1
|
Nicaise Y, Delmas C, Cohen‐Jonathan‐Moyal E, Seva C. PEA3 Transcription Factors, Role in Invasion, Proliferation and Radioresistance of Glioblastoma Stem Cells. J Cell Mol Med 2025; 29:e70533. [PMID: 40275610 PMCID: PMC12022000 DOI: 10.1111/jcmm.70533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
The presence of glioblastoma stem cells (GSCs), known for their high invasiveness and resistance to radiation, is one of the reasons for systematic recurrence. It is therefore important to understand the resistance mechanisms of these cells to optimise therapies. We focused on the PEA3 family of transcription factors, ETV1, ETV4 and ETV5, in patient-derived GSCs. We demonstrate that ETV1 is over-expressed in high invasive GSCs. In 3D invasion assays, inhibiting ETV1 expression using specific siRNAs significantly reduces cell invasion. Furthermore, we show a significant correlation between ETV1 and ZEB1, a major driver of invasion. Blocking ETV1 decreases ZEB1 expression in GSCs. The study also demonstrates the essential role of ETV1, ETV4 and ETV5 in the radioresistance of GSCs and their ability to form neurospheres. Using specific siRNAs to inhibit the expression of these transcription factors led to an increased sensitivity of GSCs to radiation and a decrease in both the number and size of neurospheres. These findings suggest that PEA3 transcription factors play a major role in GSCs aggressiveness by regulating invasion, radioresistance and the ability to form neurospheres. Trial Registration: Registry and the Registration N° of the study/trial: 12TETE01, ID-RCB No. 2012-A00585-38, approval Date: May 7, 2012.
Collapse
Affiliation(s)
- Yvan Nicaise
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
| | - Caroline Delmas
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
- IUCT‐Oncopole, Institut Claudius RegaudToulouseFrance
| | - Elizabeth Cohen‐Jonathan‐Moyal
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
- IUCT‐Oncopole, Institut Claudius RegaudToulouseFrance
| | - Catherine Seva
- Centre de Recherche en Cancérologie de Toulouse (CRCT)INSERM U1037, Université Toulouse III‐Paul SabatierToulouseFrance
| |
Collapse
|
2
|
Kandemir B, Kurnaz IA. The Role of Pea3 Transcription Factor Subfamily in the Nervous System. Mol Neurobiol 2025; 62:3293-3304. [PMID: 39269548 DOI: 10.1007/s12035-024-04432-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/08/2024] [Indexed: 09/15/2024]
Abstract
ETS domain transcription factor superfamily is highly conserved throughout metazoa and is involved in many aspects of development and tissue morphogenesis, and as such, the deregulation of ETS proteins is quite common in many diseases, including cancer. The PEA3 subfamily in particular has been extensively studied with respect to tumorigenesis and metastasis; however, they are also involved in the development of many tissues with branching morphogenesis, such as lung or kidney development. In this review, we aim to summarize findings from various studies on the role of Pea3 subfamily members in nervous system development in the embryo, as well as their functions in the adult neurons. We further discuss the different signals that were shown to regulate the function of the Pea3 family and indicate how this signal-dependent regulation of Pea3 proteins can generate neuronal circuit specificity through unique gene regulation. Finally, we discuss how these developmental roles of Pea3 proteins relate to their role in tumorigenesis.
Collapse
Affiliation(s)
- Basak Kandemir
- Department of Molecular Biology and Genetics, Baskent University, 06790, Etimesgut, Ankara, Turkey
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Isil Aksan Kurnaz
- Department of Molecular Biology and Genetics, Molecular Neurobiology Laboratory (AxanLab), Gebze Technical University, 41400, Gebze, Kocaeli, Turkey.
| |
Collapse
|
3
|
Mondal J, Zhang J, Qing F, Li S, Kumar D, Huse JT, Giancotti FG. Brd7 loss reawakens dormant metastasis initiating cells in lung by forging an immunosuppressive niche. Nat Commun 2025; 16:1378. [PMID: 39910049 PMCID: PMC11799300 DOI: 10.1038/s41467-025-56347-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025] Open
Abstract
Metastasis in cancer is influenced by epigenetic factors. Using an in vivo screen, we demonstrate that several subunits of the polybromo-associated BAF (PBAF) chromatin remodeling complex, particularly Brd7, are required for maintaining breast cancer metastatic dormancy in the lungs of female mice. Brd7 loss induces metastatic reawakening, along with modifications in epigenomic landscapes and upregulated oncogenic signaling. Breast cancer cells harboring Brd7 inactivation also reprogram the surrounding immune microenvironment by downregulating MHC-1 expression and promoting a pro-metastatic cytokine profile. Flow cytometric and single-cell analyses reveal increased levels of pro-tumorigenic inflammatory and transitional neutrophils, CD8+ exhausted T cells, and CD4+ stress response T cells in lungs from female mice harboring Brd7-deficient metastases. Finally, attenuating this immunosuppressive milieu by neutrophil depletion, neutrophil extracellular trap (NET) inhibition, or immune checkpoint therapy abrogates metastatic outgrowth. These findings implicate Brd7 and PBAF in triggering metastatic outgrowth in cancer, pointing to targetable underlying mechanisms involving specific immune cell compartments.
Collapse
Affiliation(s)
- Jayanta Mondal
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Junfeng Zhang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong Province, China.
| | - Feng Qing
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong Province, China
| | - Shunping Li
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong Province, China
| | - Dhiraj Kumar
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Johnson and Johnson Enterprise Innovations, Inc, Interventional Oncology, Spring House, PA, USA
| | - Jason T Huse
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Filippo G Giancotti
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
4
|
Ren D, Zhao F, Li J, Guo X, Ma X, Zheng Y, Shen G, Zhao J. lncRNA TCONS_00251376 promotes the proliferation and migration of gastric cancer cell through upregulating ETV1. CANCER INNOVATION 2025; 4:e156. [PMID: 39668941 PMCID: PMC11636580 DOI: 10.1002/cai2.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 12/14/2024]
Abstract
Background Although there have been significant advancements in the treatment modalities for gastric cancer (GC) in recent years, the overall prognosis remains poor, particularly for individuals in advanced stages. The absence of a sensitive tumor marker in GC is a crucial factor contributing to this challenge. Methods Our study focused on investigating a newly discovered long noncoding RNA (lncRNA) known as TCONS_00251376, which has been confirmed to exhibit differential expression in GC compared to adjacent tissues. To further validate these expression differences, we collected 22 pairs of GC and adjacent noncancerous tissues. Subsequent cell function experiments and animal studies were conducted to elucidate the role and underlying mechanisms of lncRNA TCONS_00251376 in the development of GC. Results The study revealed a significant upregulation of lncRNA TCONS_00251376 in cancer tissues (p < 0.01) and a consistent upregulation in GC cell lines (AGS, MKN45, BGC-823, and MGC-803). Furthermore, it was observed that lncRNA TCONS_00251376 played a promotive role in the proliferation, migration, and invasion of GC cells. Subsequent analysis indicated that lncRNA TCONS_00251376 could upregulate the expression of ETV1, a factor associated with the prognosis of GC. Conclusions Therefore, our findings suggest that lncRNA TCONS_00251376 functions as an oncogenic lncRNA, promoting tumorigenesis and progression by regulating the expression of ETV1 gene. This highlights its potential as an effective target for treating GC.
Collapse
Affiliation(s)
- Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| | - Jinming Li
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Graduate School, Qinghai UniversityXiningChina
| | - Xinjian Guo
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
- Department of PathologyAffiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
| | - Xinfu Ma
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
- Department of Gastrointestinal Oncology SurgeryAffiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
| | - Yonghui Zheng
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
| | - Guoshuang Shen
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University, Affiliated Cancer Hospital of Qinghai UniversityXiningChina
- Qinghai Provincial Clinical Research Center for Cancer; Qinghai Provincial Institute of Cancer ResearchXiningChina
| |
Collapse
|
5
|
Han T, Guo X, Xie J, Tong W, Zhang L. SUMO modified ETV1 promotes M2-polarized tumor-associated macrophage infiltration and cancer progression by facilitating CCL2 transcription in esophageal squamous cell carcinoma cells. Cancer Immunol Immunother 2025; 74:87. [PMID: 39891717 PMCID: PMC11787107 DOI: 10.1007/s00262-024-03914-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/02/2024] [Indexed: 02/03/2025]
Abstract
OBJECTIVE Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors with a high metastasis rate and a poor prognosis. ETS variant transcription factor 1 (ETV1) plays an important role in multiple malignancies. However, its function in ESCC progression and tumor microenvironment (TME) remains to be explored. In this study, we characterized the role of ETV1 in ESCC process and TME. METHODS Gene expression and immune infiltration in ESCC samples from the Cancer Genome Atlas (TCGA) were analyzed. The expression of ETV1 in clinical samples was detected by real-time PCR, western blot and immunohistochemistry staining. Cell growth was detected by CCK-8 and colony formation assays. Macrophage phenotypes were determined using flow cytometry. Immunofluorescence double staining was used to detect the tumor-associated macrophage (TAM) infiltration. The tumor volume was recorded and weighed. Transcriptional activity was measured using dual-luciferase assay, chromatin immunoprecipitation (ChIP) assay and DNA pull-down assay. RESULTS In this study, through analysis of ESCC samples from TCGA database and the clinic, we noticed that ETV1 was highly expressed in ESCC tumor tissues and was associated with TAM infiltration. Overexpression of ETV1 promoted ESCC cell proliferation in vitro and xenograft tumor growth in nude mice, while ETV1 knockdown elicited the opposite effects. Furthermore, ETV1 upregulation in tumor tissues was found to drive M2 macrophage infiltration both in vitro (transwell assays) and in vivo (xenograft tumor models). C-C motif chemokine ligand 2 (CCL2), a key factor inducing M2 macrophage polarization, was also found to be elevated in ESCC tumor tissues. Mechanism study demonstrated that ETV1 facilitated M2 macrophage infiltration via the transcriptional modulation of CCL2. In addition, the cause of the changes in ETV1 activity and expression was investigated. The E2 small ubiquitin-like modifier (SUMO) binding enzyme UBC9 increased ETV1 activity and expression, indicating the presence of SUMO modification in ETV1. CONCLUSIONS Our data deciphered the mechanism of ETV1-mediated M2 macrophage infiltration in the TME of ESCC, which has important implications for the development of novel prognostic and therapeutic targets to optimize current therapies against ESCC.
Collapse
Affiliation(s)
- Tianci Han
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, 44 Xiaoheyan Road, Shenyang, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, 44 Xiaoheyan Road, Shenyang, China
| | - Xiaoqi Guo
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Junwei Xie
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, 44 Xiaoheyan Road, Shenyang, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, 44 Xiaoheyan Road, Shenyang, China
| | - Wei Tong
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, 44 Xiaoheyan Road, Shenyang, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, 44 Xiaoheyan Road, Shenyang, China
| | - Liang Zhang
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, 44 Xiaoheyan Road, Shenyang, China.
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, 44 Xiaoheyan Road, Shenyang, China.
| |
Collapse
|
6
|
Qiu X, Zhang S, Zhang Y, Cai L, Li D, Lu Y. Reduction of ETV1 is Identified as a Prominent Feature of Age-Related Cataract. Curr Eye Res 2024; 49:496-504. [PMID: 38200696 DOI: 10.1080/02713683.2024.2302545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 01/01/2024] [Indexed: 01/12/2024]
Abstract
PURPOSE To identify the inactive genes in cataract lenses and explore their function in lens epithelial cells (LECs). METHODS Lens epithelium samples obtained from both age-related cataract (ARC) patients and normal donors were subjected to two forms of histone H3 immunoprecipitation: H3K9ac and H3K27me3 chromatin immunoprecipitation (ChIP), followed by ChIP-seq. The intersection set of "active genes in normal controls" and "repressed genes in cataract lenses" was identified. To validate the role of a specific gene, ETV1, within this set, quantitative polymerase chain reaction (qPCR), western blot, and immunofluorescence were performed using clinical lens epithelium samples. Small interference RNA (siRNA) was utilized to reduce the mRNA level of ETV1 in cultured LECs. Following this, transwell assay and western blot was performed to examine the migration ability of the cells. Furthermore, RNA-seq analysis was conducted on both cell samples with ETV1 knockdown and control cells. Additionally, the expression level of ETV1 in LECs was examined using qPCR under H2O2 treatment. RESULTS Six genes were identified in the intersection set of "active genes in normal controls" and "repressed genes in ARC lenses". Among these genes, ETV1 showed the most significant fold-change decrease in the cataract samples compared to the control samples. After ETV1 knockdown by siRNA in cultured LECs, reduced cell migration was observed, along with a decrease in the expression of β-Catenin and Vimentin, two specific genes associated with cell migration. In addition, under the oxidative stress induced by H2O2 treatment, the expression level of ETV1 in LECs significantly decreased. CONCLUSIONS Based on the findings of this study, it can be concluded that ETV1 is significantly reduced in human ARC lenses. The repression of ETV1 in ARC lenses appears to contribute to the disrupted differentiation of lens epithelium, which is likely caused by the inhibition of both cell differentiation and migration processes.
Collapse
Affiliation(s)
- Xiaodi Qiu
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shaohua Zhang
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Youmeng Zhang
- Department of Stomatology, Eye & ENT Hospital of Fudan University, Shanghai, China
| | - Lei Cai
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Dan Li
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yi Lu
- Eye Institute, Eye & ENT Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
7
|
Liu C, Zhou L, Chen Z. Construction of gastric cancer prognostic signature based on the E26 transcription factor and the identification of novel oncogene ELK3. Am J Cancer Res 2024; 14:1831-1849. [PMID: 38726274 PMCID: PMC11076248 DOI: 10.62347/rvbp7871] [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: 08/14/2023] [Accepted: 02/11/2024] [Indexed: 05/12/2024] Open
Abstract
The aim of the present study was to investigate the function of 29 E26 (ETS) transcription factor families in gastric cancer (GC) and determine their association with prognosis. Our analysis of the expression of the ETS family revealed that 28 genes were dysregulated in GC, and that their expression was associated with multiple clinicopathological features (P<0.05). Based on the expression signature of the ETS family, consensus clustering was performed to generate two gastric cancer subtypes. These subtypes exhibited differences in overall survival (OS, P = 0.161), disease-free survival (DFS, P<0.05) and GC grade (P<0.01). Functional enrichment analysis of the target genes associated with the ETS family indicated that these genes primarily contribute to functions that facilitate tumor progression. A systematic statistical analysis was used to construct a prognostic model related to OS and DFS in association with the ETS family. This model demonstrated that the maximum area under the curve (AUC) values for predicting OS and DFS were 0.729 and 0.670, respectively, establishing ETS as an independent prognostic factor for GC Furthermore, a nomogram was created from the prognostic signature, and its predictive accuracy was confirmed by a calibration curve. Finally, the expression and prognostic significance of the six genes comprising the model were also examined. Among these, ELK3 was found to be significantly overexpressed in GC clinical samples. Subsequent in vitro and in vivo studies verified that ELK3 regulates GC proliferation and metastasis, highlighting its potential as a therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Chenxi Liu
- School of Optometry, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| | - Liqiang Zhou
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
- Department of General Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| | - Zhiqing Chen
- Jiangxi Provincial Key Laboratory of Molecular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| |
Collapse
|
8
|
Liu C, Jiang K, Ding Y, Yang A, Cai R, Bai P, Xiong M, Fu C, Quan M, Xiong Z, Deng Y, Tian R, Wu C, Sun Y. Kindlin-2 enhances c-Myc translation through association with DDX3X to promote pancreatic ductal adenocarcinoma progression. Theranostics 2023; 13:4333-4355. [PMID: 37649609 PMCID: PMC10465218 DOI: 10.7150/thno.85421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 09/01/2023] Open
Abstract
Rationale: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive solid tumor, with extremely low survival rates. Identifying key signaling pathways driving PDAC progression is crucial for the development of therapies to improve patient response rates. Kindlin-2, a multi-functional protein, is involved in numerous biological processes including cell proliferation, apoptosis and migration. However, little is known about the functions of Kindlin-2 in pancreatic cancer progression in vivo. Methods: In this study, we employ an in vivo PDAC mouse model to directly investigate the role of Kindlin-2 in PDAC progression. Then, we utilized RNA-sequencing, the molecular and cellular assays to determine the molecular mechanisms by which Kindlin-2 promotes PDAC progression. Results: We show that loss of Kindlin-2 markedly inhibits KrasG12D-driven pancreatic cancer progression in vivo as well as in vitro. Furthermore, we provide new mechanistic insight into how Kindlin-2 functions in this process, A fraction of Kindlin-2 was localized to the endoplasmic reticulum and associated with the RNA helicase DDX3X, a key regulator of mRNA translation. Loss of Kindlin-2 blocked DDX3X from binding to the 5'-untranslated region of c-Myc and inhibited DDX3X-mediated c-Myc translation, leading to reduced c-Myc-mediated glucose metabolism and tumor growth. Importantly, restoration of the expression of either the full-length Kindlin-2 or c-Myc, but not that of a DDX3X-binding-defective mutant of Kindlin-2, in Kindlin-2 deficient PDAC cells, reversed the inhibition of glycolysis and pancreatic cancer progression induced by the loss of Kindlin-2. Conclusion: Our studies reveal a novel Kindlin-2-DDX3X-c-Myc signaling axis in PDAC progression and suggest that inhibition of this signaling axis may provide a promising therapeutic approach to alleviate PDAC progression.
Collapse
Affiliation(s)
- Chengmin Liu
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ke Jiang
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yanyan Ding
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Aihua Yang
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Renwei Cai
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Panzhu Bai
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minggang Xiong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Changying Fu
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Meiling Quan
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zailin Xiong
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yi Deng
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ruijun Tian
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chuanyue Wu
- Department of Pathology, School of Medicine and University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ying Sun
- Department of System Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China
- Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
9
|
Weng H, Feng W, Li F, Huang D, Lin L, Wang Z. Transcription factor ETV1-induced lncRNA MAFG-AS1 promotes migration, invasion, and epithelial-mesenchymal transition of pancreatic cancer cells by recruiting IGF2BP2 to stabilize ETV1 expression. Growth Factors 2023:1-13. [PMID: 37428861 DOI: 10.1080/08977194.2023.2227272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/28/2023] [Indexed: 07/12/2023]
Abstract
We investigated the mechanism of ETS-translocation variant 1 (ETV1)/lncRNA-MAFG-AS1 in pancreatic cancer (PC). MAFG-AS1 and ETV1 levels in PC cell lines and HPNE cells were determined using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blotting (WB). After transfection with sh-MAFG-AS1, PC cell invasion, migration, proliferation, and epithelial-mesenchymal transition (EMT)-related proteins were measured by 5-ethynyl-2'-deoxyuridine (EdU), Transwell assay, and WB. The binding between ETV1 and MAFG-AS1 was studied using dual-luciferase assay and chromatin immunoprecipitation. The interactions between MAFG-AS1, IGF2BP2, and ETV1 were tested. Combined experiments were further performed using sh-MAFG-AS1 and pcDNA-ETV1 simultaneously. ETV1/MAFG-AS1 was highly expressed in PC cells. Blocking MAFG-AS1 inhibited the malignant behaviors of PC cells. ETV1 induced MAFG-AS1 transcription in PC cells. MAFG-AS1 stabilized ETV1 mRNA by recruiting IGF2BP2. ETV1 overexpression partially antagonized the suppression of silencing MAFG-AS1 on PC cells. ETV1-induced MAFG-AS1 stabilized the ETV1 expression by recruiting IGF2BP2 and promoted PC cell migration, invasion, proliferation, and EMT.
Collapse
Affiliation(s)
- Hanqin Weng
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Weijian Feng
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Fengling Li
- Department of Anesthesiology, Dongguan People's Hospital, Dongguan, China
| | - Dong Huang
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Liangyi Lin
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| | - Zaiguo Wang
- Department of Hepatobiliary Surgery, Dongguan People's Hospital, Dongguan, China
| |
Collapse
|
10
|
Ohmoto M, Jyotaki M, Yee KK, Matsumoto I. A Transcription Factor Etv1/Er81 Is Involved in the Differentiation of Sweet, Umami, and Sodium Taste Cells. eNeuro 2023; 10:ENEURO.0236-22.2023. [PMID: 37045597 PMCID: PMC10131560 DOI: 10.1523/eneuro.0236-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
Taste cells are maintained by continuous turnover throughout a lifetime, yet the mechanisms of taste cell differentiation, and how taste sensations remain constant despite this continuous turnover, remain poorly understood. Here, we report that a transcription factor Etv1 (also known as Er81) is involved in the differentiation of taste cells responsible for the preference for sweet, umami, and salty tastes. Molecular analyses revealed that Etv1 is expressed by a subset of taste cells that depend on Skn-1a (also known as Pou2f3) for their generation and express T1R genes (responsible for sweet and umami tastes) or Scnn1a (responsible for amiloride-sensitive salty taste). Etv1CreERT2/CreERT2 mice express Etv1 isoform(s) but not Etv1 in putative proprioceptive neurons as comparable to wild-type mice, yet lack expression of Etv1 or an isoform in taste cells. These Etv1CreERT2/CreERT2 mice have the same population of Skn-1a-dependent cells in taste buds as wild-type mice but have altered gene expression in taste cells, with regional differences. They have markedly decreased electrophysiological responses of chorda tympani nerves to sweet and umami tastes and to amiloride-sensitive salty taste evoked by sodium cation, but they have unchanged responses to bitter or sour tastes. Our data thus show that Etv1 is involved in the differentiation of the taste cells responsible for sweet, umami, and salty taste preferences.
Collapse
Affiliation(s)
- Makoto Ohmoto
- Monell Chemical Senses Center, Philadelphia, PA 19104
| | | | - Karen K Yee
- Monell Chemical Senses Center, Philadelphia, PA 19104
| | | |
Collapse
|
11
|
Guo X, Li Y, Wan B, Lv Y, Wang X, Liu G, Wang P. ETV1 inhibition depressed M2 polarization of tumor-associated macrophage and cell process in gastrointestinal stromal tumor via down-regulating PDE3A. J Clin Biochem Nutr 2023; 72:139-146. [PMID: 36936869 PMCID: PMC10017324 DOI: 10.3164/jcbn.22-47] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/13/2022] [Indexed: 01/15/2023] Open
Abstract
M2-type polarization of tumor associated-macrophage (TAM) is involved in the malignancy of gastrointestinal stromal tumor (GIST) progression. ETS variant 1 (ETV1) has been previously validated to regulate GIST pathogenesis. Our study intended to explore the role and mechanism of ETV1 in mediating the M2-polarization of TAM in GIST progression. First, we analyzed the correlation between ETV1 expression and M2-polarization in GIST tissues. IL-4 was used to treat THP-1-derived TAM cells and IL-4-stimulated TAM were co-cultured with GIST-T1 cells to mimic the GIST microenvironment. A loss-of-function assay was performed to explore the role of ETV1. Results showed that ETV1 elevation was positively correlated with M2-polarization. IL-4-induced TAM promoted ETV1 expression, silencing ETV1 inhibited proliferation, invasion and KIT activation in IL-4-treated GIST cells, while cell apoptosis was enhanced. Besides, co-culture of ETV1-silenced GIST cells significantly depressed M2-polarization in TAM, presented as decreased levels of CD206, Agr-1 and cytokines, as well as the proportion of CD206-positive TAM. PDE3A was positively correlated with ETV1 and M2-polarization. Overexpressing PDE3A reversed the inhibitory effects of ETV1 silencing. Generally, ETV1 inhibition depressed M2-polarization of TAM in GIST and its promotion on pathological aggravation via down-regulating PDE3A. This evidence may provide a new target for GIST regulation.
Collapse
Affiliation(s)
- Xueyan Guo
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
| | - Yulong Li
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
| | - Bingbing Wan
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
| | - Yifei Lv
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
| | - Xue Wang
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
| | - Guisheng Liu
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
| | - Ping Wang
- Department of Gastroenterology, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi 710068, China
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
12
|
Jahedi H, Ramachandran A, Windsor J, Knowlton N, Blenkiron C, Print CG. Clinically Relevant Biology of Hyaluronic Acid in the Desmoplastic Stroma of Pancreatic Ductal Adenocarcinoma. Pancreas 2022; 51:1092-1104. [PMID: 37078930 DOI: 10.1097/mpa.0000000000002154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) is notorious for its poor outcome. The presence of a dense desmoplastic stroma is a hallmark of this malignancy, and abundant hyaluronic acid (HA) within this stroma is a common feature of PDAC. At the end of 2019, an HA-targeting drug, after initial promise, failed phase 3 clinical trials in PDAC. This failure in the face of such strong evidence for biological importance forces us to turn back to the research and seek a better understanding of HA biology in PDAC. Therefore, in this review, we reexamine what is known about HA biology, the methods used to detect and quantify HA, and the ability of the biological models in which HA has been investigated to recapitulate an HA-rich desmoplastic tumor stroma. The role of HA in PDAC relies on its complex interplay with a range of HA-associated molecules, which have not been as extensively investigated as HA itself. Therefore, using large genomic data sets, we cataloged the abundance and activity in PDAC of molecules that modulate HA synthesis, degradation, protein interactions, and receptor binding. Based on their association with clinical characteristics and individual patient outcomes, we suggest a small number of HA-associated molecules that warrant further investigation as biomarkers and drug targets.
Collapse
Affiliation(s)
- Hossein Jahedi
- From the Departments of Molecular Medicine and Pathology
| | | | | | | | | | | |
Collapse
|
13
|
Zhang T, Wang Y, Xie M, Ji X, Luo X, Chen X, Zhang B, Liu D, Feng Y, Sun M, Huang W, Xia L. HGF-mediated elevation of ETV1 facilitates hepatocellular carcinoma metastasis through upregulating PTK2 and c-MET. J Exp Clin Cancer Res 2022; 41:275. [PMID: 36109787 PMCID: PMC9479266 DOI: 10.1186/s13046-022-02475-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Metastasis is a major determinant of death in patients with hepatocellular carcinoma (HCC). Dissecting key molecular mediators that promote this malignant feature may help yield novel therapeutic insights. Here, we investigated the role of E-twenty-six transformation-specific variant 1 (ETV1), a member of the E-twenty-six transformation-specific (ETS) family, in HCC metastasis. Methods The clinical significance of ETV1 and its target genes in two independent cohorts of HCC patients who underwent curative resection were assessed by Kaplan–Meier analysis and Multivariate Cox proportional hazards model. Luciferase reporter assay and chromatin immunoprecipitation assay were used to detect the transcriptional regulation of target gene promoters by ETV1. The effect of ETV1 on invasiveness and metastasis of HCC were detected by transwell assays and the orthotopically metastatic model. Results ETV1 expression was frequently elevated in human HCC specimens. Increased ETV1 expression was associated with the malignant biological characteristics and poor prognosis of HCC patients. ETV1 facilitated invasion and metastasis of HCC cells in vitro and in vivo. Mechanistically, ETV1 promoted HCC metastasis via upregulating metastasis-related genes, including protein tyrosine kinase 2 (PTK2) and MET. Down-regulated the expression of PTK2 or tyrosine protein kinase Met (c-MET) decreased ETV1-mediated HCC metastasis. Hepatocyte growth factor (HGF) upregulated ETV1 expression through activating c-MET-ERK1/2-ELK1 pathway. Notably, in two independent cohorts, patients with positive coexpression of ETV1/PTK2 or ETV1/c-MET had worse prognosis. Furthermore, the combination of PTK2 inhibitor defactinib and c-MET inhibitor capmatinib significantly suppressed HCC metastasis induced by ETV1. Conclusion This study uncovers functional and prognostic roles for ETV1 in HCC and exposes a positive feedback loop of HGF-ERK1/2-ETV1-c-MET. Targeting this pathway may provide a potential therapeutic intervention for ETV1-overexpressing HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02475-2.
Collapse
|
14
|
Aberrant transcription factors in the cancers of the pancreas. Semin Cancer Biol 2022; 86:28-45. [PMID: 36058426 DOI: 10.1016/j.semcancer.2022.08.011] [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: 06/13/2022] [Revised: 08/15/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022]
Abstract
Transcription factors (TFs) are essential for proper activation of gene set during the process of organogenesis, differentiation, lineage specificity. Reactivation or dysregulation of TFs regulatory networks could lead to deformation of organs, diseases including various malignancies. Currently, understanding the mechanism of oncogenesis became necessity for the development of targeted therapeutic strategy for different cancer types. It is evident that many TFs go awry in cancers of the pancreas such as pancreatic ductal adenocarcinoma (PDAC) and pancreatic neuroendocrine neoplasms (PanNENs). These mutated or dysregulated TFs abnormally controls various signaling pathways in PDAC and PanNENs including RTK, PI3K-PTEN-AKT-mTOR, JNK, TGF-β/SMAD, WNT/β-catenin, SHH, NOTCH and VEGF which in turn regulate different hallmarks of cancer. Aberrant regulation of such pathways have been linked to the initiation, progression, metastasis, and resistance in pancreatic cancer. As of today, a number of TFs has been identified as crucial regulators of pancreatic cancer and a handful of them shown to have potential as therapeutic targets in pre-clinical and clinical settings. In this review, we have summarized the current knowledge on the role and therapeutic usefulness of TFs in PDAC and PanNENs.
Collapse
|
15
|
Wang H, Lu L, Liang X, Chen Y. Identification of prognostic genes in the pancreatic adenocarcinoma immune microenvironment by integrated bioinformatics analysis. Cancer Immunol Immunother 2022; 71:1757-1769. [PMID: 34854950 DOI: 10.1007/s00262-021-03110-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/11/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Pancreatic adenocarcinoma (PAAD) is one of the most common causes of death among solid tumors, and its pathogenesis remains to be clarified. This study aims to elucidate the value of immune/stromal-related genes in the prognosis of PAAD through comprehensive bioinformatics analysis based on the immune microenvironment and validated in Chinese pancreatic cancer patients. METHODS Gene expression profiles of pancreatic cancer patients were obtained from TCGA database. Differentially expressed genes (DEGs) were identified based on the ESTIMATE algorithm. Gene co-expression networks were constructed using WGCNA. In the key module, survival analysis was used to reveal the prognostic value. Subsequently, we performed functional enrichment analysis to construct a protein-protein interaction (PPI) network. The relationship between tumor immune infiltration and hub genes was analyzed by TIMER and CIBERSORT. Finally, it was validated in the GEO database and in tissues of Chinese pancreatic cancer patients. RESULTS In the TCGA pancreatic cancer cohort, a low immune/stromal score was associated with a good prognosis. After bioinformatic analysis, 57 genes were identified to be significantly associated with pancreatic cancer prognosis. Among them, up-regulation of four genes (COL6A3, PLAU, MMP11 and MMP14) indicated poor prognosis and was associated with multiple immune cell infiltration. IHC results showed that PLAU protein levels from Chinese pancreatic cancer tissues were significantly higher than those from adjacent non-tumor tissues and were also associated with tumor TNM stage and lymph node metastasis. CONCLUSION In conclusion, this study demonstrates that PLAU may serve as a new diagnostic and therapeutic target, which is highly expressed in Chinese pancreatic cancer tissues and associated with lymph node metastasis.
Collapse
Affiliation(s)
- Haolan Wang
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Liqing Lu
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xujun Liang
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yongheng Chen
- NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
16
|
Ammer-Herrmenau C, Wolf L, Nasrin SS, Ramu I, Roggiolani R, Goetze RG, Buchholz SM, Sendler M, Ellenrieder V, Neesse A. Activity of acute pancreatitis is modified by secreted protein acidic and rich in cysteine ablation. United European Gastroenterol J 2022; 10:544-555. [PMID: 35699570 PMCID: PMC9278565 DOI: 10.1002/ueg2.12262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
Background Acute pancreatitis (AP) is a frequent cause for hospitalization. However, molecular determinants that modulate severity of experimental pancreatitis are only partially understood. Objective To investigate the role of secreted protein acidic and rich in cysteine (SPARC) during cerulein‐induced AP in mice. Methods AP was induced by repeated cerulein injections in SPARC knock‐out mice (SPARC−/−) and control littermates (SPARC+/+). Secreted protein acidic and rich in cysteine expression and severity of AP were determined by histopathological scoring, immunohistochemistry, and biochemical assays. For functional analysis, primary murine acinar cell cultures with subsequent amylase release assays were employed. Proteome profiler assay and ELISA were conducted from pancreatic tissue lysates, and co‐immunofluorescence was performed. Results Upon cerulein induction, SPARC expression was robustly induced in pancreatic stellate cells (PSCs) but not in acinar cells. Genetic SPARC ablation resulted in attenuated severity of AP with significantly reduced levels of pancreatic necrosis, apoptosis, immune cell infiltration, and reduced fibrosis upon chronic stimulation. However, the release of amylase upon cerulein stimulation in primary acinar cell culture from SPARC+/+ and SPARC−/− was indistinguishable. Notably, immune cell derived C‐C Motif Chemokine Ligand 2 (CCL2) was highly elevated in SPARC+/+ pancreatic tissue potentially linking PSC derived SPARC with CCL2 induction in AP. Conclusion SPARC mediates the severity of AP. The potential link between SPARC and the CCL2 axis could open new avenues for tailored therapeutic interventions in AP patients and warrants further investigations.
Collapse
Affiliation(s)
- Christoph Ammer-Herrmenau
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Laurin Wolf
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Syeda S Nasrin
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Iswarya Ramu
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Roberta Roggiolani
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Robert G Goetze
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Soeren M Buchholz
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Mathias Sendler
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, Gottingen, Germany
| |
Collapse
|
17
|
Comandatore A, Immordino B, Balsano R, Capula M, Garajovà I, Ciccolini J, Giovannetti E, Morelli L. Potential Role of Exosomes in the Chemoresistance to Gemcitabine and Nab-Paclitaxel in Pancreatic Cancer. Diagnostics (Basel) 2022; 12:286. [PMID: 35204377 PMCID: PMC8871170 DOI: 10.3390/diagnostics12020286] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
In recent years, a growing number of studies have evaluated the role of exosomes in pancreatic ductal adenocarcinoma cancer (PDAC) demonstrating their involvement in a multitude of pathways, including the induction of chemoresistance. The aim of this review is to present an overview of the current knowledge on the role of exosomes in the resistance to gemcitabine and nab-paclitaxel, which are two of the most commonly used drugs for the treatment of PDAC patients. Exosomes are vesicular cargos that transport multiple miRNAs, mRNAs and proteins from one cell to another cell and some of these factors can influence specific determinants of gemcitabine activity, such as the nucleoside transporter hENT1, or multidrug resistance proteins involved in the resistance to paclitaxel. Additional mechanisms underlying exosome-mediated resistance include the modulation of apoptotic pathways, cellular metabolism, or the modulation of oncogenic miRNA, such as miR-21 and miR-155. The current status of studies on circulating exosomal miRNA and their possible role as biomarkers are also discussed. Finally, we integrated the preclinical data with emerging clinical evidence, showing how the study of exosomes could help to predict the resistance of individual tumors, and guide the clinicians in the selection of innovative therapeutic strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy;
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
| | - Benoit Immordino
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385 Marseille, France;
| | - Rita Balsano
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| | - Mjriam Capula
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
| | - Ingrid Garajovà
- Medical Oncology Unit, University Hospital of Parma, 43100 Parma, Italy;
| | - Joseph Ciccolini
- SMARTc Unit, Centre de Recherche en Cancérologie de Marseille, Inserm U1068 Aix Marseille Université, 13385 Marseille, France;
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands;
- Fondazione Pisana per La Scienza, 56124 Pisa, Italy; (B.I.); (M.C.)
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy;
| |
Collapse
|
18
|
RALY regulate the proliferation and expression of immune/inflammatory response genes via alternative splicing of FOS. Genes Immun 2022; 23:246-254. [PMID: 35941292 PMCID: PMC9758052 DOI: 10.1038/s41435-022-00178-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 01/07/2023]
Abstract
RALY is a multifunctional RNA-binding protein involved in cancer metastasis, prognosis, and chemotherapy resistance in various cancers. However, the molecular mechanism of which is still unclear. We have established RALY overexpression cell lines and studied the effect of RALY on proliferation and apoptosis in HeLa cells. Then we used RNA-seq to analyze the transcriptomes data. Lastly, RT-qPCR experiments had performed to confirm the RNA-seq results. We found that the overexpression of RALY in HeLa cells inhibited proliferation. Moreover, the overexpression of RALY changed the gene expression profile, and the significant upregulation of genes involved immune/inflammatory response related biological process by NOD-like receptor signaling pathway cytokine-cytokine receptor interaction. The significant downregulation genes involved innate immune response by the Primary immunodeficiency pathway. Notably, IFIT1, IFIT2, IFTI3, IFI44, HERC4, and OASL expression had inhibited by the overexpression of RALY. Furthermore, RALY negatively regulates the expression of transcription factors FOS and FOSB. Notably, we found that 645 alternative splicing events had regulated by overexpression of RALY, which is highly enriched in transcription regulation, RNA splicing, and cell proliferation biological process by the metabolic pathway. We show that RALY regulates the expression of immune/inflammatory response-related genes via alternative splicing of FOS in HeLa cells. The novel role of RALY in regulating immune/inflammatory gene expression may explain its function in regulating chemotherapy resistance and provides novel insights into further exploring the molecular mechanism of RALY in regulating cancer immunity and chemo/immune therapies.
Collapse
|
19
|
van Roey R, Brabletz T, Stemmler MP, Armstark I. Deregulation of Transcription Factor Networks Driving Cell Plasticity and Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:753456. [PMID: 34888306 PMCID: PMC8650502 DOI: 10.3389/fcell.2021.753456] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a very aggressive disease with 5-year survival rates of less than 10%. The constantly increasing incidence and stagnant patient outcomes despite changes in treatment regimens emphasize the requirement of a better understanding of the disease mechanisms. Challenges in treating pancreatic cancer include diagnosis at already progressed disease states due to the lack of early detection methods, rapid acquisition of therapy resistance, and high metastatic competence. Pancreatic ductal adenocarcinoma, the most prevalent type of pancreatic cancer, frequently shows dominant-active mutations in KRAS and TP53 as well as inactivation of genes involved in differentiation and cell-cycle regulation (e.g. SMAD4 and CDKN2A). Besides somatic mutations, deregulated transcription factor activities strongly contribute to disease progression. Specifically, transcriptional regulatory networks essential for proper lineage specification and differentiation during pancreas development are reactivated or become deregulated in the context of cancer and exacerbate progression towards an aggressive phenotype. This review summarizes the recent literature on transcription factor networks and epigenetic gene regulation that play a crucial role during tumorigenesis.
Collapse
Affiliation(s)
- Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
20
|
Lodestijn SC, Miedema DM, Lenos KJ, Nijman LE, Belt SC, El Makrini K, Lecca MC, Waasdorp C, van den Bosch T, Bijlsma MF, Vermeulen L. Marker-free lineage tracing reveals an environment-instructed clonogenic hierarchy in pancreatic cancer. Cell Rep 2021; 37:109852. [PMID: 34686335 DOI: 10.1016/j.celrep.2021.109852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/16/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022] Open
Abstract
Effective treatments for pancreatic ductal adenocarcinoma (PDAC) are lacking, and targeted agents have demonstrated limited efficacy. It has been speculated that a rare population of cancer stem cells (CSCs) drives growth, therapy resistance, and rapid metastatic progression in PDAC. These CSCs demonstrate high clonogenicity in vitro and tumorigenic potential in vivo. However, their relevance in established PDAC tissue has not been determined. Here, we use marker-independent stochastic clonal labeling, combined with quantitative modeling of tumor expansion, to uncover PDAC tissue growth dynamics. We find that in contrast to the CSC model, all PDAC cells display clonogenic potential in situ. Furthermore, the proximity to activated cancer-associated fibroblasts determines tumor cell clonogenicity. This means that the microenvironment is dominant in defining the clonogenic activity of PDAC cells. Indeed, manipulating the stroma by Hedgehog pathway inhibition alters the tumor growth mode, revealing that tumor-stroma crosstalk shapes tumor growth dynamics and clonal architecture.
Collapse
Affiliation(s)
- Sophie C Lodestijn
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Daniël M Miedema
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Kristiaan J Lenos
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Lisanne E Nijman
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Saskia C Belt
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Khalid El Makrini
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Maria C Lecca
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Cynthia Waasdorp
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Tom van den Bosch
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands.
| | - Louis Vermeulen
- Amsterdam UMC, University of Amsterdam, LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Oncode Institute, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands.
| |
Collapse
|
21
|
Manoukian P, Bijlsma M, van Laarhoven H. The Cellular Origins of Cancer-Associated Fibroblasts and Their Opposing Contributions to Pancreatic Cancer Growth. Front Cell Dev Biol 2021; 9:743907. [PMID: 34646829 PMCID: PMC8502878 DOI: 10.3389/fcell.2021.743907] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic tumors are known to harbor an abundant and highly desmoplastic stroma. Among the various cell types that reside within tumor stroma, cancer-associated fibroblasts (CAFs) have gained a lot of attention in the cancer field due to their contributions to carcinogenesis and tumor architecture. These cells are not a homogeneous population, but have been shown to have different origins, phenotypes, and contributions. In pancreatic tumors, CAFs generally emerge through the activation and/or recruitment of various cell types, most notably resident fibroblasts, pancreatic stellate cells (PSCs), and tumor-infiltrating mesenchymal stem cells (MSCs). In recent years, single cell transcriptomic studies allowed the identification of distinct CAF populations in pancreatic tumors. Nonetheless, the exact sources and functions of those different CAF phenotypes remain to be fully understood. Considering the importance of stromal cells in pancreatic cancer, many novel approaches have aimed at targeting the stroma but current stroma-targeting therapies have yielded subpar results, which may be attributed to heterogeneity in the fibroblast population. Thus, fully understanding the roles of different subsets of CAFs within the stroma, and the cellular dynamics at play that contribute to heterogeneity in CAF subsets may be essential for the design of novel therapies and improving clinical outcomes. Fortunately, recent advances in technologies such as microfluidics and bio-printing have made it possible to establish more advanced ex vivo models that will likely prove useful. In this review, we will present the different roles of stromal cells in pancreatic cancer, focusing on CAF origin as a source of heterogeneity, and the role this may play in therapy failure. We will discuss preclinical models that could be of benefit to the field and that may contribute to further clinical development.
Collapse
Affiliation(s)
- Paul Manoukian
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maarten Bijlsma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hanneke van Laarhoven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
22
|
Zhang Z, Qin Y, Ji S, Xu W, Liu M, Hu Q, Ye Z, Fan G, Yu X, Liu W, Xu X. FGFBP1-mediated crosstalk between fibroblasts and pancreatic cancer cells via FGF22/FGFR2 promotes invasion and metastasis of pancreatic cancer. Acta Biochim Biophys Sin (Shanghai) 2021; 53:997-1008. [PMID: 34117747 DOI: 10.1093/abbs/gmab074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Indexed: 11/12/2022] Open
Abstract
Fibroblast growth factor-binding protein 1 (FGFBP1) promotes fibroblast growth factor (FGF) activity by releasing FGFs from extracellular matrix storage. We previously reported that the tumor suppressor F-box and WD repeat domain-containing 7 suppresses FGFBP1 by reducing expression of c-Myc, which inhibits the proliferation and migration of pancreatic cancer cells. However, the potential mechanism by which FGFBP1 facilitates pancreatic ductal adenocarcinoma (PDAC) remains unexplored. In this study, we focused on the function of FGFBP1 in the interplay between cancer-associated fibroblasts (CAFs) and pancreatic cancer cells (PCCs). Decreased FGF22 expression was detected in CAFs co-cultured with PCCs with FGFBP1 abrogation, which was verified in the cell culture medium by enzyme-linked immunosorbent assay. Active cytokine FGF22 significantly facilitated the migration and invasion of PANC-1 and Mia PaCa-2 cells. The number of penetrating PCCs cocultured with CAFs with FGF22 abrogation was significantly less than that of the control group. Interestingly, higher expressions of FGF22 and fibroblast growth factor receptor 2 (FGFR2) were associated with worse prognosis of patients with PDAC and FGFR2, an independent prognostic marker of PDAC. The PANC-1 and Mia PaCa-2 cells with silenced FGFR2 showed weaker invasion and metastasis, even if these cells were simultaneously treated with cytokine FGF22. These results revealed that FGFBP1-mediated interaction between CAFs and PCCs via FGF22/FGFR2 facilitates the migration and invasion of PCCs. FGFR2 could act as a prognostic marker for patients with PDAC.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200031, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Pancreatic Cancer Institute, Shanghai 200031, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200031, China
| |
Collapse
|
23
|
Yao J, Yang M, Atteh L, Liu P, Mao Y, Meng W, Li X. A pancreas tumor derived organoid study: from drug screen to precision medicine. Cancer Cell Int 2021; 21:398. [PMID: 34315500 PMCID: PMC8314636 DOI: 10.1186/s12935-021-02044-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) one of the deadliest malignant tumor. Despite considerable progress in pancreatic cancer treatment in the past 10 years, PDAC mortality has shown no appreciable change, and systemic therapies for PDAC generally lack efficacy. Thus, developing biomarkers for treatment guidance is urgently required. This review focuses on pancreatic tumor organoids (PTOs), which can mimic the characteristics of the original tumor in vitro. As a powerful tool with several applications, PTOs represent a new strategy for targeted therapy in pancreatic cancer and contribute to the advancement of the field of personalized medicine.
Collapse
Affiliation(s)
- Jia Yao
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Man Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Lawrence Atteh
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Pinyan Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yongcui Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Wenbo Meng
- Department of General Surgery, The First Hospital of Lanzhou University, The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu, China
| |
Collapse
|
24
|
Cruz-Acuña R, Vunjak-Novakovic G, Burdick JA, Rustgi AK. Emerging technologies provide insights on cancer extracellular matrix biology and therapeutics. iScience 2021; 24:102475. [PMID: 34027324 PMCID: PMC8131321 DOI: 10.1016/j.isci.2021.102475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent engineering technologies have transformed traditional perspectives of cancer to include the important role of the extracellular matrix (ECM) in recapitulating the malignant behaviors of cancer cells. Novel biomaterials and imaging technologies have advanced our understanding of the role of ECM density, structure, mechanics, and remodeling in tumor cell-ECM interactions in cancer biology and have provided new approaches in the development of cancer therapeutics. Here, we review emerging technologies in cancer ECM biology and recent advances in engineered systems for evaluating cancer therapeutics and provide new perspectives on how engineering tools present an opportunity for advancing the modeling and treatment of cancer. This review offers the cell biology and cancer cell biology communities insight into how engineering tools can improve our understanding of cancer ECM biology and therapeutic development.
Collapse
Affiliation(s)
- Ricardo Cruz-Acuña
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
25
|
Golan T, Atias D, Stossel C, Raitses-Gurevich M. Patient-derived xenograft models of BRCA-associated pancreatic cancers. Adv Drug Deliv Rev 2021; 171:257-265. [PMID: 33617901 DOI: 10.1016/j.addr.2021.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dismal disease. The majority of patients diagnosed at an advanced, metastatic stage, and poor overall survival rates. The most clinically meaningful subtype obtained from PDAC genomic classification is represented by unstable genomes, and co-segregated with inactivation of DNA damage repair genes, e.g., Breast cancer 1/2 (BRCA1/2). The FDA and EMA has recently approved olaparib, a Poly (ADP-ribose) polymerase (PARP) inhibitor, as a maintenance strategy for platinum-sensitive advanced PDAC patients with BRCA mutations. However, susceptibility to treatment varies, and resistance may develop. Resistance can be defined as innate or acquired resistance to platinum/PARP-inhibition. Patient-derived xenograft (PDX) models have been utilized in cancer research for many years. We generated a unique PDX model, obtained from BRCA-associated PDAC patients at distinct time points of the disease recapitulating the different clinical scenario. In this review we discuss the relevant PDX-derived models for investigating BRCA-associated PDAC and drug development.
Collapse
Affiliation(s)
- Talia Golan
- Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.
| | - Dikla Atias
- Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Chani Stossel
- Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
26
|
Fu Y, Liu Q, Bao Q, Wen J, Liu Z, Hu Y, He G, Peng C, Xu Y, Zhang W. Development and analysis of long non-coding RNA-associated competing endogenous RNA network for osteosarcoma metastasis. Hereditas 2021; 158:9. [PMID: 33593435 PMCID: PMC7887822 DOI: 10.1186/s41065-021-00174-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 02/08/2021] [Indexed: 11/25/2022] Open
Abstract
Background Osteosarcoma is the primary bone malignant neoplasm that often develops metastasis. Increasing evidences have shown that non-coding RNAs (ncRNAs) relate to the progression of osteosarcoma. However, the ncRNAs’ roles in osteosarcoma metastasis are still unknown. Methods Differentially expressed (DE) RNAs were identified from Gene Expression Omnibus (GEO) database. Protein-protein interaction (PPI) of DE messenger RNAs (DEmRNAs) was built through STRING database. The target mRNAs and long ncRNAs (lncRNAs) of microRNAs (miRNA) were predicted through miRDB, Targetscan and Genecode databases, which then cross-checked with previously obtained DERNAs to construct competing endogenous RNA (ceRNA) network. All networks were visualized via Cytoscape and the hub RNAs were screened out through Cytoscape plug-in Cytohubba. The gene functional and pathway analyses were performed through DAVID and MirPath databases. The survival analyses of hub RNAs were obtained through Kaplan-Meier (KM) survival curves. Results Five hundred sixty-four DEmRNAs, 16 DElncRNAs and 22 DEmiRNAs were screened out. GO functional and KEGG pathway analyses showed that DERNAs were significantly associated with tumor metastasis. The ceRNA network including 6 lncRNAs, 55 mRNAs and 20 miRNAs were constructed and the top 10 hub RNAs were obtained. Above all, PI3K/AKT signaling pathway was identified as the most important osteosarcoma metastasis-associated pathway and its hub ceRNA module was constructed. The survival analyses showed that the RNAs in hub ceRNA module closely related to osteosarcoma patients’ prognosis. Conclusions The current study provided a new perspective on osteosarcoma metastasis. More importantly, the RNAs in hub ceRNA module might act as the novel therapeutic targets and prognostic factors for osteosarcoma patients. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-021-00174-0.
Collapse
Affiliation(s)
- Yucheng Fu
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Qi Liu
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Qiyuan Bao
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Junxiang Wen
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Zhuochao Liu
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Yuehao Hu
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Guoyu He
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Cheng Peng
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Yiqi Xu
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China
| | - Weibin Zhang
- Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, PR China.
| |
Collapse
|
27
|
Zhang T, Liu D, Wang Y, Sun M, Xia L. The E-Twenty-Six Family in Hepatocellular Carcinoma: Moving into the Spotlight. Front Oncol 2021; 10:620352. [PMID: 33585247 PMCID: PMC7873604 DOI: 10.3389/fonc.2020.620352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/08/2020] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of morbidity and mortality worldwide. Although therapeutic strategies have recently advanced, tumor metastasis and drug resistance continue to pose challenges in the treatment of HCC. Therefore, new molecular targets are needed to develop novel therapeutic strategies for this cancer. E-twenty-six (ETS) transcription family has been implicated in human malignancies pathogenesis and progression, including leukemia, Ewing sarcoma, gastrointestinal stromal tumors. Recently, increasing studies have expanded its great potential as functional players in other cancers, including HCC. This review focuses primarily on the key functions and molecular mechanisms of ETS factors in HCC. Elucidating these molecular details may provide novel potential therapeutic strategies for cancers.
Collapse
Affiliation(s)
| | | | | | | | - Limin Xia
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Zhu Y, Zeng Q, Li F, Fang H, Zhou Z, Jiang T, Yin C, Wei Q, Wang Y, Ruan J, Huang J. Dysregulated H3K27 Acetylation Is Implicated in Fatty Liver Hemorrhagic Syndrome in Chickens. Front Genet 2021; 11:574167. [PMID: 33505421 PMCID: PMC7831272 DOI: 10.3389/fgene.2020.574167] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/23/2020] [Indexed: 12/29/2022] Open
Abstract
Epigenetic regulation of gene expression has been reported in the pathogenesis of metabolic disorders such as diabetes and liver steatosis in humans. However, the molecular mechanisms of fatty liver hemorrhagic syndrome (FLHS) in chickens have been rarely studied. H3K27ac chromatin immunoprecipitation coupled with high-throughput sequencing and high-throughput RNA sequencing was performed to compare genome-wide H3K27ac profiles and transcriptomes of liver tissue between healthy and FLHS chickens. In total, 1,321 differential H3K27ac regions and 443 differentially expressed genes were identified (| log2Fold change| ≥ 1 and P-value ≤ 0.05) between the two groups. Binding motifs for transcription factors involved in immune processes and metabolic homeostasis were enriched among those differential H3K27ac regions. Differential H3K27ac peaks were associated with multiple known FLHS risk genes, involved in lipid and energy metabolism (PCK1, APOA1, ANGPTL4, and FABP1) and the immune system (FGF7, PDGFRA, and KIT). Previous studies and our current results suggested that the high-energy, low-protein (HELP) diet might have an impact on histone modification and chromatin structure, leading to the dysregulation of candidate genes and the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which causes excessive accumulation of fat in the liver tissue and induces the development of FLHS. These findings highlight that epigenetic modifications contribute to the regulation of gene expression and play a central regulatory role in FLHS. The PPAR signaling pathway and other genes implicated in FLHS are of great importance for the development of novel and specific therapies for FLHS-susceptible commercial laying hens.
Collapse
Affiliation(s)
- Yaling Zhu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China.,Department of Pathophysiology, Anhui Medical University, Hefei, China.,Laboratory Animal Research Center, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Qingjie Zeng
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Fang Li
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - Haoshu Fang
- Department of Pathophysiology, Anhui Medical University, Hefei, China.,Laboratory Animal Research Center, College of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Zhimin Zhou
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Tao Jiang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Chao Yin
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Qing Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Yujie Wang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Jiming Ruan
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| | - Jianzhen Huang
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
29
|
Nakao Y, Nakagawa S, Yamashita YI, Umezaki N, Okamoto Y, Ogata Y, Yasuda-Yoshihara N, Itoyama R, Yusa T, Yamashita K, Miyata T, Okabe H, Hayashi H, Imai K, Baba H. High ARHGEF2 (GEF-H1) Expression is Associated with Poor Prognosis Via Cell Cycle Regulation in Patients with Pancreatic Cancer. Ann Surg Oncol 2021; 28:4733-4743. [PMID: 33393038 DOI: 10.1245/s10434-020-09383-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pancreatic cancer has an extremely poor prognosis, even after curative resection. Treatment options for pancreatic cancer remain limited, therefore new therapeutic targets are urgently needed. We searched for genes predictive of poor prognosis in pancreatic cancer using a public database and validated the survival impact of the selected gene in a patient cohort. METHODS We used a public database to search for genes associated with early pancreatic cancer recurrence. As a validation cohort, 201 patients who underwent radical resection in our institution were enrolled. Expression of the target gene was evaluated using immunohistochemistry (IHC). We evaluated growth and invasiveness using small interfering RNAs, then performed pathway analysis using gene set enrichment analysis. RESULTS We extracted ARHGEF2 from GSE21501 as a gene with a high hazard ratio (HR) for early recurrence within 1 year. The high ARHGEF2 expression group had significantly poorer recurrence-free survival (RFS) and poorer overall survival (OS) than the low ARHGEF2 expression group. Multivariate analysis demonstrated that high ARHGEF2 expression was an independent poor prognostic factor for RFS (HR 1.92) and OS (HR 1.63). In vitro, ARHGEF2 suppression resulted in reduced cell growth and invasiveness. Bioinformatic analysis revealed that ARHGEF2 expression was associated with MYC, G2M, E2F, and CDC25A expression, suggesting that c-Myc and cell cycle genes are associated with high ARHGEF2 expression. IHC revealed a positive correlation between ARHGEF2 and c-Myc expression. CONCLUSIONS High ARHGEF2 expression is associated with cell cycle progression, and predicts early recurrence and poor survival in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yosuke Nakao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yo-Ichi Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoki Umezaki
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuya Okamoto
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoko Ogata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Noriko Yasuda-Yoshihara
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Rumi Itoyama
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Toshihiko Yusa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kohei Yamashita
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tatsunori Miyata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hirohisa Okabe
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
30
|
Feldmann K, Maurer C, Peschke K, Teller S, Schuck K, Steiger K, Engleitner T, Öllinger R, Nomura A, Wirges N, Papargyriou A, Jahan Sarker RS, Ranjan RA, Dantes Z, Weichert W, Rustgi AK, Schmid RM, Rad R, Schneider G, Saur D, Reichert M. Mesenchymal Plasticity Regulated by Prrx1 Drives Aggressive Pancreatic Cancer Biology. Gastroenterology 2021; 160:346-361.e24. [PMID: 33007300 DOI: 10.1053/j.gastro.2020.09.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 08/11/2020] [Accepted: 09/06/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is characterized by a fibroblast-rich desmoplastic stroma. Cancer-associated fibroblasts (CAFs) have been shown to display a high degree of interconvertible states including quiescent, inflammatory, and myofibroblastic phenotypes; however, the mechanisms by which this plasticity is achieved are poorly understood. Here, we aim to elucidate the role of CAF plasticity and its impact on PDAC biology. METHODS To investigate the role of mesenchymal plasticity in PDAC progression, we generated a PDAC mouse model in which CAF plasticity is modulated by genetic depletion of the transcription factor Prrx1. Primary pancreatic fibroblasts from this mouse model were further characterized by functional in vitro assays. To characterize the impact of CAFs on tumor differentiation and response to chemotherapy, various coculture experiments were performed. In vivo, tumors were characterized by morphology, extracellular matrix composition, and tumor dissemination and metastasis. RESULTS Our in vivo findings showed that Prrx1-deficient CAFs remain constitutively activated. Importantly, this CAF phenotype determines tumor differentiation and disrupts systemic tumor dissemination. Mechanistically, coculture experiments of tumor organoids and CAFs showed that CAFs shape the epithelial-to-mesenchymal phenotype and confer gemcitabine resistance of PDAC cells induced by CAF-derived hepatocyte growth factor. Furthermore, gene expression analysis showed that patients with pancreatic cancer with high stromal expression of Prrx1 display the squamous, most aggressive, subtype of PDAC. CONCLUSIONS Here, we define that the Prrx1 transcription factor is critical for tuning CAF activation, allowing a dynamic switch between a dormant and an activated state. This work shows that Prrx1-mediated CAF plasticity has significant impact on PDAC biology and therapeutic resistance.
Collapse
Affiliation(s)
- Karin Feldmann
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Carlo Maurer
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Katja Peschke
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Steffen Teller
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kathleen Schuck
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Thomas Engleitner
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Rupert Öllinger
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Alice Nomura
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Nils Wirges
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany
| | - Aristeidis Papargyriou
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center for Health and Environmental Research Munich, Neuherberg, Germany
| | - Rim Sabrina Jahan Sarker
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany
| | - Raphela Aranie Ranjan
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Zahra Dantes
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany; Comparative Experimental Pathology, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, Columbia University, New York, New York
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Roland Rad
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Günter Schneider
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Dieter Saur
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany
| | - Maximilian Reichert
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; German Cancer Consortium, Partner Site Munich, Germany.
| |
Collapse
|
31
|
Batzorig U, Wei PL, Wang W, Huang CY, Chang YJ. Glucose-Regulated Protein 94 Mediates the Proliferation and Metastasis through the Regulation of ETV1 and MAPK Pathway in Colorectal Cancer. Int J Med Sci 2021; 18:2251-2261. [PMID: 33967600 PMCID: PMC8100635 DOI: 10.7150/ijms.56024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/15/2021] [Indexed: 12/02/2022] Open
Abstract
Colorectal cancer (CRC) is a worldwide health problem. Glucose-regulated protein 94 (GRP94) is known as an important endoplasmic reticulum-stress response protein that shows correlation with aggressive cancer behavior. However, the role of GRP94 in CRC is still unclear. Our results showed that silencing GRP94 (GRP94-KD) reduced cell proliferation, invasion and migration of CRC cells and suppressed tumorigenesis in the xenograft mouse model. Rescue assay showed that ETV1 overexpression reversed the effect of GRP94 on cell proliferation and migration. In the molecular mechanism, we found that knockdown of GRP94 inhibited the level of MAPK pathway, including ERK/p-ERK, JNK/p-JNK, and p38/p-p38 signals. Cyclooxygenase-2 and epithelial-mesenchymal transformation biomarkers, such as N-cadherin, vimentin, and β-catenin were suppressed in GRP94 knockdown cells. Treatment of specific inhibitors of MAPK pathway showed that ERK/p-ERK, and p38/p-p38 inhibitors significantly influenced ETV1 expression as compared to JNK/p-JNK inhibitor. Our results indicated that silencing GRP94 repressed the ability of EMT process, cancer cell proliferation, metastasis, and CRC tumorigenesis. Therefore, GRP94 may play an important role in CRC by regulating ETV1 and MAPK pathway.
Collapse
Affiliation(s)
- Uyanga Batzorig
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Po-Li Wei
- Division of Colorectal Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Cancer Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110, Taiwan
| | - Weu Wang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chien-Yu Huang
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Division of Colorectal Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University.,Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Jia Chang
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
32
|
Capicua in Human Cancer. Trends Cancer 2020; 7:77-86. [PMID: 32978089 DOI: 10.1016/j.trecan.2020.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
Abstract
Capicua (CIC) is a highly conserved transcriptional repressor that is differentially regulated through mitogen-activated protein kinase (MAPK) signaling or genetic alteration across human cancer. CIC contributes to tumor progression and metastasis through direct transcriptional control of effector target genes. Recent findings indicate that CIC dysregulation is mechanistically linked and restricted to specific cancer subtypes, yet convergence on key downstream transcriptional nodes are critical for CIC-regulated oncogenesis across these cancers. In this review, we focus on how differential regulation of CIC through functional and genetic mechanisms contributes to subtype-specific cancer phenotypes and we propose new therapeutic strategies to effectively target CIC-altered cancers.
Collapse
|
33
|
Peng J, Cai Z, Zhao R, Chen J, Liu G, Dong C, Lim D, Feng Z. The intervention of valproic acid on the tumorigenesis induced by an environmental carcinogen of PAHs. Toxicol Res (Camb) 2020; 9:609-621. [PMID: 33178421 DOI: 10.1093/toxres/tfaa069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 11/14/2022] Open
Abstract
This study investigated whether valproic acid (VPA, a histone deacetylase inhibitor) can interfere with the carcinogenicity of polycyclic aromatic hydrocarbons (PAHs). A typical representative compound of PAHs, 7,12-Dimethylbenz[a]anthracene (DMBA), was used to induce rat breast cancer. The results showed that therapeutic concentration of VPA (50 and 100 mg/kg) delayed the occurrence of tumors, reduced tumor formation rate and attenuated tumors growth, and have a protective effect on normal tissues. The macrophage-mediated inflammatory response was found to be associated with the observed effect of VPA. In addition, we screened and validated a possible gene, Sema3c, which was involved in DMBA-induced breast cancer development and can be inhibited by VPA.
Collapse
Affiliation(s)
- Junxuan Peng
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| | - Zuchao Cai
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| | - Ruixue Zhao
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| | - Jiahao Chen
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| | - Guochao Liu
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| | - Chao Dong
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| | - David Lim
- School of Science and Health, Western Sydney University, Narellan Road, Campbelltown NSW 2560, Australia
| | - Zhihui Feng
- Department of Occupational Health and Occupational Medicine, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Shandong, Jinan, 250012, China
| |
Collapse
|
34
|
Bordignon P, Bottoni G, Xu X, Popescu AS, Truan Z, Guenova E, Kofler L, Jafari P, Ostano P, Röcken M, Neel V, Dotto GP. Dualism of FGF and TGF-β Signaling in Heterogeneous Cancer-Associated Fibroblast Activation with ETV1 as a Critical Determinant. Cell Rep 2020; 28:2358-2372.e6. [PMID: 31461652 PMCID: PMC6718812 DOI: 10.1016/j.celrep.2019.07.092] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/17/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022] Open
Abstract
Heterogeneity of cancer-associated fibroblasts (CAFs) can result from activation of distinct signaling pathways. We show that in primary human dermal fibroblasts (HDFs), fibroblast growth factor (FGF) and transforming growth factor β (TGF-β) signaling oppositely modulate multiple CAF effector genes. Genetic abrogation or pharmacological inhibition of either pathway results in induction of genes responsive to the other, with the ETV1 transcription factor mediating the FGF effects. Duality of FGF/TGF-β signaling and differential ETV1 expression occur in multiple CAF strains and fibroblasts of desmoplastic versus non-desmoplastic skin squamous cell carcinomas (SCCs). Functionally, HDFs with opposite TGF-β versus FGF modulation converge on promoting cancer cell proliferation. However, HDFs with increased TGF-β signaling enhance invasive properties and epithelial-mesenchymal transition (EMT) of SCC cells, whereas HDFs with increased FGF signaling promote macrophage infiltration. The findings point to a duality of FGF versus TGF-β signaling in distinct CAF populations that promote cancer development through modulation of different processes. FGF and TGF-β signaling exert opposite control over multiple CAF effector genes ETV1 transcription factor mediates FGF effects and suppresses those of TGF-β Modulation of either pathway leads to different tumor-promoting CAF populations TGF-β-activated CAFs promote EMT, but FGF-activated CAFs increase inflammation
Collapse
Affiliation(s)
- Pino Bordignon
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Giulia Bottoni
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Xiaoying Xu
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Alma S Popescu
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland
| | - Zinnia Truan
- Department of Otolaryngology-Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Emmanuella Guenova
- Department of Dermatology, University Hospital Zurich, University of Zurich, Zurich 8091, Switzerland
| | - Lukas Kofler
- Department of Dermatology, Eberhard Karls University, Tübingen 72076, Germany
| | - Paris Jafari
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; International Cancer Prevention Institute, Epalinges 1066, Switzerland
| | - Paola Ostano
- Cancer Genomics Laboratory, Edo and Elvo Tempia Valenta Foundation, Biella 13900, Italy
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen 72076, Germany
| | - Victor Neel
- Department of Dermatology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - G Paolo Dotto
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA; International Cancer Prevention Institute, Epalinges 1066, Switzerland.
| |
Collapse
|
35
|
Pancreatic Cancer Associated Fibroblasts (CAF): Under-Explored Target for Pancreatic Cancer Treatment. Cancers (Basel) 2020; 12:cancers12051347. [PMID: 32466266 PMCID: PMC7281461 DOI: 10.3390/cancers12051347] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is the 4th leading cause of cancer deaths in the United States. The pancreatic cancer phenotype is primarily a consequence of oncogenes disturbing the resident pancreas parenchymal cell repair program. Many solid tumor types including pancreatic cancer have severe tumor fibrosis called desmoplasia. Desmoplastic stroma is coopted by the tumor as a support structure and CAFs aid in tumor growth, invasion, and metastases. This stroma is caused by cancer associated fibroblasts (CAFs), which lay down extensive connective tissue in and around the tumor cells. CAFs represent a heterogeneous population of cells that produce various paracrine molecules such as transforming growth factor-beta (TGF-beta) and platelet derived growth factors (PDGFs) that aid tumor growth, local invasion, and development of metastases. The hard, fibrotic shell of desmoplasia serves as a barrier to the infiltration of both chemo- and immunotherapy drugs and host immune cells to the tumor. Although there have been recent improvements in chemotherapy and surgical techniques for management of pancreatic cancer, the majority of patients will die from this disease. Therefore, new treatment strategies are clearly needed. CAFs represent an under-explored potential therapeutic target. This paper discusses what we know about the role of CAFs in pancreatic cancer cell growth, invasion, and metastases. Additionally, we present different strategies that are being and could be explored as anti-CAF treatments for pancreatic cancer.
Collapse
|
36
|
Jinesh GG, Brohl AS. The genetic script of metastasis. Biol Rev Camb Philos Soc 2020; 95:244-266. [PMID: 31663259 DOI: 10.1111/brv.12562] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 01/24/2023]
Abstract
Metastasis is a pivotal event that changes the course of cancers from benign and treatable to malignant and difficult to treat, resulting in the demise of patients. Understanding the genetic control of metastasis is thus crucial to develop efficient and sustainable targeted therapies. Here we discuss the alterations in epigenetic mechanisms, transcription, chromosomal instability, chromosome imprinting, non-coding RNAs, coding RNAs, mutant RNAs, enhancers, G-quadruplexes, and copy number variation to dissect the genetic control of metastasis. We conclude that the genetic control of metastasis is predominantly executed through epithelial to mesenchymal transition and evasion of cell death. We discuss how genetic regulatory mechanisms can be harnessed for therapeutic purposes to achieve sustainable control over cancer metastasis.
Collapse
Affiliation(s)
- Goodwin G Jinesh
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, U.S.A.,Sarcoma Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, U.S.A
| | - Andrew S Brohl
- Sarcoma Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, U.S.A.,Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, U.S.A
| |
Collapse
|
37
|
Tian Y, Yu M, Sun L, Liu L, Wang J, Hui K, Nan Q, Nie X, Ren Y, Ren X. Distinct Patterns of mRNA and lncRNA Expression Differences Between Lung Squamous Cell Carcinoma and Adenocarcinoma. J Comput Biol 2019; 27:1067-1078. [PMID: 31750732 DOI: 10.1089/cmb.2019.0164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study aimed to assess mRNA and lncRNA expression differences between lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). Cancer tissues were obtained from three LUSC and three LUAD patients, followed by RNA-seq. Differentially expressed mRNAs (DE-mRNAs) and lncRNAs (DE-lncRNAs) were identified between LUSC and LUAD, after which functional enrichment analysis and protein-protein interaction (PPI) network construction was performed on DEGs. Coexpression analysis of lncRNA-gene and prediction of DEG-related miRNAs as well as function enrichment analysis, and construction of competing endogenous RNAs (ceRNA) regulatory network were then conducted. Moreover, survival analysis on differentially expressed RNAs was performed based on data downloaded from The Cancer Genome Atlas (TCGA) database. In this study, 518 DEGs and 117 DE-lncRNAs were identified between LUSC and LUAD. The DEGs were mainly associated with cell adhesion, PI3K-Akt signaling pathway, and focal adhesion. PPI network analysis indicated several genes with highest connectivity, such as CCND1. DE-lncRNAs that coexpressed with DEGs were also associated with tight junction and DE-lncRNAs that had more corepressed relationships with DEGs included GSEC, NKX2-1-AS1, LINC01415, and LINC00839. Moreover, the genes and lncRNAs with higher connectivity in the ceRNA network included NEAT1, SLC5A3, LINC00839, ETV1, CMTM4, and SNX30. Several genes were significantly related to the survival of patients with LUSC and LUAD, including ETV1, RTKN2, SNX30, PAK2, and CCND1. Genes and lncRNAs associated with cell junction have specific patterns in two major histological subtypes of NSCLC. GSEC, NKX2-1-AS1, NEAT1, CCND1, and ETV1 may be potential novel biomarkers for personalized treatment strategies of NSCLC.
Collapse
Affiliation(s)
- Yingxuan Tian
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China.,Department of Elderly Medicine, the Affiliated Shaanxi Provincial People's Hospital, Xi'an Medical College, Xi'an, China
| | - Min Yu
- Department of Oncology Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Li Sun
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China.,Department of Elderly Medicine, the Affiliated Shaanxi Provincial People's Hospital, Xi'an Medical College, Xi'an, China
| | - Linghua Liu
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jun Wang
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Ke Hui
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Qiaofeng Nan
- Department of Elderly Medicine, the Affiliated Shaanxi Provincial People's Hospital, Xi'an Medical College, Xi'an, China
| | - Xinyu Nie
- Graduate School of Medical College, Yan'an University, Yan'an, China
| | - Yajuan Ren
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiaoping Ren
- Department of Elderly Respiratory Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
38
|
Monkman JH, Thompson EW, Nagaraj SH. Targeting Epithelial Mesenchymal Plasticity in Pancreatic Cancer: A Compendium of Preclinical Discovery in a Heterogeneous Disease. Cancers (Basel) 2019; 11:E1745. [PMID: 31703358 PMCID: PMC6896204 DOI: 10.3390/cancers11111745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a particularly insidious and aggressive disease that causes significant mortality worldwide. The direct correlation between PDAC incidence, disease progression, and mortality highlights the critical need to understand the mechanisms by which PDAC cells rapidly progress to drive metastatic disease in order to identify actionable vulnerabilities. One such proposed vulnerability is epithelial mesenchymal plasticity (EMP), a process whereby neoplastic epithelial cells delaminate from their neighbours, either collectively or individually, allowing for their subsequent invasion into host tissue. This disruption of tissue homeostasis, particularly in PDAC, further promotes cellular transformation by inducing inflammatory interactions with the stromal compartment, which in turn contributes to intratumoural heterogeneity. This review describes the role of EMP in PDAC, and the preclinical target discovery that has been conducted to identify the molecular regulators and effectors of this EMP program. While inhibition of individual targets may provide therapeutic insights, a single 'master-key' remains elusive, making their collective interactions of greater importance in controlling the behaviours' of heterogeneous tumour cell populations. Much work has been undertaken to understand key transcriptional programs that drive EMP in certain contexts, however, a collaborative appreciation for the subtle, context-dependent programs governing EMP regulation is needed in order to design therapeutic strategies to curb PDAC mortality.
Collapse
Affiliation(s)
- James H. Monkman
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Erik W. Thompson
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Shivashankar H. Nagaraj
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4059, Australia
- Translational Research Institute, Brisbane, QLD 4102, Australia
| |
Collapse
|
39
|
Yuzhalin AE, Lim SY, Gordon-Weeks AN, Fischer R, Kessler BM, Yu D, Muschel RJ. Proteomics analysis of the matrisome from MC38 experimental mouse liver metastases. Am J Physiol Gastrointest Liver Physiol 2019; 317:G625-G639. [PMID: 31545917 PMCID: PMC6879896 DOI: 10.1152/ajpgi.00014.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 01/31/2023]
Abstract
Dissemination of primary tumors to distant anatomical sites has a substantial negative impact on patient prognosis. The liver is a common site for metastases from colorectal cancer, and patients with hepatic metastases have generally much shorter survival, raising a need to develop and implement novel strategies for targeting metastatic disease. The extracellular matrix (ECM) is a meshwork of highly crosslinked, insoluble high-molecular-mass proteins maintaining tissue integrity and establishing cell-cell interactions. Emerging evidence identifies the importance of the ECM in cancer cell migration, invasion, intravasation, and metastasis. Here, we isolated the ECM from MC38 mouse liver metastases using our optimized method of mild detergent solubilization followed by biochemical enrichment. The matrices were subjected to label-free quantitative mass spectrometry analysis, revealing proteins highly abundant in the metastatic matrisome. The resulting list of proteins upregulated in the ECM significantly predicted survival in patients with colorectal cancer but not other cancers with strong involvement of the ECM component. One of the proteins upregulated in liver metastatic ECM, annexin A1, was not previously studied in the context of cancer-associated matrisome. Here, we show that annexin A1 was markedly upregulated in colon cancer cell lines compared with cancer cells of other origin and also over-represented in human primary colorectal lesions, as well as hepatic metastases, compared with their adjacent healthy tissue counterparts. In conclusion, our study provides a comprehensive ECM characterization of MC38 experimental liver metastases and proposes annexin A1 as a putative target for this disease.NEW & NOTEWORTHY Here, the authors provide an extensive proteomics characterization of murine colorectal cancer liver metastasis matrisome (the ensemble of all extracellular matrix molecules). The findings presented in this study may enable identification of therapeutic targets or biomarkers of hepatic metastases.
Collapse
Affiliation(s)
- Arseniy E Yuzhalin
- Cancer Research United Kingdom/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - Su Yin Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia
| | - Alex N Gordon-Weeks
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Roman Fischer
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ruth J Muschel
- Cancer Research United Kingdom/Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Ramu I, Buchholz SM, Patzak MS, Goetze RG, Singh SK, Richards FM, Jodrell DI, Sipos B, Ströbel P, Ellenrieder V, Hessmann E, Neesse A. SPARC dependent collagen deposition and gemcitabine delivery in a genetically engineered mouse model of pancreas cancer. EBioMedicine 2019; 48:161-168. [PMID: 31597597 PMCID: PMC6838446 DOI: 10.1016/j.ebiom.2019.09.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/07/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is characterised by extensive matrix deposition that has been implicated in impaired drug delivery and therapeutic resistance. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular protein that regulates collagen deposition and is highly upregulated in the activated stroma subtype with poor prognosis in PDAC patients. METHODS KrasG12D;p48-Cre;SPARC-/- (KC-SPARC-/-) and KrasG12D;p48-Cre;SPARCWT (KC-SPARCWT) were generated and analysed at different stages of carcinogenesis by histological grading, immunohistochemistry for epithelial and stromal markers, survival and preclinical analysis. Pharmacokinetic and pharmacodynamic studies were conducted by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and immunohistochemistry following gemcitabine treatment (100 mg/kg) in vivo. FINDINGS Global genetic ablation of SPARC in a KrasG12D driven mouse model resulted in significantly reduced overall and mature collagen deposition around early and advanced pancreatic intraepithelial neoplasia (PanIN) lesions and in invasive PDAC (p < .001). However, detailed pathological scoring and molecular analysis showed no effects on PanIN to PDAC progression, vessel density (CD31), tumour incidence, grading or metastatic frequency. Despite comparable tumour kinetics, ablation of SPARC resulted in a significantly shortened survival in KC-SPARC-/- mice (280 days versus 485 days, p < .03, log-rank-test). Using LC-MS/MS, we show that SPARC dependent collagen deposition does not affect intratumoural gemcitabine accumulation or immediate therapeutic response in tumour bearing KC-SPARCWT and KC-SPARC-/-mice. INTERPRETATION Global SPARC ablation reduces the collagen-rich microenvironment in murine PDAC. Moreover, global SPARC depletion did not affect tumour growth kinetics, grading or metastatic frequency. Notably, the dense-collagen matrix did not restrict access of gemcitabine to the tumour. These findings may have direct translational implications in clinical trial design.
Collapse
Affiliation(s)
- Iswarya Ramu
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Sören M Buchholz
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Melanie S Patzak
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Robert G Goetze
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Shiv K Singh
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Frances M Richards
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, The University of Cambridge, United Kingdom
| | - Duncan I Jodrell
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, The University of Cambridge, United Kingdom
| | - Bence Sipos
- Institute of Pathology and Neuropathology, University Clinic Tübingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Centre Göttingen, Germany
| | - Volker Ellenrieder
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Elisabeth Hessmann
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany
| | - Albrecht Neesse
- Department of Gastroenterology and Gastrointestinal Oncology, University Medical Centre Göttingen, Germany.
| |
Collapse
|
41
|
Matjašič A, Zupan A, Boštjančič E, Pižem J, Popović M, Kolenc D. A novel PTPRZ1-ETV1 fusion in gliomas. Brain Pathol 2019; 30:226-234. [PMID: 31381204 DOI: 10.1111/bpa.12776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 07/26/2019] [Indexed: 12/17/2022] Open
Abstract
The aggressive nature of malignant gliomas and their genetic and clinical heterogeneity present a major challenge in their diagnosis and treatment. Development of targeted therapy brought attention on detecting novel gene fusions, since they represent promising therapeutic targets (eg, TRK inhibitors in NTRK fusion-positive tumors). Using targeted next-generation sequencing, we prospectively analyzed 205 primary brain tumors and detected a novel PTPRZ1-ETV1 fusion transcript in 11 of 191 (5.8%) gliomas, including nine glioblastomas, one anaplastic oligodendroglioma and one pilocytic astrocytoma. PTPRZ1-ETV1 fusion was confirmed by RT-PCR followed by Sanger sequencing, and in-silico analysis predicted a potential driver role. The newly detected fusion consists of the PTPRZ1 promoter in frame with the highly conserved DNA-binding domain of ETV1 transcription factor. The ETV1 and PTPRZ1 genes are known oncogenes, involved in processes of tumor development. ETV1 is a member of the ETS family of transcription factors, already known oncogenic drivers in Ewing sarcoma, prostate cancer and gastrointestinal stromal tumors, but not in gliomas. Its overexpression contributes to tumor growth and more aggressive tumor behavior. PTPRZ1 is already considered to be a tumor growth promoting oncogene in gliomas. In 8%-16% of gliomas, PTPRZ1 is fused to the MET oncogene, resulting in a PTPRZ1-MET fusion, which is associated with poorer prognosis but is also a positive predictive biomarker for treatment with kinase inhibitors. In view of the oncogenic role that the two fusion partners, PTPRZ1 and ETV1, exhibit in other malignancies, PTPRZ1-ETV1 fusion might present a novel potential therapeutic target in gliomas. Although histopathological examination of PTPRZ1-ETV1 fusion-positive gliomas did not reveal any specific or unique pathological features, and the follow-up period was too short to assess prognostic value of the fusion, careful monitoring of patients and their response to therapy might provide additional insights into the prognostic and predictive value of this novel fusion.
Collapse
Affiliation(s)
- Alenka Matjašič
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Andrej Zupan
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Emanuela Boštjančič
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jože Pižem
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mara Popović
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Danijela Kolenc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
42
|
Xiao Y, Zhang H, Ma Q, Huang R, Lu J, Liang X, Liu X, Zhang Z, Yu L, Pang J, Zhou L, Liu T, Wu H, Liang Z. YAP1-mediated pancreatic stellate cell activation inhibits pancreatic cancer cell proliferation. Cancer Lett 2019; 462:51-60. [PMID: 31352078 DOI: 10.1016/j.canlet.2019.07.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 07/09/2019] [Accepted: 07/20/2019] [Indexed: 12/11/2022]
Abstract
Pancreatic stellate cells (PSCs) are activated in pancreatic ductal adenocarcinoma (PDAC) and are responsible for dense desmoplastic stroma. Yes-associated protein 1 (YAP1) can induce cancer-associated fibroblast activation in liver and breast tumors, but its effect on PSCs is unknown. In the present study, we determined that YAP1 was highly expressed in the nuclei of PDAC-derived activated PSCs. RNAi-mediated or pharmacological inhibition of YAP1 led to PSC deactivation. In addition, YAP1 stimulated the expression of secreted protein acidic and cysteine rich (SPARC) in PSCs, which was inhibited by RUNX1. SPARC secreted from PSCs inhibited pancreatic cancer cell (PCC) proliferation. High expression of nuclear YAP1 in tumor stroma was significantly correlated with SPARC expression and fibrosis degree in human PDAC tissues. Our study revealed a critical role for YAP1 in the regulation of PSC activation and paracrine signaling. Our findings provide insights into a novel rationale for targeting YAP1 to reprogram the PDAC microenvironment.
Collapse
Affiliation(s)
- Ying Xiao
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Department of Pathology, Beijing Tsinghua Changgung Hospital, School of Clinic Medicine, Tsinghua University, Beijing, PR China
| | - Hui Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Qiang Ma
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Rui Huang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Junliang Lu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Xiaolong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Xuguang Liu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Zhiwen Zhang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Lianyuan Yu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Junyi Pang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Liangrui Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China
| | - Tonghua Liu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China.
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China.
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China; Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
43
|
Weng CC, Hsieh MJ, Wu CC, Lin YC, Shan YS, Hung WC, Chen LT, Cheng KH. Loss of the transcriptional repressor TGIF1 results in enhanced Kras-driven development of pancreatic cancer. Mol Cancer 2019; 18:96. [PMID: 31109321 PMCID: PMC6526617 DOI: 10.1186/s12943-019-1023-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/02/2019] [Indexed: 02/23/2023] Open
Abstract
Background The TG-interacting factor 1 (TGIF1) gene, which encodes a nuclear transcriptional corepressor of the TGFβ1/Smad signaling pathway, has been implicated in the pathogenesis of various types of human cancer; however, its role in pancreatic ductal adenocarcinoma (PDAC) has yet to be elucidated. Methods The expression of TGIF1 in human and murine PDAC specimens were detected by IHC analysis. The functions of TGIF1 in in vivo PDAC growth, dissemination, and metastasis were assessed using conditional inactivation of TGIF1 in well-established autochthonous mouse models of PDAC. Primary cells from TGIF1 null or wild type PDAC mice were examined by assays for cell proliferation, migration, invasion, soft agar and xenograft tumorigenesis. Gene expression profiling, pathway analyses, epigenetic changes associated with TGIF1 loss, and in vitro and in vivo effects of 4-MU were assessed. Results Conditional deletion of TGIF1 in the mouse pancreas had no discernible effect on pancreatic development or physiology. Notably, TGIF1 loss induced KrasG12D-driven PDAC models exhibited shorter latency and greater propensity for distant metastases. Deciphering the molecular mechanisms highlighted the TGIF1 loss-induced activation of the hyaluronan synthase 2 (HAS2)-CD44 signaling pathway and upregulation of the immune checkpoint regulator PD-L1 to facilitate the epithelial–mesenchymal transition (EMT) and tumor immune suppression. We also founded that TGIF1 might function as an epigenetic regulator and response for aberrant EMT gene expression during PDAC progression. Conclusions Our results imply that targeting the HAS2 pathway in TGIF1 loss of PDAC could be a promising therapeutic strategy for improving the clinical efficacy against PDAC metastasis. Electronic supplementary material The online version of this article (10.1186/s12943-019-1023-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ching-Chieh Weng
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Mei-Jen Hsieh
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.,Division of Neurology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, 802, Taiwan
| | - Chia-Chen Wu
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Yu-Chun Lin
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan
| | - Kuang-Hung Cheng
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan. .,National Institute of Cancer Research, National Health Research Institutes, Tainan, 704, Taiwan. .,Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
| |
Collapse
|
44
|
Das KK, Heeg S, Pitarresi JR, Reichert M, Bakir B, Takano S, Kopp JL, Wahl-Feuerstein A, Hicks P, Sander M, Rustgi AK. ETV5 regulates ductal morphogenesis with Sox9 and is critical for regeneration from pancreatitis. Dev Dyn 2018. [PMID: 29532564 DOI: 10.1002/dvdy.24626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The plasticity of pancreatic acinar cells to undergo acinar to ductal metaplasia (ADM) has been demonstrated to contribute to the regeneration of the pancreas in response to injury. Sox9 is critical for ductal cell fate and important in the formation of ADM, most likely in concert with a complex hierarchy of, as yet, not fully elucidated transcription factors. RESULTS By using a mouse model of acute pancreatitis and three dimensional organoid culture of primary pancreatic ductal cells, we herein characterize the Ets-transcription factor Etv5 as a pivotal regulator of ductal cell identity and ADM that acts upstream of Sox9 and is essential for Sox9 expression in ADM. Loss of Etv5 is associated with increased severity of acute pancreatitis and impaired ADM formation leading to delayed tissue regeneration and recovery in response to injury. CONCLUSIONS Our data provide new insights in the regulation of ADM with implications in our understanding of pancreatic homeostasis, pancreatitis and epithelial plasticity. Developmental Dynamics 247:854-866, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Koushik K Das
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.,Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Steffen Heeg
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine II, Medical Center, University of Freiburg, Freiburg, Germany
| | - Jason R Pitarresi
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maximilian Reichert
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.,II. Medizinische Klinik, Technical University of Munich, Munich, Germany
| | - Basil Bakir
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shigetsugu Takano
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Anja Wahl-Feuerstein
- Department of Medicine II, Medical Center, University of Freiburg, Freiburg, Germany
| | - Philip Hicks
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maike Sander
- Department of Pediatrics, Department of Cellular and Molecular Medicine, University of California San Diego School of Medicine, San Diego, California
| | - Anil K Rustgi
- Division of Gastroenterology, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Zeleniak AE, Huang W, Brinkman MK, Fishel ML, Hill R. Loss of MTSS1 results in increased metastatic potential in pancreatic cancer. Oncotarget 2017; 8:16473-16487. [PMID: 28146435 PMCID: PMC5369978 DOI: 10.18632/oncotarget.14869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 01/19/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of 7%. This dismal prognosis is largely due to the inability to diagnose the disease before metastasis occurs. Tumor cell dissemination occurs early in PDAC. While it is known that inflammation facilitates this process, the underlying mechanisms responsible for this progression have not been fully characterized. Here, we functionally test the role of metastasis suppressor 1 (MTSS1) in PDAC. Despite evidence showing that MTSS1 could be important for regulating metastasis in many different cancers, its function in PDAC has not been studied. Here, we show that loss of MTSS1 leads to increased invasion and migration in PDAC cell lines. Moreover, PDAC cells treated with cancer-associated fibroblast-conditioned media also have increased metastatic potential, which is augmented by loss of MTSS1. Finally, overexpression of MTSS1 in PDAC cell lines leads to a loss of migratory potential in vitro and an increase in overall survival in vivo. Collectively, our data provide insight into an important role for MTSS1 in suppressing tumor cell invasion and migration driven by the tumor microenvironment and suggest that therapeutic strategies aimed at increasing MTSS1 levels may effectively slow the development of metastatic lesions, increasing survival of patients with PDAC.
Collapse
Affiliation(s)
- Ann E Zeleniak
- Integrated Biomedical Sciences Program, University of Notre Dame, South Bend, Indiana, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA
| | - Wei Huang
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Mary K Brinkman
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Melissa L Fishel
- Indiana University School of Medicine, Department of Pharmacology and Toxicology, Indianapolis, Indiana, USA.,Indiana University School of Medicine, Department of Pediatrics, Wells Center for Pediatric Research, Indianapolis, Indiana, USA.,Pancreatic Cancer Signature Center, Indianapolis, Indiana, USA
| | - Reginald Hill
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana, USA.,Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| |
Collapse
|
46
|
Tajima H, Makino I, Ohbatake Y, Nakanuma S, Hayashi H, Nakagawara H, Miyashita T, Takamura H, Ohta T. Neoadjuvant chemotherapy for pancreatic cancer: Effects on cancer tissue and novel perspectives. Oncol Lett 2017; 13:3975-3981. [PMID: 28599404 DOI: 10.3892/ol.2017.6008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 02/17/2017] [Indexed: 01/05/2023] Open
Abstract
Chemotherapy for pancreatic cancer has diversified following the addition of more treatment regimens; however, in spite of this, pancreatic cancer remains a fatal disease. Preoperative (neoadjuvant) chemotherapy (NAC) or neoadjuvant chemoradiation therapy (NACRT) has been developed and implemented. For patients with borderline resectable pancreatic cancer (BRPC) and locally advanced pancreatic cancer (LAPC), a number of clinical trials have been conducted; NACRT was demonstrated to improve resectability, R0 resection rate, overall survival rate, disease-free survival rate and even an LAPC and BRPC survival advantage over NAC. However, from the knowledge obtained from resected specimens following preoperative treatment, residual pancreatic cancer tissues following NAC are rich in chemoresistant cancer stem-like cells and epithelial-mesenchymal transition (EMT) markers. Conversely, metformin, angiotensin receptor blocker, statins and low-dose paclitaxel are well-known as drugs that inhibit EMT, which is associated with cancer stem cell-like characteristics. Although clinical effectiveness is unlikely to be achieved using one of these as an anticancer agent, it is reasonable to use these drugs for patients with comorbidities in the treatment of pancreatic cancer. Furthermore, gemcitabine (GEM) affects antitumor immunity by stimulating the expression of major histocompatibility complex class I-related chain A on the surface of cancer cells to enhance the cytotoxicity of natural killer cells. Considering EMT and antitumor immunity, there is a possibility that GEM and nanoparticle albumin-bound paclitaxel therapy is the most suitable regimen for treating pancreatic cancer. However, even as preoperative treatment progresses, R0 resection is the most important factor for the long-term survival of pancreatic cancer patients.
Collapse
Affiliation(s)
- Hidehiro Tajima
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Isamu Makino
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Yoshinao Ohbatake
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Shinichi Nakanuma
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hironori Hayashi
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hisatoshi Nakagawara
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medicine Science, Kanazawa University, Kanazawa 920-8641, Japan
| |
Collapse
|
47
|
von Ahrens D, Bhagat TD, Nagrath D, Maitra A, Verma A. The role of stromal cancer-associated fibroblasts in pancreatic cancer. J Hematol Oncol 2017; 10:76. [PMID: 28351381 PMCID: PMC5371211 DOI: 10.1186/s13045-017-0448-5] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/23/2017] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer generally refractory to conventional treatments. Cancer-associated fibroblasts (CAFs) are cellular components of the desmoplastic stroma characteristic to the tumor that contributes to this treatment resistance. Various markers for CAFs have been explored including palladin and CD146 that have prognostic and functional roles in the pathobiology of PDAC. Mechanisms of CAF-tumor cell interaction have been described including exosomal transfer and paracrine signaling mediated by cytokines such as GM-CSF and IL-6. The role of downstream signaling pathways including JAK/STAT, mTOR, sonic hedge hog (SHH), and NFkB have also been shown to play an important function in PDAC-CAF cross talk. The role of autophagy and other metabolic effects on each cell type within the tumor have also been proposed to play roles in facilitating CAF secretory function and enhancing tumor growth in a low-glucose microenvironment. Targeting the stroma has gained interest with multiple preclinical and clinical trials targeting SHH, JAK2, and methods of either exploiting the secretory capability of CAFs to enhance drug delivery or inhibiting it to prevent its influence on cancer cell chemoresistance. This review summarizes the most recent progress made in understanding stromal formation; its contribution to tumor proliferation, invasion, and metastasis; its role in chemoresistance; and potential therapeutic strategies on the horizon.
Collapse
Affiliation(s)
- Dagny von Ahrens
- Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Tushar D Bhagat
- Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA
| | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Anirban Maitra
- The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Amit Verma
- Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, 10461, USA.
| |
Collapse
|
48
|
Träger MM, Dhayat SA. Epigenetics of epithelial-to-mesenchymal transition in pancreatic carcinoma. Int J Cancer 2017; 141:24-32. [DOI: 10.1002/ijc.30626] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/09/2017] [Accepted: 01/25/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Max M. Träger
- Department of General and Visceral Surgery; University Hospital of Muenster; Muenster Germany
| | - Sameer A. Dhayat
- Department of General and Visceral Surgery; University Hospital of Muenster; Muenster Germany
| |
Collapse
|