1
|
Ye C, Xia L, Gong R, Chang J, Sun Q, Xu J, Li F. Integrating plasma proteome with genome reveals novel protein biomarkers in colorectal cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03616-z. [PMID: 39017955 DOI: 10.1007/s12094-024-03616-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Biomarkers for colorectal cancer (CRC) can complement population screening methods, but so far, few plasma proteins have been identified as biomarkers for CRC. This study aims to identify potential protein biomarkers and therapeutic targets for CRC within the proteome range. METHODS We extracted summary-level data of circulating protein from 7 published genome-wide association studies (GWASs) of plasma proteome for Mendelian randomization (MR), summary-data-based MR (SMR), and co-localization analyses to screen and validate proteins with causal effects in CRC. In addition, we further conducted druggability evaluation, prognosis analysis at the transcriptional level, and enrichment expression at the single-cell level, highlighting the important role of these plasma protein biomarkers in CRC. RESULTS We identified 117 plasma protein biomarkers associated with CRC risk, with 9 proteins showing stronger genetic correlations in Bayesian co-localization (PP.H4 > 0.70). Further, we found 26 protein-coding genes already used in targeted drug development and may potentially become therapeutic targets for CRC. In prognosis analysis, the encoding genes of plasma proteins exhibited consistent effects with MR analysis and can serve as prognostic biomarkers for CRC. Additionally, we also found that the differentially expressed proteins are mainly expressed in fibroblasts, endothelial cells, macrophages, and T cells. CONCLUSION Our study has identified plasma protein biomarkers associated with CRC risk, which may complement population screening methods for CRC and achieve more precise treatment for patients.
Collapse
Affiliation(s)
- Changchun Ye
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Leizhou Xia
- Department of General Surgery, Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Ruimin Gong
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingbo Chang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qi Sun
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiaxi Xu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University Health Science Center, Xi'an, China.
| | - Fanni Li
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Isshiki T, Naiel S, Vierhout M, Otsubo K, Ali P, Tsubouchi K, Yazdanshenas P, Kumaran V, Dvorkin-Gheva A, Kolb MRJ, Ask K. Therapeutic strategies to target connective tissue growth factor in fibrotic lung diseases. Pharmacol Ther 2024; 253:108578. [PMID: 38103794 DOI: 10.1016/j.pharmthera.2023.108578] [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: 06/04/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The treatment of interstitial lung diseases, including idiopathic pulmonary fibrosis (IPF), remains challenging as current available antifibrotic agents are not effective in halting disease progression. Connective tissue growth factor (CTGF), also known as cellular communication factor 2 (CCN2), is a member of the CCN family of proteins that regulates cell signaling through cell surface receptors such as integrins, the activity of cytokines/growth factors, and the turnover of extracellular matrix (ECM) proteins. Accumulating evidence indicates that CTGF plays a crucial role in promoting lung fibrosis through multiple processes, including inducing transdifferentiation of fibroblasts to myofibroblasts, epithelial-mesenchymal transition (EMT), and cooperating with other fibrotic mediators such as TGF-β. Increased expression of CTGF has been observed in fibrotic lungs and inhibiting CTGF signaling has been shown to suppress lung fibrosis in several animal models. Thus, the CTGF signaling pathway is emerging as a potential therapeutic target in IPF and other pulmonary fibrotic conditions. This review provides a comprehensive overview of the current evidence on the pathogenic role of CTGF in pulmonary fibrosis and discusses the current therapeutic agents targeting CTGF using a systematic review approach.
Collapse
Affiliation(s)
- Takuma Isshiki
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada; Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omori Nisi, Ota-ku, Tokyo 143-8541, Japan
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Kohei Otsubo
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Pareesa Ali
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Kazuya Tsubouchi
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka 812-8582, Japan
| | - Parichehr Yazdanshenas
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Vaishnavi Kumaran
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Anna Dvorkin-Gheva
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada
| | - Martin R J Kolb
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, 5o Charlton Avenue East, Hamilton, ON, L8N 4A6, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 48L, Canada.
| |
Collapse
|
3
|
Fatemi M, Feng E, Sharma C, Azher Z, Goel T, Ramwala O, Palisoul SM, Barney RE, Perreard L, Kolling FW, Salas LA, Christensen BC, Tsongalis GJ, Vaickus LJ, Levy JJ. Inferring spatial transcriptomics markers from whole slide images to characterize metastasis-related spatial heterogeneity of colorectal tumors: A pilot study. J Pathol Inform 2023; 14:100308. [PMID: 37114077 PMCID: PMC10127126 DOI: 10.1016/j.jpi.2023.100308] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
Over 150 000 Americans are diagnosed with colorectal cancer (CRC) every year, and annually over 50 000 individuals will die from CRC, necessitating improvements in screening, prognostication, disease management, and therapeutic options. Tumor metastasis is the primary factor related to the risk of recurrence and mortality. Yet, screening for nodal and distant metastasis is costly, and invasive and incomplete resection may hamper adequate assessment. Signatures of the tumor-immune microenvironment (TIME) at the primary site can provide valuable insights into the aggressiveness of the tumor and the effectiveness of various treatment options. Spatially resolved transcriptomics technologies offer an unprecedented characterization of TIME through high multiplexing, yet their scope is constrained by cost. Meanwhile, it has long been suspected that histological, cytological, and macroarchitectural tissue characteristics correlate well with molecular information (e.g., gene expression). Thus, a method for predicting transcriptomics data through inference of RNA patterns from whole slide images (WSI) is a key step in studying metastasis at scale. In this work, we collected tissue from 4 stage-III (pT3) matched colorectal cancer patients for spatial transcriptomics profiling. The Visium spatial transcriptomics (ST) assay was used to measure transcript abundance for 17 943 genes at up to 5000 55-micron (i.e., 1-10 cells) spots per patient sampled in a honeycomb pattern, co-registered with hematoxylin and eosin (H&E) stained WSI. The Visium ST assay can measure expression at these spots through tissue permeabilization of mRNAs, which are captured through spatially (i.e., x-y positional coordinates) barcoded, gene specific oligo probes. WSI subimages were extracted around each co-registered Visium spot and were used to predict the expression at these spots using machine learning models. We prototyped and compared several convolutional, transformer, and graph convolutional neural networks to predict spatial RNA patterns at the Visium spots under the hypothesis that the transformer- and graph-based approaches better capture relevant spatial tissue architecture. We further analyzed the model's ability to recapitulate spatial autocorrelation statistics using SPARK and SpatialDE. Overall, the results indicate that the transformer- and graph-based approaches were unable to outperform the convolutional neural network architecture, though they exhibited optimal performance for relevant disease-associated genes. Initial findings suggest that different neural networks that operate on different scales are relevant for capturing distinct disease pathways (e.g., epithelial to mesenchymal transition). We add further evidence that deep learning models can accurately predict gene expression in whole slide images and comment on understudied factors which may increase its external applicability (e.g., tissue context). Our preliminary work will motivate further investigation of inference for molecular patterns from whole slide images as metastasis predictors and in other applications.
Collapse
Affiliation(s)
- Michael Fatemi
- Department of Computer Science, University of Virginia, Charlottesville, VA, USA
| | - Eric Feng
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, USA
| | - Cyril Sharma
- Department of Computer Science, Purdue University, West Lafayette, IN, USA
| | - Zarif Azher
- Thomas Jefferson High School for Science and Technology, Alexandria, VA, USA
| | - Tarushii Goel
- Department of Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ojas Ramwala
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Scott M. Palisoul
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, USA
| | - Rachael E. Barney
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, USA
| | | | | | - Lucas A. Salas
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Department of Molecular and Systems Biology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Integrative Neuroscience at Dartmouth (IND) graduate program, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
| | - Brock C. Christensen
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Department of Molecular and Systems Biology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Department of Community and Family Medicine, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
| | - Gregory J. Tsongalis
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, USA
| | - Louis J. Vaickus
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, USA
| | - Joshua J. Levy
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, USA
- Department of Epidemiology, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
- Department of Dermatology, Dartmouth Health, Lebanon, NH, USA
- Program in Quantitative Biomedical Sciences, Dartmouth College Geisel School of Medicine, Hanover, NH, USA
| |
Collapse
|
4
|
Ding JT, Zhou HN, Huang YF, Peng J, Huang HY, Yi H, Zong Z, Ning ZK. TGF-β Pathways Stratify Colorectal Cancer into Two Subtypes with Distinct Cartilage Oligomeric Matrix Protein (COMP) Expression-Related Characteristics. Biomolecules 2022; 12:1877. [PMID: 36551305 PMCID: PMC9775768 DOI: 10.3390/biom12121877] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Colorectal cancers (CRCs) continue to be the leading cause of cancer-related deaths worldwide. The exact landscape of the molecular features of TGF-β pathway-inducing CRCs remains uncharacterized. METHODS Unsupervised hierarchical clustering was performed to stratify samples into two clusters based on the differences in TGF-β pathways. Weighted gene co-expression network analysis was applied to identify the key gene modules mediating the different characteristics between two subtypes. An algorithm integrating the least absolute shrinkage and selection operator (LASSO), XGBoost, and random forest regression was performed to narrow down the candidate genes. Further bioinformatic analyses were performed focusing on COMP-related immune infiltration and functions. RESULTS The integrated machine learning algorithm identified COMP as the hub gene, which exhibited a significant predictive value for two subtypes with an area under the curve (AUC) value equaling 0.91. Further bioinformatic analysis revealed that COMP was significantly upregulated in various cancers, especially in advanced CRCs, and regulated the immune infiltration, especially M2 macrophages and cancer-associated fibroblasts in CRCs. CONCLUSIONS Comprehensive immune analysis and experimental validation demonstrate that COMP is a reliable signature for subtype prediction. Our results could provide a new point for TGFβ-targeted anticancer drugs and contribute to guiding clinical decision making for CRC patients.
Collapse
Affiliation(s)
- Jia-Tong Ding
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medicine School, Nanchang University, Nanchang 330006, China
| | - Hao-Nan Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Ying-Feng Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jie Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- The Second Clinical Medicine School, Nanchang University, Nanchang 330006, China
| | - Hao-Yu Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hao Yi
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Zhi-Kun Ning
- Department of Day Ward, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
5
|
Oncogenic Role of Connective Tissue Growth Factor Is Associated with Canonical TGF-β Cascade in Colorectal Cancer. Genes (Basel) 2022; 13:genes13040689. [PMID: 35456495 PMCID: PMC9031605 DOI: 10.3390/genes13040689] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/28/2022] [Accepted: 04/05/2022] [Indexed: 01/27/2023] Open
Abstract
TGF-β signaling pathways promote tumour development and control several downstream genes such as CTGF and MMPs. This study aimed to investigate the association between CTGF and MMP-1 mRNA expressions with clinicopathological status and survival rate in colorectal cancer patients. We investigated expression levels of CTGF and MMP-1 genes in paraffin-embedded tumours and adjacent normal tissue blocks (ADJ) by Real Time-PCR. Then, the expression of Smad2 and Smad4 proteins in the TGF-β canonical pathway was evaluated by immunohistochemistry. Finally, the correlation between CTGF, MMP-1, and the canonical TGF-β-signalling pathway with the clinicopathological features was investigated. Expression levels of MMP-1and CTGF were higher in tumours compared with adjacent normal tissues. Overexpression levels of MMP-1 and CTGF were associated with lymph node metastasis, distant metastasis, tumour histopathological grading, advanced stage, and poor survival (p < 0.05). Additionally, a significant association between the upregulation of MMP-1 and tumour location was noted. Upregulation of Smad2 and Smad4 proteins were also significantly correlated with lymph node metastasis, distant metastasis, advanced stage, and poor survival (p < 0.0001). This study showed that canonical TGF-β signalling regulates both CTGF and MMP-1 expression and CRC progression. Moreover, TGF-β signalling and its downstream genes could be used as novel biomarkers and novel approaches for targeted therapy in CRC.
Collapse
|
6
|
Yeger H, Perbal B. The CCN axis in cancer development and progression. J Cell Commun Signal 2021; 15:491-517. [PMID: 33877533 PMCID: PMC8642525 DOI: 10.1007/s12079-021-00618-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Since the authors first reviewed this subject in 2016 significant progress has been documented in the CCN field with advances made in the understanding of how members of the CCN family of proteins, CCN1-6, contribute to the pathogenesis and progression, positive and negative, of a larger variety of cancers. As termed matricellular proteins, and more recently the connective communication network, it has become clearer that members of the CCN family interact complexly with other proteins in the extracellular microenvironment, membrane signaling proteins, and can also operate intracellularly at the transcriptional level. In this review we expand on this earlier information providing new detailed information and insights that appropriate a much greater involvement and importance of their role in multiple aspects of cancer. Despite all the new information many more questions have been raised and intriguing results generated that warrant greater investigation. In order to permit the reader to smoothly integrate the new information we discuss all relevant CCN members in the context of cancer subtypes. We have harmonized the nomenclature with CCN numbering for easier comparisons. Finally, we summarize what new has been learned and provide a perspective on how our knowledge about CCN1-6 is being used to drive new initiatives on cancer therapeutics.
Collapse
Affiliation(s)
- Herman Yeger
- Program in Developmental and Stem Cell Biology Research Institute, SickKids, Toronto, Canada
| | | |
Collapse
|
7
|
Jia Q, Xu B, Zhang Y, Ali A, Liao X. CCN Family Proteins in Cancer: Insight Into Their Structures and Coordination Role in Tumor Microenvironment. Front Genet 2021; 12:649387. [PMID: 33833779 PMCID: PMC8021874 DOI: 10.3389/fgene.2021.649387] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
The crosstalk between tumor cells and the tumor microenvironment (TME), triggers a variety of critical signaling pathways and promotes the malignant progression of cancer. The success rate of cancer therapy through targeting single molecule of this crosstalk may be extremely low, whereas co-targeting multiple components could be complicated design and likely to have more side effects. The six members of cellular communication network (CCN) family proteins are scaffolding proteins that may govern the TME, and several studies have shown targeted therapy of CCN family proteins may be effective for the treatment of cancer. CCN protein family shares similar structures, and they mutually reinforce and neutralize each other to serve various roles that are tightly regulated in a spatiotemporal manner by the TME. Here, we review the current knowledge on the structures and roles of CCN proteins in different types of cancer. We also analyze CCN mRNA expression, and reasons for its diverse relationship to prognosis in different cancers. In this review, we conclude that the discrepant functions of CCN proteins in different types of cancer are attributed to diverse TME and CCN truncated isoforms, and speculate that targeting CCN proteins to rebalance the TME could be a potent anti-cancer strategy.
Collapse
Affiliation(s)
- Qingan Jia
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Binghui Xu
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Yaoyao Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Arshad Ali
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xia Liao
- Department of Nutrition, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Stromal microenvironment promoted infiltration in esophageal adenocarcinoma and squamous cell carcinoma: a multi-cohort gene-based analysis. Sci Rep 2020; 10:18589. [PMID: 33122682 PMCID: PMC7596515 DOI: 10.1038/s41598-020-75541-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 10/15/2020] [Indexed: 12/23/2022] Open
Abstract
The stromal microenvironment has been shown to affect the infiltration of esophageal carcinoma (ESCA), which is linked to prognosis. However, the complicated mechanism of how infiltration is influenced by the stromal microenvironment is not well-defined. In this study, a stromal activation classifier was established with ridge cox regression to calculate stroma scores for training (n = 182) and validation cohorts (n = 227) based on the stroma-related 32 hub genes identified by sequential bioinformatics algorithms. Patients with high stromal activation were associated with high T stage and poor prognosis in both esophagus adenocarcinoma and esophagus squamous cell carcinoma. Besides, comprehensive multi-omics analysis was used to outline stromal characterizations of 2 distinct stromal groups. Patients with activated tumor stoma showed high stromal cell infiltration (fibroblasts, endothelial cells, and monocyte macrophages), epithelial-mesenchymal transition, tumor angiogenesis and M2 macrophage polarization (CD163 and CD206). Tumor mutation burden of differential stromal groups was also depicted. In addition, a total of 6 stromal activation markers in ESCA were defined and involved in the function of carcinoma-associated fibroblasts that were crucial in the differentiation of distinct stromal characterizations. Based on these studies, a practical classifier for the stromal microenvironment was successfully proposed to predict the prognosis of ESCA patients.
Collapse
|
9
|
Reprogramming of Mesothelial-Mesenchymal Transition in Chronic Peritoneal Diseases by Estrogen Receptor Modulation and TGF-β1 Inhibition. Int J Mol Sci 2020; 21:ijms21114158. [PMID: 32532126 PMCID: PMC7312018 DOI: 10.3390/ijms21114158] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
In chronic peritoneal diseases, mesothelial-mesenchymal transition is determined by cues from the extracellular environment rather than just the cellular genome. The transformation of peritoneal mesothelial cells and other host cells into myofibroblasts is mediated by cell membrane receptors, Transforming Growth Factor β1 (TGF-β1), Src and Hypoxia-inducible factor (HIF). This article provides a narrative review of the reprogramming of mesothelial mesenchymal transition in chronic peritoneal diseases, drawing on the similarities in pathophysiology between encapsulating peritoneal sclerosis and peritoneal metastasis, with a particular focus on TGF-β1 signaling and estrogen receptor modulators. Estrogen receptors act at the cell membrane/cytosol as tyrosine kinases that can phosphorylate Src, in a similar way to other receptor tyrosine kinases; or can activate the estrogen response element via nuclear translocation. Tamoxifen can modulate estrogen membrane receptors, and has been shown to be a potent inhibitor of mesothelial-mesenchymal transition (MMT), peritoneal mesothelial cell migration, stromal fibrosis, and neoangiogenesis in the treatment of encapsulating peritoneal sclerosis, with a known side effect and safety profile. The ability of tamoxifen to inhibit the transduction pathways of TGF-β1 and HIF and achieve a quiescent peritoneal stroma makes it a potential candidate for use in cancer treatments. This is relevant to tumors that spread to the peritoneum, particularly those with mesenchymal phenotypes, such as colorectal CMS4 and MSS/EMT gastric cancers, and pancreatic cancer with its desmoplastic stroma. Morphological changes observed during mesothelial mesenchymal transition can be treated with estrogen receptor modulation and TGF-β1 inhibition, which may enable the regression of encapsulating peritoneal sclerosis and peritoneal metastasis.
Collapse
|
10
|
Okusha Y, Eguchi T, Tran MT, Sogawa C, Yoshida K, Itagaki M, Taha EA, Ono K, Aoyama E, Okamura H, Kozaki KI, Calderwood SK, Takigawa M, Okamoto K. Extracellular Vesicles Enriched with Moonlighting Metalloproteinase Are Highly Transmissive, Pro-Tumorigenic, and Trans-Activates Cellular Communication Network Factor ( CCN2/CTGF): CRISPR against Cancer. Cancers (Basel) 2020; 12:cancers12040881. [PMID: 32260433 PMCID: PMC7226423 DOI: 10.3390/cancers12040881] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/24/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinase 3 (MMP3) plays multiple roles in extracellular proteolysis as well as intracellular transcription, prompting a new definition of moonlighting metalloproteinase (MMP), according to a definition of protein moonlighting (or gene sharing), a phenomenon by which a protein can perform more than one function. Indeed, connective tissue growth factor (CTGF, aka cellular communication network factor 2 (CCN2)) is transcriptionally induced as well as cleaved by MMP3. Moreover, several members of the MMP family have been found within tumor-derived extracellular vesicles (EVs). We here investigated the roles of MMP3-rich EVs in tumor progression, molecular transmission, and gene regulation. EVs derived from a rapidly metastatic cancer cell line (LuM1) were enriched in MMP3 and a C-terminal half fragment of CCN2/CTGF. MMP3-rich, LuM1-derived EVs were disseminated to multiple organs through body fluid and were pro-tumorigenic in an allograft mouse model, which prompted us to define LuM1-EVs as oncosomes in the present study. Oncosome-derived MMP3 was transferred into recipient cell nuclei and thereby trans-activated the CCN2/CTGF promoter, and induced CCN2/CTGF production in vitro. TRENDIC and other cis-elements in the CCN2/CTGF promoter were essential for the oncosomal responsivity. The CRISPR/Cas9-mediated knockout of MMP3 showed significant anti-tumor effects such as the inhibition of migration and invasion of tumor cells, and a reduction in CCN2/CTGF promoter activity and fragmentations in vitro. A high expression level of MMP3 or CCN2/CTGF mRNA was prognostic and unfavorable in particular types of cancers including head and neck, lung, pancreatic, cervical, stomach, and urothelial cancers. These data newly demonstrate that oncogenic EVs-derived MMP is a transmissive trans-activator for the cellular communication network gene and promotes tumorigenesis at distant sites.
Collapse
Affiliation(s)
- Yuka Okusha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
- Division of Molecular and Cellular Biology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (E.A.); (M.T.)
- Correspondence: or
| | - Manh T. Tran
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
| | - Chiharu Sogawa
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
| | - Kaya Yoshida
- Department of Oral Healthcare Education, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8504, Japan;
| | - Mami Itagaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
- Research program for undergraduate students, Okayama University Dental School, Okayama 700-8525, Japan
| | - Eman A. Taha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
- Department of Medical Bioengineering, Graduate School of Natural Science and Technology, Okayama University, Okayama 700-8530, Japan
- Department of Biochemistry, Ain Shams University Faculty of Science, Cairo 11566, Egypt
| | - Kisho Ono
- Department of Oral and Maxillofacial Surgery, Okayama University Hospital, Okayama 700-0914, Japan;
| | - Eriko Aoyama
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (E.A.); (M.T.)
| | - Hirohiko Okamura
- Department of Oral Morphology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama 700-8525, Japan;
| | - Ken-ichi Kozaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
| | - Stuart K. Calderwood
- Division of Molecular and Cellular Biology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA;
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (E.A.); (M.T.)
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8525, Japan; (Y.O.); (M.T.T.); (C.S.); (M.I.); (E.A.T.); (K.-i.K.); (K.O.)
| |
Collapse
|
11
|
Cui WQ, Wang ST, Pan D, Chang B, Sang LX. Caffeine and its main targets of colorectal cancer. World J Gastrointest Oncol 2020; 12:149-172. [PMID: 32104547 PMCID: PMC7031145 DOI: 10.4251/wjgo.v12.i2.149] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/25/2019] [Accepted: 11/13/2019] [Indexed: 02/05/2023] Open
Abstract
Caffeine is a purine alkaloid and is widely consumed in coffee, soda, tea, chocolate and energy drinks. To date, a growing number of studies have indicated that caffeine is associated with many diseases including colorectal cancer. Caffeine exerts its biological activity through binding to adenosine receptors, inhibiting phosphodiesterases, sensitizing calcium channels, antagonizing gamma-aminobutyric acid receptors and stimulating adrenal hormones. Some studies have indicated that caffeine can interact with signaling pathways such as transforming growth factor β, phosphoinositide-3-kinase/AKT/mammalian target of rapamycin and mitogen-activated protein kinase pathways through which caffeine can play an important role in colorectal cancer pathogenesis, metastasis and prognosis. Moreover, caffeine can act as a general antioxidant that protects cells from oxidative stress and also as a regulatory factor of the cell cycle that modulates the DNA repair system. Additionally, as for intestinal homeostasis, through the interaction with receptors and cytokines, caffeine can modulate the immune system mediating its effects on T lymphocytes, B lymphocytes, natural killer cells and macrophages. Furthermore, caffeine can not only directly inhibit species in the gut microbiome, such as Escherichia coli and Candida albicans but also can indirectly exert inhibition by increasing the effects of other antimicrobial drugs. This review summarizes the association between colorectal cancer and caffeine that is being currently studied.
Collapse
Affiliation(s)
- Wen-Qi Cui
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
- China Medical University 101K class 87, Shenyang 110001, Liaoning Province, China
| | - Shi-Tong Wang
- Department of Cardiovascular Ultrasound, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
- China Medical University 101K class 87, Shenyang 110001, Liaoning Province, China
| | - Dan Pan
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li-Xuan Sang
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
12
|
Yao Y, Wang Y, Li L, Xiang X, Li J, Chen J, Liu Z, Huang S, Xiong J, Deng J. Down-regulation of interferon regulatory factor 2 binding protein 2 suppresses gastric cancer progression by negatively regulating connective tissue growth factor. J Cell Mol Med 2019; 23:8076-8089. [PMID: 31559693 PMCID: PMC6851004 DOI: 10.1111/jcmm.14677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 08/24/2019] [Indexed: 12/16/2022] Open
Abstract
Interferon regulatory factor 2 binding protein 2 (IRF2BP2) is a transcriptional repressor involved in regulating gene expression and other biological processes, including tumorigenesis. However, the clinical significance and roles of IRF2BP2 in human gastric cancer (GC) remain uncertain. Clinical GC tissues were obtained from GC patients at the First Affiliated Hospital of Nanchang University. Immunohistochemistry (IHC) was conducted to detect the IRF2BP2 protein in clinical paraffin specimens. Cell proliferation, migration and invasion were evaluated by MTT, colony formation assays and transwell assays. Co-immunoprecipitation was conducted to detect the interaction between TEA domain family members 4 (TEAD4) and vestigial-like family member 4 (VGLL4) or Yes-associated protein 1 (YAP1). Dual-luciferase reporter assay was used to confirm the binding of miR-101-3p to the 3'-UTR. The expression of IRF2BP2 was significantly higher in GC tissues than in normal tissues. Patients with higher IRF2BP2 protein expression had lower survival. IRF2BP2 knockdown inhibited proliferation, migration, invasion and epithelial-mesenchymal transition in GC cells. IRF2BP2 knockdown decreased the mRNA and protein levels of connective tissue growth factor (CTGF). The interaction between IRF2BP2 and VGLL4 increased the binding of TEAD4 to YAP1, resulting in the transcriptional coactivation of CTGF. In addition, miR-101-3p suppressed the expression of CTGF by directly targeting the 3'-UTR of IRF2BP2. Taken together, these findings provide a model for the role of miR-101-3p-IRF2BP2-CTGF signalling axis in GC and a novel insight into the mechanism of GC progression and metastasis.
Collapse
Affiliation(s)
- Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Yi Wang
- Radiotherapy&Chemotherapy Department, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang Province, China
| | - Li Li
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaojun Xiang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Junhe Li
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhen Liu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Shanshan Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
13
|
The Role of CTGF in Inflammatory Responses Induced by Silica Particles in Human Bronchial Epithelial Cells. Lung 2019; 197:783-791. [DOI: 10.1007/s00408-019-00272-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/04/2019] [Indexed: 01/31/2023]
|
14
|
Glatzel-Plucińska N, Piotrowska A, Dzięgiel P, Podhorska-Okołów M. The Role of SATB1 in Tumour Progression and Metastasis. Int J Mol Sci 2019; 20:E4156. [PMID: 31450715 PMCID: PMC6747166 DOI: 10.3390/ijms20174156] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/16/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022] Open
Abstract
Carcinogenesis is a long-drawn, multistep process, in which metastatic spread is an unequivocal hallmark of a poor prognosis. The progression and dissemination of epithelial cancers is commonly thought to rely on the epidermal-mesenchymal transition (EMT) process. During EMT, epithelial cells lose their junctions and apical-basal polarity, and they acquire a mesenchymal phenotype with its migratory and invasive capabilities. One of the proteins involved in cancer progression and EMT may be SATB1 (Special AT-Rich Binding Protein 1)-a chromatin organiser and a global transcriptional regulator. SATB1 organizes chromatin into spatial loops, providing a "docking site" necessary for the binding of further transcription factors and chromatin modifying enzymes. SATB1 has the ability to regulate whole sets of genes, even those located on distant chromosomes. SATB1 was found to be overexpressed in numerous malignancies, including lymphomas, breast, colorectal, prostate, liver, bladder and ovarian cancers. In the solid tumours, an elevated SATB1 level was observed to be associated with an aggressive phenotype, presence of lymph node, distant metastases, and a poor prognosis. In this review, we briefly describe the prognostic significance of SATB1 expression in most common human cancers, and analyse its impact on EMT and metastasis.
Collapse
Affiliation(s)
- Natalia Glatzel-Plucińska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland.
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland
| | | |
Collapse
|
15
|
Jacenik D, Beswick EJ, Krajewska WM, Prossnitz ER. G protein-coupled estrogen receptor in colon function, immune regulation and carcinogenesis. World J Gastroenterol 2019; 25:4092-4104. [PMID: 31435166 PMCID: PMC6700692 DOI: 10.3748/wjg.v25.i30.4092] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 02/06/2023] Open
Abstract
Estrogens play important roles in the development and progression of multiple tumor types. Accumulating evidence points to the significance of estrogen action not only in tumors of hormonally regulated tissues such as the breast, endometrium and ovary, but also in the development of colorectal cancer (CRC). The effects of estrogens in physiological and pathophysiological conditions are mediated by the nuclear estrogen receptors α and β, as well as the membrane-bound G protein-coupled estrogen receptor (GPER). The roles of GPER in CRC development and progression, however, remain poorly understood. Studies on the functions of GPER in the colon have shown that this estrogen receptor regulates colonic motility as well as immune responses in CRC-associated diseases, such as Crohn’s disease and ulcerative colitis. GPER is also involved in cell cycle regulation, endoplasmic reticulum stress, proliferation, apoptosis, vascularization, cell migration, and the regulation of fatty acid and estrogen metabolism in CRC cells. Thus, multiple lines of evidence suggest that GPER may play an important role in colorectal carcinogenesis. In this review, we present the current state of knowledge regarding the contribution of GPER to colon function and CRC.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
- Department of Internal Medicine, School of Medicine, and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, United States
| | - Ellen J Beswick
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, UT 84132, United States
| | - Wanda M Krajewska
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-236, Poland
| | - Eric R Prossnitz
- Department of Internal Medicine, School of Medicine, and UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, United States
| |
Collapse
|
16
|
Hosseinipour M, Wan F, Altomare D, Creek KE, Pirisi L. HPV16-transformed human keratinocytes depend on SIX1 expression for proliferation and HPV E6/E7 gene expression. Virology 2019; 537:20-30. [PMID: 31425971 DOI: 10.1016/j.virol.2019.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
The homeodomain transcription factor SIX1 plays a critical role in embryogenesis, is not expressed in normal adult tissue, but is expressed in many malignancies, including cervical cancer. SIX1 drives the progression of HPV16-immortalized human keratinocytes (HKc/HPV16) toward malignancy: HKc/HPV16 express high levels of SIX1 mRNA and protein; overexpression of SIX1 in HKc/HPV16 produces pre-malignant, differentiation-resistant lines (HKc/DR); SIX1 overexpression in HKc/DR induces tumorigenicity. In this paper, we explore the consequences of inhibition of SIX1 expression in premalignant HKc/DR. Only partial inhibition of SIX1 expression could be obtained in HKc/DR by RNA interference. Decreased SIX1 expression (up to 80%) in HKc/DR resulted in slower proliferation, decreased HPV16-E6/E7 mRNA levels, and increased p53 protein levels. Gene expression changes induced in HKc/DR by anti-SIX1 shRNA were indicative of mesenchymal-epithelial transition (MET) and changes in TGF-beta signaling. We conclude that HPV16-transformed cells depend on SIX1 for survival, HPV16 E6/E7 gene expression and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Maria Hosseinipour
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine at Columbia, Columbia, SC 29208, USA
| | - Fang Wan
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine at Columbia, Columbia, SC 29208, USA; Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, 29208, USA
| | - Kim E Creek
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, 29208, USA
| | - Lucia Pirisi
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine at Columbia, Columbia, SC 29208, USA.
| |
Collapse
|
17
|
High Expression of LTBP2 Contributes to Poor Prognosis in Colorectal Cancer Patients and Correlates with the Mesenchymal Colorectal Cancer Subtype. DISEASE MARKERS 2019; 2019:5231269. [PMID: 30956730 PMCID: PMC6431450 DOI: 10.1155/2019/5231269] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/07/2019] [Indexed: 01/12/2023]
Abstract
Colorectal cancer (CRC) is a complex and heterogeneous disease with four consensus molecular subtypes (CMS1-4). LTBP2 is a member of the fibrillin/LTBP super family and plays a critical role in tumorigenesis by activating TGF-β in the CMS4 CRC subtype. So far, the expression and prognostic significance of LTBP2 in CRC remains obscure. In this study, we aimed to analyze the mRNA and protein expression levels of LTBP2 in CRC tissues and then estimate their values as a potential prognostic biomarker. We detected the mRNA expression of LTBP2 in 28 cases of fresh CRC tissues and 4 CRC cell lines and the protein expression of LTBP2 in 483 samples of CRC tissues, matched tumor-adjacent tissues, and benign colorectal diseases. LTBP2 protein expression was then correlated to patients' clinical features and overall survival. Both LTBP2 mRNA and protein expression levels in CRC tissues were remarkably superior to those in adjacent normal colorectal tissues (P = 0.0071 and P < 0.001, respectively), according to TCGA dataset of CRC. High LTBP2 protein expression was correlated with TNM stage (P < 0.001), T stage (P < 0.001), N stage (P < 0.001), and M stage (P < 0.001). High LTBP2 protein expression was related to poor overall survival in CRC patients and was an independent prognostic factor for CRC. LTBP2 mRNA expression was especially higher in the CMS4 subtype (P < 0.001), which was confirmed in CRC cell lines. Our data suggested that LTBP2 may act as an oncogene in the development of colorectal cancer and have important significance in predicting CRC prognosis. LTBP2 could be a novel biomarker and potential therapeutic target for mesenchymal colorectal cancer and can improve the outcome of high-risk CRC.
Collapse
|
18
|
Ma T, Dong LJ, Du XL, Niu R, Hu BJ. Research progress on the role of connective tissue growth factor in fibrosis of diabetic retinopathy. Int J Ophthalmol 2018; 11:1550-1554. [PMID: 30225233 DOI: 10.18240/ijo.2018.09.20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 03/15/2018] [Indexed: 01/03/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the most important types of diabetic microangiopathy, which is a specific change of fundus lesions and is one of the most serious complications of diabetes. When DR develops to proliferative DR, the main factors of decreasing vision, and even blindness, include retinal detachment and vitreous hemorrhage caused by contraction of blood vessels by fiber membrane. Recent studies reported that the formation of fiber vascular membrane is closely related to retinal fibrosis. The connective tissue growth factor (CTGF) is a cytokine that is closely related to DR fibrosis. However, its mechanism is poorly understood. This paper summarizes the recent studies about CTGF on DR fibrosis for a comprehensive understanding of the role and mechanism of CTGF in PDR.
Collapse
Affiliation(s)
- Teng Ma
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, Ophthalmology College of Tianjin Medical University, Tianjin 300384, China
| | - Li-Jie Dong
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, Ophthalmology College of Tianjin Medical University, Tianjin 300384, China
| | - Xue-Li Du
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, Ophthalmology College of Tianjin Medical University, Tianjin 300384, China
| | - Rui Niu
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, Ophthalmology College of Tianjin Medical University, Tianjin 300384, China
| | - Bo-Jie Hu
- Tianjin Medical University Eye Hospital, Tianjin Medical University Eye Institute, Ophthalmology College of Tianjin Medical University, Tianjin 300384, China
| |
Collapse
|
19
|
Ramazani Y, Knops N, Elmonem MA, Nguyen TQ, Arcolino FO, van den Heuvel L, Levtchenko E, Kuypers D, Goldschmeding R. Connective tissue growth factor (CTGF) from basics to clinics. Matrix Biol 2018; 68-69:44-66. [DOI: 10.1016/j.matbio.2018.03.007] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
|
20
|
Emon B, Bauer J, Jain Y, Jung B, Saif T. Biophysics of Tumor Microenvironment and Cancer Metastasis - A Mini Review. Comput Struct Biotechnol J 2018; 16:279-287. [PMID: 30128085 PMCID: PMC6097544 DOI: 10.1016/j.csbj.2018.07.003] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 07/20/2018] [Accepted: 07/21/2018] [Indexed: 02/07/2023] Open
Abstract
The role of tumor microenvironment in cancer progression is gaining significant attention. It is realized that cancer cells and the corresponding stroma co-evolve with time. Cancer cells recruit and transform the stromal cells, which in turn remodel the extra cellular matrix of the stroma. This complex interaction between the stroma and the cancer cells results in a dynamic feed-forward/feed-back loop with biochemical and biophysical cues that assist metastatic transition of the cancer cells. Although biochemistry has long been studied for the understanding of cancer progression, biophysical signaling is emerging as a critical paradigm determining cancer metastasis. In this mini review, we discuss the role of one of the biophysical cues, mostly the mechanical stiffness of tumor microenvironment, in cancer progression and its clinical implications.
Collapse
Key Words
- ADAMs, Adamalysins
- ANGPT2, Angiopoietin 2
- Activin/TGFβ
- CAF, Cancer associated fibroblast
- CSF-1, Colony stimulating factor 1
- CTGF, Connective tissue growth factor
- CYR61/CCN1, Cysteine-rich angiogenic inducer 61/CCN family member 1
- Cancer
- ECM stiffness
- ECM, Extracellular matrix
- EGF, Epidermal growth factor
- EMT, Epithelial to mesenchymal transition
- FGF, Fibroblast growth factor
- Growth factors
- HGF/SF, Hepatocyte growth factor/Scatter factor
- IGFs, Insulin-like growth factors
- IL-13, Interleukin-13
- IL-33, Interleukin-33
- IL-6, Interleukin-6
- KGF, Keratinocyte growth factor, also FGF7
- LOX, Lysyl Oxidase
- MMPs, Matrix metalloproteinases
- Metastasis
- NO, Nitric oxide
- SDF-1/CXCL12, Stromal cell-derived factor 1/C-X-C motif chemokine 12
- TACs, Tumor-associated collagen signatures
- TGFβ, Transforming growth factor β
- TNF-α, Tumor necrosis factor-α
- Tumor biophysics
- VEGF, Vascular endothelial growth factor
- α-SMA, α-Smooth muscle actin
Collapse
Affiliation(s)
- Bashar Emon
- Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, United States
| | - Jessica Bauer
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, United States
| | - Yasna Jain
- Department of Architecture, BRAC University, Dhaka
| | - Barbara Jung
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, United States
| | - Taher Saif
- Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, United States
- Bioengineering, University of Illinois at Urbana-Champaign, United States
| |
Collapse
|
21
|
Hou CH, Yang RS, Tsao YT. Connective tissue growth factor stimulates osteosarcoma cell migration and induces osteosarcoma metastasis by upregulating VCAM-1 expression. Biochem Pharmacol 2018; 155:71-81. [PMID: 29909077 DOI: 10.1016/j.bcp.2018.06.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/13/2018] [Indexed: 12/22/2022]
Abstract
Osteosarcoma is the most common bone malignancy that occurs in the young population. After osteosarcoma cells metastasize to the lung, prognosis is very poor owing to difficulties in early diagnosis and effective treatment. Recently, connective tissue growth factor (CTGF) was reported to be a critical contributor to osteosarcoma metastasis. However, the detailed mechanism associated with CTGF-directed migration in bone neoplasms is still mostly unknown. Through the in vivo and in vitro examination of osteosarcoma cells, this study suggests that VCAM-1 up-regulation and increased osteosarcoma cell migration are involved in this process. Antagonizing αvβ3 integrin inhibited cell migration. Moreover, FAK, PI3K, Akt and NF-κB activation were also shown to be involved in CTGF-mediated osteosarcoma metastasis. Taken together, CTGF promotes VCAM-1 production and further induces osteosarcoma metastasis via the αvβ3 integrin/FAK/PI3K/Akt/NF-κB signaling pathway, which could represent a promising clinical target to improve patient outcome.
Collapse
Affiliation(s)
- Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, NO 1, Jen-Ai Road, Taipei 100, Taiwan.
| | - Rong-Sen Yang
- Department of Orthopedic Surgery, National Taiwan University Hospital, NO 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Ya-Ting Tsao
- Department of Orthopedic Surgery, National Taiwan University Hospital, NO 1, Jen-Ai Road, Taipei 100, Taiwan
| |
Collapse
|
22
|
Lun W, Wu X, Deng Q, Zhi F. MiR-218 regulates epithelial-mesenchymal transition and angiogenesis in colorectal cancer via targeting CTGF. Cancer Cell Int 2018; 18:83. [PMID: 29977158 PMCID: PMC5994014 DOI: 10.1186/s12935-018-0575-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 05/26/2018] [Indexed: 12/24/2022] Open
Abstract
Background Endothelial-to-mesenchymal transition (EMT) and angiogenesis play important roles in colorectal cancer (CRC) development. Connective tissue growth factor (CTGF) has been reported to promote several kinds of cancer progression and miR-218 has been identified as a tumor suppressor miRNA. However, little is known about the function of miR-218 in CRC. Here we investigated the effects of miR-218 on EMT and angiogenesis process in CRC cells. As well, the relation between miR-218 and CTGF was identified. The mechanism of miR-218’s function was illustrated. Methods CRC cell lines were transfected with miR-218 mimics. Proliferation, migration and angiogenesis were identified by MTT assay, Transwell assay, colony formation assay and tube formation assay. Protein and mRNA expression levels of associated genes were measured by Western blotting and RT-PCR. Dual luciferase assay was used to determine the relation of miR-218 and CTGF. Results miR-218 was down-regulated in CRC cell lines and over expression of miR-218 could significantly inhibit EMT and angiogenesis. CTGF was a direct target of miR-218. Up regulation of CTGF level after miR-218 transfection could sufficiently rescue the suppression effects on EMT and angiogenesis. Conclusion miR-218 directly targets CTGF and inhibits its expression, leading to suppression on EMT and angiogenesis of CRC cells. miR-218 might be used as potential therapeutic strategy for CRC treatment.
Collapse
Affiliation(s)
- Weijian Lun
- Guangdong Provincial Key Laboratory of Gastroenterology, Inst. of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Xiongjian Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Inst. of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Qiliang Deng
- Guangdong Provincial Key Laboratory of Gastroenterology, Inst. of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Inst. of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
23
|
Santolla MF, Lappano R, Cirillo F, Rigiracciolo DC, Sebastiani A, Abonante S, Tassone P, Tagliaferri P, Di Martino MT, Maggiolini M, Vivacqua A. miR-221 stimulates breast cancer cells and cancer-associated fibroblasts (CAFs) through selective interference with the A20/c-Rel/CTGF signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:94. [PMID: 29716623 PMCID: PMC5930435 DOI: 10.1186/s13046-018-0767-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/20/2018] [Indexed: 01/19/2023]
Abstract
Background MicroRNA (miRNAs) are non-coding small RNA molecules that regulate gene expression by inhibiting the translation of target mRNAs. Among several dysregulated miRNAs in human cancer, the up-regulation of miR-221 has been associated with development of a variety of hematologic and solid malignancies. In this study, we investigated the involvement of miR-221 in breast cancer. Methods TaqMan microRNA assay was used to detect the miR-221 levels in normal cells and in MDA-MB 231 and SkBr3 breast cancer cells as well as in main players of the tumor microenvironment, namely cancer-associated fibroblasts (CAFs). miR-221 mimic sequence and locked nucleic acid (LNA)-i-miR-221 construct were used to induce or inhibit, respectively, the miR-221 expression in cells used. Quantitative PCR and western blotting analysis were performed to evaluate the levels of the miR-221 target gene A20 (TNFAIP3), as well as the member of the NF-kB complex namely c-Rel and the connective tissue growth factor (CTGF). Chromatin immunoprecipitation (ChIP) assay was performed to ascertain the recruitment of c-Rel to the CTFG promoter. Finally, the cell growth and migration in the presence of LNA-i-miR-221 or silencing c-Rel and CTGF by specific short hairpin were assessed by cell count, colony formation and boyden chambers assays. Statistical analysis was performed by ANOVA. Results We first demonstrated that LNA-i-miR-221 inhibits both endogenous and ectopic expression of miR-221 in our experimental models. Next, we found that the A20 down-regulation, as well as the up-regulation of c-Rel induced by miR-221 were no longer evident using LNA-i-miR-221. Moreover, we established that the miR-221 dependent recruitment of c-Rel to the NF-kB binding site located within the CTGF promoter region is prevented by using LNA-i-miR-221. Furthermore, we determined that the up-regulation of CTGF mRNA and protein levels by miR-221 is no longer evident using LNA-i-miR221 and silencing c-Rel. Finally, we assessed that cell growth and migration induced by miR-221 in MDA-MB 231 and SkBr3 breast cancer cells as well as in CAFs are abolished by LNAi-miR-221 and silencing c-Rel or CTGF. Conclusions Overall, these data provide novel insights into the stimulatory action of miR-221 in breast cancer cells and CAFs, suggesting that its inhibition may be considered toward targeted therapeutic approaches in breast cancer patients.
Collapse
Affiliation(s)
| | - Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | - Anna Sebastiani
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Adele Vivacqua
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
24
|
Druliner BR, Wang P, Bae T, Baheti S, Slettedahl S, Mahoney D, Vasmatzis N, Xu H, Kim M, Bockol M, O'Brien D, Grill D, Warner N, Munoz-Gomez M, Kossick K, Johnson R, Mouchli M, Felmlee-Devine D, Washechek-Aletto J, Smyrk T, Oberg A, Wang J, Chia N, Abyzov A, Ahlquist D, Boardman LA. Molecular characterization of colorectal adenomas with and without malignancy reveals distinguishing genome, transcriptome and methylome alterations. Sci Rep 2018; 8:3161. [PMID: 29453410 PMCID: PMC5816667 DOI: 10.1038/s41598-018-21525-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 02/06/2018] [Indexed: 12/19/2022] Open
Abstract
The majority of colorectal cancer (CRC) arises from precursor lesions known as polyps. The molecular determinants that distinguish benign from malignant polyps remain unclear. To molecularly characterize polyps, we utilized Cancer Adjacent Polyp (CAP) and Cancer Free Polyp (CFP) patients. CAPs had tissues from the residual polyp of origin and contiguous cancer; CFPs had polyp tissues matched to CAPs based on polyp size, histology and dysplasia. To determine whether molecular features distinguish CAPs and CFPs, we conducted Whole Genome Sequencing, RNA-seq, and RRBS on over 90 tissues from 31 patients. CAPs had significantly more mutations, altered expression and hypermethylation compared to CFPs. APC was significantly mutated in both polyp groups, but mutations in TP53, FBXW7, PIK3CA, KIAA1804 and SMAD2 were exclusive to CAPs. We found significant expression changes between CAPs and CFPs in GREM1, IGF2, CTGF, and PLAU, and both expression and methylation alterations in FES and HES1. Integrative analyses revealed 124 genes with alterations in at least two platforms, and ERBB3 and E2F8 showed aberrations specific to CAPs across all platforms. These findings provide a resource of molecular distinctions between polyps with and without cancer, which have the potential to enhance the diagnosis, risk assessment and management of polyps.
Collapse
Affiliation(s)
- Brooke R Druliner
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Panwen Wang
- Health Sciences Research, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Taejeong Bae
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Saurabh Baheti
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Seth Slettedahl
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Douglas Mahoney
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nikolaos Vasmatzis
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Hang Xu
- Center for Genomic Sciences & School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Minsoo Kim
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew Bockol
- Information Technology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Daniel O'Brien
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Diane Grill
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nathaniel Warner
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Miguel Munoz-Gomez
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Kimberlee Kossick
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ruth Johnson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Mohamad Mouchli
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Donna Felmlee-Devine
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jill Washechek-Aletto
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Thomas Smyrk
- Anatomic Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ann Oberg
- Department of Health Sciences Research, Cancer Center Statistics Mayo Clinic, Rochester, MN, 55905, USA
| | - Junwen Wang
- Health Sciences Research, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Nicholas Chia
- Department of Health Sciences Research, Center for Individualized Medicine, College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.,Department of Surgery, College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA.,Department of Bioengineering and Physiology, College of Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alexej Abyzov
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - David Ahlquist
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lisa A Boardman
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
25
|
Takigawa M. An early history of CCN2/CTGF research: the road to CCN2 via hcs24, ctgf, ecogenin, and regenerin. J Cell Commun Signal 2017; 12:253-264. [PMID: 29076115 DOI: 10.1007/s12079-017-0414-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023] Open
Abstract
The principal aim of this historical review is to present the processes by which the different aspects of CCN2/CTGF/Hcs24 were discovered by different groups and how much CCN2/CTGF, by being integrated into CCN family, has contributed to the establishment of the basic concepts regarding the role and functions of this new class of proteins. This review should be particularly useful to new investigators who have recently entered this exciting field of study and also provides a good opportunity to acknowledge the input of those individuals who participated in the development of this scientific field.
Collapse
Affiliation(s)
- Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences (ARCOCS), Okayama University Dental School/Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Okayama, 700-8525, Japan.
| |
Collapse
|
26
|
Wang WH, Deng AJ, He SG. A key role of microRNA-26a in the scar formation after glaucoma filtration surgery. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:831-837. [PMID: 28685590 DOI: 10.1080/21691401.2017.1345926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Wei-Hong Wang
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ai-Jun Deng
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Sheng-Guang He
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
27
|
陈 会, 周 升, 辛 瑞, 刘 钰, 王 凯, 刘 鲁. LAPTM4B-35、CTGF蛋白在食管胃交界部腺癌中的表达及其意义. Shijie Huaren Xiaohua Zazhi 2017; 25:1240-1247. [DOI: 10.11569/wcjd.v25.i14.1240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
目的 探讨溶酶体相关4次跨膜蛋白β(lysosome-associated protein transmembrane-4β, LAPTM4B)基因-35及结缔组织生长因子(connective tissue growth factor, CTGF)蛋白在食管胃交界部腺癌组织中的表达及其意义.
方法 采用免疫组织化学染色及蛋白印迹分析检测137例食管胃交界部腺癌组织中LAPTM4B-35及CTGF蛋白的表达情况, 分析蛋白表达与患者临床病理参数的关系及对患者预后的影响.
结果 LAPTM4B-35蛋白在89.1%的食管胃交界部腺癌组织中呈高表达状态, LAPTM4B-35高表达组癌细胞发生远处转移(P = 0.011)、肿瘤进展至TNM Ⅲ/Ⅳ期(P = 0.026)显著多于低表达组. CTGF蛋白在51.1%的癌组织中呈高表达状态, CTGF高表达组发生远处转移(P = 0.033)、肿瘤直径>5 cm(P = 0.021)均多于低表达组. 癌组织中LAPTM4B-35蛋白与CTGF蛋白表达之间呈正相关关系(r = 0.218, P = 0.010). 单因素生存分析显示LAPTM4B-35、CTGF蛋白高表达的患者其术后生存时间分别显著低于LAPTM4B-35、CTGF蛋白低表达的患者(双侧log-rank检验, P<0.001). LAPTM4B-35蛋白表达(P<0.001)、发生远处转移(P<0.001)、淋巴结转移(P = 0.007)、查见脉管癌栓(P = 0.022)及患者性别(P<0.001)是影响食管胃交界部腺癌患者的独立预后因子.
结论 LAPTM4B-35及CTGF蛋白的高表达与肿瘤的侵袭转移及不良预后密切相关.
Collapse
|