1
|
Christofidis K, Pergaris A, Fioretzaki R, Charalampakis N, Kapetanakis EΙ, Kavantzas N, Schizas D, Sakellariou S. Annexin A2 in Tumors of the Gastrointestinal Tract, Liver, and Pancreas. Cancers (Basel) 2024; 16:3764. [PMID: 39594718 PMCID: PMC11592865 DOI: 10.3390/cancers16223764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Annexin A2 (ANXA2) is a protein that is involved in many physiological and pathological cellular processes. There is compelling evidence that its dysregulated expression, be it up- or downregulation, contributes to the oncogenesis of various neoplasms, including those of the digestive system. The present review summarizes the current knowledge on the role of ANXA2 in the main tumors of the digestive system. The clinical significance of ANXA2 is primordial, due to its potential use as a diagnostic and prognostic biomarker, and as a part of therapeutic protocols. Certain preclinical studies have shown that inhibiting ANXA2 or disrupting its interactions with key molecules can suppress tumor growth, invasion, and metastasis, as well as increase the cancer cells' sensitivity to treatment in various cancers. Further research is needed to fully elucidate the complex role of ANXA2 in the carcinogenesis of tumors of the digestive system, and to translate these findings into clinical applications for improved diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Konstantinos Christofidis
- Cytopathology Laboratory, Laiko General Hospital of Athens, 11527 Athens, Greece; (K.C.); (N.K.); (S.S.)
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Alexandros Pergaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Rodanthi Fioretzaki
- First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital of Athens, 11527 Athens, Greece; (R.F.); (D.S.)
| | - Nikolaos Charalampakis
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, 18537 Piraeus, Greece;
| | - Emmanouil Ι. Kapetanakis
- Department of Thoracic Surgery, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Nikolaos Kavantzas
- Cytopathology Laboratory, Laiko General Hospital of Athens, 11527 Athens, Greece; (K.C.); (N.K.); (S.S.)
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital of Athens, 11527 Athens, Greece; (R.F.); (D.S.)
| | - Stratigoula Sakellariou
- Cytopathology Laboratory, Laiko General Hospital of Athens, 11527 Athens, Greece; (K.C.); (N.K.); (S.S.)
| |
Collapse
|
2
|
Waldron RT, Lugea A, Chang HH, Su HY, Quiros C, Lewis MS, Che M, Ramanujan VK, Rozengurt E, Eibl G, Pandol SJ. Upregulated Matrisomal Proteins and Extracellular Matrix Mechanosignaling Underlie Obesity-Associated Promotion of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:1593. [PMID: 38672675 PMCID: PMC11048773 DOI: 10.3390/cancers16081593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Diet-induced obesity (DIO) promotes pancreatic ductal adenocarcinoma (PDAC) in mice expressing KRasG12D in the pancreas (KC mice), but the precise mechanisms remain unclear. Here, we performed multiplex quantitative proteomic and phosphoproteomic analysis by liquid chromatography-tandem mass spectrometry and further bioinformatic and spatial analysis of pancreas tissues from control-fed versus DIO KC mice after 3, 6, and 9 months. Normal pancreatic parenchyma and associated proteins were steadily eliminated and the novel proteins, phosphoproteins, and signaling pathways associated with PDAC tumorigenesis increased until 6 months, when most males exhibited cancer, but females did not. Differentially expressed proteins and phosphoproteins induced by DIO revealed the crucial functional role of matrisomal proteins, which implies the roles of upstream regulation by TGFβ, extracellular matrix-receptor signaling to downstream PI3K-Akt-mTOR-, MAPK-, and Yap/Taz activation, and crucial effects in the tumor microenvironment such as metabolic alterations and signaling crosstalk between immune cells, cancer-associated fibroblasts (CAFs), and tumor cells. Staining tissues from KC mice localized the expression of several prognostic PDAC biomarkers and elucidated tumorigenic features, such as robust macrophage infiltration, acinar-ductal metaplasia, mucinous PanIN, distinct nonmucinous atypical flat lesions (AFLs) surrounded by smooth muscle actin-positive CAFs, invasive tumors with epithelial-mesenchymal transition arising close to AFLs, and expanding deserted areas by 9 months. We next used Nanostring GeoMX to characterize the early spatial distribution of specific immune cell subtypes in distinct normal, stromal, and PanIN areas. Taken together, these data richly contextualize DIO promotion of Kras-driven PDAC tumorigenesis and provide many novel insights into the signaling pathways and processes involved.
Collapse
Affiliation(s)
- Richard T. Waldron
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Aurelia Lugea
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hui-Hua Chang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Hsin-Yuan Su
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Crystal Quiros
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael S. Lewis
- Department of Medicine and Department of Pathology & Laboratory Medicine, VA Greater Los Angeles Health System, Cedars-Sinai Medical Center, Los Angeles, CA 90073, USA;
| | - Mingtian Che
- Biobank and Research Pathology Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - V. Krishnan Ramanujan
- Biobank and Research Pathology Resource, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Stephen J. Pandol
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
3
|
Fernandes S, Oliver-De La Cruz J, Morazzo S, Niro F, Cassani M, Ďuríková H, Caravella A, Fiore P, Azzato G, De Marco G, Lauria A, Izzi V, Bosáková V, Fric J, Filipensky P, Forte G. TGF-β induces matrisome pathological alterations and EMT in patient-derived prostate cancer tumoroids. Matrix Biol 2024; 125:12-30. [PMID: 37944712 DOI: 10.1016/j.matbio.2023.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Extracellular matrix (ECM) tumorigenic alterations resulting in high matrix deposition and stiffening are hallmarks of adenocarcinomas and are collectively defined as desmoplasia. Here, we thoroughly analysed primary prostate cancer tissues obtained from numerous patients undergoing radical prostatectomy to highlight reproducible structural changes in the ECM leading to the loss of the glandular architecture. Starting from patient cells, we established prostate cancer tumoroids (PCTs) and demonstrated they require TGF-β signalling pathway activity to preserve phenotypical and structural similarities with the tissue of origin. By modulating TGF-β signalling pathway in PCTs, we unveiled its role in ECM accumulation and remodelling in prostate cancer. We also found that TGF-β-induced ECM remodelling is responsible for the initiation of prostate cell epithelial-to-mesenchymal transition (EMT) and the acquisition of a migratory, invasive phenotype. Our findings highlight the cooperative role of TGF-β signalling and ECM desmoplasia in prompting prostate cell EMT and promoting tumour progression and dissemination.
Collapse
Affiliation(s)
- Soraia Fernandes
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic.
| | - Jorge Oliver-De La Cruz
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic
| | - Sofia Morazzo
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic; Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, Brno 62500, Czech Republic
| | - Francesco Niro
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic; Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, Brno 62500, Czech Republic
| | - Marco Cassani
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic
| | - Helena Ďuríková
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic
| | - Alessio Caravella
- Department of Computer Engineering, Modelling, Electronics and Systems Engineering (DIMES), University of Calabria (UNICAL), Via P. Bucci, Cubo 42C, Rende (CS) 87036, Italy
| | - Piergiuseppe Fiore
- Department of Computer Engineering, Modelling, Electronics and Systems Engineering (DIMES), University of Calabria (UNICAL), Via P. Bucci, Cubo 42C, Rende (CS) 87036, Italy
| | - Giulia Azzato
- Department of Computer Engineering, Modelling, Electronics and Systems Engineering (DIMES), University of Calabria (UNICAL), Via P. Bucci, Cubo 42C, Rende (CS) 87036, Italy
| | - Giuseppe De Marco
- Information Technology Center (ICT), University of Calabria (UNICAL), Via P. Bucci, Cubo 22B, Rende (CS) 87036, Italy
| | - Agostino Lauria
- Department of Engineering for Innovation, University of Salento (UNISALENTO), Corpo Z, Campus Ecotekne, SP.6 per Monteroni, Lecce (LE), Italy
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu FI-90014, Finland; Faculty of Medicine, BioIM Research Unit, University of Oulu, Oulu FI-90014, Finland; Foundation for the Finnish Cancer Institute, Tukholmankatu 8, Helsinki, Finland
| | - Veronika Bosáková
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic; Faculty of Medicine, Department of Biomedical Sciences, Masaryk University, Brno 62500, Czech Republic
| | - Jan Fric
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic; Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petr Filipensky
- Department of Urology, St. Anne's University Hospital, Brno 60200, Czech Republic
| | - Giancarlo Forte
- International Clinical Research Center, St. Anne's University Hospital, Brno 60200, Czech Republic; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London SE5 9NU, UK.
| |
Collapse
|
4
|
Tsutsumi K, Otsuka M. Clinical application of pancreatic juice‐derived small extracellular vesicles of pancreatic ductal adenocarcinoma. CLINICAL AND TRANSLATIONAL DISCOVERY 2023; 3. [DOI: 10.1002/ctd2.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/08/2023] [Indexed: 02/07/2025]
Abstract
AbstractBackgroundRecent imaging modalities have helped inthe early detection of pancreatic ductal adenocarcinoma (PDAC), resulting inimproved survival rates for patients with early‐stage PDAC. However, preoperative pathological diagnosis of early‐stage PDAC remains a challenge, particularly for small PDAC that is difficult to diagnose through standardendoscopic ultrasound‐guided fine‐needle biopsy. In this context, pancreaticjuice cytology has been re‐evaluated as an important tool for the preoperativediagnosis of early‐stage PDAC.MainPancreatic juice (PJ) comes in directcontact with PDAC lesions in the pancreatic duct and thus may contain a fewHG‐PanIN/PDAC cells and specific molecules. Additionally, the PJ may containconcentrated small extracellular vesicles (sEVs) that are released from cancerlesions. sEVs are double‐layered lipid‐bound particles that contain cargoassociated with the cell‐of‐origin, including proteins, microRNA, and RNA. sEVsreleased from cancer lesions found in body fluids, such as blood, urine, andsaliva, have already been studied as potential sources of diagnostic biomarkersfor cancer. PJ‐derived sEVs could serve as a “liquid biopsy” for theearly diagnosis of PDAC. However, little is known about the existence,physiological status, and function of PJ‐derived sEVs and their potentialutility as biomarkers for diagnostic, surveillance, and monitoring purposes oras therapeutic targets.ConclusionPJ‐derived sEVs represent a promisingavenue for the early diagnosis of PDAC. The utility of these particles as biomarkersfor diagnostic, surveillance, and monitoring purposes, or as therapeutictargets, warrants further research. Understanding the existence, physiologicalstatus, and function of PJ‐derived sEVs is crucial to unlocking their potentialas a valuable tool for overcoming PDAC.
Collapse
Affiliation(s)
- Koichiro Tsutsumi
- Department of Gastroenterology Okayama University Hospital Okayama Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology Okayama University Hospital Okayama Japan
- Department of Gastroenterology and Hepatology Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Science Okayama Japan
| |
Collapse
|
5
|
Schlensog M, Ruehlmann AC, Haeberle L, Opitz F, Becher AK, Goering W, Buth J, Knoefel WT, Ladage D, Meyer A, Esposito I. Tenascin-C affects invasiveness of EGFR-mutated lung adenocarcinoma through a putative paracrine loop. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166684. [PMID: 36878305 DOI: 10.1016/j.bbadis.2023.166684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/26/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Tenascin C (TNC) is an extracellular matrix (ECM) protein and a potential biomarker affecting progression of different tumor types, such as pancreatic and lung cancer. Alternative splicing variants of TNC are known to have an impact on interaction partners like other ECM proteins or cell surface receptors, including epidermal growth factor receptor (EGFR), leading to numerous and sometimes opposite roles of TNC in tumor cell dissemination and proliferation. Only little is known about the impact of TNC on biologic characteristics of lung cancer, such as invasion and metastatic potential. In the present study, we could link an increased expression of TNC in lung adenocarcinoma (LUAD) tissues with an unfavorable clinical outcome of patients. Furthermore, we investigated the functional role of TNC in LUAD. Immunohistochemical staining of TNC revealed a significant increase of TNC levels in primary tumours and metastases compared to normal lung tissue. Additionally, a significant correlation between TNC mRNA expression and EGFR copy number and protein expression levels has been determined. Moreover, inhibition of TNC in lung fibroblasts led to reduced invasiveness of LUAD cells harboring EGFR-activating mutations and to a shorter lamellipodia perimeter and a reduced lamellipodia area on the surface of LUAD cells. This study provides the evidence that TNC expression might be a biological relevant factor in LUAD progression in an EGFR-dependent manner and that it regulates tumor cell invasion by rearrangement of the actin cytoskeleton, especially affecting lamellipodia formation.
Collapse
Affiliation(s)
- Martin Schlensog
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Ann-Cathrin Ruehlmann
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Lena Haeberle
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Friederike Opitz
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Ann-Kathrin Becher
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Wolfgang Goering
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Juliane Buth
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Wolfram Trudo Knoefel
- Department of General, Visceral and Pediatric Surgery, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany
| | - Dennis Ladage
- Department of Pneumology, Kliniken Maria Hilf GmbH, Moenchengladbach, Germany
| | - Andreas Meyer
- Department of Pneumology, Kliniken Maria Hilf GmbH, Moenchengladbach, Germany
| | - Irene Esposito
- Institute of Pathology, Medical Faculty of the Heinrich Heine University, Duesseldorf, Germany.
| |
Collapse
|
6
|
Siddhartha R, Garg M. Interplay Between Extracellular Matrix Remodeling and Angiogenesis in Tumor Ecosystem. Mol Cancer Ther 2023; 22:291-305. [PMID: 36861362 DOI: 10.1158/1535-7163.mct-22-0595] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/27/2022] [Accepted: 12/16/2022] [Indexed: 03/03/2023]
Abstract
Studying the complex mechanisms of tumorigenesis and examining the interactions of neoplastic cells within tumor ecosystem are critical to explore the possibility of effective cancer treatment modalities. Dynamic tumor ecosystem is constantly evolving and is composed of tumor cells, extracellular matrix (ECM), secreted factors, and stromal cancer-associated fibroblasts (CAF), pericytes, endothelial cells (EC), adipocytes, and immune cells. ECM remodeling by synthesis, contraction, and/or proteolytic degradation of ECM components and release of matrix-sequestered growth factors create a microenvironment that promotes EC proliferation, migration, and angiogenesis. Stromal CAFs release multiple angiogenic cues (angiogenic growth factors, cytokines, and proteolytic enzymes) which interact with ECM proteins, thus contribute to enhance proangiogenic/promigratory properties and support aggressive tumor growth. Targeting angiogenesis brings about vascular changes including reduced adherence junction proteins, basement membrane and pericyte coverage, and increased leakiness. This facilitates ECM remodeling, metastatic colonization and chemoresistance. Owing to significant role of denser and stiffer ECM in inducing chemoresistance, direct or indirect targeting of ECM components is being reported as major axis of anticancer treatment. Exploring the agents targeting angiogenesis and ECM in a context specific manner may lead to reduced tumor burden by promoting conventional therapeutic effectiveness and overcoming the hurdles of therapy resistance.
Collapse
Affiliation(s)
- Rohit Siddhartha
- Department of Biochemistry, University of Lucknow, Lucknow, India
| | - Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow, India
| |
Collapse
|
7
|
Prognostic role of annexin A2 and cancer-associated fibroblasts in advanced non-small cell lung cancer: Implication in epithelial-mesenchymal transition and gefitinib resistance. Pathol Res Pract 2023; 241:154293. [PMID: 36586309 DOI: 10.1016/j.prp.2022.154293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/18/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite advances in treatment of non-small cell lung cancer (NSCLC), its prognosis remains dismal. Development of drug resistance is a major obstacle against success of targeted epidermal growth factor receptor (EGFR) -tyrosine kinase inhibitors (TKI) therapy. This study aimed to assess the prognostic role of annexin A2 (ANXA2) expression, within both tumor cells and stroma, as well as cancer associated fibroblasts (CAFs) in NSCLC and to investigate their potential role in induction of epithelial mesenchymal transition (EMT) and resistance to gefitinib. METHOD Immunohistochemistry was performed to evaluate tumoral and stromal ANXA2 expression and α-SMA-stained CAFs in 110 advanced NSCLC patients. Furthermore, STAT3 and E-cadherin mRNA expression was studied by quantitative reverse transcription PCR (qRT-PCR). RESULTS Both tumoral and stromal ANXA2 as well as CAFs were significantly related to clinical stage IV and malignant pleural effusion, while tumoral ANXA2 was significantly related to poor tumor differentiation. EGFR mutation and high tumoral ANXA2 were independent factors for poor overall survival, whereas high stromal and tumoral ANXA2 and high CAFs were independent predictors for poor progression-free survival. Moreover, high ANXA2 and CAFs were significantly associated with high STAT3 and low E-cadherin mRNA expression. Focusing on EGFR mutated cases, gefitinib resistance was significantly associated with high tumoral and stromal ANXA2, high CAFs, high STAT3 and low E-cadherin. CONCLUSION CAFs and ANXA2 could be considered as poor prognostic parameters in advanced NSCLC and are potential factors for gefitinib therapy resistance through EMT induction.
Collapse
|
8
|
Marangio A, Biccari A, D’Angelo E, Sensi F, Spolverato G, Pucciarelli S, Agostini M. The Study of the Extracellular Matrix in Chronic Inflammation: A Way to Prevent Cancer Initiation? Cancers (Basel) 2022; 14:cancers14235903. [PMID: 36497384 PMCID: PMC9741172 DOI: 10.3390/cancers14235903] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
Bidirectional communication between cells and their microenvironment has a key function in normal tissue homeostasis, and in disease initiation, progression and a patient's prognosis, at the very least. The extracellular matrix (ECM), as an element of all tissues and cellular microenvironment, is a frequently overlooked component implicated in the pathogenesis and progression of several diseases. In the inflammatory microenvironment (IME), different alterations resulting from remodeling processes can affect ECM, progressively inducing cancer initiation and the passage toward a tumor microenvironment (TME). Indeed, it has been demonstrated that altered ECM components interact with a variety of surface receptors triggering intracellular signaling that affect cellular pathways in turn. This review aims to support the notion that the ECM and its alterations actively participate in the promotion of chronic inflammation and cancer initiation. In conclusion, some data obtained in cancer research with the employment of decellularized ECM (dECM) models are described. The reported results encourage the application of dECM models to investigate the short circuits contributing to the creation of distinct IME, thus representing a potential tool to avoid the progression toward a malignant lesion.
Collapse
Affiliation(s)
- Asia Marangio
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Andrea Biccari
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Edoardo D’Angelo
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Francesca Sensi
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy
| | - Gaya Spolverato
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Marco Agostini
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
- Correspondence: ; Tel.: +39-049-964-0160
| |
Collapse
|
9
|
Shichi Y, Gomi F, Sasaki N, Nonaka K, Arai T, Ishiwata T. Epithelial and Mesenchymal Features of Pancreatic Ductal Adenocarcinoma Cell Lines in Two- and Three-Dimensional Cultures. J Pers Med 2022; 12:jpm12050746. [PMID: 35629168 PMCID: PMC9146102 DOI: 10.3390/jpm12050746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/01/2022] [Accepted: 05/02/2022] [Indexed: 02/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an intractable cancer that is difficult to diagnose early, and there is no cure other than surgery. PDAC is classified as an adenocarcinoma that has limited effective anticancer drug and molecular-targeted therapies compared to adenocarcinoma found in other organs. A large number of cancer cell lines have been established from patients with PDAC that have different genetic abnormalities, including four driver genes; however, little is known about the differences in biological behaviors among these cell lines. Recent studies have shown that PDAC cell lines can be divided into epithelial and mesenchymal cell lines. In 3D cultures, morphological and functional differences between epithelial and mesenchymal PDAC cell lines were observed as well as the drug effects of different anticancer drugs. These effects included gemcitabine causing an increased growth inhibition of epithelial PDAC cells, while nab-paclitaxel caused greater mesenchymal PDAC cell inhibition. Thus, examining the characteristics of epithelial or mesenchymal PDAC cells with stromal cells using a 3D co-culture may lead to the development of new anticancer drugs.
Collapse
Affiliation(s)
- Yuuki Shichi
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan; (Y.S.); (F.G.); (K.N.)
| | - Fujiya Gomi
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan; (Y.S.); (F.G.); (K.N.)
| | - Norihiko Sasaki
- Research Team for Geriatric Medicine (Vascular Medicine), Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan;
| | - Keisuke Nonaka
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan; (Y.S.); (F.G.); (K.N.)
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Hospital and Institute of Gerontology, Tokyo 173-0015, Japan;
| | - Toshiyuki Ishiwata
- Division of Aging and Carcinogenesis, Research Team for Geriatric Pathology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan; (Y.S.); (F.G.); (K.N.)
- Correspondence: ; Tel.: +81-3-3964-1141 (ext. 4414)
| |
Collapse
|
10
|
Geleta B, Tout FS, Lim SC, Sahni S, Jansson PJ, Apte MV, Richardson DR, Kovačević Ž. Targeting Wnt/tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. J Biol Chem 2022; 298:101608. [PMID: 35065073 PMCID: PMC8881656 DOI: 10.1016/j.jbc.2022.101608] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
A major barrier to successful pancreatic cancer (PC) treatment is the surrounding stroma, which secretes growth factors/cytokines that promote PC progression. Wnt and tenascin C (TnC) are key ligands secreted by stromal pancreatic stellate cells (PSCs) that then act on PC cells in a paracrine manner to activate the oncogenic β-catenin and YAP/TAZ signaling pathways. Therefore, therapies targeting oncogenic Wnt/TnC cross talk between PC cells and PSCs constitute a promising new therapeutic approach for PC treatment. The metastasis suppressor N-myc downstream-regulated gene-1 (NDRG1) inhibits tumor progression and metastasis in numerous cancers, including PC. We demonstrate herein that targeting NDRG1 using the clinically trialed anticancer agent di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) inhibited Wnt/TnC-mediated interactions between PC cells and the surrounding PSCs. Mechanistically, NDRG1 and DpC markedly inhibit secretion of Wnt3a and TnC by PSCs, while also attenuating Wnt/β-catenin and YAP/TAZ activation and downstream signaling in PC cells. This antioncogenic activity was mediated by direct inhibition of β-catenin and YAP/TAZ nuclear localization and by increasing the Wnt inhibitor, DKK1. Expression of NDRG1 also inhibited transforming growth factor (TGF)-β secretion by PC cells, a key mechanism by which PC cells activate PSCs. Using an in vivo orthotopic PC mouse model, we show DpC downregulated β-catenin, TnC, and YAP/TAZ, while potently increasing NDRG1 expression in PC tumors. We conclude that NDRG1 and DpC inhibit Wnt/TnC-mediated interactions between PC cells and PSCs. These results further illuminate the antioncogenic mechanism of NDRG1 and the potential of targeting this metastasis suppressor to overcome the oncogenic effects of the PC-PSC interaction.
Collapse
Affiliation(s)
- Bekesho Geleta
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Faten S Tout
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Department of Medical Laboratory Science, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Syer Choon Lim
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia; Cancer Drug Resistance & Stem Cell Program, Faculty of Medicine and Health, School of Medical Science, University of Sydney, Sydney, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, UNSW Sydney, Sydney, New South Wales, Australia; Pancreatic Research Group, Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Žaklina Kovačević
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
11
|
Opitz FV, Haeberle L, Daum A, Esposito I. Tumor Microenvironment in Pancreatic Intraepithelial Neoplasia. Cancers (Basel) 2021; 13:cancers13246188. [PMID: 34944807 PMCID: PMC8699458 DOI: 10.3390/cancers13246188] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive neoplasm with a poor survival rate. This is mainly due to late detection, which substantially limits therapy options. A better understanding of the early phases of pancreatic carcinogenesis is fundamental for improving patient prognosis in the future. In this article, we focused on the tumor microenvironment (TME), which provides the biological niche for the development of PDAC from its most common precursor lesions, PanIN (pancreatic intraepithelial neoplasias). Abstract Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive tumors with a poor prognosis. A characteristic of PDAC is the formation of an immunosuppressive tumor microenvironment (TME) that facilitates bypassing of the immune surveillance. The TME consists of a desmoplastic stroma, largely composed of cancer-associated fibroblasts (CAFs), immunosuppressive immune cells, immunoregulatory soluble factors, neural network cells, and endothelial cells with complex interactions. PDAC develops from various precursor lesions such as pancreatic intraepithelial neoplasia (PanIN), intraductal papillary mucinous neoplasms (IPMN), mucinous cystic neoplasms (MCN), and possibly, atypical flat lesions (AFL). In this review, we focus on the composition of the TME in PanINs to reveal detailed insights into the complex restructuring of the TME at early time points in PDAC progression and to explore ways of modifying the TME to slow or even halt tumor progression.
Collapse
|
12
|
Fujita M, Suzuki H, Fukai F. Involvement of integrin-activating peptides derived from tenascin-C in colon cancer progression. World J Gastrointest Oncol 2021; 13:980-994. [PMID: 34616507 PMCID: PMC8465449 DOI: 10.4251/wjgo.v13.i9.980] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/03/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Tenascin-C (TNC) is an adhesion modulatory protein present in the extracellular matrix that is highly expressed in several malignancies, including colon cancer. Although TNC is considered a negative prognostic factor for cancer patients, the substantial role of the TNC molecule in colorectal carcinogenesis and its malignant progression is poorly understood. We previously found that TNC has a cryptic functional site and that a TNC peptide containing this site, termed TNIIIA2, can potently and persistently activate beta1-integrins. In contrast, the peptide FNIII14, which contains a cryptic bioactive site within the fibronectin molecule, can inactivate beta1-integrins. This review presents the role of TNC in the development of colitis-associated colorectal cancer and in the malignant progression of colon cancer, particularly the major involvement of its cryptic functional site TNIIIA2. We propose new possible prophylactic and therapeutic strategies based on inhibition of the TNIIIA2-induced beta1-integrin activation by peptide FNIII14.
Collapse
Affiliation(s)
- Motomichi Fujita
- Department of Molecular Patho-Physiology, Tokyo University of Science, Noda 278-8510, Chiba, Japan
| | - Hideo Suzuki
- Department of Gastroenterology, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Fumio Fukai
- Department of Molecular Patho-Physiology, Tokyo University of Science, Noda 278-8510, Chiba, Japan
| |
Collapse
|
13
|
Li Z, Yu L, Hu B, Chen L, Jv M, Wang L, Zhou C, Wei M, Zhao L. Advances in cancer treatment: a new therapeutic target, Annexin A2. J Cancer 2021; 12:3587-3596. [PMID: 33995636 PMCID: PMC8120175 DOI: 10.7150/jca.55173] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Annexin A2 (ANXA2) is a calcium regulated phospholipid-binding protein. It is expressed in some tumor cells, endothelial cells, macrophages, and mononuclear cells, affecting cell survival and mediating interactions between intercellular and extracellular microenvironment. Aberrant expression of ANXA2 can be used as a potential predictive factor, diagnostic biomarker and therapeutic target in cancer therapy. Investigators used various technologies to target ANXA2 in a preclinical model of human cancers and demonstrated encouraging results. In this review article, we discuss the diagnosis and prognosis latent capacity of ANXA2 in progressive cancers, focus on the exploration of restorative interventions targeting ANXA2 in cancer treatment. Further, we comment on a promising candidate therapy that is conceivable for clinical translation.
Collapse
Affiliation(s)
- Zinan Li
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Baohui Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lianze Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Mingyi Jv
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lin Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Chenyi Zhou
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Medical Diagnosis and Treatment Center, Liaoning Province, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| |
Collapse
|
14
|
Liot S, Balas J, Aubert A, Prigent L, Mercier-Gouy P, Verrier B, Bertolino P, Hennino A, Valcourt U, Lambert E. Stroma Involvement in Pancreatic Ductal Adenocarcinoma: An Overview Focusing on Extracellular Matrix Proteins. Front Immunol 2021; 12:612271. [PMID: 33889150 PMCID: PMC8056076 DOI: 10.3389/fimmu.2021.612271] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide and is predicted to become second in 2030 in industrialized countries if no therapeutic progress is made. Among the different types of pancreatic cancers, Pancreatic Ductal Adenocarcinoma (PDAC) is by far the most represented one with an occurrence of more than 90%. This specific cancer is a devastating malignancy with an extremely poor prognosis, as shown by the 5-years survival rate of 2–9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Pancreatic tumors progress with few specific symptoms and are thus at an advanced stage at diagnosis in most patients. This malignancy is characterized by an extremely dense stroma deposition around lesions, accompanied by tissue hypovascularization and a profound immune suppression. Altogether, these combined features make access to cancer cells almost impossible for conventional chemotherapeutics and new immunotherapeutic agents, thus contributing to the fatal outcomes of the disease. Initially ignored, the Tumor MicroEnvironment (TME) is now the subject of intensive research related to PDAC treatment and could contain new therapeutic targets. In this review, we will summarize the current state of knowledge in the field by focusing on TME composition to understand how this specific compartment could influence tumor progression and resistance to therapies. Attention will be paid to Tenascin-C, a matrix glycoprotein commonly upregulated during cancer that participates to PDAC progression and thus contributes to poor prognosis.
Collapse
Affiliation(s)
- Sophie Liot
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Jonathan Balas
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Alexandre Aubert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laura Prigent
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Philippe Bertolino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ana Hennino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ulrich Valcourt
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| |
Collapse
|
15
|
Winkler J, Abisoye-Ogunniyan A, Metcalf KJ, Werb Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat Commun 2020; 11:5120. [PMID: 33037194 PMCID: PMC7547708 DOI: 10.1038/s41467-020-18794-x] [Citation(s) in RCA: 1223] [Impact Index Per Article: 244.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023] Open
Abstract
Tissues are dynamically shaped by bidirectional communication between resident cells and the extracellular matrix (ECM) through cell-matrix interactions and ECM remodelling. Tumours leverage ECM remodelling to create a microenvironment that promotes tumourigenesis and metastasis. In this review, we focus on how tumour and tumour-associated stromal cells deposit, biochemically and biophysically modify, and degrade tumour-associated ECM. These tumour-driven changes support tumour growth, increase migration of tumour cells, and remodel the ECM in distant organs to allow for metastatic progression. A better understanding of the underlying mechanisms of tumourigenic ECM remodelling is crucial for developing therapeutic treatments for patients. Tumors are more than cancer cells — the extracellular matrix is a protein structure that organizes all tissues and is altered in cancer. Here, the authors review recent progress in understanding how the cancer cells and tumor-associated stroma cells remodel the extracellular matrix to drive tumor growth and metastasis.
Collapse
Affiliation(s)
- Juliane Winkler
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA.
| | - Abisola Abisoye-Ogunniyan
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Kevin J Metcalf
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| | - Zena Werb
- Department of Anatomy, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, 94143, USA
| |
Collapse
|
16
|
Armacki M, Polaschek S, Waldenmaier M, Morawe M, Ruhland C, Schmid R, Lechel A, Tharehalli U, Steup C, Bektas Y, Li H, Kraus JM, Kestler HA, Kruger S, Ormanns S, Walther P, Eiseler T, Seufferlein T. Protein Kinase D1, Reduced in Human Pancreatic Tumors, Increases Secretion of Small Extracellular Vesicles From Cancer Cells That Promote Metastasis to Lung in Mice. Gastroenterology 2020; 159:1019-1035.e22. [PMID: 32446697 DOI: 10.1053/j.gastro.2020.05.052] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 04/21/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Pancreatic tumor cells release small extracellular vesicles (sEVs, exosomes) that contain lipids and proteins, RNA, and DNA molecules that might promote formation of metastases. It is not clear what cargo these vesicles contain and how they are released. Protein kinase D1 (PRKD1) inhibits cell motility and is believed to be dysregulated in pancreatic ductal adenocarcinomas. We investigated whether it regulates production of sEVs in pancreatic cancer cells and their ability to form premetastatic niches for pancreatic cancer cells in mice. METHODS We analyzed data from UALCAN and human pancreatic tissue microarrays to compare levels of PRKD1 between tumor and nontumor tissues. We studied mice with pancreas-specific disruption of Prkd1 (PRKD1KO mice), mice that express oncogenic KRAS (KC mice), and KC mice with disruption of Prkd1 (PRKD1KO-KC mice). Subcutaneous xenograft tumors were grown in NSG mice from Panc1 cells; some mice were then given injections of sEVs. Pancreata and lung tissues from mice were analyzed by histology, immunohistochemistry, and/or quantitative polymerase chain reaction; we performed nanoparticle tracking analysis of plasma sEVs. The Prkd1 gene was disrupted in Panc1 cells using CRISPR-Cas9 or knocked down with small hairpin RNAs, or PRKD1 activity was inhibited with the selective inhibitor CRT0066101. Pancreatic cancer cell lines were analyzed by gene-expression microarray, quantitative polymerase chain reaction, immunoblot, and immunofluorescence analyses. sEVs secreted by Panc1 cell lines were analyzed by flow cytometry, transmission electron microscopy, and mass spectrometry. RESULTS Levels of PRKD1 were reduced in human pancreatic ductal adenocarcinoma tissues compared with nontumor tissues. PRKD1KO-KC mice developed more pancreatic intraepithelial neoplasia, at a faster rate, than KC mice, and had more lung metastases and significantly shorter average survival time. Serum from PRKD1KO-KC mice had increased levels of sEVs compared with KC mice. Pancreatic cancer cells with loss or inhibition of PRKD1 increased secretion of sEVs; loss of PRKD1 reduced phosphorylation of its substrate, cortactin, resulting in increased F-actin levels at the plasma membrane. sEVs from cells with loss or reduced expression of PRKD1 had altered content, and injection of these sEVs into mice increased metastasis of xenograft tumors to lung, compared with sEVs from pancreatic cells that expressed PRKD1. PRKD1-deficient pancreatic cancer cells showed increased loading of integrin α6β4 into sEVs-a process that required CD82. CONCLUSIONS Human pancreatic ductal adenocarcinoma has reduced levels of PRKD1 compared with nontumor pancreatic tissues. Loss of PRKD1 results in reduced phosphorylation of cortactin in pancreatic cancer cell lines, resulting in increased in F-actin at the plasma membrane and increased release of sEVs, with altered content. These sEVs promote metastasis of xenograft and pancreatic tumors to lung in mice.
Collapse
Affiliation(s)
- Milena Armacki
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Sandra Polaschek
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | - Mareen Morawe
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Claudia Ruhland
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Rebecca Schmid
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - André Lechel
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Umesh Tharehalli
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Christoph Steup
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Yasin Bektas
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Hongxia Li
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Johann M Kraus
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Stephan Kruger
- Department of Medicine III, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Steffen Ormanns
- Institute of Pathology, Faculty of Medicine, Ludwig Maximilian University of Munich, Munich, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, University of Ulm, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany.
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
17
|
Hadden M, Mittal A, Samra J, Zreiqat H, Sahni S, Ramaswamy Y. Mechanically stressed cancer microenvironment: Role in pancreatic cancer progression. Biochim Biophys Acta Rev Cancer 2020; 1874:188418. [PMID: 32827581 DOI: 10.1016/j.bbcan.2020.188418] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/21/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies in the world due to its insensitivity to current therapies and its propensity to metastases from the primary tumor mass. This is largely attributed to its complex microenvironment composed of unique stromal cell populations and extracellular matrix (ECM). The recruitment and activation of these cell populations cause an increase in deposition of ECM components, which highly influences the behavior of malignant cells through disrupted forms of signaling. As PDAC progresses from premalignant lesion to invasive carcinoma, this dynamic landscape shields the mass from immune defenses and cytotoxic intervention. This microenvironment influences an invasive cell phenotype through altered forms of mechanical signaling, capable of enacting biochemical changes within cells through activated mechanotransduction pathways. The effects of altered mechanical cues on malignant cell mechanotransduction have long remained enigmatic, particularly in PDAC, whose microenvironment significantly changes over time. A more complete and thorough understanding of PDAC's physical surroundings (microenvironment), mechanosensing proteins, and mechanical properties may help in identifying novel mechanisms that influence disease progression, and thus, provide new potential therapeutic targets.
Collapse
Affiliation(s)
- Matthew Hadden
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia
| | - Anubhav Mittal
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Australian Pancreatic Centre, St Leonards, Sydney, Australia
| | - Jaswinder Samra
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Australian Pancreatic Centre, St Leonards, Sydney, Australia
| | - Hala Zreiqat
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia; ARC Training Centre for Innovative Bioengineering, The University of Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sumit Sahni
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Australia; Kolling Institute of Medical Research, University of Sydney, Australia; Australian Pancreatic Centre, St Leonards, Sydney, Australia.
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
18
|
Starzyńska T, Karczmarski J, Paziewska A, Kulecka M, Kuśnierz K, Żeber-Lubecka N, Ambrożkiewicz F, Mikula M, Kos-Kudła B, Ostrowski J. Differences between Well-Differentiated Neuroendocrine Tumors and Ductal Adenocarcinomas of the Pancreas Assessed by Multi-Omics Profiling. Int J Mol Sci 2020; 21:E4470. [PMID: 32586046 PMCID: PMC7352720 DOI: 10.3390/ijms21124470] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023] Open
Abstract
Most pancreatic neuroendocrine tumors (PNETs) are indolent, while pancreatic ductal adenocarcinomas (PDACs) are particularly aggressive. To elucidate the basis for this difference and to establish the biomarkers, by using the deep sequencing, we analyzed somatic variants across coding regions of 409 cancer genes and measured mRNA/miRNA expression in nine PNETs, eight PDACs, and four intestinal neuroendocrine tumors (INETs). There were 153 unique somatic variants considered pathogenic or likely pathogenic, found in 50, 57, and 24 genes in PDACs, PNETs, and INETs, respectively. Ten and 11 genes contained a pathogenic mutation in at least one sample of all tumor types and in PDACs and PNETs, respectively, while 28, 34, and 11 genes were found to be mutated exclusively in PDACs, PNETs, and INETs, respectively. The mRNA and miRNA transcriptomes of PDACs and NETs were distinct: from 54 to 1659 differentially expressed mRNAs and from 117 to 250 differentially expressed miRNAs exhibited high discrimination ability and resulted in models with an area under the receiver operating characteristics curve (AUC-ROC) >0.9 for both miRNA and mRNA. Given the miRNAs high stability, we proposed exploring that class of RNA as new pancreatic tumor biomarkers.
Collapse
Affiliation(s)
- Teresa Starzyńska
- Department of Gastroenterology, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland;
| | - Jakub Karczmarski
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (A.P.); (M.K.); (F.A.); (M.M.)
| | - Agnieszka Paziewska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (A.P.); (M.K.); (F.A.); (M.M.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
| | - Maria Kulecka
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (A.P.); (M.K.); (F.A.); (M.M.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
| | - Katarzyna Kuśnierz
- Department of Gastrointestinal Surgery, Medical University of Silesia, 40-514 Katowice, Poland;
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
| | - Filip Ambrożkiewicz
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (A.P.); (M.K.); (F.A.); (M.M.)
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (A.P.); (M.K.); (F.A.); (M.M.)
| | - Beata Kos-Kudła
- Department of Endocrinology and Neuroendocrine Tumors, ENETS Center of Excelence, Department of Pathophysiology and Endocrinology, Medical University of Silesia, 40-514 Katowice, Poland;
| | - Jerzy Ostrowski
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (J.K.); (A.P.); (M.K.); (F.A.); (M.M.)
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland;
| |
Collapse
|
19
|
Häberle L, Esposito I. Circumferential resection margin (CRM) in pancreatic cancer. SURGERY IN PRACTICE AND SCIENCE 2020. [DOI: 10.1016/j.sipas.2020.100006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
20
|
Qin YY, Huang SN, Chen G, Pang YY, Li XJ, Xing WW, Wei DM, He Y, Rong MH, Tang XZ. Clinicopathological value and underlying molecular mechanism of annexin A2 in 992 cases of thyroid carcinoma. Comput Biol Chem 2020; 86:107258. [PMID: 32304977 DOI: 10.1016/j.compbiolchem.2020.107258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/30/2019] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Thyroid carcinoma (THCA) is one of the most frequent endocrine cancers and has increasing morbidity. Annexin A2 (ANXA2) has been found to be highly expressed in various cancers; however, its expression level and potential mechanism in THCA remain unknown. This study investigated the clinicopathological value and primary molecular machinery of ANXA2 in THCA. MATERIAL AND METHODS Public RNA-sequencing and microarray data were obtained and analyzed with ANXA2 expression in THCA and corresponding non-cancerous thyroid tissue. A Pearson correlation coefficient calculation was used for the acquisition of ANXA2 coexpressed genes, while edgR, limma, and Robust Rank Aggregation were employed for differentially expressed gene (DEG) in THCA. The probable mechanism of ANXA2 in THCA was predicted by gene ontology and pathway enrichment. A dual-luciferase reporter assay was employed to confirm the targeting relationships between ANXA2 and its predicted microRNA (miRNA). RESULTS Expression of ANXA2 was significantly upregulated in THCA tissues with a summarized standardized mean difference of 1.09 (P < 0.0001) based on 992 THCA cases and 589 cases of normal thyroid tissue. Expression of ANXA2 was related to pathologic stage. Subsequently, 1442 genes were obtained when overlapping 4542 ANXA2 coexpressed genes with 2248 DEGs in THCA; these genes were mostly enriched in pathways of extracellular matrix-receptor interaction, cell adhesion molecules, and complement and coagulation cascades. MiR-23b-3p was confirmed to target ANXA2 by dual-luciferase reporter assay. CONCLUSIONS Upregulated expression of ANXA2 may promote the malignant biological behavior of THCA by affecting the involving pathways or being targeted by miR-23b-3p.
Collapse
Affiliation(s)
- Yong-Ying Qin
- Department of Head and Neck Tumor Surgery, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Su-Ning Huang
- Department of Radiotherapy, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Yu-Yan Pang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Xiao-Jiao Li
- Department of PET/CT, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Wen-Wen Xing
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Dan-Ming Wei
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Yun He
- Department of Ultrasound, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Min-Hua Rong
- Department of Research, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China.
| | - Xiao-Zhun Tang
- Department of Head and Neck Tumor Surgery, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning, Guangxi Zhuang Autonomous Region, PR China.
| |
Collapse
|
21
|
Tenascin C in the Tumor-Nerve Microenvironment Enhances Perineural Invasion and Correlates With Locoregional Recurrence in Pancreatic Ductal Adenocarcinoma. Pancreas 2020; 49:442-454. [PMID: 32132519 DOI: 10.1097/mpa.0000000000001506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Perineural invasion is common in pancreatic ductal adenocarcinoma (PDAC) and worsens the postoperative prognosis. Tenascin C (TNC), an extracellular matrix glycoprotein, modulates tumor progression. We evaluated the functional roles of TNC, especially in perineural invasion of PDAC. METHODS We examined immunohistochemical TNC expression in 78 resected PDAC specimens. The relationships between TNC expression and clinicopathological features were retrospectively analyzed. Interactions between cancer cells and nerves with TNC supplementation were investigated using an in vitro coculture model with PDAC cell line and mouse dorsal root ganglion (DRG). RESULTS Tenascin C expression was predominant in perineural sites at the invasive tumor front. High perineural TNC expression in 30 patients (38%) was associated with perineural invasion, pathological T stage ≥3, and postoperative locoregional recurrence. High TNC expression was independently associated with postoperative, poor recurrence-free survival by multivariate analysis. In the in vitro coculture model, a TNC-rich matrix enhanced both PDAC cell colony extensions toward nerves and DRG axonal outgrowth toward cancer cell colonies, whereas TNC did not affect axonal outgrowth or cancer cell proliferation in separately cultured DRG and PDAC cells. CONCLUSIONS Strong perineural TNC expression indicated poor prognosis with locoregional recurrence. The neurotropism of TNC-induced PDAC suggests that TNC is a potential PDAC therapeutic target.
Collapse
|
22
|
Huang Y, Chen Y, Zhou S, Chen L, Wang J, Pei Y, Xu M, Feng J, Jiang T, Liang K, Liu S, Song Q, Jiang G, Gu X, Zhang Q, Gao X, Chen J. Dual-mechanism based CTLs infiltration enhancement initiated by Nano-sapper potentiates immunotherapy against immune-excluded tumors. Nat Commun 2020; 11:622. [PMID: 32001695 PMCID: PMC6992734 DOI: 10.1038/s41467-020-14425-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/06/2020] [Indexed: 12/29/2022] Open
Abstract
The failure of immunotherapies in immune-excluded tumor (IET) is largely ascribed to the void of intratumoral cytotoxic T cells (CTLs). The major obstacles are the excessive stroma, defective vasculatures and the deficiency of signals recruiting CTLs. Here we report a dual-mechanism based CTLs infiltration enhancer, Nano-sapper, which can simultaneously reduce the physical obstacles in tumor microenvironment and recruiting CTLs to potentiate immunotherapy in IET. Nano-sapper consists a core that co-loaded with antifibrotic phosphates-modified α-mangostin and plasmid encoding immune-enhanced cytokine LIGHT. Through reversing the abnormal activated fibroblasts, decreasing collagen deposition, normalizing the intratumoral vasculatures, and in situ stimulating the lymphocyte-recruiting chemoattractants expression, Nano-sapper paves the road for the CTLs infiltration, induces the intratumoral tertiary lymphoid structures, thus reshapes tumor microenvironment and potentiates checkpoint inhibitor against IET. This study demonstrates that the combination of antifibrotic agent and immune-enhanced cytokine might represent a modality in promoting immunotherapy against IET. The exclusion of cytotoxic T cells remains an important barrier to the efficacy of immunotherapies. Here the authors demonstrate that the combination anti-fibrosis agents and immune-enhanced cytokines can enhance T cell infiltration in a mouse model of pancreatic cancer.
Collapse
Affiliation(s)
- Yukun Huang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Yu Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Songlei Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Liang Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Jiahao Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Yuanyuan Pei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Jingxian Feng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Tianze Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Kaifan Liang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Shanshan Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China
| | - Qingxiang Song
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Gan Jiang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Xiao Gu
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Qian Zhang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China
| | - Xiaoling Gao
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, P.R. China.
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Lane 826, Zhangheng Road, Shanghai, 201203, P.R. China. .,Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, 2800 Gongwei Road, Shanghai, 201399, P.R. China.
| |
Collapse
|
23
|
Hagiwara K, Harimoto N, Yokobori T, Muranushi R, Hoshino K, Gantumur D, Yamanaka T, Ishii N, Tsukagoshi M, Igarashi T, Tanaka H, Watanabe A, Kubo N, Araki K, Hosouchi Y, Shirabe K. High Co-expression of Large Tenascin C Splice Variants in Stromal Tissue and Annexin A2 in Cancer Cell Membranes is Associated with Poor Prognosis in Pancreatic Cancer. Ann Surg Oncol 2019; 27:924-930. [PMID: 31463696 DOI: 10.1245/s10434-019-07708-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Pancreatic cancer tissue contains abundant stromal components, including extracellular matrix proteins such as tenascin C (TNC), which exists as large (TNC-L) and non-large splice variants. Here, we examined human pancreatic cancer specimens for the expression of total TNC (TNC-ALL) and TNC-L in the stroma and annexin A2 (ANXA2), a cell surface receptor for TNC, and evaluated their significance as prognostic markers for pancreatic cancer. METHODS Expression of ANXA2, TNC-ALL, and TNC-L was examined in 106 pancreatic cancer tissues from patients who underwent curative resection and who had not received prior therapy or surgery. Protein expression was measured by immunohistochemistry and scored on a semi-quantitative scale. The relationships between protein expression, clinicopathological factors, and prognosis were evaluated by Cox proportional hazards analysis. RESULTS TNC-ALL and TNC-L were detected mainly in the stroma, whereas ANXA2 was predominantly expressed in cancer cell membranes. TNC-ALL was also expressed in non-tumor pancreatic tissue. High levels of stromal TNC-L and membranous ANXA2, but not stromal TNC-ALL, were independently associated with cancer progression and poor prognosis. Moreover, high co-expression of stromal TNC-L and membranous ANXA2 was a superior indicator of poor prognosis compared with detection of TNC-ALL, TNC-L, or ANXA2 alone. CONCLUSIONS Our data suggest that co-expression of stromal TNC-L and membranous ANXA2 is a poor prognostic marker compared with detection of TNC-L or ANXA2 alone for pancreatic cancer patients. Additionally, targeting of crosstalk between stromal TNC and cancer cell ANXA2 could be a promising therapeutic strategy to overcome refractory pancreatic cancer.
Collapse
Affiliation(s)
- Kei Hagiwara
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Norifumi Harimoto
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan.
| | - Takehiko Yokobori
- Department of Innovative Cancer Immunotherapy, Gunma University, Maebashi, Japan.,Gunma University Initiative for Advanced Research (GIAR), Maebashi, Japan
| | - Ryo Muranushi
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Kouki Hoshino
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Dorgormaa Gantumur
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Takahiro Yamanaka
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Norihiro Ishii
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Mariko Tsukagoshi
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan.,Department of Innovative Cancer Immunotherapy, Gunma University, Maebashi, Japan
| | - Takamichi Igarashi
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Hiroshi Tanaka
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Akira Watanabe
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Norio Kubo
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Kenichiro Araki
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| | - Yasuo Hosouchi
- Department of Surgery and Laparoscopic Surgery, Gunma Prefecture Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Ken Shirabe
- Department of Hepatobiliary and Pancreatic Surgery, Gunma University, Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
24
|
He H, Xiao L, Cheng S, Yang Q, Li J, Hou Y, Song F, Su X, Jin H, Liu Z, Dong J, Zuo R, Song X, Wang Y, Zhang K, Duan W, Hou Y. Annexin A2 Enhances the Progression of Colorectal Cancer and Hepatocarcinoma via Cytoskeleton Structural Rearrangements. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2019; 25:950-960. [PMID: 31172894 DOI: 10.1017/s1431927619000679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Annexin A2 (ANXA2) is reported to be associated with cancer development. To investigate the roles ANXA2 plays during the development of cancer, the RNAi method was used to inhibit the ANXA2 expression in caco2 (human colorectal cancer cell line) and SMMC7721 (human hepatocarcinoma cell line) cells. The results showed that when the expression of ANXA2 was efficiently inhibited, the growth and motility of both cell lines were significantly decreased, and the development of the motility relevant microstructures, such as pseudopodia, filopodia, and the polymerization of microfilaments and microtubules were obviously inhibited. The cancer cell apoptosis was enhanced without obvious significance. The possible regulating pathway in the process was also predicted and discussed. Our results suggested that ANXA2 plays important roles in maintaining the malignancy of colorectal and hepatic cancer by enhancing the cell proliferation, motility, and development of the motility associated microstructures of cancer cells based on a possible complicated signal pathway.
Collapse
Affiliation(s)
- Huimin He
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Li Xiao
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Sinan Cheng
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Qian Yang
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Jinmei Li
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Yifan Hou
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Fengying Song
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Xiaorong Su
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Huijuan Jin
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Zheng Liu
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Jing Dong
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Ruiye Zuo
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Xigui Song
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Yanyan Wang
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Kun Zhang
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| | - Wei Duan
- School of Medicine, Deakin University,Waurn Ponds, VIC 3216,Australia
| | - Yingchun Hou
- Department of Cell Biology,College of Life Sciences, Shaanxi Normal University,620 West Chang-An Ave, Xi'an, Shaanxi 710119,China
| |
Collapse
|
25
|
Haeberle L, Esposito I. Pathology of pancreatic cancer. Transl Gastroenterol Hepatol 2019; 4:50. [PMID: 31304427 DOI: 10.21037/tgh.2019.06.02] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/04/2019] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy and estimated to become the second leading cause of cancer-related deaths by 2030. Although overall 5-year survival rates have constantly remained below 10% for the last decades, several key points important for accurate patient stratification have emerged during recent years. These key points include a highly standardized gross examination of PDAC resection specimens, using an axial slicing technique and inking of the circumferential resection margin (CRM), as well as a meticulous microscopic examination, taking into account the prognostic relevance of factors such as the exact resection status (R0 vs. R1 1-mm vs. R1 resection), histopathological tumor grading and the so-called lymph node ratio (LNR). With increasing use of neoadjuvant therapy in PDAC, tumor regression grading (TRG) for PDAC is currently rising in relevance in order to stratify and manage pre-operatively treated PDAC patients. As all current TRG systems for PDAC are unsatisfactory, new standardized international protocols are urgently needed. Several morphological subtypes of PDAC exist, some of which share the same molecular background with classical PDAC, while others are characterized by a distinct molecular pathogenesis. While some show a prognosis similar to classical PDAC, other subtypes stand out due to a better or even worse prognosis than classical PDAC. Prognostic relevant molecular subtypes of PDAC have been proposed as well, however, limitations of used cohorts and the lacking correlation of molecular subtypes with histomorphological subtypes limit the translation of these findings into valuable clinical applications. Lastly, several macroscopic and microscopic precursor lesions of PDAC have been described in genetically engineered mouse models (GEMM) and humans in recent times, providing further insight into PDAC carcinogenesis. In addition, improved diagnosis of PDAC precursors represents a chance to select patients for resection before invasive PDAC is present.
Collapse
Affiliation(s)
- Lena Haeberle
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Germany
| | - Irene Esposito
- Institute of Pathology, Heinrich Heine University and University Hospital of Duesseldorf, Germany
| |
Collapse
|
26
|
Leu SJJ, Lee TY, Cheng SW, Tsai MY, Lin YS, Chiou TJ, Huang KY, Chiang AN. Structural and functional characterization of β 2 -glycoprotein I domain 1 in anti-melanoma cell migration. Cancer Sci 2019; 110:1974-1986. [PMID: 31012976 PMCID: PMC6549912 DOI: 10.1111/cas.14030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
We previously found that circulating β2‐glycoprotein I inhibits human endothelial cell migration, proliferation, and angiogenesis by diverse mechanisms. In the present study, we investigated the antitumor activities of β2‐glycoprotein I using structure‐function analysis and mapped the critical region within the β2‐glycoprotein I peptide sequence that mediates anticancer effects. We constructed recombinant cDNA and purified different β2‐glycoprotein I polypeptide domains using a baculovirus expression system. We found that purified β2‐glycoprotein I, as well as recombinant β2‐glycoprotein I full‐length (D12345), polypeptide domains I‐IV (D1234), and polypeptide domain I (D1) significantly inhibited melanoma cell migration, proliferation and invasion. Western blot analyses were used to determine the dysregulated expression of proteins essential for intracellular signaling pathways in B16‐F10 treated with β2‐glycoprotein I and variant recombinant polypeptides. Using a melanoma mouse model, we found that D1 polypeptide showed stronger potency in suppressing tumor growth. Structural analysis showed that fragments A and B within domain I would be the critical regions responsible for antitumor activity. Annexin A2 was identified as the counterpart molecule for β2‐glycoprotein I by immunofluorescence and coimmunoprecipitation assays. Interaction between specific amino acids of β2‐glycoprotein I D1 and annexin A2 was later evaluated by the molecular docking approach. Moreover, five amino acid residues were selected from fragments A and B for functional evaluation using site‐directed mutagenesis, and P11A, M42A, and I55P mutations were shown to disrupt the anti‐melanoma cell migration ability of β2‐glycoprotein I. This is the first study to show the therapeutic potential of β2‐glycoprotein I D1 in the treatment of melanoma progression.
Collapse
Affiliation(s)
- Shr-Jeng Jim Leu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Tzong-Yi Lee
- Warshel Institute for Computational Biology, Chinese University of Hong Kong, Shenzhen, China
| | - Shu-Wei Cheng
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Meng-Ying Tsai
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Shan Lin
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Tzeon-Jye Chiou
- Division of Transfusion Medicine, Taipei Veterans General Hospital, and School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Kai-Yao Huang
- Warshel Institute for Computational Biology, Chinese University of Hong Kong, Shenzhen, China
| | - An-Na Chiang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
27
|
Abstract
OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is characterized by abundant stroma with increased expression of tenascin C and fibronectin. Their role and tumor-stroma ratio in PDAC are not well known. The aim of this study was to evaluate tenascin C and fibronectin expression and tumor-stroma ratio and their prognostic relevance in PDAC. METHODS Ninety-five resected PDACs were immunohistochemically stained for tenascin C and fibronectin, and the expression was separately assessed in tumor bulk and front. Tumor-stroma ratio was determined with sections stained with hematoxylin-eosin. RESULTS Tenascin C and fibronectin were abundantly expressed in the stroma of PDAC, but absent in adjacent normal pancreatic tissue. Fibronectin expression of the bulk was associated with high T class (P = 0.045). In the main analysis, tenascin C and fibronectin expression and tumor-stroma ratio were not associated with patient survival. In a subgroup analysis of early-stage PDAC (T1-T2 tumors), high tenascin C expression in the tumor bulk was associated with poor prognosis (hazard ratio, 8.23; 95% confidence interval, 2.71-24.96). CONCLUSIONS Tenascin C and fibronectin are abundantly expressed in PDAC, but they seem to have no major association with patient survival. However, in early-stage PDAC, tenascin C expression of the tumor bulk may have prognostic impact. Tumor-stroma ratio has no prognostic value in PDAC.
Collapse
|
28
|
Reciprocal regulation of pro-inflammatory Annexin A2 and anti-inflammatory Annexin A1 in the pathogenesis of rheumatoid arthritis. Mol Biol Rep 2018; 46:83-95. [PMID: 30426384 DOI: 10.1007/s11033-018-4448-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023]
Abstract
Annexin A2 has been implicated in several immune modulated diseases including Rheumatoid arthritis (RA) pannus formation. The most relied treatment option for RA pathogenesis is glucocorticoids. Glucocorticoids regulate the synthesis, phosphorylation and cellular deposition of Annexin A1. This annexin mediates the anti-inflammatory actions of glucocorticoids. These two first characterized members of annexin superfamily proteins acts reciprocally, one as an anti-inflammatory and the other proinflammatory in nature. The possibility of these molecules as soluble biomarkers and as an upstream regulator of major cytokine devastation at RA microenvironment has not been previously explored. Current study elucidates the reciprocal regulation of these two annexins in RA pathogenesis. These Annexin A2/A1 and downstream cytokines in RA serum were analysed by ELISA. Western blot, Immunocytochemistry, immunoprecipitation and Immunohistochemistry were adapted to analyse these molecules in tissue and synovial fibroblasts and also in different experimental conditions. Significant increase in the level of Annexin A2 was noticed in naïve RA patients compared to controls (14.582 ± 1.766 ng/ml vs. 7.37 ± 1.450 ng/ml; p ≤ 0.001). In remission cases significant low levels was detected. On the contrary, significant decrease in the level of Annexin A1 was noticed in naïve RA patients compared to healthy controls (12.322 ± 2.91 vs. 16.998 ± 4.298 ng/ml; p ≤ 0.001), wherein remission cases serum Annexin A1 was significantly high. The knockdown of proinflammatory Annexin A2 by siRNA/antibody treatment could mimic the glucocorticoid treatment as which induced cellular Annexin A1 and membrane translocation resulting in the terminal action. Current data elucidating the regulatory interplay between Annexin A2 and Annexin A1 in RA pathogenesis.
Collapse
|
29
|
Nielsen MFB, Mortensen MB, Detlefsen S. Typing of pancreatic cancer-associated fibroblasts identifies different subpopulations. World J Gastroenterol 2018; 24:4663-4678. [PMID: 30416314 PMCID: PMC6224473 DOI: 10.3748/wjg.v24.i41.4663] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 10/12/2018] [Accepted: 10/21/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To determine whether it is possible to identify different immune phenotypic subpopulations of cancer-associated fibroblasts (CAFs) in pancreatic cancer (PC).
METHODS We defined four different stromal compartments in surgical specimens with PC: The juxtatumoural, peripheral, lobular and septal stroma. Tissue microarrays were produced containing all pre-defined PC compartments, and the expression of 37 fibroblast (FB) and 8 extracellular matrix (ECM) markers was evaluated by immunohistochemistry, immunofluorescence (IF), double-IF, and/or in situ hybridization. The compartment-specific mean labelling score was determined for each marker using a four-tiered scoring system. DOG1 gene expression was examined by quantitative reverse transcription PCR (qPCR).
RESULTS CD10, CD271, cytoglobin, DOG1, miR-21, nestin, and tenascin C exhibited significant differences in expression profiles between the juxtatumoural and peripheral compartments. The expression of CD10, cytoglobin, DOG1, nestin, and miR-21 was moderate/strong in juxtatumoural CAFs (j-CAFs) and barely perceptible/weak in peripheral CAFs (p-CAFs). The upregulation of DOG1 gene expression in PC compared to normal pancreas was verified by qPCR. Tenascin C expression was strong in the juxtatumoural ECM and barely perceptible/weak in the peripheral ECM. CD271 expression was barely perceptible in j-CAFs but moderate in the other compartments. Galectin-1 was stronger expressed in j-CAFs vs septal fibroblasts, PDGF-Rβ, tissue transglutaminase 2, and hyaluronic acid were stronger expressed in lobular fibroblasts vs p-CAFs, and plectin-1 was stronger expressed in j-CAFs vs l-FBs. The expression of the remaining 33 markers did not differ significantly when related to the quantity of CAFs/FBs or the amount of ECM in the respective compartments.
CONCLUSION Different immune phenotypic CAF subpopulations can be identified in PC, using markers such as cytoglobin, CD271, and miR-21. Future studies should determine whether CAF subpopulations have different functional properties.
Collapse
Affiliation(s)
- Michael Friberg Bruun Nielsen
- Department of Pathology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Odense Pancreas Center (OPAC), Odense C 5000, Denmark
| | - Michael Bau Mortensen
- Department of Surgery, HPB Section, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Odense Pancreas Center (OPAC), Odense C 5000, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Odense Pancreas Center (OPAC), Odense C 5000, Denmark
| |
Collapse
|
30
|
Cai J, Lu W, Du S, Guo Z, Wang H, Wei W, Shen X. Tenascin-C Modulates Cell Cycle Progression to Enhance Tumour Cell Proliferation through AKT/FOXO1 Signalling in Pancreatic Cancer. J Cancer 2018; 9:4449-4462. [PMID: 30519351 PMCID: PMC6277647 DOI: 10.7150/jca.25926] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a disease with an extremely poor prognosis that is characterized by a rich extracellular matrix (ECM). Tenascin-C (TNC) is a component of the ECM and plays a role in tumour progression. In this study, we reported that TNC is overexpressed in PDAC tissues and is correlated with tumour stage and cyclin D1 expression. Cyclin D1 is key regulator of the cell cycle G1/S transition. Further experiments revealed that TNC promotes G1/S transition through AKT signalling. TNC/AKT increases the expression of cyclin D1 by enhancing the transcriptional activity of β-catenin, whereas the translocation of FOXO1 from the nucleus results in the downregulation of p27Kip1. Cyclin D1 and p27Kip1 regulate the activity of cyclin D1-CDK4 complexes and retinoblastoma (Rb), and then they stimulate the progression of G1/S transition and tumour cell proliferation. In conclusion, TNC exerts its activating effect on the proliferation of pancreatic cancer cells in vitro and in vivo through its functional target AKT/FOXO1/β-catenin. The molecular mechanisms that drive PDAC progression will be useful for the development of molecular markers and the evaluation of patient prognosis.
Collapse
Affiliation(s)
- Jun Cai
- School of medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wenli Lu
- Department of Health Statistics, School of Public Health, Tianjin Medical University, Tianjin 300000, China
| | - Shaoxia Du
- School of medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Zhongkui Guo
- School of medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Hui Wang
- School of medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Wei Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Huanhu West Road, Tianjin 300060, China
| | - Xiaohong Shen
- School of medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| |
Collapse
|
31
|
Sharma MC. Annexin A2 (ANX A2): An emerging biomarker and potential therapeutic target for aggressive cancers. Int J Cancer 2018; 144:2074-2081. [PMID: 30125343 DOI: 10.1002/ijc.31817] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
ANX A2 is an important member of annexin family of proteins expressed on surface of endothelial cells (ECs), macrophages, mononuclear cells and various types of cancer cells. It exhibits high affinity binding for calcium (Ca++ ) and phospholipids. ANX A2 plays an important role in many biological processes such as endocytosis, exocytosis, autophagy, cell-cell communications and biochemical activation of plasminogen. On the cell surface ANX A2 organizes the assembly of plasminogen (PLG) and tissue plasminogen activator (tPA) for efficient conversion of PLG to plasmin, a serine protease. Proteolytic activity of plasmin is required for activation of inactive pro-metalloproteases (pro-MMPs) and latent growth factors for their biological actions. These activation steps are critical for degradation of extracellular matrix (ECM) and basement proteins (BM) for cancer cell invasion and metastasis. Increased expression of ANX A2 protein/gene has been correlated with invasion and metastasis in a variety of human cancers. Moreover, clinical studies have positively correlated ANX A2 protein expression with aggressive cancers and with resistance to anticancer drugs, shorter disease-free survival (DFS), and worse overall survival (OS). The mechanism(s) by which ANX A2 regulates cancer invasion and metastasis are beginning to emerge. Investigators used various technologies to target ANX A2 in preclinical model of human cancers and demonstrated exciting results. In this review article, we analyzed existing literature concurrent with our own findings and provided a critical overview of ANX A2-dependent mechanism(s) of cancer invasion and metastasis.
Collapse
Affiliation(s)
- Mahesh C Sharma
- Research Service, Veterans Affairs Medical Center, Washington, DC.,Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC
| |
Collapse
|
32
|
Mirzaei R, Sarkar S, Dzikowski L, Rawji KS, Khan L, Faissner A, Bose P, Yong VW. Brain tumor-initiating cells export tenascin-C associated with exosomes to suppress T cell activity. Oncoimmunology 2018; 7:e1478647. [PMID: 30288344 DOI: 10.1080/2162402x.2018.1478647] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 05/01/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
The dismal prognosis of glioblastoma is attributed in part to the existence of stem-like brain tumor-initiating cells (BTICs) that are highly radio- and chemo-resistant. New approaches such as therapies that reprogram compromised immune cells against BTICs are needed. Effective immunotherapies in glioblastoma, however, remain elusive unless the mechanisms of immunosuppression by the tumor are better understood. Here, we describe that while the conditioned media of activated T lymphocytes reduce the growth capacity of BTICs, this growth suppression was abrogated in live co-culture of BTICs with T cells. We present evidence that BTICs produce the extracellular matrix protein tenascin-C (TNC) to inhibit T cell activity in live co-culture. In human glioblastoma brain specimens, TNC was widely deposited in the vicinity of T cells. Mechanistically, TNC inhibited T cell proliferation through interaction with α5β1 and αvβ6 integrins on T lymphocytes associated with reduced mTOR signaling. Strikingly, TNC was exported out of BTICs associated with exosomes, and TNC-depleted exosomes suppressed T cell responses to a significantly lesser extent than control. Finally, we found that circulating exosomes from glioblastoma patients contained more TNC and T cell-suppressive activity than those from control individuals. Taken together, our study establishes a novel immunosuppressive role for TNC associated with BTIC-secreted exosomes to affect local and distal T lymphocyte immunity.
Collapse
Affiliation(s)
- Reza Mirzaei
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Susobhan Sarkar
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Lauren Dzikowski
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Khalil S Rawji
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Lubaba Khan
- Departments of Biochemistry and Molecular Biology, Oncology and Surgery, Ohlson Research Initiative and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Pinaki Bose
- Departments of Biochemistry and Molecular Biology, Oncology and Surgery, Ohlson Research Initiative and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
33
|
Stromal heterogeneity in pancreatic cancer and chronic pancreatitis. Pancreatology 2018; 18:536-549. [PMID: 29778400 DOI: 10.1016/j.pan.2018.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 05/09/2018] [Accepted: 05/11/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES An abundant stromal reaction is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and chronic pancreatitis (CP). The cells mainly responsible for the stromal reaction are activated pancreatic stellate cells (PSCs). Despite their crucial role, PSCs are not well characterized. PSCs share characteristics with the better-known hepatic stellate cells (HSCs). The aim of this study was a detailed analysis of PSCs in PDAC and CP. METHODS Whole-slide specimens of CP (n = 12) and PDAC (n = 10) were studied by histochemistry and immunohistochemistry. The stroma was evaluated using Movat's pentachrome stain. PSCs were tested by immunohistochemistry for PSC markers (α-SMA, CD34, desmin, NGFR, SPARC and tenascin C) and HSC markers (α-crystallin B, CD56, NGF, NT-3, synaptophysin and TrkC). Alpha-SMA, tenascin C, SPARC and NT-3 staining were verified on tissue micro arrays (TMAs) from a well-characterized cohort of 223 PDAC patients. PSCs isolated from human PDAC and CP tissue samples as well as HSCs were evaluated by immunofluorescence. RESULTS While the stroma of CP cases was characterized by a collagen-rich fibrosis, PDAC stroma displayed higher mucin content (p = 0.0002). PSCs showed variable expression of tested markers. In PDAC samples, staining of most markers was found around tumor complexes, while CP samples showed a greater variety of localizations. Alpha-SMA staining correlated with collagen-rich fibrosis (p = 0.012), while NT-3 staining correlated with mucin-rich stroma (p = 0.008). A peritumoral staining was confirmed for α-SMA, tenascin C, SPARC and NT-3 in the PDAC TMA cohort (n = 223). In a subgroup of patients with pancreatic head tumors and UICC 2009 IIB (n = 144), α-SMA staining intensity was a prognostic factor for overall survival at uni- and multivariate analysis (p = 0.036 and p = 0.002). CONCLUSIONS The close similarities between PSCs and HSCs were confirmed. Heterogeneous expression patterns of the tested markers might reflect different levels of activation or differentiation, or even multiple subpopulations of PSCs. Survival analysis suggests an impact of stromal composition on survival.
Collapse
|
34
|
Zheng J, Hernandez JM, Doussot A, Bojmar L, Zambirinis CP, Costa-Silva B, van Beek EJ, Mark MT, Molina H, Askan G, Basturk O, Gonen M, Kingham TP, Allen PJ, D’Angelica MI, DeMatteo RP, Lyden D, Jarnagin WR. Extracellular matrix proteins and carcinoembryonic antigen-related cell adhesion molecules characterize pancreatic duct fluid exosomes in patients with pancreatic cancer. HPB (Oxford) 2018; 20:597-604. [PMID: 29339034 PMCID: PMC6779041 DOI: 10.1016/j.hpb.2017.12.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/27/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Exosomes are nanovesicles that have been shown to mediate carcinogenesis in pancreatic ductal adenocarcinoma (PDAC). Given the direct communication of pancreatic duct fluid with the tumor and its relative accessibility, we aimed to determine the feasibility of isolating and characterizing exosomes from pancreatic duct fluid. METHODS Pancreatic duct fluid was collected from 26 patients with PDAC (n = 13), intraductal papillary mucinous neoplasm (IPMN) (n = 8) and other benign pancreatic diseases (n = 5) at resection. Exosomes were isolated by serial ultracentrifugation, proteins were identified by mass spectrometry, and their expression was evaluated by immunohistochemistry. RESULTS Exosomes were isolated from all specimens with a mean concentration of 5.9 ± 1 × 108 particles/mL and most frequent size of 138 ± 9 nm. Among the top 35 proteins that were significantly associated with PDAC, multiple carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) and extracellular matrix (ECM) proteins were identified. Interestingly, CEACAM 1/5 expression by immunohistochemistry was seen only on tumor epithelia whereas tenascin C positivity was restricted to stroma, suggesting that both tumor and stromal cells contributed to exosomes. CONCLUSION This is the first study showing that exosome isolation is feasible from pancreatic duct fluid, and that exosomal proteins may be utilized to diagnose patients with PDAC.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Alexandre Doussot
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Linda Bojmar
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | | | - Bruno Costa-Silva
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Elke J.A.H. van Beek
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Milica Tesic Mark
- Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Henrik Molina
- Proteomics Resource Center, The Rockefeller University, New York, NY, USA
| | - Gokce Askan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T. Peter Kingham
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter J. Allen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Ronald P. DeMatteo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Lyden
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - William R. Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Corresponding author: William R. Jarnagin, MD, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue C-891, New York, NY 10065, Phone: 212-639-3624; Fax: 917-432-2387,
| |
Collapse
|
35
|
Yoneura N, Takano S, Yoshitomi H, Nakata Y, Shimazaki R, Kagawa S, Furukawa K, Takayashiki T, Kuboki S, Miyazaki M, Ohtsuka M. Expression of annexin II and stromal tenascin C promotes epithelial to mesenchymal transition and correlates with distant metastasis in pancreatic cancer. Int J Mol Med 2018; 42:821-830. [PMID: 29749431 PMCID: PMC6034933 DOI: 10.3892/ijmm.2018.3652] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/30/2018] [Indexed: 12/24/2022] Open
Abstract
The interaction between cancer cells and stromal components contributes to cancer invasion and metastasis in pancreatic ductal adenocarcinoma (PDAC). The present study investigated the role of the correlation between annexin II (ANX2) and stromal tenascin C (TNC) with the progression of PDAC. The functions of the expression ANX2 and TNC were assessed in in vitro experiments using mouse and human PDAC cells, and the clinical effect was analyzed using immunohistochemistry with surgically resected PDAC tissues. The effects on epithelial to mesenchymal transition (EMT), invasion, putative cancer stemness, and anoikis resistance were examined in vitro using murine precancerous pancreatic intraepithelial neoplasia (PanIN) cells and murine and human invasive PDAC cells with ANX2 knockdown using specific small interfering RNA (siRNA)s and recombinant TNC (rTNC). ANX2 was expressed at a high level in primary PanIN cells and invasive PDAC cells, compared with the levels in liver metastatic PDAC cells. In the ANX2-knockdown cells, there were fewer cells with a morphological mesenchymal appearance in three-dimensional culture and invasion was reduced compared with that in the control cells. Morphological change into the mesenchymal phenotype and invasion were enhanced by rTNC treatment in the control PDAC cells but not in the ANX2-knockdown cells. Pancreatosphere formation assays showed that ANX2 and TNC facilitated the maintenance of stem-like characters in PDAC cells. Furthermore, anoikis assays indicated that the interaction of ANX2-TNC contributed to anoikis resistance in PDAC cells. In the immunohistochemistry analyses, the group with a high expression of ANX2 and high stromal TNC was significantly correlated with distant metastasis, and was associated with hematogenous/peritoneal recurrence and poor outcomes following surgery in resected human primary PDAC tissues. In conclusion, the results demonstrated that ANX2 and stromal TNC regulated invasion in addition to stemness and anoikis resistance, which are crucial for metastasis in the progression of PDAC. These results indicate the potential of the ANX2-TNC axis as a therapeutic target for PDAC metastasis.
Collapse
Affiliation(s)
- Naoko Yoneura
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Hideyuki Yoshitomi
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Yasuyuki Nakata
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Reiri Shimazaki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Shingo Kagawa
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Katsunori Furukawa
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Satoshi Kuboki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Masaru Miyazaki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba 260‑8677, Japan
| |
Collapse
|
36
|
Hayasaki A, Murata Y, Usui M, Hibi T, Ito T, Iizawa Y, Kato H, Tanemura A, Azumi Y, Kuriyama N, Kishiwada M, Mizuno S, Sakurai H, Yoshida T, Isaji S. Clinical Significance of Histological Effect and Intratumor Stromal Expression of Tenascin-C in Resected Specimens After Chemoradiotherapy for Initially Locally Advanced Unresectable Pancreatic Ductal Adenocarcinoma. Pancreas 2018; 47:390-399. [PMID: 29517632 DOI: 10.1097/mpa.0000000000001022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Tenascin-C (TN-C) is an extracellular matrix protein that is up-regulated in pancreatic ductal adenocarcinoma (PDAC) stroma and associated with tumor invasion. We examined intratumor stromal expression of TN-C in resected specimens and the histologic effect of chemoradiotherapy (CRT) as prognostic indicators in initially locally advanced unresectable (UR-LA) PDAC. METHODS Among 110 UR-LA PDAC patients enrolled in the CRT protocol from February 2005 to December 2015, 46 who underwent curative-intent resection were classified as high (tumor destruction >50%) and low (≤50%) responders according to the Evans grading system. Tenascin-C expression was immunohistologically evaluated in all patients except one with complete response. RESULTS The 12 high responders achieved a significantly higher R0 rate than did the 34 low responders (83.3 vs 47.1%), but disease-specific survival (DSS) time was not significantly different (median survival time, 29.8 vs 21.0 months). Tenascin-C expression was inversely correlated with histologic effect of CRT. The 22 patients with negative TN-C had significantly longer DSS time than did the 23 with positive TN-C (29.3 vs 17.1 months). In multivariate analysis, only TN-C expression was a significant prognostic factor for DSS. CONCLUSIONS Intratumor stromal expression of TN-C is a strong prognostic indicator in UR-LA PDAC patients with resection after CRT.
Collapse
|
37
|
Yang CT, Li JM, Li LF, Ko YS, Chen JT. Stomatin-like protein 2 regulates survivin expression in non-small cell lung cancer cells through β-catenin signaling pathway. Cell Death Dis 2018; 9:425. [PMID: 29556045 PMCID: PMC5859036 DOI: 10.1038/s41419-018-0461-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 01/15/2023]
Abstract
The overexpression of stomatin-like protein-2 (SLP-2) is commonly observed in non-small cell lung cancer (NSCLC) cells. In the present study, we transfected a number of NSCLC cells with an SLP-2 shRNA-expressing vector (AdSLP2i) and examined its possible effects on cell growth and apoptosis. We found that suppression of SLP-2 expression inhibited cell growth, and that the apoptosis induced by SLP-2 suppression was correlated with decreased survivin protein expression. Moreover, the reduced survivin expression was found to be associated with reduced β-catenin nuclear localization and appeared not to be modulated through the AKT signaling pathway. By using immunoprecipitation and proteomics to analyze protein-protein interactions in A549 cells with SLP-2 overexpression, we found that annexin A2 interacted with SLP-2 and β-catenin directly. Our data further suggested that the knockdown of SLP-2 gene affected the SLP-2/Annexin A2/β-catenin cascade formation, reduced the translocation of cytoplasmic β-catenin into nucleus, and downregulated downstream target genes. The results presented in this study, together with our previous findings, suggest that SLP-2 promotes NSCLC cell proliferation by enhancing survivin expression mediated via β-catenin pathway.
Collapse
Affiliation(s)
- Cheng-Ta Yang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Jhy-Ming Li
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Fu Li
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Shien Ko
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Cardiology, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Jeng-Ting Chen
- Department of Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
38
|
Barrett AS, Maller O, Pickup MW, Weaver VM, Hansen KC. Compartment resolved proteomics reveals a dynamic matrisome in a biomechanically driven model of pancreatic ductal adenocarcinoma. JOURNAL OF IMMUNOLOGY AND REGENERATIVE MEDICINE 2018; 1:67-75. [PMID: 36908331 PMCID: PMC10003644 DOI: 10.1016/j.regen.2018.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a severe fibrotic component that compromises treatment, alters the immune cell profile and contributes to patient mortality. It has been shown that early on in this process, dynamic changes in tissue biomechanics play an integral role in supporting pancreatic cancer development and progression. Despite the acknowledgement of its importance, a granular view of how stromal composition changes during the course of PDAC progression remains largely unknown. To mimic the quasi-mesenchymal phenotype and pronounced desmoplastic response observed clinically, we utilized a genetically engineered mouse model of PDAC that is driven by a KrasG12D mutation and loss of Tgfbr2 expression. Application of compartment resolved proteomics revealed that PDAC progression in this KTC model is associated with dynamic stromal alterations that are indicative of a wound healing program. We identified an early provisional matricellular fibrosis that was accompanied by markers of macrophage activation and infiltration, consistent with the inflammatory phase of wound healing. At 20 weeks a proliferative phenotype was observed with increased fibroblast markers, further collagen deposition and loss of basement membrane and native cell markers.
Collapse
Affiliation(s)
- Alexander S Barrett
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Ori Maller
- Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Michael W Pickup
- Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Valerie M Weaver
- Department of Surgery, University of California San Francisco, San Francisco, California, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
39
|
Experimental models of pancreatic cancer desmoplasia. J Transl Med 2018; 98:27-40. [PMID: 29155423 DOI: 10.1038/labinvest.2017.127] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/12/2017] [Accepted: 10/12/2017] [Indexed: 01/18/2023] Open
Abstract
Desmoplasia is a fibro-inflammatory process and a well-established feature of pancreatic cancer. A key contributor to pancreatic cancer desmoplasia is the pancreatic stellate cell. Various in vitro and in vivo methods have emerged for the isolation, characterization, and use of pancreatic stellate cells in models of cancer-associated fibrosis. In addition to cell culture models, genetically engineered animal models have been established that spontaneously develop pancreatic cancer with desmoplasia. These animal models are currently being used for the study of pancreatic cancer pathogenesis and for evaluating therapeutics against pancreatic cancer. Here, we review various in vitro and in vivo models that are being used or have the potential to be used to study desmoplasia in pancreatic cancer.
Collapse
|
40
|
Ji T, Lang J, Wang J, Cai R, Zhang Y, Qi F, Zhang L, Zhao X, Wu W, Hao J, Qin Z, Zhao Y, Nie G. Designing Liposomes To Suppress Extracellular Matrix Expression To Enhance Drug Penetration and Pancreatic Tumor Therapy. ACS NANO 2017; 11:8668-8678. [PMID: 28806504 DOI: 10.1021/acsnano.7b01026] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
During pancreatic tumor development, pancreatic stellate cells (PSCs) proliferate exuberantly to secrete extracellular matrix (ECM) in the tumor stroma, which presents major barriers for drug delivery and penetration in tumor tissue. Thus, down-regulating ECM levels via regulation of the PSCs may allow enhanced penetration of therapeutic drugs and thereby enhancing their therapeutic efficacy. To regulate the PSCs, a matrix metalloproteinase-2 (MMP-2) responsive peptide-hybrid liposome (MRPL) was constructed via coassembly of a tailor-designed MMP-2 responsive amphiphilic peptide and phospholipids. By utilizing the MMP-2-rich pathological environment, the pirfenidone (PFD) loaded MRPL (MRPL-PFD) can specifically release PFD at the pancreatic tumor site and down-regulate the multiple components of ECM expressed by the PSCs. This resulted in a significant increase in the penetration of gemcitabine into the tumor tissue and enhanced the efficacy of gemcitabine for pancreatic tumor. Our design tailored for antifibrosis of pancreatic cancer may provide a practical approach to build functional liposomes through supramolecular assembly, and regulation of ECM may be a promising adjuvant therapeutic strategy for pancreatic and other ECM-rich tumors.
Collapse
Affiliation(s)
- Tianjiao Ji
- The First Affiliated Hospital, Zhengzhou University , Zhengzhou 450052, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jiayan Lang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- College of Pharmaceutical Science, Jilin University , Changchun 130021, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Feifei Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Lijing Zhang
- The First Affiliated Hospital, Zhengzhou University , Zhengzhou 450052, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Wenjing Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- College of Pharmaceutical Science, Jilin University , Changchun 130021, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Jihui Hao
- Department of Pancreatic Carcinoma Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy , Tianjin 300060, China
| | - Zhihai Qin
- The First Affiliated Hospital, Zhengzhou University , Zhengzhou 450052, China
| | - Ying Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Beijing 100190, China
- University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
41
|
Abstract
Inflammatory and fibrotic events that drive chronic pancreatitis (CP) are likely orchestrated via signaling of soluble cytokines and chemokines systemically and within the pancreas. However, a comprehensive summary of the expression of such factors during CP has not been reported to date. This information is important given continued interest in targeting cytokines that influence CP pathogenesis. Reported data on the expression change of soluble immunomodulatory factors in human CP patients were identified via a literature search using a single search term. Thirty-one articles meeting the prespecified inclusion criteria were identified to generate a compiled data summary. Compiled data demonstrated up-regulation of several factors in the blood or pancreas microenvironment of CP patients. Nine factors were elevated in both compartments, including fractalkine, IFN-γ, interleukin 1β, IL-6, IL-8, macrophage inhibitory cytokine 1, neutrophil gelatinase-associated lipocalin, transforming growth factor β, and tumor necrosis factor α. Most up-regulated factors could be classified into one of several functional groups, including inflammation, chemotaxis, angiogenesis, bone remodeling, extracellular matrix remodeling, and pain. After further validation, these factors may be used as biomarkers for disease diagnosis and identification of comorbidities, or as potential therapeutic targets.
Collapse
|
42
|
Cai J, Du S, Wang H, Xin B, Wang J, Shen W, Wei W, Guo Z, Shen X. Tenascin-C induces migration and invasion through JNK/c-Jun signalling in pancreatic cancer. Oncotarget 2017; 8:74406-74422. [PMID: 29088796 PMCID: PMC5650351 DOI: 10.18632/oncotarget.20160] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 06/20/2017] [Indexed: 12/21/2022] Open
Abstract
Tenascin-C (TNC), a large extracellular matrix glycoprotein, has been reported to be associated with metastasis and poor prognosis in pancreatic cancer. However, the effects and mechanisms of TNC in pancreatic cancer metastasis largely remain unclear. We performed Transwell assays to investigate the effects of TNC on Capan-2, AsPC-1 and PANC-1 cells. In addition, western blot and RT-qPCR assays were used to examine potential TNC metastasis-associated targets, such as JNK/c-Jun, Paxillin/FAK, E-cadherin, N-cadherin, Vimentin, and MMP9/2. Lastly, we utilized a variety of methods, such as immunofluorescence, gelatin zymography and immunoprecipitation, to determine the molecular mechanisms of TNC in pancreatic cancer cell motility. The present study showed that TNC induced migration and invasion in pancreatic cancer cells and regulated a number of metastasis-associated proteins, including the EMT markers, MMP9 and Paxillin. Moreover, our data showed that TNC induced pancreatic cancer cells to generate an EMT phenotype and acquire motility potential through the activation of JNK/c-Jun signalling. In addition, TNC increased the DNA binding activity of c-Jun to the MMP9 promoter, an action likely resulting in increased MMP9 expression and activity. TNC/JNK also markedly induced the phosphorylation of Paxillin on serine 178, which is critical for the association between FAK and Paxillin and promoted the formation of focal adhesions. TNC/JNK initiates cell migration and invasion of pancreatic cancer cells through the promotion of EMT, the transactivation of MMP9 and the phosphorylation of Paxillin on serine 178. TNC may be a potential therapeutic target for treating pancreatic cancer metastasis.
Collapse
Affiliation(s)
- Jun Cai
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Shaoxia Du
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Hui Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Beibei Xin
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Juan Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Wenyuan Shen
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Wei Wei
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhongkui Guo
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaohong Shen
- School of Medicine, Nankai University, Tianjin 300071, China
| |
Collapse
|
43
|
Intracellular targeting of annexin A2 inhibits tumor cell adhesion, migration, and in vivo grafting. Sci Rep 2017; 7:4243. [PMID: 28652618 PMCID: PMC5484684 DOI: 10.1038/s41598-017-03470-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 05/03/2017] [Indexed: 12/22/2022] Open
Abstract
Cytoskeletal-associated proteins play an active role in coordinating the adhesion and migration machinery in cancer progression. To identify functional protein networks and potential inhibitors, we screened an internalizing phage (iPhage) display library in tumor cells, and selected LGRFYAASG as a cytosol-targeting peptide. By affinity purification and mass spectrometry, intracellular annexin A2 was identified as the corresponding binding protein. Consistently, annexin A2 and a cell-internalizing, penetratin-fused version of the selected peptide (LGRFYAASG-pen) co-localized and specifically accumulated in the cytoplasm at the cell edges and cell-cell contacts. Functionally, tumor cells incubated with LGRFYAASG-pen showed disruption of filamentous actin, focal adhesions and caveolae-mediated membrane trafficking, resulting in impaired cell adhesion and migration in vitro. These effects were paralleled by a decrease in the phosphorylation of both focal adhesion kinase (Fak) and protein kinase B (Akt). Likewise, tumor cells pretreated with LGRFYAASG-pen exhibited an impaired capacity to colonize the lungs in vivo in several mouse models. Together, our findings demonstrate an unrecognized functional link between intracellular annexin A2 and tumor cell adhesion, migration and in vivo grafting. Moreover, this work uncovers a new peptide motif that binds to and inhibits intracellular annexin A2 as a candidate therapeutic lead for potential translation into clinical applications.
Collapse
|
44
|
Zhu J, Wu J, Pei X, Tan Z, Shi J, Lubman DM. Annexin A10 is a candidate marker associated with the progression of pancreatic precursor lesions to adenocarcinoma. PLoS One 2017; 12:e0175039. [PMID: 28369074 PMCID: PMC5378402 DOI: 10.1371/journal.pone.0175039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/20/2017] [Indexed: 12/22/2022] Open
Abstract
Annexins are a multigene family of calcium and phospholipid-binding proteins that play important roles in calcium signaling, cell motility, differentiation and proliferation. Our previous mass spectrometry-based proteomics study revealed that annexin A10 (ANXA10) was uniquely overexpressed in pancreatic CD24+ adenocarcinoma cells that were dissected from clinical PDAC tissues but was absent in CD24- adjacent normal cells. The correlation between ANXA10 expression and the progression of pancreatic cancer remains unknown. In this study, we performed an immunostaining assay to evaluate ANXA10 expression in 155 primary human tissue specimens, including normal pancreas, chronic pancreatitis (CP), pancreatic adenocarcinoma (PDAC), pancreatic intraepithelial neoplasia (PanIN, the most important precursor of PDAC), and intraductal papillary mucinous neoplasm (IPMN). The immunostaining result showed that ANXA10 was significantly overexpressed in PanINs, IPMNs, and PDACs but negative in normal pancreas and the majority of chronic pancreatitis tissues. Statistical analysis revealed that ANXA10 expression was significantly associated with PDAC and its precursor lesions (p<0.0001). Abundant ANXA10 expression was predominantly present in pancreatic ductal epithelial cells of PanINs, IPMNs, and tumor cells of PDACs. Since PDAC develops through a series of PanINs which in turn arise from pancreatic ducts, the consistent overexpression of ANXA10 in ductal epithelial cells in PanINs and PDACs but negative in normal pancreatic ducts suggests that ANXA10 could serve as a potential marker indicating the presence of PDAC at its earliest precancerous stages. Double immunostaining of ANXA10 and CD24 showed that there was a large overlap between these two markers in PDAC and high-grade neoplasia lesions. The statistical analysis showed that the coexpression of ANXA10 and CD24 was significantly correlated with the progression of pancreatic precursor lesions towards PDACs.
Collapse
Affiliation(s)
- Jianhui Zhu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States of America
| | - Jing Wu
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States of America
| | - Xiucong Pei
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States of America
- Department of Toxicology, School of Public Health, Shenyang Medical College, Liaoning, China
| | - Zhijing Tan
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States of America
| | - Jiaqi Shi
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - David M. Lubman
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI, United States of America
| |
Collapse
|
45
|
Rath N, Morton JP, Julian L, Helbig L, Kadir S, McGhee EJ, Anderson KI, Kalna G, Mullin M, Pinho AV, Rooman I, Samuel MS, Olson MF. ROCK signaling promotes collagen remodeling to facilitate invasive pancreatic ductal adenocarcinoma tumor cell growth. EMBO Mol Med 2017; 9:198-218. [PMID: 28031255 PMCID: PMC5286371 DOI: 10.15252/emmm.201606743] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 01/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer death; identifying PDAC enablers may reveal potential therapeutic targets. Expression of the actomyosin regulatory ROCK1 and ROCK2 kinases increased with tumor progression in human and mouse pancreatic tumors, while elevated ROCK1/ROCK2 expression in human patients, or conditional ROCK2 activation in a KrasG12D/p53R172H mouse PDAC model, was associated with reduced survival. Conditional ROCK1 or ROCK2 activation promoted invasive growth of mouse PDAC cells into three-dimensional collagen matrices by increasing matrix remodeling activities. RNA sequencing revealed a coordinated program of ROCK-induced genes that facilitate extracellular matrix remodeling, with greatest fold-changes for matrix metalloproteinases (MMPs) Mmp10 and Mmp13 MMP inhibition not only decreased collagen degradation and invasion, but also reduced proliferation in three-dimensional contexts. Treatment of KrasG12D/p53R172H PDAC mice with a ROCK inhibitor prolonged survival, which was associated with increased tumor-associated collagen. These findings reveal an ancillary role for increased ROCK signaling in pancreatic cancer progression to promote extracellular matrix remodeling that facilitates proliferation and invasive tumor growth.
Collapse
Affiliation(s)
- Nicola Rath
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Linda Julian
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Lena Helbig
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | | | | | | | - Margaret Mullin
- Electron Microscopy Facility, School of Life Sciences, University of Glasgow, Glasgow, UK
| | - Andreia V Pinho
- Cancer Research Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Ilse Rooman
- Oncology Research Centre, Free University Brussels (VUB), Brussels, Belgium
| | - Michael S Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, SA, Australia
| | - Michael F Olson
- Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
46
|
Choi CH, Chung JY, Chung EJ, Sears JD, Lee JW, Bae DS, Hewitt SM. Prognostic significance of annexin A2 and annexin A4 expression in patients with cervical cancer. BMC Cancer 2016; 16:448. [PMID: 27402115 PMCID: PMC4940752 DOI: 10.1186/s12885-016-2459-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/23/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The annexins (ANXs) have diverse roles in tumor development and progression, however, their clinical significance in cervical cancer has not been elucidated. The present study was to investigate the clinical significance of annexin A2 (ANXA2) and annexin A4 (ANXA4) expression in cervical cancer. METHODS ANXA2 and ANXA4 immunohistochemical staining were performed on a cervical cancer tissue microarray consisting of 46 normal cervical epithelium samples and 336 cervical cancer cases and compared the data with clinicopathological variables, including the survival of cervical cancer patients. RESULTS ANXA2 expression was lower in cancer tissue (p = 0.002), whereas ANXA4 staining increased significantly in cancer tissues (p < 0.001). ANXA2 expression was more prominent in squamous cell carcinoma (p < 0.001), whereas ANXA4 was more highly expressed in adeno/adenosquamous carcinoma (p < 0.001). ANXA2 overexpression was positively correlated with advanced cancer phenotypes, whereas ANXA4 expression was associated with resistance to radiation with or without chemotherapy (p = 0.029). Notably, high ANXA2 and ANXA4 expression was significantly associated with shorter disease-free survival (p = 0.004 and p = 0.033, respectively). Multivariate analysis indicated that ANXA2+ (HR = 2.72, p = 0.003) and ANXA2+/ANXA4+ (HR = 2.69, p = 0.039) are independent prognostic factors of disease-free survival in cervical cancer. Furthermore, a random survival forest model using combined ANXA2, ANXA4, and clinical variables resulted in improved predictive power (mean C-index, 0.76) compared to that of clinical-variable-only models (mean C-index, 0.70) (p = 0.006). CONCLUSIONS These findings indicate that detecting ANXA2 and ANXA4 expression may aid the evaluation of cervical carcinoma prognosis.
Collapse
Affiliation(s)
- Chel Hun Choi
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MSC 1500, Bethesda, MD, 20892, USA.,Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MSC 1500, Bethesda, MD, 20892, USA
| | - Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, 20892, USA
| | - John D Sears
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MSC 1500, Bethesda, MD, 20892, USA
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, 135-710, Republic of Korea
| | - Duk-Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-dong, Gangnam-gu, Seoul, 135-710, Republic of Korea.
| | - Stephen M Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, MSC 1500, Bethesda, MD, 20892, USA.
| |
Collapse
|
47
|
Kita K, Sugita K, Sato C, Sugaya S, Sato T, Kaneda A. Extracellular Release of Annexin A2 is Enhanced upon Oxidative Stress Response via the p38 MAPK Pathway after Low-Dose X-Ray Irradiation. Radiat Res 2016; 186:79-91. [DOI: 10.1667/rr14277.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Kazuko Kita
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba 260-8670, Japan and
| | - Katsuo Sugita
- Department of Clinical Medicine, Faculty of Education, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Chihomi Sato
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba 260-8670, Japan and
| | - Shigeru Sugaya
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba 260-8670, Japan and
| | - Tetsuo Sato
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba 260-8670, Japan and
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Inohana, Chuou-ku, Chiba 260-8670, Japan and
| |
Collapse
|
48
|
Corrado C, Saieva L, Raimondo S, Santoro A, De Leo G, Alessandro R. Chronic myelogenous leukaemia exosomes modulate bone marrow microenvironment through activation of epidermal growth factor receptor. J Cell Mol Med 2016; 20:1829-39. [PMID: 27196940 PMCID: PMC4876029 DOI: 10.1111/jcmm.12873] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/08/2016] [Indexed: 12/13/2022] Open
Abstract
Chronic myelogenous leukaemia (CML) is a clonal myeloproliferative disorder. Recent evidence indicates that altered crosstalk between CML and mesenchymal stromal cells may affect leukaemia survival; moreover, vesicles released by both tumour and non‐tumour cells into the microenvironment provide a suitable niche for cancer cell growth and survival. We previously demonstrated that leukaemic and stromal cells establish an exosome‐mediated bidirectional crosstalk leading to the production of IL8 in stromal cells, thus sustaining the survival of CML cells. Human cell lines used are LAMA84 (CML cells), HS5 (stromal cells) and bone marrow primary stromal cells; gene expression and protein analysis were performed by real‐time PCR and Western blot. IL8 and MMP9 secretions were evaluated by ELISA. Exosomes were isolated from CML cells and blood samples of CML patients. Here, we show that LAMA84 and CML patients’ exosomes contain amphiregulin (AREG), thus activating epidermal growth factor receptor (EGFR) signalling in stromal cells. EGFR signalling increases the expression of SNAIL and its targets, MMP9 and IL8. We also demonstrated that pre‐treatment of HS5 with LAMA84 exosomes increases the expression of annexin A2 that promotes the adhesion of leukaemic cells to the stromal monolayer, finally supporting the growth and invasiveness of leukaemic cells. Leukaemic and stromal cells establish a bidirectional crosstalk: exosomes promote proliferation and survival of leukaemic cells, both in vitro and in vivo, by inducing IL8 secretion from stromal cells. We propose that this mechanism is activated by a ligand–receptor interaction between AREG, found in CML exosomes, and EGFR in bone marrow stromal cells.
Collapse
Affiliation(s)
- Chiara Corrado
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università degli studi di Palermo, sezione di Biologia e Genetica, Palermo, Italy
| | - Laura Saieva
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università degli studi di Palermo, sezione di Biologia e Genetica, Palermo, Italy
| | - Stefania Raimondo
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università degli studi di Palermo, sezione di Biologia e Genetica, Palermo, Italy
| | - Alessandra Santoro
- Divisione di Ematologia, A.O. Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Giacomo De Leo
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università degli studi di Palermo, sezione di Biologia e Genetica, Palermo, Italy
| | - Riccardo Alessandro
- Dipartimento di Biopatologia e Biotecnologie Mediche, Università degli studi di Palermo, sezione di Biologia e Genetica, Palermo, Italy.
| |
Collapse
|
49
|
Laklai H, Miroshnikova YA, Pickup MW, Collisson EA, Kim GE, Barrett AS, Hill RC, Lakins JN, Schlaepfer DD, Mouw JK, LeBleu VS, Roy N, Novitskiy SV, Johansen JS, Poli V, Kalluri R, Iacobuzio-Donahue CA, Wood LD, Hebrok M, Hansen K, Moses HL, Weaver VM. Genotype tunes pancreatic ductal adenocarcinoma tissue tension to induce matricellular fibrosis and tumor progression. Nat Med 2016; 22:497-505. [PMID: 27089513 PMCID: PMC4860133 DOI: 10.1038/nm.4082] [Citation(s) in RCA: 459] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/11/2016] [Indexed: 12/13/2022]
Abstract
Fibrosis compromises pancreatic ductal carcinoma (PDAC) treatment and contributes to patient mortality, yet antistromal therapies are controversial. We found that human PDACs with impaired epithelial transforming growth factor-β (TGF-β) signaling have high epithelial STAT3 activity and develop stiff, matricellular-enriched fibrosis associated with high epithelial tension and shorter patient survival. In several KRAS-driven mouse models, both the loss of TGF-β signaling and elevated β1-integrin mechanosignaling engaged a positive feedback loop whereby STAT3 signaling promotes tumor progression by increasing matricellular fibrosis and tissue tension. In contrast, epithelial STAT3 ablation attenuated tumor progression by reducing the stromal stiffening and epithelial contractility induced by loss of TGF-β signaling. In PDAC patient biopsies, higher matricellular protein and activated STAT3 were associated with SMAD4 mutation and shorter survival. The findings implicate epithelial tension and matricellular fibrosis in the aggressiveness of SMAD4 mutant pancreatic tumors and highlight STAT3 and mechanics as key drivers of this phenotype.
Collapse
Affiliation(s)
- Hanane Laklai
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Yekaterina A. Miroshnikova
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michael W. Pickup
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Eric A. Collisson
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Grace E. Kim
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Alex S. Barrett
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, USA
| | - Ryan C. Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, USA
| | - Johnathon N. Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - David D. Schlaepfer
- Department of Reproductive Medicine, University of California, San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Janna K. Mouw
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Valerie S. LeBleu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston–Medical School, Houston, TX, USA
| | - Nilotpal Roy
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA USA
| | - Sergey V. Novitskiy
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Julia S. Johansen
- Department of Oncology, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Raghu Kalluri
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston–Medical School, Houston, TX, USA
| | - Christine A. Iacobuzio-Donahue
- Department of Pathology, David Rubenstein Center for Pancreatic Cancer Research, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Laura D. Wood
- Gastrointestinal and Liver Pathology Department, Johns Hopkins University, Baltimore, MD, USA
| | - Matthias Hebrok
- Diabetes Center, Department of Medicine, University of California, San Francisco, CA USA
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, USA
| | - Harold L. Moses
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Valerie M. Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
50
|
The Annexin a2 Promotes Development in Arthritis through Neovascularization by Amplification Hedgehog Pathway. PLoS One 2016; 11:e0150363. [PMID: 26963384 PMCID: PMC4786284 DOI: 10.1371/journal.pone.0150363] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/12/2016] [Indexed: 02/08/2023] Open
Abstract
The neovascularization network of pannus formation plays a crucial role in the development of rheumatoid arthritis (RA). Annexin a2 (Axna2) is an important mediating agent that induces angiogenesis in vascular diseases. The correlation between Axna2 and pannus formation has not been studied. Here, we provided evidence that compared to osteoarthritis (OA) patients and healthy people, the expression of Axna2 and Axna2 receptor (Axna2R) were up-regulated in patients with RA. Joint swelling, inflammation and neovascularization were increased significantly in mice with collagen-induced arthritis (CIA) that were exogenously added Axna2. Cell experiments showed that Axna2 promoted HUVEC proliferation by binding Axna2R, and could activate Hedgehog (HH) signaling and up-regulate the expression of Ihh and Gli. Besides, expression of Ihh, Patched (Ptc), Smoothened (Smo) and Gli and matrix metalloproteinase-2 (MMP-2), vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang-2), angiogenic growth factor of HH signaling downstream, were down-regulated after inhibition of expression Axna2R on HUVEC. Together, our research definitely observed that over-expression of Axna2 could promote the development of CIA, especially during the process of pannus formation for the first time. Meanwhile, Axna2 depended on combining Axna2R to activate and enlarge HH signaling and the expression of its downstream VEGF, Ang-2 and MMP-2 to promote HUVEC proliferation, and eventually caused to angiogenesis. Therefore, the role of Axna2 is instructive for understanding the development of RA, suppress the effect of Axna2 might provide a new potential measure for treatment of RA.
Collapse
|