1
|
Jo Y, Sim HI, Yun B, Park Y, Jin HS. Revisiting T-cell adhesion molecules as potential targets for cancer immunotherapy: CD226 and CD2. Exp Mol Med 2024; 56:2113-2126. [PMID: 39349829 PMCID: PMC11541569 DOI: 10.1038/s12276-024-01317-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/26/2024] [Accepted: 07/04/2024] [Indexed: 10/03/2024] Open
Abstract
Cancer immunotherapy aims to initiate or amplify immune responses that eliminate cancer cells and create immune memory to prevent relapse. Immune checkpoint inhibitors (ICIs), which target coinhibitory receptors on immune effector cells, such as CTLA-4 and PD-(L)1, have made significant strides in cancer treatment. However, they still face challenges in achieving widespread and durable responses. The effectiveness of anticancer immunity, which is determined by the interplay of coinhibitory and costimulatory signals in tumor-infiltrating immune cells, highlights the potential of costimulatory receptors as key targets for immunotherapy. This review explores our current understanding of the functions of CD2 and CD226, placing a special emphasis on their potential as novel agonist targets for cancer immunotherapy. CD2 and CD226, which are present mainly on T and NK cells, serve important functions in cell adhesion and recognition. These molecules are now recognized for their costimulatory benefits, particularly in the context of overcoming T-cell exhaustion and boosting antitumor responses. The importance of CD226, especially in anti-TIGIT therapy, along with the CD2‒CD58 axis in overcoming resistance to ICI or chimeric antigen receptor (CAR) T-cell therapies provides valuable insights into advancing beyond the current barriers of cancer immunotherapy, underscoring their promise as targets for novel agonist therapy.
Collapse
Affiliation(s)
- Yunju Jo
- Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Hye-In Sim
- Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Bohwan Yun
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yoon Park
- Chemical and Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea.
| | - Hyung-Seung Jin
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| |
Collapse
|
2
|
Shen K, Chen B, Gao W. Integrated single-cell RNA sequencing analysis reveals a mesenchymal stem cell-associated signature for estimating prognosis and drug sensitivity in gastric cancer. J Cancer Res Clin Oncol 2023; 149:11829-11847. [PMID: 37410142 DOI: 10.1007/s00432-023-05058-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/28/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) play an important role in regulating all stages of the immune response, angiogenesis, and transformation of matrix components in the tumor microenvironment. The aim of this study was to identify the prognostic value of MSC-related signatures in patients with gastric cancer (GC). METHODS MSC marker genes were identified by analyzing single-cell RNA sequencing (scRNA-seq) data for GC from the Gene Expression Omnibus (GEO) database. Using bulk sequencing data from the Cancer Genome Atlas-Stomach adenocarcinoma (TCGA-STAD), as a training cohort, and data from GEO, as a validation cohort, we developed a risk model consisting of MSC prognostic signature genes, and classified GC patients into high- and low-MSC risk subgroups. Multifactorial Cox regression was used to evaluate whether MSC prognostic signature was an independent prognostic factor. An MSC nomogram was constructed combining clinical information and risk grouping. Subsequently, we evaluated the effect of MSC prognostic signature on immune cell infiltration, antitumor drugs and immune checkpoints and verified the expression of MSC prognostic signature by in vitro cellular assays. RESULTS In this study, 174 MSC marker genes were identified by analyzing scRNA-seq data. We identified seven genes (POSTN, PLOD2, ITGAV, MMP11, SDC2, MARCKS, ANXA5) to construct MSC prognostic signature. MSC prognostic signature was an independent risk factor in the TCGA and GEO cohorts. GC patients in the high-MSC risk group had worse prognoses. In addition, the MSC nomogram has a high clinical application value. Notably, the MSC signature can induce the development of a poor immune microenvironment. GC patients in the high MSC-risk group were more sensitive to anticancer drugs and tended to have higher levels of immune checkpoint markers. In qRT-PCR assays, the MSC signature was more highly expressed in GC cell lines. CONCLUSIONS The MSC marker gene-based risk signature developed in this study can not only be used to predict the prognosis of GC patients, but also has the potential to reflect the efficacy of antitumor therapies.
Collapse
Affiliation(s)
- Kaiyu Shen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Binyu Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wencang Gao
- Department of Oncology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| |
Collapse
|
3
|
de Joode K, van de Geer WS, van Leenders GJLH, Hamberg P, Westgeest HM, Beeker A, Oosting SF, van Rooijen JM, Beerepoot LV, Labots M, Mathijssen RHJ, Lolkema MP, Cuppen E, Sleijfer S, van de Werken HJG, van der Veldt AAM. The genomic and transcriptomic landscape of advanced renal cell cancer for individualized treatment strategies. Sci Rep 2023; 13:10720. [PMID: 37400554 DOI: 10.1038/s41598-023-37764-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
Differences in the clinical course and treatment responses in individual patients with advanced renal cell carcinoma (RCC) can largely be explained by the different genomics of this disease. To improve the personalized treatment strategy and survival outcomes for patients with advanced RCC, the genomic make-up in patients with advanced RCC was investigated to identify putative actionable variants and signatures. In this prospective multicenter study (NCT01855477), whole-genome sequencing (WGS) data of locally advanced and metastatic tissue biopsies and matched whole-blood samples were collected from 91 patients with histopathologically confirmed RCC. WGS data were analyzed for small somatic variants, copy-number alterations and structural variants. For a subgroup of patients, RNA sequencing (RNA-Seq) data could be analyzed. RNA-Seq data were clustered on immunogenic and angiogenic gene expression patterns according to a previously developed angio-immunogenic gene signature. In all patients with papillary and clear cell RCC, putative actionable drug targets were detected by WGS, of which 94% were on-label available. RNA-Seq data of clear cell and papillary RCC were clustered using a previously developed angio-immunogenic gene signature. Analyses of driver mutations and RNA-Seq data revealed clear differences among different RCC subtypes, showing the added value of WGS and RNA-Seq over clinicopathological data. By improving both histological subtyping and the selection of treatment according to actionable targets and immune signatures, WGS and RNA-Seq may improve therapeutic decision making for most patients with advanced RCC, including patients with non-clear cell RCC for whom no standard treatment is available to data. Prospective clinical trials are needed to evaluate the impact of genomic and transcriptomic diagnostics on survival outcome for advanced RCC patients.
Collapse
Affiliation(s)
- K de Joode
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - W S van de Geer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Internal Postal Address NA-1218, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | - P Hamberg
- Department of Internal Medicine, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | - H M Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands
| | - A Beeker
- Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - S F Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J M van Rooijen
- Department of Internal Medicine, Martini Hospital, Groningen, The Netherlands
| | - L V Beerepoot
- Department of Internal Medicine, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - M Labots
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - R H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - M P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - E Cuppen
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
- Hartwig Medical Foundation, Amsterdam, The Netherlands
| | - S Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Center for Personalized Cancer Treatment, Rotterdam, The Netherlands
| | - H J G van de Werken
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Internal Postal Address NA-1218, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands.
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands.
| | - A A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- Departments of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Kwon MJ. Matrix metalloproteinases as therapeutic targets in breast cancer. Front Oncol 2023; 12:1108695. [PMID: 36741729 PMCID: PMC9897057 DOI: 10.3389/fonc.2022.1108695] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/28/2022] [Indexed: 01/22/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are the most prominent proteinases involved in tumorigenesis. They were initially recognized to promote tumor progression by remodeling the extracellular matrix through their proteolytic activity. However, accumulating evidence has revealed that some MMPs have protective roles in cancer progression, and the same MMP can exert opposing roles depending on the cell type in which it is expressed or the stage of cancer. Moreover, studies have shown that MMPs are involved in cancer progression through their roles in other biological processes such as cell signaling and immune regulation, independent of their catalytic activity. Despite the prognostic significance of tumoral or stromal expression of MMPs in breast cancer, their roles and molecular mechanisms in breast cancer progression remain unclear. As the failures of early clinical trials with broad-spectrum MMP inhibitors were mainly due to a lack of drug specificity, substantial efforts have been made to develop highly selective MMP inhibitors. Some recently developed MMP inhibitory monoclonal antibodies demonstrated promising anti-tumor effects in preclinical models of breast cancer. Importantly, anti-tumor effects of these antibodies were associated with the modulation of tumor immune microenvironment, suggesting that the use of MMP inhibitors in combination with immunotherapy can improve the efficacy of immunotherapy in HER2-positive or triple-negative breast cancer. In this review, the current understanding of the roles of tumoral or stromal MMPs in breast cancer is summarized, and recent advances in the development of highly selective MMP inhibitors are discussed.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- Vessel-Organ Interaction Research Center (MRC), College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea,BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea,*Correspondence: Mi Jeong Kwon,
| |
Collapse
|
5
|
Martin‐Morales L, Manzano S, Rodrigo‐Faus M, Vicente‐Barrueco A, Lorca V, Núñez‐Moreno G, Bragado P, Porras A, Caldes T, Garre P, Gutierrez‐Uzquiza A. Germline gain-of-function MMP11 variant results in an aggressive form of colorectal cancer. Int J Cancer 2023; 152:283-297. [PMID: 36093604 PMCID: PMC9827992 DOI: 10.1002/ijc.34289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 01/12/2023]
Abstract
Matrix metalloproteinase-11 (MMP11) is an enzyme with proteolytic activity against matrix and nonmatrix proteins. Although most MMPs are secreted as inactive proenzymes and are later activated extracellularly, MMP11 is activated intracellularly by furin within the constitutive secretory pathway. It is a key factor in physiological tissue remodeling and its alteration may play an important role in the progression of epithelial malignancies and other diseases. TCGA colon and colorectal adenocarcinoma data showed that upregulation of MMP11 expression correlates with tumorigenesis and malignancy. Here, we provide evidence that a germline variant in the MMP11 gene (NM_005940: c.232C>T; p.(Pro78Ser)), identified by whole exome sequencing, can increase the tumorigenic properties of colorectal cancer (CRC) cells. P78S is located in the prodomain region, which is responsible for blocking MMP11's protease activity. This variant was detected in the proband and all the cancer-affected family members analyzed, while it was not detected in healthy relatives. In silico analyses predict that P78S could have an impact on the activation of the enzyme. Furthermore, our in vitro analyses show that the expression of P78S in HCT116 cells increases tumor cell invasion and proliferation. In summary, our results show that this variant could modify the structure of the MMP11 prodomain, producing a premature or uncontrolled activation of the enzyme that may contribute to an early CRC onset in these patients. The study of this gene in other CRC cases will provide further information about its role in CRC development, which might improve patient treatment in the future.
Collapse
Affiliation(s)
- Lorena Martin‐Morales
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Laboratory of Cancer Stemness, GIGA‐InstituteUniversity of LiegeLiegeBelgium
| | - Sara Manzano
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain,Biodonostia Health Research InstituteSan Sebastian/DonostiaSpain
| | - Maria Rodrigo‐Faus
- Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Adrian Vicente‐Barrueco
- Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain,Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Victor Lorca
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Gonzalo Núñez‐Moreno
- Department of Genetics, Health Research Institute‐Fundación Jiménez Díaz University HospitalUniversidad Autónoma de Madrid (IIS‐FJD, UAM)MadridSpain,Bioinformatics Unit, Health Research Institute‐Fundación Jiménez Díaz University HospitalUniversidad Autónoma de Madrid (IIS‐FJD, UAM)MadridSpain
| | - Paloma Bragado
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Almudena Porras
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Trinidad Caldes
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Pilar Garre
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Clinical Analysis Service, Molecular Diagnostic UnitIML, Hospital Clínico San CarlosMadridSpain
| | - Alvaro Gutierrez‐Uzquiza
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| |
Collapse
|
6
|
Shao X, Zheng Y, Huang Y, Li G, Zou W, Shi L. Hsa-miR-221-3p promotes proliferation and migration in HER2-positive breast cancer cells by targeting LASS2 and MBD2. Histol Histopathol 2022; 37:1099-1112. [PMID: 35734966 DOI: 10.14670/hh-18-483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Human epidermal growth factor receptor (HER2)-positive breast cancers account for nearly 20% of all breast cancer cases and microRNAs (miRNAs) play crucial roles in disease progression. The study was aimed to explore the role of miR-221-3p in HER2-positive breast cancer. METHODS Differentially expressed miRNAs were identified by high-throughput sequencing. Quantitative real-time PCR was used to evaluate mRNA levels of corresponding genes. CKK8 and transwell assays were performed to evaluate cell viability and migration. The translation binding was assessed by luciferase assay. RESULTS Hsa-miR-221-3p was highly upregulated in HER2-positive breast cancer samples, particularly in patients with advanced or metastatic disease, as compared to healthy controls. miR-221-3p upregulation using mimics promoted cell proliferation and migration in HER2-positive cell lines, whereas miR-221-3p suppression had the opposite effect. Additionally, miR-221-3p mimics reduced the expression levels of LASS2 and MBD2 in HER2-positive breast cancer cells; conversely, miR-221-3p inhibition upregulated LASS2 and MBD2. miR-221-3p inhibited the translation of LASS2 and MBD2 by directly binding to their 3'-untranslated regions. Forced expression of LASS2 and MBD2 significantly attenuated the ability of miR-221-3p mimics to enhance cell growth and migration in HER2-positive but not in HER2-negative breast cancer cells. In HER-2-positive breast cancer patients, the levels of miR-221-3p were negatively correlated with the mRNA levels of LASS2 and MBD2. CONCLUSIONS Upregulation of hsa-miR-221-3 in HER2-positive breast cancer contributes to cancer cell proliferation and migration by directly targeting the tumor suppressors LASS2 and MBD2. Therefore, the hsa-miR-221-3 may serve as a promising and actionable therapeutic target in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Xiying Shao
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Zhejiang, PR China
| | - Yabing Zheng
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Zhejiang, PR China.,Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Zhejiang, PR China.
| | - Yuan Huang
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Zhejiang, PR China
| | - Guangliang Li
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Zhejiang, PR China
| | - Weibin Zou
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Zhejiang, PR China
| | - Lei Shi
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang, PR China.,Department of Breast Medical Oncology, Zhejiang Cancer Hospital, Zhejiang, PR China
| |
Collapse
|
7
|
Lin Y, Zhou H, Li S. BTN3A2 Expression Is Connected With Favorable Prognosis and High Infiltrating Immune in Lung Adenocarcinoma. Front Genet 2022; 13:848476. [PMID: 35873496 PMCID: PMC9298880 DOI: 10.3389/fgene.2022.848476] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Butyrophilin subfamily 3 member A2 (BTN3A2) is an important mediator in immune activation, and it is reported to be linked to many cancer progresses. However, the relation with infiltrating immune and prognosis of BTN3A2 in lung adenocarcinoma are not clear. Methods: In our study, we checked the mRNA expression and protein expression profile of BTN3A2 in lung adenocarcinoma (LUAD) and its relation to clinical outcomes using TIMER and UALCAN databases. In addition, we analyzed the survival of BTN3A2 in LUAD using the Kaplan–Meier Plotter database and PrognoScan database. Moreover, we analyzed gene set enrichment analysis (GSEA) of the BTN3A2. Next, we explored the relation of BTN3A2 expression with the immune infiltration by TIMER. At last, in order to enrich the regulatory mechanism of BTN3A2, we used miRarbase, starbase, and miRDB databases to look for miRNA targets of BTN3A2. Results: The mRNA along with the protein expression of BTN3A2 in the LUAD group was lower than that in the normal group. In addition, high BTN3A2 expression was connected with good first progression (FP) and overall survival (OS) in LUAD. Then, the GSEA analysis demonstrated that T-cell receptor signaling cascade, B-cell receptor signaling cascade, natural killer cell–mediated cytotoxicity, immune receptor activity, immunological synapse, and T-cell activation were enriched differentially in the BTN3A2 high expression phenotype of LUAD. Moreover, BTN3A2 expression is a remarkable positive correlation with invading levels of tumor purity, B cells, neutrophils, CD4+ T cells, dendritic cells, macrophages, and CD8+ T cells in LUAD, and B cells and dendritic cells were linked with a good prognosis of LUAD. To further enrich the possible regulatory mechanisms of BTN3A2, we analyzed the miRNA targets. The results showed that hsa-miR-17-5p may be miRNA targets of BTN3A2. Conclusion: Taking together, we provide evidence of BTN3A2 as possible prognosis biomarkers of LUAD. In addition, high BTN3A2 expression in LUAD may influence the prognosis because of immune invasion. Moreover, our findings provide a potential mechanism that hsa-miR-17-5p may be miRNA targets of BTN3A2.
Collapse
Affiliation(s)
- Yuansheng Lin
- Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Hao Zhou
- Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shengjun Li
- Suzhou Science and Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
8
|
Jin Y, Liang ZY, Zhou WX, Zhou L. An MMP-based risk score strongly distinguishes prognosis in hepatocellular carcinoma after resection. Future Oncol 2022; 18:2903-2917. [PMID: 35861053 DOI: 10.2217/fon-2021-1558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To first explore the prognostic value of MMP11 and MMP15 in hepatocellular carcinoma. Methods: MMP11/MMP15 expression was immunohistochemically detected and correlated with clinicopathologic variables and survival and confirmed in publicly available databases. An MMP-based risk score (MMPRS) was established. Results: Tumoral MMP11/MMP15 expression was higher and univariately associated with crucial clinicopathologic parameters, overall survival and disease-free survival in all patients and/or many subsets. Multivariately, MMP11/MMP15 expression remained significant. Their overexpression and prognostic value were confirmed in the Ualcan and Kaplan-Meier plotter databases. Critically, the novel MMPRS integrating MMP11, MMP15 and tumor-node-metastasis stage identified subgroups with the best and worst prognoses, with much higher predictive power. Conclusion: MMP11 and MMP15 served as prognosticators in hepatocellular carcinoma. MMPRS might work more accurately.
Collapse
Affiliation(s)
- Ye Jin
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100730, China
| |
Collapse
|
9
|
Kang SU, Cho SY, Jeong H, Han J, Chae HY, Yang H, Sung CO, Choi YL, Shin YK, Kwon MJ. Matrix metalloproteinase 11 (MMP11) in macrophages promotes the migration of HER2-positive breast cancer cells and monocyte recruitment through CCL2-CCR2 signaling. J Transl Med 2022; 102:376-390. [PMID: 34775491 DOI: 10.1038/s41374-021-00699-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/15/2021] [Accepted: 10/29/2021] [Indexed: 12/21/2022] Open
Abstract
Matrix metalloproteinase 11 (MMP11), a member of the MMP family involved in the degradation of the extracellular matrix, has been implicated in cancer progression. Despite the stromal expression of MMP11 in breast cancer, the prognostic significance and role of MMP11 in immune or stromal cells of breast cancer remain unclear. Based on the immunohistochemical analysis of breast cancer tissues from 497 patients, we demonstrated that MMP11 expression in mononuclear inflammatory cells (predominantly macrophages) is an independent negative prognostic factor in breast cancer, whereas MMP11 expression in tumor cells and fibroblasts is not associated with patient survival. Enforced MMP11 expression in breast cancer cells did not promote cell proliferation and migration. However, MMP11-overexpressing macrophages enhanced the migration of HER2-positive (HER2+) breast cancer cells, recruitment of monocytes, and tube formation of endothelial cells. Furthermore, we found that the chemokine CCL2 secreted from MMP11-overexpressing macrophages activated the MAPK pathway via its receptor CCR2 in breast cancer cells, thereby promoting the migration of HER2+ breast cancer cells through MMP9 upregulation. We also found that MMP11 expression in macrophages was stimulated by MMP11-overepressing HER2+ breast cancer cells. Collectively, our findings provide evidence that MMP11 in macrophages may play a pro-tumoral role in HER2+ breast cancer through interaction with cancer cells, monocytes, and endothelial cells.
Collapse
Affiliation(s)
- Shin Ung Kang
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Soo Youn Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyojin Jeong
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea.,Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jinil Han
- Gencurix, Inc, Seoul, Republic of Korea
| | - Ha Yeong Chae
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Hobin Yang
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Chang Ohk Sung
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Laboratory of Cancer Genomics and Molecular Pathology, Samsung Biomedical Research Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Mi Jeong Kwon
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea. .,Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
10
|
Gene Expression Profiling in Early Breast Cancer-Patient Stratification Based on Molecular and Tumor Microenvironment Features. Biomedicines 2022; 10:biomedicines10020248. [PMID: 35203458 PMCID: PMC8869155 DOI: 10.3390/biomedicines10020248] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/19/2022] [Indexed: 11/17/2022] Open
Abstract
Patients with early-stage hormone receptor-positive, human epidermal growth factor receptor 2-negative (HER2−) breast cancer (BC) are typically treated with surgery, followed by adjuvant systemic endocrine therapy with or without adjuvant chemotherapy and radiation therapy. Current guidelines regarding the use of adjuvant systemic therapy depend on clinical and pathological factors, such as the morphological assessment of tumor subtype; histological grade; tumor size; lymphovascular invasion; and lymph node status combined with estrogen receptor, progesterone receptor, and HER2 biomarker profiles assessed using immunohistochemistry and in situ hybridization. Additionally, the prognostic and predictive value of tumor-infiltrating lymphocytes and their composition is emerging as a key marker in triple negative (TNBC) and HER2-enriched molecular breast tumor subtypes. However, all these factors do not necessarily reflect the molecular heterogeneity and complexity of breast cancer. In the last two decades, gene expression signatures or profiling (GEP) tests have been developed to predict the risk of disease recurrence and estimate the potential benefit of receiving adjuvant systemic chemotherapy in patients with luminal breast cancer. GEPs have been utilized to help physicians to refine decision-making process, complementing clinicopathological parameters, and can now be used to classify the risk of recurrence and tailoring personalized treatments. Several clinical trials using GEPs validate the increasing value of such assays in different clinical settings, addressing relevant clinical endpoints. Finally, the recent approval of immune checkpoint inhibitors in TNBC and the increasing use of immunotherapy in different molecular BC populations highlight the opportunity to refine current GEPs by including a variety of immune-related genes that may help to improve predicting drug response and finetune prognosis.
Collapse
|
11
|
The tamoxifen-regulated, long non-coding RNA LINC00992 affects proliferation, migration, and expression of tamoxifen resistance-associated genes in MCF-7 breast cancer cells. Contemp Oncol (Pozn) 2022; 26:294-305. [PMID: 36816389 PMCID: PMC9933353 DOI: 10.5114/wo.2023.125000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Tamoxifen-adapted MCF-7 breast cancer cells (MCF-7-TAM-R) are a model for acquired tamoxifen resistance in oestrogen receptor-positive breast cancer. In this system, the expression of long-non-coding RNA LINC00992 is decreased. LINC00992 might therefore contribute to tamoxifen adaption and associated gene expres-sion changes. Here, we investigated whether a modulation of LINC00992 modifies gene expression, proliferation, and migration. Material and methods Up- and down-- regulation of LINC00992 was performed using plasmid vectors and siRNA. Gene expression was measured via nCounter® and quantitative real-time polymerase chain reaction. Database analysis was performed using GEPIA2 and cBioportal. Furthermore, we performed scratch assays, colony-forming assays, and proliferation assays with MCF-7 and MCF-7-TAM-R after up-regulation of LINC00992. Results Up- and down-regulation of LINC00992 caused gene expression changes in 4 of the 42 tamoxifen-regulated genes tested. Especially ubiquitin D, single-minded homologue 1 (SIM1) carcinoembryonic antigen-related cell adhesion molecule 5 and the G-protein coupled oestrogen receptor 1 were affected. In tamoxifen-adapted MCF-7-TAM-R cells, LINC00992 overexpression resulted in augmented viability and proliferation and enhanced migration. Database analyses revealed that luminal breast cancers have increased expression of LINC00992 compared to Her2-type/neu- or basal type. Furthermore, higher expression of LINC00992 was associated with poor prognosis in luminal-A carcinomas. Conclusions Changes in the expression of tamoxifen-regulated genes could be induced by manipulating LINC00992's abundance, suggesting that it is at least partially involved in the establishment of the tamoxifen-induced gene expression pattern. LINC00992 may also serve as a prognostic biomarker and may indicate the development of tamoxifen resistance.
Collapse
|
12
|
Wang T, Zhang Y, Bai J, Xue Y, Peng Q. MMP1 and MMP9 are potential prognostic biomarkers and targets for uveal melanoma. BMC Cancer 2021; 21:1068. [PMID: 34587931 PMCID: PMC8482640 DOI: 10.1186/s12885-021-08788-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/14/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Uveal melanoma (UVM) is the leading cause of eye-related mortality worldwide. This study aimed to explore the expression and prognostic value of matrix metalloproteinases (MMPs) in UVM. METHODS Gene expression levels were obtained from the Gene Expression Omnibus (GEO) and Oncomine databases. Functional and pathway enrichment analyses were performed using the Metascape database. GeneMANIA was then applied to construct a protein-protein interaction network and identify the hub genes. Moreover, overall survival (OS) and disease-free survival (DFS) analysis for the hub genes was performed using the UALCAN and Gene Expression Profiling Interactive Analysis (GEPIA) online tool. Furthermore, TRRUST was used to predict the targets of the MMPs. RESULTS Our results revealed that the transcriptional levels of MMP1, MMP9, MMP10, MMP11, MMP13, MMP14, and MMP17 were upregulated in UVM tissues compared to normal tissues. A protein-protein interaction (PPI) network was constructed and the top 50 hub genes were identified. The functions of MMPs and their neighboring proteins are mainly associated with ECM-receptor interaction, proteoglycans in cancer, the IL-17 signaling pathway, and microRNAs in cancer. Among the MMPs, MMP1/2/9/11/14/15/16/17/24 played significant roles in the progression of UVM from stage 3 to stage 4. We also found that the expression of MMP1, MMP2, MMP9, and MMP16 positively correlated with OS and DFS in patients with UVM. Additionally, 18 transcription factors associated with nine MMPs were identified. CONCLUSIONS The results of this study may provide potential biomarkers and targets for UVM. However, further studies are required to confirm these results.
Collapse
Affiliation(s)
- Tianyu Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yuanyuan Zhang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jianhao Bai
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yawen Xue
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Qing Peng
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
13
|
Chen S, Li Z, Huang W, Wang Y, Fan S. Prognostic and Therapeutic Significance of BTN3A Proteins in Tumors. J Cancer 2021; 12:4505-4512. [PMID: 34149914 PMCID: PMC8210570 DOI: 10.7150/jca.57831] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023] Open
Abstract
The Butyrophilin 3A (BTN3A) family is a type I transmembrane protein belonging to the immunoglobulin (Ig) superfamily. The family contains three members: BTN3A1, BTN3A2 and BTN3A3, which share 95% homology in the extracellular domain. The expression of BTN3A family members is different in different types of tumors, which plays an important role in tumor prognosis. Among them, there are many studies on tumor immunity of BTN3A1, which shows that it is essential for the activation of Vγ9Vδ2 T cells, while BTN3A3 is expected to become a potential therapeutic target for breast cancer. Recent studies have shown that the BTN3A family is closely related to the occurrence and development of tumors. Now the BTN3A family has become one of the research hotspots and is expected to become new tumor prediction and treatment targets.
Collapse
Affiliation(s)
- Sihan Chen
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China.,College of Health Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| | - Zhangyun Li
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| | - Wenyi Huang
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| | - Yanyan Wang
- Department of Ultrasonic Medicine, Xuzhou First People's Hospital, Jiangsu, China
| | - Shaohua Fan
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu, China
| |
Collapse
|
14
|
Kim HS, Kim MG, Min KW, Jung US, Kim DH. High MMP-11 expression associated with low CD8+ T cells decreases the survival rate in patients with breast cancer. PLoS One 2021; 16:e0252052. [PMID: 34038440 PMCID: PMC8153507 DOI: 10.1371/journal.pone.0252052] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Matrix metalloproteinase-11 (MMP-11) promote cancer invasion and metastasis through degrading the extracellular matrix. Protein degradation by MMP-11 in tumor cells may progressively suppress cancer surveillance activities with blocking immune response in breast cancer. The aim of study is to analyze clinicopathological parameters, molecular interactions and anticancer immune response in patients with MMP-11 expression and to provide candidate target drugs. We investigated the clinicopathologic parameters, specific gene sets, tumor antigenicity, and immunologic relevance according to MMP-11 expression in 226 and 776 breast cancer patients from the Hanyang University Guri Hospital (HUGH) cohort and The Cancer Genome Atlas (TCGA) data, respectively. We analyzed pathway networks and in vitro drug response. High MMP-11 expression was associated with worse survival rate in breast cancer from HUGH cohort and TCGA data (all p < 0.05). In analysis of immunologic gene sets, high MMP-11 expression was related to low immune response such as CD8+T cell, CD4+T cell and B cell. In silico cytometry, there was a decrease of cancer testis antigen and low tumor infiltrating lymphocyte in patient with high MMP-11 expression: activated dendritic cell, CD8+T cell, CD4+ memory T cell, and memory B cell. In pathway networks, MMP-11 was linked to the pathways including low immune response, response to growth hormone and catabolic process. We found that pictilisib and AZ960 effectively inhibited the breast cancer cell lines with high MMP-11 expression. Strategies making use of MMP-11-related hub genes could contribute to better clinical management/research for patients with breast cancer.
Collapse
Affiliation(s)
- Hyung Suk Kim
- Division of Breast Surgery, Department of Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Min Gyu Kim
- Department of Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Un Suk Jung
- Department of Obstetrics and Gynecology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
- * E-mail: (USJ); (DHK)
| | - Dong-Hoon Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- * E-mail: (USJ); (DHK)
| |
Collapse
|
15
|
Chen Y, Meng Z, Zhang L, Liu F. CD2 Is a Novel Immune-Related Prognostic Biomarker of Invasive Breast Carcinoma That Modulates the Tumor Microenvironment. Front Immunol 2021; 12:664845. [PMID: 33968066 PMCID: PMC8102873 DOI: 10.3389/fimmu.2021.664845] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/07/2021] [Indexed: 12/18/2022] Open
Abstract
Female breast cancer (BCa) is the most commonly occurring cancer worldwide. The tumor microenvironment (TME) plays an essential role in tumor invasion, angiogenesis, unlimited proliferation, and even immune escape, but we know little about the TME of BCa. In this study, we aimed to find a TME-related biomarker for BCa, especially for invasive breast carcinoma (BRCA), that could predict prognosis and immunotherapy efficacy. Based on RNA-seq transcriptome data and the clinical characteristics of 1222 samples (113 normal and 1109 tumor samples) from The Cancer Genome Atlas (TCGA) database, we used the ESTIMATE algorithm to calculate the ImmuneScore and StromalScore and then identified differentially expressed genes (DEGs) between the high and low ImmuneScore groups and the high and low StromalScore groups. Thereafter, a protein–protein interaction (PPI) network analysis and univariate Cox regression analyses of overall survival were used to identify potential key genes. Five candidate genes were identified, comprising CD2, CCL19, CD52, CD3E, and ITK. Thereafter, we focused on CD2, analyzing CD2 expression and its association with survival. CD2 expression was associated with tumor size (T stage) to some extent, but not with overall TNM stage, lymph node status (N stage), or distant metastasis (M stage). High CD2 expression was associated with longer survival. METABRIC data were used to validate the survival result (n = 276). Gene set enrichment analysis (GSEA) showed that the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways that were significantly associated with high CD2 expression were mainly immune-related pathways. Furthermore, CD2 expression was correlated with 16 types of tumor-infiltrating immune cells (TICs). Hence, CD2 might be a novel biomarker in terms of molecular typing, and it may serve as a complementary approach to TNM staging to improve clinical outcome prediction for BCa patients.
Collapse
Affiliation(s)
- Yanzhu Chen
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhishang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhang
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Feng Liu
- Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
16
|
Zhuang Y, Li X, Zhan P, Pi G, Wen G. MMP11 promotes the proliferation and progression of breast cancer through stabilizing Smad2 protein. Oncol Rep 2021; 45:16. [PMID: 33649832 PMCID: PMC7876999 DOI: 10.3892/or.2021.7967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/08/2020] [Indexed: 11/17/2022] Open
Abstract
Breast cancer (BC) is one of the most common malignant tumours in women. The matrix metalloproteinase (MMP) enzyme family plays a complex role in the development of BC. There is increasing evidence that MMP11 plays a major role in BC; however, the underlying mechanisms are not clear. The present study confirmed by analysing clinical samples and TCGA data sets, that high expression of MMP11 in clinical samples of BC was strongly associated with a poor prognosis in BC patients. In addition, MTT and colony formation assays indicated that the proliferative capacity of BC was affected when MMP11 expression changed. Furthermore, pathway enrichment analysis was performed and it was revealed that the TGF‑β signalling pathway was a potential downstream target of MMP11. In the TGF‑β signalling pathway, MMP11 could significantly regulate the protein expression levels of Smad2 and Smad3 and inhibit the degradation of Smad2 through the ubiquitin proteasome pathway as determined by western blotting. In vivo, it was further verified that MMP11 knockdown could inhibit tumour proliferation and growth. Collectively, the present results demonstrated that MMP11 inhibited the degradation of Smad2 in the TGF‑β signalling pathway, thereby promoting the development of BC. Thus, MMP11 expression was not only revealed to be an important indicator of BC prognosis but may also be an important therapeutic target for further prevention of BC growth and proliferation. The present study indicated that MMP11‑targeted therapy may provide new solutions for BC treatment.
Collapse
Affiliation(s)
- Ying Zhuang
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Xiang Li
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Peng Zhan
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Guoliang Pi
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| | - Gu Wen
- Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, P.R. China
| |
Collapse
|
17
|
Wu HJ, Chu PY. Recent Discoveries of Macromolecule- and Cell-Based Biomarkers and Therapeutic Implications in Breast Cancer. Int J Mol Sci 2021; 22:ijms22020636. [PMID: 33435254 PMCID: PMC7827149 DOI: 10.3390/ijms22020636] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer type and the leading cause of cancer-related mortality in women worldwide. Breast cancer is fairly heterogeneous and reveals six molecular subtypes: luminal A, luminal B, HER2+, basal-like subtype (ER−, PR−, and HER2−), normal breast-like, and claudin-low. Breast cancer screening and early diagnosis play critical roles in improving therapeutic outcomes and prognosis. Mammography is currently the main commercially available detection method for breast cancer; however, it has numerous limitations. Therefore, reliable noninvasive diagnostic and prognostic biomarkers are required. Biomarkers used in cancer range from macromolecules, such as DNA, RNA, and proteins, to whole cells. Biomarkers for cancer risk, diagnosis, proliferation, metastasis, drug resistance, and prognosis have been identified in breast cancer. In addition, there is currently a greater demand for personalized or precise treatments; moreover, the identification of novel biomarkers to further the development of new drugs is urgently needed. In this review, we summarize and focus on the recent discoveries of promising macromolecules and cell-based biomarkers for the diagnosis and prognosis of breast cancer and provide implications for therapeutic strategies.
Collapse
Affiliation(s)
- Hsing-Ju Wu
- Department of Biology, National Changhua University of Education, Changhua 500, Taiwan;
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
- Department of Medical Research, Chang Bing Show Chwan Memorial Hospital, Lukang Town, Changhua County 505, Taiwan
| | - Pei-Yi Chu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 231, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, No. 542, Sec. 1 Chung-Shan Rd., Changhua 500, Taiwan
- Department of Health Food, Chung Chou University of Science and Technology, Changhua 510, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
- Correspondence: ; Tel.: +886-975-611-855; Fax: +886-4-7227-116
| |
Collapse
|
18
|
Lee H, Kwon MJ, Koo BM, Park HG, Han J, Shin YK. A novel immune prognostic index for stratification of high-risk patients with early breast cancer. Sci Rep 2021; 11:128. [PMID: 33420250 PMCID: PMC7794340 DOI: 10.1038/s41598-020-80274-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022] Open
Abstract
The prognostic value of current multigene assays for breast cancer is limited to hormone receptor-positive, human epidermal growth factor receptor 2-negative early breast cancer. Despite the prognostic significance of immune response-related genes in breast cancer, immune gene signatures have not been incorporated into most multigene assays. Here, using public gene expression microarray datasets, we classified breast cancer patients into three risk groups according to clinical risk and proliferation risk. We then developed the immune prognostic index based on expression of five immune response-related genes (TRAT1, IL2RB, CTLA4, IGHM and IL21R) and lymph node status to predict the risk of recurrence in the clinical and proliferation high-risk (CPH) group. The 10-year probability of disease-free survival (DFS) or distant metastasis-free survival (DMFS) of patients classified as high risk according to the immune prognostic index was significantly lower than those of patients classified as intermediate or low risk. Multivariate analysis revealed that the index is an independent prognostic factor for DFS or DMFS. Moreover, the C-index revealed that it is superior to clinicopathological variables for predicting prognosis. Its prognostic significance was also validated in independent datasets. The immune prognostic index identified low-risk patients among patients classified as CPH, regardless of the molecular subtype of breast cancer, and may overcome the limitations of current multigene assays.
Collapse
Affiliation(s)
- Hannah Lee
- Interdisciplinary Program in Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, 41566, Republic of Korea
| | - Mi Jeong Kwon
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.,Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Beom-Mo Koo
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hee Geon Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinil Han
- Gencurix, Inc., Seoul, 08394, Republic of Korea
| | - Young Kee Shin
- Interdisciplinary Program in Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, 41566, Republic of Korea. .,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea. .,Laboratory of Molecular Pathology and Cancer Genomics, Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
19
|
Sathyanarayanan A, Natarajan A, Paramasivam OR, Gopinath P, Gopal G. Comprehensive analysis of genomic alterations, clinical outcomes, putative functions and potential therapeutic value of MMP11 in human breast cancer. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
20
|
Li Z, Chen Z, Hu G, Zhang Y, Feng Y, Jiang Y, Wang J. Profiling and integrated analysis of differentially expressed circRNAs as novel biomarkers for breast cancer. J Cell Physiol 2020; 235:7945-7959. [PMID: 31943203 DOI: 10.1002/jcp.29449] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022]
Abstract
Breast cancer (BC) is a globally common cancer with the highest and increasing morbidity and mortality among females. Novel biomarkers are warranted to be discovered for the early detection, treatment, and prognosis of BC. In this study, we investigated the profiles of differentially expressed (DE) circular RNAs (circRNAs) by competing endogenous RNAs (ceRNA) microarray to construct a genome-wide circRNA profile. Then, we performed Gene Ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analysis of the host genes (HGs) of circRNAs. A total of 4,370 DE circRNAs were detected and GO and KEGG analysis showed that they were significantly associated with cell cycle, DNA replication, BC, and familial BC. We validated the differential circRNAs and relevant HGs through quantitative real-time polymerase chain reaction and constructed a putative circRNA-microRNA-messenger RNA regulatory network. Eight circRNAs, including hsa_circ_0069094, hsa_circ_0062558, hsa_circ_0074026, hsa_circ_0079876, hsa_circ_0017536, hsa_circ_0023302, hsa_circ_0017650, and hsa_circ_0017545, were validated significantly DE in BC tissue and associated with TNM staging, lymph node infiltration, and Ki67. Hsa_circ_0069094, hsa_circ_0079876, hsa_circ_0017650, and hsa_circ_0017526 were upregulated in plasma. This study revealed the general expression characteristics of specific DE circRNAs in BC and hsa_circ_0069094, hsa_circ_0079876, hsa_circ_0017650, and hsa_circ_0017526 might be promising candidate targets.
Collapse
Affiliation(s)
- Zehuan Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhanghan Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guohua Hu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanling Feng
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ying Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jin Wang
- Scientific Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Wu H, Zhang XY, Niu M, Li FF, Gao S, Wei W, Li SW, Zhang XD, Liu SL, Pang D. Isobaric Tags for Relative and Absolute Quantitation in Proteomic Analysis of Potential Biomarkers in Invasive Cancer, Ductal Carcinoma In Situ, and Mammary Fibroadenoma. Front Oncol 2020; 10:574552. [PMID: 33194682 PMCID: PMC7640741 DOI: 10.3389/fonc.2020.574552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Breast malignancy is a serious threat to women's health around the world. Following the rapid progress in the field of cancer diagnostics and identification of pathological markers, breast tumor treatment methods have been greatly improved. However, for invasive, ductal carcinomas and mammary fibroadenoma, there is an urgent demand for better breast tumor-linked biomarkers. The current study was designed to identify diagnostic and/or therapeutic protein biomarkers for breast tumors. METHODS A total of 140 individuals were included, comprising 35 healthy women, 35 invasive breast cancers (IBC), 35 breast ductal carcinomas in situ (DCIS), and 35 breast fibroadenoma patients. Isobaric tags for relative and absolute quantitation (iTRAQ) proteomic analysis was employed to characterize differentially expressed proteins for potential biomarkers in IBC, DCIS, and fibroadenomas by comparisons with their matched adjacent tissues and/or normal breast tissues. The public databases Metascape and String were used for bioinformatic analyses. RESULTS Using the proteomics approach, we identified differentially expressed proteins in tissues of different breast tumors compared to normal/adjacent breast tissues, including 100 in IBC, 52 in DCIS, and 44 in fibroadenoma. Among the 100 IBC differentially expressed proteins, 37 were found to be specific to this type of cancer only. Additionally, four proteins were specifically expressed in DCIS and four in fibroadenoma. Compared to corresponding adjacent tissues and normal breast tissues, 18 step-changing proteins were differentially expressed in IBC, 14 in DCIS, and 13 in fibroadenoma, respectively. Compared to DCIS and normal breast tissues, 65 proteins were differentially expressed in IBC with growing levels of malignancy. CONCLUSIONS The identified potential protein biomarkers may be used as diagnostic and/or therapeutic targets in breast tumors.
Collapse
Affiliation(s)
- Hao Wu
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xian-Yu Zhang
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ming Niu
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fei-Feng Li
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Song Gao
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei Wei
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Si-Wei Li
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xing-Da Zhang
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shu-Lin Liu
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- HMU-UCCSM Centre for Infection and Genomics, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | - Da Pang
- Genomics Research Center, College of Pharmacy, State-Province Laboratory of Biomedicine and Pharmaceutics of China, Harbin Medical University, Harbin, China
- Sino-Russian Medical Research Center, Harbin Medical University Cancer Hospital, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, China
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
22
|
Feng Q, Hu Q, Liu Y, Yang T, Yin Z. Diagnosis of triple negative breast cancer based on radiomics signatures extracted from preoperative contrast-enhanced chest computed tomography. BMC Cancer 2020; 20:579. [PMID: 32571245 PMCID: PMC7309976 DOI: 10.1186/s12885-020-07053-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Background To explore the diagnostic value of radiomics features of preoperative computed tomography (CT) for triple negative breast cancer (TNBC) for better treatment of patients with breast cancer. Methods A total of 890 patients with breast cancer admitted to our hospital from June 2016 to January 2018 were analyzed. They were diagnosed by surgery and pathology to have mass and invasive breast cancer and had contrast-enhanced chest CT examination before operation. 300 patients were randomly selected for the study, including 100 TNBC and 200 non-TNBC (NTNBC) patients. Among them 180 were used in discovery group and 120 were used in validation group. The molecular subtypes of breast cancer in the patients were determined immunohistochemistrially. Radiomics features were extracted from three dimensional CT-images. The LASSO logistic method was used to select image features and calculate radiomics scores. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic value of radiomics scores for TNBC. Results Five image features were found to be related to TNBC subtype (P < 0.001). These image features based-radiomic signatures had good predictive values for TNBC with the areas under ROC curve (AUC) of 0.881 (95% CI: 0.781–0.921) in the discovery group and 0.851 (95% CI: 0.761–0.961) in the validation group, respectively. The sensitivities and specificities were 0.767, and 0.873 in the discovery group and 0.785 and 0.915 in the validation group. Conclusions Radiomic signature based on preoperative CT is capable of distinguishing patients with TNBC and NTNBC. It adds additional value for conventional chest contrast-enhanced CT and helps plan the strategy for clinical treatment of the patients.
Collapse
Affiliation(s)
- Qingliang Feng
- Department of Radiology, Linyi Central Hospital, Linyi, China
| | - Qiang Hu
- Department of Radiology, Linyi Central Hospital, Linyi, China
| | - Yan Liu
- Department of Healthcare, Linyi Central Hospital, Linyi, China
| | - Tao Yang
- Department of Radiology, Linyi Central Hospital, Linyi, China
| | - Ziyi Yin
- Department of Surgery, Beijing Tiantan Hospital, Capital Medical University, 119 South 4th Ring West Road, Beijing, 100070, China.
| |
Collapse
|
23
|
Screening of a Novel Upregulated lncRNA, A2M-AS1, That Promotes Invasion and Migration and Signifies Poor Prognosis in Breast Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9747826. [PMID: 32352014 PMCID: PMC7171613 DOI: 10.1155/2020/9747826] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/26/2019] [Accepted: 10/11/2019] [Indexed: 12/25/2022]
Abstract
Understanding of prognostic factors and therapeutic targets for breast cancer is imperative for guidance of patient care. We studied 1203 tumour samples from the Gene Expression Omnibus (GEO) to evaluate potential genes related to breast cancer. R software was used to analyse differentially expressed long noncoding RNAs (lncRNAs) in the RNA microarray expression profiles GSE45827 and GSE65216 and to identify a series of differentially expressed lncRNAs associated with human breast cancer. Of these lncRNAs, A2M-AS1, a lncRNA that has not been previously reported, was significantly upregulated in human breast cancer tissues compared with adjacent nontumour tissues. Importantly, A2M-AS1 upregulation was significantly associated with ER-negative, HER2-positive, and basal-like breast cancer and with poor recurrence-free survival and metastasis-free survival in breast cancer patients. After validating these results in 96 collected human breast cancer tissues and 64 paired adjacent noncancerous tissues, we further investigated the roles of A2M-AS1 in human ER-negative and basal-like breast cancer cells. The results revealed that A2M-AS1 significantly promotes human breast cancer cell proliferation, invasion, and migration. Additionally, bioinformatics analysis of genes coexpressed with A2M-AS1 in the context of human breast cancer combined with qRT-PCR and Western blot assays revealed that A2M-AS1 exerts regulatory effects on downstream factors in the cell adhesion molecule pathway, including CD2 and SELL. These results imply that A2M-AS1 might be a promising candidate prognostic factor and therapeutic target for breast cancer.
Collapse
|
24
|
Identification of gene modules associated with survival of diffuse large B-cell lymphoma treated with CHOP-based chemotherapy. THE PHARMACOGENOMICS JOURNAL 2020; 20:705-716. [PMID: 32042095 DOI: 10.1038/s41397-020-0161-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Diffuse Large B-cell Lymphoma (DLBCL), a heterogeneous disease, is influenced by complex network of gene interactions. Most previous studies focused on individual genes, but ignored the importance of intergenic correlations. In current study, we aimed to explore the association between gene networks and overall survival (OS) of DLBCL patients treated with CHOP-based chemotherapy (cyclophosphamide combination with doxorubicin, vincristine and prednisone). Weighted gene co-expression network analysis was conducted to obtain insights into the molecular characteristics of DLBCL. Ten co-expression gene networks (modules) were identified in training dataset (n = 470), and their associations with patients' OS after chemotherapy were tested. The results were validated in four independent datasets (n = 802). Gene ontology (GO) biological function enrichment analysis was conducted with Metascape. Three modules (purple, brown and red), which were enriched in T-cell immune, cell-cell adhesion and extracellular matrix (ECM), respectively, were found to be related to longer OS. Higher expression of several hub genes within these three co-expression modules, for example, LCP2 (HR = 0.77, p = 5.40 × 10-2), CD2 (HR = 0.87, p = 6.31 × 10-2), CD3D (HR = 0.83, p = 6.94 × 10-3), FYB (HR = 0.82, p = 1.40 × 10-2), GZMK (HR = 0.92, p = 1.19 × 10-1), FN1 (HR = 0.88, p = 7.06 × 10-2), SPARC (HR = 0.82, p = 2.06 × 10-2), were found to be associated with favourable survival. Moreover, the associations of the modules and hub genes with OS in different molecular subtypes and different chemotherapy groups were also revealed. In general, our research revealed the key gene modules and several hub genes were upregulated correlated with good survival of DLBCL patients, which might provide potential therapeutic targets for future clinical research.
Collapse
|
25
|
Kim YJ, Lee G, Han J, Song K, Choi JS, Choi YL, Shin YK. UBE2C Overexpression Aggravates Patient Outcome by Promoting Estrogen-Dependent/Independent Cell Proliferation in Early Hormone Receptor-Positive and HER2-Negative Breast Cancer. Front Oncol 2020; 9:1574. [PMID: 32039034 PMCID: PMC6989552 DOI: 10.3389/fonc.2019.01574] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/30/2019] [Indexed: 12/17/2022] Open
Abstract
We previously showed that UBE2C mRNA expression is significantly associated with poor prognosis only in patients with hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2)– breast cancer. In this study, we further reanalyzed the correlation between UBE2C mRNA expression and clinical outcomes in patients with HR+/HER2– breast cancer, and we investigated the molecular mechanism underlying the role of UBE2C modulation in disease progression in this subgroup of patients. Univariate and multivariate analyses showed that high UBE2C expression was associated with significantly shorter survival of breast cancer patients with pN0 and pN1 tumors but not pN2/N3 tumors (P < 0.05). In vitro functional experiments in HR+/HER2– breast cancer cells showed that UBE2C expression is a tumorigenic factor, and that estrogen upregulated UBE2C mRNA and protein by directly binding to the UBE2C promoter region. UBE2C knockdown inhibited cell proliferation by affecting cell cycle progression, and UBE2C overexpression was associated with estrogen-independent growth. UBE2C depletion markedly increased the cytotoxicity of tamoxifen by inducing apoptosis. The present findings suggest that UBE2C overexpression is correlated with relapse and promotes estrogen-dependent/independent proliferation in early HR+/HER2– breast cancer.
Collapse
Affiliation(s)
- Yu-Jin Kim
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Gyunghwa Lee
- Laboratory of Molecular Pathology and Cancer Genomics, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, South Korea
| | | | - Kyoung Song
- The Center for Companion Diagnostics, LOGONE Bio Convergence Research Foundation, Seoul, South Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan-si, South Korea
| | - Yoon-La Choi
- Laboratory of Cancer Genomics and Molecular Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, South Korea.,The Center for Anti-cancer Companion Diagnostics, BioMAX/N-Bio, Seoul National University, Seoul, South Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
Luo ZB, Lai GE, Jiang T, Cao CL, Peng T, Liu FE. A Competing Endogenous RNA Network Reveals Novel lncRNA, miRNA and mRNA Biomarkers With Diagnostic and Prognostic Value for Early Breast Cancer. Technol Cancer Res Treat 2020; 19:1533033820983293. [PMID: 33371806 PMCID: PMC7871288 DOI: 10.1177/1533033820983293] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/04/2020] [Accepted: 11/30/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND This study aims to reveal early breast cancer (BC) specific competing endogenous RNA (ceRNA) network through the expression profiles of microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and mRNAs. METHODS Based on The Cancer Genome Atlas (TCGA), we obtained the differentially expressed mRNAs, miRNAs, and lncRNAs (DEmRNAs, DEmiRNAs and DElncRNAs) between early BC and normal samples. The lncRNA-miRNA-mRNA interaction network was constructed using Cytoscape. Functional enrichment were performed using GeneCoDis3. The expression of selected genes were validated by qRT-PCR. Based on the published dataset, we validated the result of TCGA integration analysis. The diagnostic and prognostic value of candidate genes was evaluated by ROC curve analysis and survival analysis, respectively. RESULTS Totally, 1207 DEmRNAs, 194 DElncRNAs and 37 DEmiRNAs were obtained. Functional enrichment analysis results showed that all of DEmRNAs were enriched in pathway of cytokine-cytokine receptor interaction, PPAR signaling pathway and pathways in cancer. The DEmRNA-DEmiRNA-DElncRNA interaction network in early BC was consisted of 23 DEmiRNAs, 95 DElncRNAs and 309 DEmRNAs. Among ceRNA network, IL-6-hsa-miR-182-5p-ADAMTS9-AS1 interactions, LIFR-hsa-miR-21-5p-ADAMTS9-AS1 interactions and MMP1/MMP11-hsa-miR-145-5p-CDKN2B-AS1 interactions were speculated to involve in the development of early BC. The qRT-PCR results were consistent with our integrated analysis. Except for ADAMTS9-AS1 and CDKN2B-AS1, expression of the others results in the Gene Expression Omnibus (GEO) dataset were generally consistent with TCGA integrated analysis. The area under curve (AUC) of the ADAMTS9-AS1, CDKN2B-AS1, IL-6, MMP11, hsa-miR-145-5p and hsa-miR-182-5p were 0.947, 0.862, 0.842, 0.993, 0.960 and 0.944, and the specificity and sensitivity of the 6 biomarkers were 83.4% and 95.6%, 72.2% and 90.3%, 80.1% and 74.3%, 96.2% and 96.5%, 90.1% and 92.3%, and 88.7% and 90.4%, respectively. In addition, IL-6 had potential prognostic value for early BC. CONCLUSION These findings may provide novel insights into the lncRNA-miRNA-mRNA network and uncover potential therapeutic targets in early BC.
Collapse
Affiliation(s)
- Zhong-Bing Luo
- Department of Breast Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou of Jiangxi Province, China
| | - Gui-E Lai
- Department of Breast Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou of Jiangxi Province, China
| | - Tao Jiang
- Department of Breast Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou of Jiangxi Province, China
| | - Chuan-Lin Cao
- Department of Breast Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou of Jiangxi Province, China
| | - Tao Peng
- Department of Breast Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou of Jiangxi Province, China
| | - Feng-En Liu
- Department of Breast Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou of Jiangxi Province, China
| |
Collapse
|
27
|
Na H, Han J, Ka NL, Lee MH, Choi YL, Shin YK, Lee MO. High expression of NR1D1 is associated with good prognosis in triple-negative breast cancer patients treated with chemotherapy. Breast Cancer Res 2019; 21:127. [PMID: 31779659 PMCID: PMC6883674 DOI: 10.1186/s13058-019-1197-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
Background Nuclear receptor subfamily 1, group D, member 1 (NR1D1) is a ligand-regulated nuclear receptor and transcriptional factor. Although recent studies have implicated NR1D1 as a regulator of DNA repair and proliferation in breast cancers, its potential as a therapeutic target for breast cancer has not been assessed in terms of clinical outcomes. Thus, this study aims to analyze NR1D1 expression in breast cancer patients and to evaluate its potential prognostic value. Methods NR1D1 expression was analyzed by immunohistochemistry using an anti-NR1D1 antibody in 694 breast cancer samples. Survival analyses were performed using the Kaplan–Meier method with the log-rank test to investigate the association of NR1D1 expression with clinical outcome. Results One hundred thirty-nine of these samples exhibited high NR1D1 expression, mostly in the nucleus of breast cancer cells. NR1D1 expression correlated significantly with histological grade and estrogen receptor status. Overall survival (OS) and disease-free survival (DFS) did not correlate significantly with NR1D1 expression in breast cancer patients regardless of whether they had received chemotherapy. Subgroup analysis performed according to molecular subtype of breast cancer showed a significant influence of high NR1D1 expression on OS (P = 0.002) and DFS (P = 0.007) in patients with triple-negative breast cancer (TNBC) treated with chemotherapy. Conclusions High NR1D1 expression level had a favorable impact on OS and DFS in patients with TNBC treated with chemotherapy. NR1D1 should be investigated further as a possible prognostic marker in TNBC patients receiving chemotherapeutic treatment and as a target in the development of chemotherapeutic approaches to treating TNBC.
Collapse
Affiliation(s)
- Hyelin Na
- College of Pharmacy, Bio-MAX, and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinil Han
- Gencurix, Inc, Seoul, 08394, Republic of Korea
| | - Na-Lee Ka
- College of Pharmacy, Bio-MAX, and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Ho Lee
- College of Pharmacy, Bio-MAX, and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Kee Shin
- College of Pharmacy, Bio-MAX, and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Mi-Ock Lee
- College of Pharmacy, Bio-MAX, and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
28
|
Emerging immune gene signatures as prognostic or predictive biomarkers in breast cancer. Arch Pharm Res 2019; 42:947-961. [PMID: 31707598 DOI: 10.1007/s12272-019-01189-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/01/2019] [Indexed: 12/20/2022]
Abstract
Several multigene assays have been developed to predict the risk of distant recurrence and response to adjuvant therapy in early breast cancer. However, the prognostic or predictive value of current proliferation gene signature-based assays are limited to hormone receptor-positive, human epidermal growth factor receptor 2-negative (HR+/HER2-) early breast cancer. Considerable discordance between the different assays in classifying patients into risk groups has also been reported, thus raising questions about the clinical utility of these assays for individual patients. Therefore, there still remains a need for better prognostic or predictive biomarkers for breast cancer. The role of immune cells comprising tumor microenvironment in tumor progression has been recognized. Accumulating evidences have shown that immune gene signatures and tumor-infiltrating lymphocytes (TILs) can be prognostic or predictive factors in breast cancer, particularly with regard to HER2+ and triple-negative breast cancer. In this review, I summarize current multigene assays for breast cancer and discuss recent progress in identifying novel breast cancer biomarkers, focusing on the emerging importance of immune gene signatures and TILs as prognostic or predictive biomarkers.
Collapse
|
29
|
Eiro N, Cid S, Fernández B, Fraile M, Cernea A, Sánchez R, Andicoechea A, DeAndrés Galiana EJ, González LO, Fernández‐Muñiz Z, Fernández‐Martínez JL, Vizoso FJ. MMP11 expression in intratumoral inflammatory cells in breast cancer. Histopathology 2019; 75:916-930. [DOI: 10.1111/his.13956] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Noemi Eiro
- Research Unit Fundación Hospital de Jove Gijón Spain
| | - Sandra Cid
- Research Unit Fundación Hospital de Jove Gijón Spain
| | - Berta Fernández
- Department of Surgery Hospital Universitario Central de Asturias Oviedo Spain
| | - Maria Fraile
- Research Unit Fundación Hospital de Jove Gijón Spain
| | - Ana Cernea
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
| | - Rosario Sánchez
- Department of Surgery Fundación Hospital de Jove Gijón Spain
| | | | - Enrique J DeAndrés Galiana
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
- Department of Informatics and Computer Science University of Oviedo Oviedo Spain
| | - Luis O González
- Department of Anatomical Pathology Fundación Hospital de Jove Gijón Spain
| | - Zulima Fernández‐Muñiz
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
| | - Juan L Fernández‐Martínez
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
| | - Francisco J Vizoso
- Research Unit Fundación Hospital de Jove Gijón Spain
- Department of Surgery Fundación Hospital de Jove Gijón Spain
| |
Collapse
|
30
|
Yang H, Jiang P, Liu D, Wang HQ, Deng Q, Niu X, Lu L, Dai H, Wang H, Yang W. Matrix Metalloproteinase 11 Is a Potential Therapeutic Target in Lung Adenocarcinoma. Mol Ther Oncolytics 2019; 14:82-93. [PMID: 31024988 PMCID: PMC6477516 DOI: 10.1016/j.omto.2019.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 03/27/2019] [Indexed: 12/29/2022] Open
Abstract
Lung cancer is one of the leading causes of cancer-associated death, with the etiology largely unknown. The aim of this study was to identify key driver genes with therapeutic potentials in lung adenocarcinoma (LUAD). Transcriptome microarray data from four GEO datasets (GEO: GSE7670, GSE10072, GSE68465, and GSE43458) were jointly analyzed for differentially expressed genes (DEGs). Ontologic analysis showed that most of the upregulated DEGs enriched in collagen catabolic and fibril organization processes were regulated by matrix metalloproteinases (MMPs). Matrix metalloproteinase 11 (MMP11), the highest upregulated MMP family member in LUAD-transformed cells, acted in an autocrine manner and was significantly increased in sera of LUAD patients. MMP11 depletion severely impaired LUAD cell proliferation, migration, and invasion in vitro, in line with retarded tumor growth in xenograft models. Treatment of different human LUAD cell lines with anti-MMP11 antibody significantly retarded cell growth and migration. Administration of anti-MMP11 antibody at a dose of 1 μg/g body weight significantly suppressed tumor growth in xenograft models. These findings indicate that MMP11 is a key cancer driver gene in LUAD and is an appealing target for antibody therapy.
Collapse
Affiliation(s)
- Haoran Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Peng Jiang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Dongyan Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- University of Science and Technology of China, Hefei 230026, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Hong-Qiang Wang
- Biological Molecular Information System Lab., Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Qingmei Deng
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Xiaojie Niu
- Department of Anatomy, Shanxi Medical University, Taiyuan 030024, China
| | - Li Lu
- Department of Anatomy, Shanxi Medical University, Taiyuan 030024, China
| | - Haiming Dai
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Center of Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Cancer Hospital, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
31
|
Xia H, Yu W, Liu M, Li H, Pang W, Wang L, Zhang Y. An integrated bioinformatics analysis of potential therapeutic targets among matrix metalloproteinases in breast cancer. Oncol Lett 2019; 18:2985-2994. [PMID: 31452777 PMCID: PMC6704324 DOI: 10.3892/ol.2019.10669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/14/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is one of the most aggressive malignancies worldwide among females. Matrix metalloproteinases (MMPs), as the most abundant class of non-serine proteases present in invasive and metastatic tumors, can regulate a variety of alterations in the microenvironment during tumor progression. However, the differential expression of MMPs and its prognostic values in BC is yet to be elucidated. In this research, using the ONCOMINE dataset, The Cancer Genome Atlas, Breast Cancer Gene-Expression Miner v4.1 (Bc-GenExMiner), Kaplan-Meier Plotter and cBioPortal, the transcriptional MMPs and survival outcome data of patients with BC was compared. It was indicated that mRNA levels of MMP1/3/9/10/11/12/13 were increased compared with non-tumor tissues, whereas mRNA expression of MMP2/16/19/23B/28 was lower in BC tissues. Kaplan-Meier plots showed that high mRNA levels of MMP2/10/16/19/20/23B/27 in patients with BC were associated with better recurrence-free survival. In contrast, high MMP1/8/9/11/12 conferred worse RFS rate. Meanwhile, high transcription levels of MMP1/3/11/12/13 predicted shorter distant metastasis-free survival, while high levels of MMP1/12 demonstrated worse overall survival in patients with BC. From Bc-GenExMiner, it was indicated that high expression of MMP16/20 was correlated with better prognosis, while MMP1/9/11/12/13/14/15 exerted a negative effect on patient prognosis. The integrative bioinformatics analysis performed in the present study suggests that MMP1/9/12/16, compared with other MMPs, are potentially appropriate targets for targeted therapy in patients with BC.
Collapse
Affiliation(s)
- Haiqun Xia
- Department of Radiation Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| | - Weixuan Yu
- Department of Surgical Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| | - Ming Liu
- Department of Surgical Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| | - Hong Li
- Department of Surgical Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| | - Wei Pang
- Department of Radiation Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| | - Libin Wang
- Department of Surgical Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| | - Yunda Zhang
- Department of Surgical Oncology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, Guangdong 523000, P.R. China
| |
Collapse
|
32
|
Shen Y, Shen Y, Liu J, Shi JM, Ding J. Significance of expression of EIF3h, MMP-10 and MMP-11 in colonic adenocarcinoma. Shijie Huaren Xiaohua Zazhi 2018; 26:834-841. [DOI: 10.11569/wcjd.v26.i14.834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of eukaryotic translation initiation factor 3h (EIF3h), matrix metallopeptidase 10 (MMP-10), and matrix metallopeptidase 11 (MMP-11) in colonic adenocarcinoma and to analyze their clinical significance.
METHODS One hundred and ten colonic adenocarcinoma tissues, 43 high-grade colorectal intraepithelial neoplasia tissues, 43 low-grade colorectal intraepithelial neoplasia tissues, and 43 normal colonic mucosal tissues were collected. The expression of EIF3h, MMP-10, and MMP-11 in the above tissues was detected by immunohistochemistry.
RESULTS The expression of EIF3h, MMP-10, and MMP-11 differed significantly among the four groups. Expression of EIF3h, MMP-10, and MMP-11 was correlated with lymph node metastasis. Expression of EIF3h was correlated with tumor size, differentiation, proliferation index, and TNM stage. Expression of MMP-10 and MMP-11 was correlated with vascular invasion. There was a positive correlation between MMP-10 and MMP-11 expression in colonic adenocarcinoma.
CONCLUSION High expression and synergy of EIF3h, MMP-10, and MMP-11 can promote tumor formation and progression in colonic adenocarcinoma. EIF3h may be associated with the prognosis of colonic adenocarcinoma.
Collapse
Affiliation(s)
- Yuan Shen
- Department of Internal Medicine, Huzhou Sixth Hospital, Huzhou 313000, Zhejiang Province, China
| | - Ying Shen
- Department of Internal Medicine, Huzhou Sixth Hospital, Huzhou 313000, Zhejiang Province, China
| | - Jiang Liu
- Department of Gastroenterology, Huzhou Central Hospital, Huzhou 313000, Zhejiang Province, China
| | - Jie-Min Shi
- Department of Gastroenterology, Huzhou Central Hospital, Huzhou 313000, Zhejiang Province, China
| | - Jian Ding
- Department of Gastroenterology, Huzhou Central Hospital, Huzhou 313000, Zhejiang Province, China
| |
Collapse
|