1
|
Polushkina TM, Porubaeva EE, Danilova NV. [The phenomenon of tumor budding in gastric cancer]. Arkh Patol 2025; 87:79-87. [PMID: 40289438 DOI: 10.17116/patol20258702179] [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] [Indexed: 04/30/2025]
Abstract
Gastric cancer remains the fifth most common malignant neoplasm in the world and ranks fifth among the causes associated with cancer. The TNM system remains the gold standard for predictive stratification of patients with gastric cancer, but the search for new sensitive, specific and reproducible biomarkers to develop a personalized approach to the management of patients with gastric cancer does not lose its relevance. The phenomenon of tumor budding is a well-established independent prognostic factor in colorectal cancer. In 2017, the first guideline on the method of calculating tumor budding for colorectal cancer was published. Despite the promising potential of using tumor budding in gastric cancer this parameter is still not evaluated in everyday practice. This lection provides data on various methods of counting tumor budding in gastric carcinomas, describes the molecular mechanisms of interaction between tumor cells and the immune microenvironment, and summarizes the available data on the relationship of clinical and morphological characteristics of gastric cancer with the degree of tumor budding. The relationship between the degree of tumor budding and the prognostic characteristics of gastric cancer and the prospects for its use is also described.
Collapse
Affiliation(s)
| | | | - N V Danilova
- Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
2
|
Wolosowicz M, Prokopiuk S, Kaminski TW. The Complex Role of Matrix Metalloproteinase-2 (MMP-2) in Health and Disease. Int J Mol Sci 2024; 25:13691. [PMID: 39769454 PMCID: PMC11728377 DOI: 10.3390/ijms252413691] [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: 11/29/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Matrix metalloproteinase-2 (MMP-2), a zinc-dependent enzyme, plays a critical role in the degradation and remodeling of the extracellular matrix (ECM). As a member of the gelatinase subgroup of matrix metalloproteinases, MMP-2 is involved in a variety of physiological processes, including tissue repair, wound healing, angiogenesis, and embryogenesis. It is primarily responsible for the degradation of type IV and V collagen, fibronectin, laminin, and elastin, which are essential components of the ECM. MMP-2 is secreted as an inactive pro-enzyme (proMMP-2) and activated through proteolytic cleavage, with its activity being precisely regulated by tissue inhibitors of metalloproteinases (TIMPs). Dysregulation of MMP-2 has been linked to a variety of pathological conditions, including cardiovascular diseases, diabetic complications, kidney diseases, and cancer. In cardiovascular diseases, it contributes to vascular remodeling, atherosclerosis, and aneurysms, while in fibrotic diseases, it mediates excessive ECM degradation leading to tissue scarring. In diabetes, elevated MMP-2 activity exacerbates complications such as nephropathy, retinopathy, and cardiovascular disease. In cancer, MMP-2 facilitates tumor invasion and metastasis by degrading ECM components and promoting angiogenesis. Despite its essential roles in both physiological and pathological processes, targeting MMP-2 for therapeutic purposes presents challenges due to its dual functions in tissue remodeling and repair, raising concerns about unplanned consequences such as impaired tissue healing or excessive tissue damage. These challenges underscore the need for future research to focus on developing selective modulators that can precisely balance their activity under specific disease environments. Clinical trials targeting MMP-2 modulation highlight the potential of gelatinase inhibitors, including those targeting MMP-2, to reduce tumor progression in fibrosarcoma, breast, and lung cancers. This paper reviews the structure, function, and regulation of MMP-2, its involvement in disease pathogenesis, and the potential challenges in the therapeutic implications of modulating its activity.
Collapse
Affiliation(s)
- Marta Wolosowicz
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Slawomir Prokopiuk
- Faculty of Health Sciences, University of Lomza, 14 Akademicka St., 18-400 Łomża, Poland;
| | - Tomasz W. Kaminski
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Thrombosis and Hemostasis Program, VERSITI Blood Research Institute, Milwaukee, WI 53226, USA
| |
Collapse
|
3
|
Marques-Ramos A, Cervantes R. Expression of mTOR in normal and pathological conditions. Mol Cancer 2023; 22:112. [PMID: 37454139 PMCID: PMC10349476 DOI: 10.1186/s12943-023-01820-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/08/2023] [Indexed: 07/18/2023] Open
Abstract
The mechanistic/mammalian target of rapamycin (mTOR), a protein discovered in 1991, integrates a complex pathway with a key role in maintaining cellular homeostasis. By comprising two functionally distinct complexes, mTOR complex 1 (mTORC1) and mTORC2, it is a central cellular hub that integrates intra- and extracellular signals of energy, nutrient, and hormone availability, modulating the molecular responses to acquire a homeostatic state through the regulation of anabolic and catabolic processes. Accordingly, dysregulation of mTOR pathway has been implicated in a variety of human diseases. While major advances have been made regarding the regulators and effectors of mTOR signaling pathway, insights into the regulation of mTOR gene expression are beginning to emerge. Here, we present the current available data regarding the mTOR expression regulation at the level of transcription, translation and mRNA stability and systematize the current knowledge about the fluctuations of mTOR expression observed in several diseases, both cancerous and non-cancerous. In addition, we discuss whether mTOR expression changes can be used as a biomarker for diagnosis, disease progression, prognosis and/or response to therapeutics. We believe that our study will contribute for the implementation of new disease biomarkers based on mTOR as it gives an exhaustive perspective about the regulation of mTOR gene expression in both normal and pathological conditions.
Collapse
Affiliation(s)
- A Marques-Ramos
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal.
| | - R Cervantes
- H&TRC-Health & Technology Research Center, ESTeSL-Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
- Public Health Research Centre, NOVA National School of Public Health, Universidade Nova de Lisboa, Lisbon, Portugal
- Comprehensive Health Research Center (CHRC), Lisbon, Portugal
| |
Collapse
|
4
|
Xiao SM, Li J. Tumor budding in gastric cancer. World J Gastrointest Surg 2023; 15:578-591. [PMID: 37206064 PMCID: PMC10190737 DOI: 10.4240/wjgs.v15.i4.578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/04/2023] [Accepted: 03/21/2023] [Indexed: 04/22/2023] Open
Abstract
The tumor, nodes, metastasis (TNM) staging system has long been the gold standard for the classification and prognosis of solid tumors. However, the TNM staging system is not without limitations. Prognostic heterogeneity exists within patients at the same stage. Therefore, the pursuit of other biomarkers with the potential to classify patients with cancer has never stopped. One of them, tumor budding (TB), has gained much success in colorectal cancer. In recent years, TB in gastric cancer has attracted much attention from researchers, beginning to reveal the molecular and biological aspects of this phenomenon in gastric cancer, and has emerged as a promising prognostic biomarker in gastric cancer, predicting disease progression and unfavorable survival. Therefore, it is time and essential to provide a holistic overview of TB in gastric cancer, which has not been achieved and is the aim of this review.
Collapse
Affiliation(s)
- Shuo-Meng Xiao
- Department of Gastric Surgery, Sichuan Cancer Hospital, Chengdu 610041, Sichuan Province, China
| | - Jian Li
- Department of General Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang 621000, Sichuan Province, China
| |
Collapse
|
5
|
Shan MJ, Meng LB, Guo P, Zhang YM, Kong D, Liu YB. Screening and Identification of Key Biomarkers of Gastric Cancer: Three Genes Jointly Predict Gastric Cancer. Front Oncol 2021; 11:591893. [PMID: 34485109 PMCID: PMC8416116 DOI: 10.3389/fonc.2021.591893] [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: 08/05/2020] [Accepted: 06/17/2021] [Indexed: 12/12/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common cancers all over the world, causing high mortality. Gastric cancer screening is one of the effective strategies used to reduce mortality. We expect that good biomarkers can be discovered to diagnose and treat gastric cancer as early as possible. Methods We download four gene expression profiling datasets of gastric cancer (GSE118916, GSE54129, GSE103236, GSE112369), which were obtained from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) between gastric cancer and adjacent normal tissues were detected to explore biomarkers that may play an important role in gastric cancer. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of overlap genes were conducted by the Metascape online database; the protein-protein interaction (PPI) network was constructed by the STRING online database, and we screened the hub genes of the PPI network using the Cytoscape software. The survival curve analysis was conducted by km-plotter and the stage plots of hub genes were created by the GEPIA online database. PCR, WB, and immunohistochemistry were used to verify the expression of hub genes. A neural network model was established to quantify the predictors of gastric cancer. Results The relative expression level of cadherin-3 (CDH3), lymphoid enhancer-binding factor 1 (LEF1), and matrix metallopeptidase 7 (MMP7) were significantly higher in gastric samples, compared with the normal groups (p<0.05). Receiver operator characteristic (ROC) curves were constructed to determine the effect of the three genes’ expression on gastric cancer, and the AUC was used to determine the degree of confidence: CDH3 (AUC = 0.800, P<0.05, 95% CI =0.857-0.895), LEF1 (AUC=0.620, P<0.05, 95%CI=0.632-0.714), and MMP7 (AUC=0.914, P<0.05, 95%CI=0.714-0.947). The high-risk warning indicator of gastric cancer contained 8<CDH3<15 and 10<expression of LEF1<16. Conclusions CDH3, LEF1, and MMP7 can be used as candidate biomarkers to construct a neural network model from hub genes, which may be helpful for the early diagnosis of gastric cancer.
Collapse
Affiliation(s)
- Meng-Jie Shan
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing, China.,Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ling-Bing Meng
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Cardiology Department, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Peng Guo
- Department of Orthopedics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuan-Meng Zhang
- Department of Cardiology, The Third Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Dexian Kong
- Department of Endocrinology, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ya-Bin Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
6
|
Song R, Qiao W, He J, Huang J, Luo Y, Yang T. Proteases and Their Modulators in Cancer Therapy: Challenges and Opportunities. J Med Chem 2021; 64:2851-2877. [PMID: 33656892 DOI: 10.1021/acs.jmedchem.0c01640] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteostasis is the process of regulating intracellular proteins to maintain the balance of the cell proteome, which is crucial for cancer cell survival. Several proteases located in the cytoplasm, mitochondria, lysosome, and extracellular environment have been identified as potential antitumor targets because of their involvement in proteostasis. Although the discovery of small-molecule inhibitors targeting proteases faces particular challenges, rapid advances in chemical biology and structural biology, and the new technology of drug discovery have facilitated the development of promising protease modulators. In this review, the protein structure and function of important tumor-related proteases and their inhibitors are presented. We also provide a prospective on advances and the outlook of new drug strategies that target these proteases.
Collapse
Affiliation(s)
- Rao Song
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenliang Qiao
- Lung Cancer Center, Laboratory of Lung Cancer, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Jun He
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiasheng Huang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Jafari E, Safinejad S, Dabiri S, Naghibzadeh-Tahami A. Study of the Relationship between MMP-2 and MMP-9 and Her2/neu Overexpression in Gastric Cancer: Clinico- Pathological Correlations. Asian Pac J Cancer Prev 2021; 22:811-817. [PMID: 33773545 PMCID: PMC8286688 DOI: 10.31557/apjcp.2021.22.3.811] [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: 10/06/2020] [Indexed: 12/09/2022] Open
Abstract
Background: The relationship between the expressions of matrix metalloproteinases with clinico-pathological data on gastric cancer has been investigated in many countries, but this relationship remains unexplored in Iranian patients. Also, the correlation of the MMPs and the HER-2/neu proto-oncogene with other clinic-pathological variables has been evaluated for several other malignancies, but little effort has been made to shed light on the relationship with gastric cancer. Methods: We investigated MMP-2 and MMP-9 expression and HERE-2/neu overexpression in 48 gastric cancer patients referred to Afzalipour Hospital, associated with Kerman Medical University. Immunohistochemistry staining with rabbit polyclonal antibodies was used. Data statistical analysis was done by SPSS software (Version 20.0). Results: The mean age was 59, most of the patients were male (79.2%), and the average tumor size was larger than 5 centimeters in its greatest diameter. The majority of tumors were of the intestinal subtype and were located in the pyloric and antrum regions (43.8%). Invasion to muscularis properia was seen in 87.5% of the tumors (T3). MMP-2 and MMP-9 were highly expressed in 58.3% and 50% of cases, respectively, and Her-2/neu positivity was 10.4%. MMP-2, MMP-9 and HER-2 were found to have no relation with any clinicopathological parameters. Conclusion: According to the results of this study, MMP-2 and MMP-9 were highly expressed in gastric cancer, but there was no significant association with other clinicopathological variables.
Collapse
Affiliation(s)
- Elham Jafari
- Pathology and Stem Cells Research Center, Department of Pathology, Kerman University of Medical Science, Kerman, Iran
| | - Somaye Safinejad
- Pathology and Stem Cells Research Center, Department of Pathology, Kerman University of Medical Science, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cells Research Center, Department of Pathology, Kerman University of Medical Science, Kerman, Iran
| | - Ahmad Naghibzadeh-Tahami
- Health Services Management Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
8
|
Zhao C, Yuan G, Jiang Y, Xu J, Ye L, Zhan W, Wang J. Capn4 contributes to tumor invasion and metastasis in gastric cancer via activation of the Wnt/β-catenin/MMP9 signalling pathways. Exp Cell Res 2020; 395:112220. [PMID: 32777225 DOI: 10.1016/j.yexcr.2020.112220] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/03/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
Capn4, a small regulatory subunit of the calpain proteolytic system, functions as a potential tumor promoter in several cancers. However, the biological functions and molecular mechanisms of Capn4 in gastric cancer (GC) remain poorly understood. In the current study, we found that upregulation of Capn4 was detected frequently in GC tissues, and was associated with significantly worse survival among the GC patients. Multivariate analyses revealed that abundance of Capn4 was an independent predictive marker for the poor prognosis of GC. Further, Capn4 knockdown notably suppressed GC invasion and metastasis in vitro. Consistently, a xenograft assay showed that silencing of Capn4 in GC cells suppressed their dissemination to lung tissue in vivo. Moreover, our results indicated that Capn4 promotes gastric cancer metastasis by increasing MMP9 expression, and demonstrated that MMP9 is crucial for the pro-metastasis role of Capn4 in GC cells. Further investigation revealed that Capn4 regulated MMP9 expression via activation of Wnt/β-catenin signaling pathway. Mechanistically, we found that Capn4 can decreased β-catenin ubiquitination to enhance the protein stability of β-catenin in GC cells. Collectively, Capn4 has a central role in gastric cancer metastasis, which could be a potential diagnostic and therapeutic target for GC.
Collapse
Affiliation(s)
- Chuanwen Zhao
- Department of General Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi 332000, China
| | - Guohui Yuan
- Department of Hepatopancreatobiliary Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi 332000, China
| | - Yuemei Jiang
- Department of prosthodontics, The Affiliated Stomatological Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Jianfeng Xu
- Department of Hepatopancreatobiliary Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi 332000, China
| | - Lin Ye
- Department of General Surgery, Jiujiang First People's Hospital, Jiujiang, Jiangxi 332000, China
| | - Wenhui Zhan
- Department of Maxillofacial Surgery, The Affiliated Stomatological Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Junfu Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
9
|
The Role of Matrix Metalloproteinase-2 Expression in Gastric Cancer Susceptibility: A Systematic Review. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2019. [DOI: 10.5812/ijcm.94185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
10
|
Tafrihi M, Golbabaei M, Shokrzadeh M. Association of the −181 G→A polymorphism in the MMP-7 gene promoter and gastric cancer: A case-control study. Meta Gene 2019. [DOI: 10.1016/j.mgene.2019.100594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
11
|
Li Y, Sun Q, Jiang M, Li S, Zhang J, Xu Z, Guo D, Gu T, Wang B, Xiao L, Zhou T, Zhuo W. KLF9 suppresses gastric cancer cell invasion and metastasis through transcriptional inhibition of MMP28. FASEB J 2019; 33:7915-7928. [PMID: 30913394 DOI: 10.1096/fj.201802531r] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Gastric cancer (GC) is among the most lethal human malignancies, and the leading cause of GC mortality is metastasis. However, the precise mechanism of GC metastasis remains unclear. To screen key transcriptional factors (TFs) involved in GC metastasis, we performed bioinformatics analysis of The Cancer Genome Atlas database and found that Krüppel-like factor 9 (KLF9) is a GC metastasis-associated TF. KLF9 is significantly decreased in patients with GC with distant metastasis compared with those patients without distant metastasis. Ectopic expression of KLF9 evidently inhibited the migration and invasion capabilities of GC cells. Conversely, knockdown of KLF9 endowed GC cells with stronger invasive capacity. Moreover, tail intravenous injection confirmed that KLF9 strongly inhibits the lung metastasis process of GC in vivo. Mechanistically, chromatin immunoprecipitation coupled with high-throughput sequencing data from Encyclopedia of DNA Elements revealed that KLF9 specifically binds to the promoter region of matrix metalloproteinase (MMP)28. Further quantitative real-time PCR and dual-luciferase assay indicated that KLF9 directly inhibited MMP28 transcription. Importantly, decreased invasion and metastasis capability of GC cells caused by ectopic KLF9 expression could be rescued via reinforcing MMP28 expression in vivo. Collectively, our study indicates that KLF9 significantly suppresses GC cell invasion and metastasis through inhibiting MMP28 transcription.-Li, Y., Sun, Q., Jiang, M., Li, S., Zhang, J., Xu, Z., Guo, D., Gu, T., Wang, B., Xiao, L., Zhou, T., Zhuo, W. KLF9 suppresses gastric cancer cell invasion and metastasis through transcriptional inhibition of MMP28.
Collapse
Affiliation(s)
- Yang Li
- College of Animal Science, Zhejiang University, Hangzhou, China
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Sun
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingchun Jiang
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuang Li
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiayu Zhang
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhangqi Xu
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongyang Guo
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianning Gu
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Boya Wang
- Department of Pharmacy, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xiao
- College of Animal Science, Zhejiang University, Hangzhou, China
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Wei Zhuo
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
12
|
Huang YK, Kang WM, Ma ZQ, Liu YQ, Zhou L, Yu JC. NUCKS1 promotes gastric cancer cell aggressiveness by upregulating IGF-1R and subsequently activating the PI3K/Akt/mTOR signaling pathway. Carcinogenesis 2018; 40:370-379. [PMID: 30371738 DOI: 10.1093/carcin/bgy142] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/17/2018] [Indexed: 01/13/2023] Open
Affiliation(s)
- Ya-Kai Huang
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, P.R. China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Wei-Ming Kang
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Zhi-Qiang Ma
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Yu-Qin Liu
- Cell Culture Centre, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P.R. China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| | - Jian-Chun Yu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, P.R. China
| |
Collapse
|
13
|
Zhang S, Zhang H, Yu L. HMGA2 promotes glioma invasion and poor prognosis via a long-range chromatin interaction. Cancer Med 2018; 7:3226-3239. [PMID: 29733521 PMCID: PMC6051173 DOI: 10.1002/cam4.1534] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/27/2018] [Accepted: 04/09/2018] [Indexed: 12/15/2022] Open
Abstract
To identify the function and underlying mechanisms of HMGA2 on the prognosis and invasion of gliomas, HMGA2 was detected by immunohistochemistry. The Kaplan‐Meier and Cox's regression analysis results showed that higher HMGA2 level predicted the poorer outcomes of glioma patients. ChIP‐qPCR, DNA electrophoretic mobility shift assay, chromosome conformation capture, and co‐immunoprecipitation were applied to identify HMGA2‐activated target sites, which were further verified by mRNA and protein expression detection. Transwell and orthotopic implantation were used to investigate the roles of HMGA2 in glioma cells. HMGA2 shRNA transfection inhibited glioblastoma invasion. Mechanistically, we first discovered that HMGA2, together with GCN5, facilitated the invasion of glioma cells via inducing chromatin conformational remodeling of the MMP2 gene promoter and epigenetically activating MMP2 gene transcription. Our results indicated that HMGA2, as a novel GCN5 recognition partner and histone acetylation modulator, may be novel prognostic indicator and promising glioma treatment target.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huibian Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lin Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
14
|
Winerdal ME, Krantz D, Hartana CA, Zirakzadeh AA, Linton L, Bergman EA, Rosenblatt R, Vasko J, Alamdari F, Hansson J, Holmström B, Johansson M, Winerdal M, Marits P, Sherif A, Winqvist O. Urinary Bladder Cancer Tregs Suppress MMP2 and Potentially Regulate Invasiveness. Cancer Immunol Res 2018; 6:528-538. [DOI: 10.1158/2326-6066.cir-17-0466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 11/13/2017] [Accepted: 03/13/2018] [Indexed: 11/16/2022]
|
15
|
Fayyad A, Lapp S, Risha E, Pfankuche VM, Rohn K, Barthel Y, Schaudien D, Baumgärtner W, Puff C. Matrix metalloproteinases expression in spontaneous canine histiocytic sarcomas and its xenograft model. Vet Immunol Immunopathol 2018; 198:54-64. [PMID: 29571518 DOI: 10.1016/j.vetimm.2018.03.002] [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] [Received: 10/24/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 12/31/2022]
Abstract
Canine histiocytic sarcoma (HS) represents a malignant neoplastic disorder often with a rapid and progressive clinical course. A better understanding of the interaction between tumor cells and the local microenvironment may provide new insights into mechanisms of tumor growth and metastasis. The influence of matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) on tumor angiogenesis, invasion and metastasis has been detailed in previous studies. In addition, inflammatory cells infiltrating neoplasms especially tumor associated macrophages (TAM) may contribute significantly to tumor progression. Due to the high variability of spontaneously occurring canine HS, standardized models are highly required to investigate tumor progression and interaction with its microenvironment. Therefore, the present study comparatively characterized the intratumoral macrophage infiltration as well as the expression of MMP-2, MMP-9, MMP-14 and TIMP-1 in spontaneous canine HS and its murine model. In spontaneous canine HS, scattered MAC 387-positive macrophages were randomly found in tumor center and periphery, whereas tumor cells were negative for this marker. Interestingly, quantitative analysis revealed that MMPs and TIMP-1 were mainly expressed at the invasive front while tumor centers exhibited significantly reduced immunoreactivity. Similar findings were obtained in xenotransplanted HS. Interestingly, murine tumor associated macrophages (TAM), characterized by Mac3 expression (CD107b/LAMP2), which was not present in xenotransplanted histiocytic sarcoma cells, strongly express MMPs and TIMP-1. In addition, MMPs are known to regulate angiogenesis and a positive correlation between MMP-14 expression and microvessel density was demonstrated in xenotransplanted histiocytic sarcomas. Summarized similar findings with respect to MMP and TIMP distribution and the role of macrophages in spontaneously-occurring and xenotransplanted HS indicate the high suitability of this murine model to further investigate HS under standardized conditions. Moreover results indicate that MMP expression contributes to tumor progression and invasion and TAMs seem to be major players in the interaction between neoplastic cells, the microenvironment and vessel formation indicating that therapeutic approaches modulating TAM associated molecules might represent promising future treatment options.
Collapse
Affiliation(s)
- Adnan Fayyad
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Stefanie Lapp
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Engy Risha
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany; Clinical Pathology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Vanessa M Pfankuche
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Karl Rohn
- Institute for Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Bünteweg 2, 30559, Hannover, Germany
| | - Yvonne Barthel
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany
| | - Christina Puff
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
| |
Collapse
|
16
|
Microbial Antigens Stimulate Metalloprotease-7 Secretion in Human B-Lymphocytes Using mTOR-Dependent and Independent Pathways. Sci Rep 2017. [PMID: 28634370 PMCID: PMC5478602 DOI: 10.1038/s41598-017-04199-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Metalloproteinases (MMPs) contribute to tissue remodeling and acute inflammation not only by degrading extracellular matrix proteins but also by controlling the influx of chemokines through the regulation and shedding of syndecans. B-lymphocytes, in addition to their well-known function as antibody producing cells, participate in the innate immune response by secreting inflammatory cytokines and chemokines. However, there is little information about the role of B-lymphocytes in the regulation of MMPs; consequently, herein we investigated whether activated human circulating B-lymphocytes contributed to the secretion of MMPs. We demonstrate that B-lymphocytes activated by un-methylated CpG motifs, found in bacterial DNA, and β-glucans, found in the cell wall of fungi, both induced MMP-7. Interestingly, while CpG-stimulated cells activated the mTOR pathway via TLR9 receptor to induced MMP-7, β-glucan-stimulated cells were mTOR-independent and used Dectin-1 receptor. B-lymphocytes did not seem to have a major role in the secretion of tissue inhibitors of metalloproteinases (TIMPs). However, secreted MMP-7 participated in the shedding of Syndecan-4 from the surface of B-lymphocytes. In conclusion, circulating human B-lymphocytes contribute to the regulation of the innate immune system by participating in the secretion of MMP-7 which in turn is important for the shedding of Syndecan-4 in response to infectious stimuli.
Collapse
|
17
|
Milovancev M, Russell DS. Surgical margins in the veterinary cancer patient. Vet Comp Oncol 2017; 15:1136-1157. [PMID: 28194921 DOI: 10.1111/vco.12284] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
In veterinary oncologic specimens, histopathology is the gold standard for determining adequacy of excision. Despite limitations of this technique, the pathologist's interpretation of margin status significantly impacts patient management, including indications for adjuvant therapy. This article aims to summarize peer-reviewed literature as it relates to histologic margin evaluation in veterinary cancer patients. The value of histologic tumour-free margins and technical factors influencing histopathologic margin outcomes are also discussed. We review alternative strategies for determining excisional status, and discuss how an evolving understanding of tumour biology might inform clinical and research perspectives on surgical margins. In doing so, we aim to provide context and a stimulus for future investigations into this important yet incompletely understood topic.
Collapse
Affiliation(s)
- M Milovancev
- Department of Veterinary Clinical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - D S Russell
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
18
|
Isaacson KJ, Martin Jensen M, Subrahmanyam NB, Ghandehari H. Matrix-metalloproteinases as targets for controlled delivery in cancer: An analysis of upregulation and expression. J Control Release 2017; 259:62-75. [PMID: 28153760 DOI: 10.1016/j.jconrel.2017.01.034] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/18/2017] [Accepted: 01/26/2017] [Indexed: 02/07/2023]
Abstract
While commonly known for degradation of the extracellular matrix, matrix metalloproteinases (MMPs) exhibit broad potential for use in targeting of bioactive and imaging agents in cancer treatment. MMPs are upregulated at all stages of expression in cancers. A comprehensive analysis of published literature on expression of all MMP subtypes at the genetic, protein, and activity levels in normal and diseased tissues indicate targeting applicability in a variety of cancers. This expression significantly increases at advanced cancer stages, providing an improved opportunity for controlled release in higher-stage patients. Since MMPs are integral at every stage of metastasis, MMP roles in cancer are discussed with a focus on MMP distribution and mobility within cells and tumors for cancer targeting applications. Several strategies for MMP utilization in targeting - such as matrix degradation, MMP cleavage, MMP binding, and MMP-induced environmental changes - are addressed.
Collapse
Affiliation(s)
- Kyle J Isaacson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - M Martin Jensen
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Nithya B Subrahmanyam
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA
| | - Hamidreza Ghandehari
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|