to reveal biological features and to diagnose various diseases, including cancers. Most existing approaches improve the performance of learning models by removing most irrelevant and redundant genes from the data. They select the marker genes which are expressed differently in normal and tumor tissues. These techniques ignore the importance of the complex functional-dependencies between genes. In this paper, we propose a new method for cancer classification which uses distinguished variations of gene-gene correlation in two sample groups. The cancer specific genetic network composed of these gene pairs contains many literature-curated prostate cancer genes, and we were successful in identifying new candidate prostate cancer genes inferred by them. Furthermore, this method achieved a high accuracy with a small number of genes in cancer classification.
Collapse
Affiliation(s)
- Eunji Shin
- Department of Computer Science, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul 120-749, South Korea
| | | | | | | |
Collapse
|
42
|
Friedl P, Alexander S. Cancer Invasion and the Microenvironment: Plasticity and Reciprocity. Cell 2011; 147:992-1009. [DOI: 10.1016/j.cell.2011.11.016] [Citation(s) in RCA: 1419] [Impact Index Per Article: 101.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Indexed: 02/07/2023]
|
43
|
Cheng A, Guo J, Henderson-Jackson E, Kim D, Malafa M, Coppola D. IκB Kinase ε expression in pancreatic ductal adenocarcinoma. Am J Clin Pathol 2011; 136:60-6. [PMID: 21685032 DOI: 10.1309/ajcp2jjgyniuas2v] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
IκB kinase (IKKε) is a serine/threonine protein kinase that belongs to the IKK kinase family. Recent studies have shown that IKKε functions as a breast and ovarian cancer oncogene. We demonstrated frequent overexpression of IKKε in pancreatic ductal adenocarcinoma (PDA). We immunohistochemically evaluated 78 PDAs using the avidin-biotin-peroxidase method and the anti-IKKε rabbit polyclonal antibody. Elevated IKKε reactivity (immunohistochemical score, 4-9) was observed in 64% of PDAs (50/78), but in 0.0% of nonneoplastic pancreatic ductal epithelium (0/113; P < .001). Kaplan-Meier analysis of overall survival revealed that patients with high IKKε-immunohistochemical scores (4-9) had significantly shorter survival than did patients with low IKKε immunohistochemical scores (0-3; P = .023; log-rank test) independent of tumor stage or grade. These data indicate that deregulation of IKKε is a common event in PDA and might have an important role in the pathogenesis of this deadly disease. In addition, IKKε could serve as a prognostic marker and potential therapeutic target for PDA intervention.
Collapse
|
44
|
Chiu CC, Liu PL, Huang KJ, Wang HM, Chang KF, Chou CK, Chang FR, Chong IW, Fang K, Chen JS, Chang HW, Wu YC. Goniothalamin inhibits growth of human lung cancer cells through DNA damage, apoptosis, and reduced migration ability. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:4288-93. [PMID: 21391609 DOI: 10.1021/jf200566a] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
We evaluated the possible anticancer performance of a natural compound, goniothalamin (GTN), against human lung cancer using as a non-small cell lung cancer (NSCLC) cell line, H1299, as the model system. Cellular proliferation was significantly inhibited by GTN. Using an improved alkaline comet-nuclear extract (comet-NE) assay, GTN was found to induce a significant increase in the tail DNA. Wound healing and zymography assays showed that GTN attenuated cell migration and caused a reduction in the activity level of two major migration-associated matrix metalloproteinases, MMP-2 and MMP-9. It can be concluded that the DNA-damaging effect of GTN against lung cancer cells leads to growth inhibition as well as a depression in migration ability. Therefore, GTN has potential as a chemotherapeutic agent against lung cancer.
Collapse
Affiliation(s)
- Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dormoy V, Béraud C, Lindner V, Thomas L, Coquard C, Barthelmebs M, Jacqmin D, Lang H, Massfelder T. LIM-class homeobox gene Lim1, a novel oncogene in human renal cell carcinoma. Oncogene 2010; 30:1753-63. [PMID: 21132009 DOI: 10.1038/onc.2010.557] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Human clear cell renal cell carcinoma (CCC) remains resistant to therapies. The transcription factor LIM-class homeobox gene Lim1 is required for normal organogenesis, including nephrogenesis, by regulating cell movements, differentiation and growth. Its expression is controlled partly by the sonic hedgehog-Gli signaling pathway, which we have recently shown to be reactivated in human CCC. So far, no study has assessed whether Lim1 may be associated with tumorigenesis. Using a panel of human CCC cell lines expressing or not the von Hippel-Lindau tumor suppressor gene and 44 tumor/normal tissues pairs, we found that Lim1 is constitutively and exclusively reexpressed in tumors. Through Lim1 silencing or overexpressing, we show that Lim1 is a growth and survival factor in human CCC, at least through the activation of oncogenic pathways including the phosphoinositide kinase-3/Akt and nuclear factor-kappaB pathways. More importantly, in nude mice bearing human CCC tumors, Lim1 silencing abolished tumor growth through the same mechanism as in vitro. In Lim1-depleted cells and tumors, cell movements were substantially impaired because of the inhibition of expression of various proteins involved in metastatic spread, such as paxillin or tenascin-C. These findings establish that the developmental marker Lim1 acts as an oncogene in cancer cells and targeting Lim1 may constitute an innovative therapeutic intervention in human CCC.
Collapse
Affiliation(s)
- V Dormoy
- INSERM U682, Section of Kidney Cancer and Renal Physiopathology, University of Strasbourg, School of Medicine, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Liu X, Zhang M, Go VLW, Hu S. Membrane proteomic analysis of pancreatic cancer cells. J Biomed Sci 2010; 17:74. [PMID: 20831833 PMCID: PMC2949717 DOI: 10.1186/1423-0127-17-74] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 09/13/2010] [Indexed: 01/08/2023] Open
Abstract
Background Pancreatic cancer is one of the most aggressive human tumors due to its high potential of local invasion and metastasis. The aim of this study was to characterize the membrane proteomes of pancreatic ductal adenocarcinoma (PDAC) cells of primary and metastatic origins, and to identify potential target proteins related to metastasis of pancreatic cancer. Methods Membrane/membrane-associated proteins were isolated from AsPC-1 and BxPC-3 cells and identified with a proteomic approach based on SDS-PAGE, in-gel tryptic digestion and liquid chromatography with tandem mass spectrometry (LC-MS/MS). X! Tandem was used for database searching against the SwissProt human protein database. Results We identified 221 & 208 proteins from AsPC-1 and BxPC-3 cells, respectively, most of which are membrane or membrane-associated proteins. A hundred and nine proteins were found in both cell lines while the others were present in either AsPC-1 or BxPC-3 cells. Differentially expressed proteins between two cell lines include modulators of cell adhesion, cell motility or tumor invasion as well as metabolic enzymes involved in glycolysis, tricarboxylic acid cycle, or nucleotide/lipid metabolism. Conclusion Membrane proteomes of AsPC-1 (metastatic) and BxPC-3 (primary) cells are remarkably different. The differentially expressed membrane proteins may serve as potential targets for diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Xiaojun Liu
- UCLA School of Dentistry & Dental Research Institute, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
47
|
Kruegel J, Miosge N. Basement membrane components are key players in specialized extracellular matrices. Cell Mol Life Sci 2010; 67:2879-95. [PMID: 20428923 PMCID: PMC2921489 DOI: 10.1007/s00018-010-0367-x] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 03/17/2010] [Accepted: 03/24/2010] [Indexed: 01/11/2023]
Abstract
More than three decades ago, basement membranes (BMs) were described as membrane-like structures capable of isolating a cell from and connecting a cell to its environment. Since this time, it has been revealed that BMs are specialized extracellular matrices (sECMs) with unique components that support important functions including differentiation, proliferation, migration, and chemotaxis of cells during development. The composition of these sECM is as unique as the tissues to which they are localized, opening the possibility that such matrices can fulfill distinct functions. Changes in BM composition play significant roles in facilitating the development of various diseases. Furthermore, tissues have to provide sECM for their stem cells during development and for their adult life. Here, we briefly review the latest research on these unique sECM and their components with a special emphasis on embryonic and adult stem cells and their niches.
Collapse
Affiliation(s)
- Jenny Kruegel
- Tissue Regeneration Work Group, Department of Prosthodontics, Georg August University, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Nicolai Miosge
- Tissue Regeneration Work Group, Department of Prosthodontics, Georg August University, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| |
Collapse
|
48
|
Neural invasion in pancreatic cancer: the past, present and future. Cancers (Basel) 2010; 2:1513-27. [PMID: 24281170 PMCID: PMC3837319 DOI: 10.3390/cancers2031513] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 06/30/2010] [Accepted: 07/07/2010] [Indexed: 12/20/2022] Open
Abstract
In the past 15 years, invasion of nerves by cancer cells has escaped from its role as a mere bystander in cancer biology and turned into an attractive niche to study the heterotypic interaction between cancer cells and neurons. Today, neural invasion (NI) in pancreatic cancer (PCa) stands out due to the recent demonstration of its association with tumor progression, local recurrence and neuropathic pain. Accordingly, recent research on NI in PCa revealed the critical involvement of numerous nerve- or cancer cell-derived molecules in several novel in vitro and in vivo models of NI, which, however, still need further major improvement.
Collapse
|
49
|
Sroka IC, Anderson TA, McDaniel KM, Nagle RB, Gretzer MB, Cress AE. The laminin binding integrin alpha6beta1 in prostate cancer perineural invasion. J Cell Physiol 2010; 224:283-8. [PMID: 20432448 DOI: 10.1002/jcp.22149] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastasizing prostate tumor cells invade along nerves innervating the encapsulated human prostate gland in a process known as perineural invasion. The extracellular matrix laminin class of proteins line the neural route and tumor cells escaping from the gland express the laminin binding integrin alpha6beta1 as a prominent cell surface receptor. Integrin alpha6beta1 promotes aggressive disease and supports prostate tumor cell metastasis to bone. Laminins and their integrin receptors are necessary for the development and maintenance of the peripheral nervous system, indicating the potential role for integrin receptors in directing prostate tumor cell invasion on nerves during perineural invasion.
Collapse
Affiliation(s)
- Isis C Sroka
- Department of Pharmacology, The University of Arizona, Tucson, Arizona, USA
| | | | | | | | | | | |
Collapse
|