1
|
Hong J, Jin HJ, Choi MR, Lim DWT, Park JE, Kim YS, Lim SB. Matrisomics: Beyond the extracellular matrix for unveiling tumor microenvironment. Biochim Biophys Acta Rev Cancer 2024; 1879:189178. [PMID: 39241895 DOI: 10.1016/j.bbcan.2024.189178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
The matrisome, a group of proteins constituting or interacting with the extracellular matrix (ECM), has garnered attention as a potent regulator of cancer progression. An increasing number of studies have focused on cancer matrisome utilizing diverse -omics approaches. Here, we present diverse patterns of matrisomal populations within cancer tissues, exploring recent -omics studies spanning different '-omics' levels (epigenomics, genomics, transcriptomics, and proteomics), as well as newly developed sequencing techniques such as single-cell RNA sequencing and spatial transcriptomics. Some matrisome genes showed uniform patterns of upregulated or downregulated expression across various cancers, while others displayed different expression patterns according to the cancer types. This matrisomal dysregulation in cancer was further examined according to their originating cell type and spatial location in the tumor tissue. Experimental studies were also collected to demonstrate the identified roles of matrisome genes during cancer progression. Interestingly, many studies on cancer matrisome have suggested matrisome genes as effective biomarkers in cancer research. Although the specific mechanisms and clinical applications of cancer matrisome have not yet been fully elucidated, recent techniques and analyses on cancer matrisomics have emphasized their biological importance in cancer progression and their clinical implications in deciding the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Jiwon Hong
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Hyo Joon Jin
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Mi Ran Choi
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Darren Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre, Singapore 168583, Singapore
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-Ro, Yuseong-Gu, Daejeon 34141, Republic of Korea
| | - You-Sun Kim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
2
|
Capuano A, Vescovo M, Canesi S, Pivetta E, Doliana R, Nadin MG, Yamamoto M, Tsukamoto T, Nomura S, Pilozzi E, Palumbo A, Canzonieri V, Cannizzaro R, Scanziani E, Baldassarre G, Mongiat M, Spessotto P. The extracellular matrix protein EMILIN-1 impacts on the microenvironment by hampering gastric cancer development and progression. Gastric Cancer 2024; 27:1016-1030. [PMID: 38941035 PMCID: PMC11335817 DOI: 10.1007/s10120-024-01528-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND The contribution of the tumor microenvironment and extracellular matrix to the aggressive biology of Gastric Cancer (GC) has been recently characterized; however, the role of EMILIN-1 in this context is unknown. EMILIN-1 is an essential structural element for the maintenance of lymphatic vessel (LV) integrity and displays anti-proliferative properties as demonstrated in skin and colon cancer. Given the key role of LVs in GC progression, the aim of this study was to investigate the role of EMILIN-1 in GC mouse models. METHODS We used the syngeneic YTN16 cells which were injected subcutaneously and intraperitoneally in genetically modified EMILIN-1 mice. In alternative, carcinogenesis was induced using N-Methyl-N-nitrosourea (MNU). Mouse-derived samples and human biopsies were analyzed by IHC and IF to the possible correlation between EMILIN-1 expression and LV pattern. RESULTS Transgenic mice developed tumors earlier compared to WT animals. 20 days post-injection tumors developed in EMILIN-1 mutant mice were larger and displayed a significant increase of lymphangiogenesis. Treatment of transgenic mice with MNU associated with an increased number of tumors, exacerbated aggressive lesions and higher levels of LV abnormalities. A significant correlation between the levels of EMILIN-1 and podoplanin was detected also in human samples, confirming the results obtained with the pre-clinical models. CONCLUSIONS This study demonstrates for the first time that loss of EMILIN-1 in GC leads to lymphatic dysfunction and proliferative advantages that sustain tumorigenesis, and assess the use of our animal model as a valuable tool to verify the fate of GC upon loss of EMILIN-1.
Collapse
Affiliation(s)
- Alessandra Capuano
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Maddalena Vescovo
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Simone Canesi
- Dipartimento di Medicina Veterinaria e Scienze Animali (DIVAS), Università Degli Studi di Milano, Milan, Italy
| | - Eliana Pivetta
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
- Clinical Pathology Unit, Ospedale Santa Maria Degli Angeli, Pordenone, Italy
| | - Roberto Doliana
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Maria Grazia Nadin
- Oncological Gastroenterology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Department of Pathology, Graduate School of Medicine, Fujita Health University, Toyoake, Japan
| | - Sachiyo Nomura
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Azienda Ospedaliero-Universitaria Sant'Andrea, Rome, Italy
| | - Antonio Palumbo
- Pathology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Renato Cannizzaro
- Oncological Gastroenterology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Eugenio Scanziani
- Dipartimento di Medicina Veterinaria e Scienze Animali (DIVAS), Università Degli Studi di Milano, Milan, Italy
| | - Gustavo Baldassarre
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Maurizio Mongiat
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy
| | - Paola Spessotto
- Molecular Oncology Unit, Centro di Riferimento Oncologico Aviano, (CRO) IRCCS, Via Franco Gallini 2, 33081, Aviano, PN, Italy.
| |
Collapse
|
3
|
Xu W, Goreczny GJ, Forsythe I, Brennan G, Stowell T, Brock K, Capella B, Turner CE. Hic-5 regulates extracellular matrix-associated gene expression and cytokine secretion in cancer associated fibroblasts. Exp Cell Res 2024; 435:113930. [PMID: 38237846 PMCID: PMC10923124 DOI: 10.1016/j.yexcr.2024.113930] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
The focal adhesion protein, Hic-5 plays a key role in promoting extracellular matrix deposition and remodeling by cancer associated fibroblasts within the tumor stroma to promote breast tumor cell invasion. However, whether stromal matrix gene expression is regulated by Hic-5 is still unknown. Utilizing a constitutive Hic-5 knockout, Mouse Mammary Tumor Virus-Polyoma Middle T-Antigen spontaneous breast tumor mouse model, bulk RNAseq analysis was performed on cancer associated fibroblasts isolated from Hic-5 knockout mammary tumors. Functional network analysis highlighted a key role for Hic-5 in extracellular matrix organization, with both structural matrix genes, as well as matrix remodeling genes being differentially expressed in relation to Hic-5 expression. The subcellular distribution of the MRTF-A transcription factor and expression of a subset of MRTF-A responsive genes was also impacted by Hic-5 expression. Additionally, cytokine array analysis of conditioned media from the Hic-5 and Hic-5 knockout cancer associated fibroblasts revealed that Hic-5 is important for the secretion of several key factors that are associated with matrix remodeling, angiogenesis and immune evasion. Together, these data provide further evidence of a central role for Hic-5 expression in cancer associated fibroblasts in regulating the composition and organization of the tumor stroma microenvironment to promote breast tumor progression.
Collapse
Affiliation(s)
- Weiyi Xu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Gregory J Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Jnana Therapeutics, Boston, MA, USA
| | - Ian Forsythe
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA; Zymo Research Corp, Huntington Beach, CA, USA
| | - Grant Brennan
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Theresa Stowell
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Katia Brock
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Benjamin Capella
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
4
|
Zhang W, Zhou D, Song S, Hong X, Xu Y, Wu Y, Li S, Zeng S, Huang Y, Chen X, Liang Y, Guo S, Pan H, Li H. Prediction and verification of the prognostic biomarker SLC2A2 and its association with immune infiltration in gastric cancer. Oncol Lett 2024; 27:70. [PMID: 38192676 PMCID: PMC10773219 DOI: 10.3892/ol.2023.14203] [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: 07/01/2023] [Accepted: 11/15/2023] [Indexed: 01/10/2024] Open
Abstract
Gastric cancer (GC) is the fifth most common cause of cancer-associated deaths; however, its treatment options are limited. Despite clinical improvements, chemotherapy resistance and metastasis are major challenges in improving the prognosis and quality of life of patients with GC. Therefore, effective prognostic biomarkers and targets associated with immunological interventions need to be identified. Solute carrier family 2 member 2 (SLC2A2) may serve a role in tumor development and invasion. The present study aimed to evaluate SLC2A2 as a prospective prognostic marker and chemotherapeutic target for GC. SLC2A2 expression in several types of cancer and GC was analyzed using online databases, and the effects of SLC2A2 expression on survival prognosis in GC were investigated. Clinicopathological parameters were examined to explore the association between SLC2A2 expression and overall survival (OS). Associations between SLC2A2 expression and immune infiltration, immune checkpoints and IC50 were estimated using quantification of the tumor immune contexture from human RNA-seq data, the Tumor Immune Estimation Resource 2.0 database and the Genomics of Drug Sensitivity in Cancer database. Differential SLC2A2 expression and the predictive value were validated using the Human Protein Atlas, Gene Expression Omnibus, immunohistochemistry and reverse transcription-quantitative PCR. SLC2A2 expression was downregulated in most types of tumor but upregulated in GC. Functional enrichment analysis revealed an association between SLC2A2 expression and lipid metabolism and the tumor immune microenvironment. According to Gene Ontology term functional enrichment analysis, SLC2A2-related differentially expressed genes were enriched predominantly in 'chylomicron assembly', 'plasma lipoprotein particle assembly', 'high-density lipoprotein particle', 'chylomicron', 'triglyceride-rich plasma lipoprotein particle', 'very-low-density lipoprotein particle'. 'intermembrane lipid transfer activity', 'lipoprotein particle receptor binding', 'cholesterol transporter activity' and 'intermembrane cholesterol transfer activity'. In addition, 'cholesterol metabolism', and 'fat digestion and absorption' were significantly enriched in the Kyoto Encyclopedia of Genes and Genomes pathway analysis. Patients with GC with high SLC2A2 expression had higher levels of neutrophil and M2 macrophage infiltration and a significant inverse correlation was observed between SLC2A2 expression and MYC targets, tumor mutation burden, microsatellite instability and immune checkpoints. Furthermore, patients with high SLC2A2 expression had worse prognosis, including OS, disease-specific survival and progression-free interval. Multivariate regression analysis demonstrated that SLC2A2 could independently prognosticate GC and the nomogram model showed favorable performance for survival prediction. SLC2A2 may be a prospective prognostic marker for GC. The prediction model may improve the prognosis of patients with GC in clinical practice, and SLC2A2 may serve as a novel therapeutic target to provide immunotherapy plans for GC.
Collapse
Affiliation(s)
- Weijian Zhang
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Dishu Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Shuya Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Xinxin Hong
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yifei Xu
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yuqi Wu
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Shiting Li
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Sihui Zeng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yanzi Huang
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Xinbo Chen
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yizhong Liang
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Shaoju Guo
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Haiwen Li
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|
5
|
Salimian N, Peymani M, Ghaedi K, Hashemi M, Rahimi E. Collagen 1A1 (COL1A1) and Collagen11A1(COL11A1) as diagnostic biomarkers in Breast, colorectal and gastric cancers. Gene 2024; 892:147867. [PMID: 37783295 DOI: 10.1016/j.gene.2023.147867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
PURPOSE Collagen family genes (CFGs) play a significant role in the pathogenesis of cancers. This study aimed to evaluate changes in the expression levels (Els) of CFGs related to epithelial-mesenchymal transition (EMT) and metastasis in gastric (GC), breast (BC), and colorectal (CRC) cancers to introduce these genes as potential diagnostic biomarkers for these three types of cancer. METHODS The Cancer Genome Atlas (TCGA) examined ELS changes in CFGs associated with EMT and metastasis to determine their diagnostic value for GC, BC, and CRC. InteractiVenn was used to find genes shared by these three cancers. The biomarker role of CFGs was determined using the receiver operating characteristic (ROC) analysis. GC, BC, and CRC samples were analyzed using the RT-qPCR method to verify the bioinformatics results and evaluate the EL of the selected genes as biomarkers for these cancers. RESULTS The in-silico results showed a significant increase in the EL of several CFGs involved in EMT and metastasis in GC, BC, and CRC samples compared to healthy samples. Six common genes (COL11A1, COL12A1, COL1A1, COL1A2, COL5A1, and COL5A2) showed significantly increased in these three cancers, therebysupporting their oncogenic role. Furthermore, the biomarker-related analyses indicated that COL11A1 and COL1A1 were common diagnostic biomarkers for the three cancers. The RT-qPCR method confirmed that the ELs of COL11A1 and COL1A1 in the GC, BC, and CRC samples increased significantly compared to the adjacent normal samples. CONCLUSION CFGs in EMT and metastasis of GC, BC, and CRC are strong common diagnostic biomarkers for these cancers.
Collapse
Affiliation(s)
- Niloufar Salimian
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ebrahim Rahimi
- Department of Food Hygiene, Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
6
|
Guo KS, Brodsky AS. Tumor collagens predict genetic features and patient outcomes. NPJ Genom Med 2023; 8:15. [PMID: 37414817 DOI: 10.1038/s41525-023-00358-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 06/14/2023] [Indexed: 07/08/2023] Open
Abstract
The extracellular matrix (ECM) is a critical determinant of tumor fate that reflects the output from myriad cell types in the tumor. Collagens constitute the principal components of the tumor ECM. The changing collagen composition in tumors along with their impact on patient outcomes and possible biomarkers remains largely unknown. The RNA expression of the 43 collagen genes from solid tumors in The Cancer Genome Atlas (TCGA) was clustered to classify tumors. PanCancer analysis revealed how collagens by themselves can identify the tissue of origin. Clustering by collagens in each cancer type demonstrated strong associations with survival, specific immunoenvironments, somatic gene mutations, copy number variations, and aneuploidy. We developed a machine learning classifier that predicts aneuploidy, and chromosome arm copy number alteration (CNA) status based on collagen expression alone with high accuracy in many cancer types with somatic mutations, suggesting a strong relationship between the collagen ECM context and specific molecular alterations. These findings have broad implications in defining the relationship between cancer-related genetic defects and the tumor microenvironment to improve prognosis and therapeutic targeting for patient care, opening new avenues of investigation to define tumor ecosystems.
Collapse
Affiliation(s)
- Kevin S Guo
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Alexander S Brodsky
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
7
|
Zhang Z, Ning M, Li L, Li Z, Wang Y, Zhao J. Knockdown of ASF1B inhibits cell proliferation, migration, invasion and cisplatin resistance in gastric cancer through the Myc pathway. Oncol Lett 2023; 25:242. [PMID: 37153049 PMCID: PMC10161352 DOI: 10.3892/ol.2023.13828] [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: 11/30/2022] [Accepted: 03/07/2023] [Indexed: 05/09/2023] Open
Abstract
Gastric cancer (GC) is a prevalent malignancy in the digestive system that poses a serious threat to human health. Anti-silencing function 1B (ASF1B) performs an important role in the progression of numerous tumors; however, its function in GC still requires further elucidation. Using data from The Cancer Genome Atlas, the expression levels of ASF1B in GC tissues were analyzed and a survival curve for high-ASF1B expression and low-ASF1B expression groups was plotted using the Kaplan-Meier method. Reverse transcription-quantitative PCR was performed to evaluate ASF1B expression in GC tissues and cells. Small interfering RNAs targeting ASF1B were transfected into HGC-27 and AGS cells to silence ASF1B expression. Cell viability, proliferation, migration, invasion, and apoptosis in HGC-27 and AGS cells was assessed using cell counting kit-8 assay, colony formation assay, wound healing assay, Transwell assay and flow cytometry, respectively. The protein changes were assessed using western blotting. Gene Set Enrichment Analysis (GSEA) was used to identify ASF1B related pathways. The results demonstrated that ASF1B expression was increased in GC tissues and cells compared with adjacent healthy tissues and normal cells (GES-1), and high expression of ASF1B was associated with poor survival outcomes in patients with GC. Silencing ASF1B inhibited cell viability, colony formation, migration, invasion and cisplatin resistance, while also attenuating the apoptotic capability of HGC-27 and AGS cells. GSEA showed that ASF1B could activate the Myc-targets-v1 and Myc-targets-v2 pathways. Moreover, silencing ASF1B inhibited the Myc pathway-related proteins Myc, minichromosome maintenance (MCM)4 and MCM5. Overexpression of Myc reversed the inhibitory effect of ASF1B silencing on AGS cell proliferation, invasion and cisplatin resistance. In conclusion, the results indicate that knockdown of ASF1B may suppress GC cell proliferation, migration and invasion, and promote cell apoptosis and cisplatin sensitivity by modulating the Myc pathway, thereby offering novel possibilities for reversing cisplatin resistance in GC.
Collapse
Affiliation(s)
- Zao Zhang
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
- Correspondence to: Dr Zao Zhang, Department of Pharmacy, Cangzhou Central Hospital, 50 Xinhua Middle Road, Cangzhou, Hebei 061000, P.R. China, E-mail:
| | - Meiying Ning
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Li Li
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Zhuangzhuang Li
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yanrong Wang
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Jing Zhao
- Department of Pharmacy, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
8
|
De Martino D, Bravo-Cordero JJ. Collagens in Cancer: Structural Regulators and Guardians of Cancer Progression. Cancer Res 2023; 83:1386-1392. [PMID: 36638361 PMCID: PMC10159947 DOI: 10.1158/0008-5472.can-22-2034] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/29/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Collagen is one of the most abundant proteins in animals and a major component of the extracellular matrix (ECM) in tissues. Besides playing a role as a structural building block of tissues, collagens can modulate the behavior of cells, and their deregulation can promote diseases such as cancer. In tumors, collagens and many other ECM molecules are mainly produced by fibroblasts, and recent evidence points toward a role of tumor-derived collagens in tumor progression and metastasis. In this review, we focus on the newly discovered functions of collagens in cancer. Novel findings have revealed the role of collagens in tumor dormancy and immune evasion, as well as their interplay with cancer cell metabolism. Collagens could serve as prognostic markers for patients with cancer, and therapeutic strategies targeting the collagen ECM have the potential to prevent tumor progression and metastasis.
Collapse
Affiliation(s)
- Daniela De Martino
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
9
|
Chivu-Economescu M, Necula LG, Matei L, Dragu D, Bleotu C, Sorop A, Herlea V, Dima S, Popescu I, Diaconu CC. Collagen Family and Other Matrix Remodeling Proteins Identified by Bioinformatics Analysis as Hub Genes Involved in Gastric Cancer Progression and Prognosis. Int J Mol Sci 2022; 23:ijms23063214. [PMID: 35328635 PMCID: PMC8950589 DOI: 10.3390/ijms23063214] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer has remained in the top five cancers for over ten years, both in terms of incidence and mortality due to the shortage of biomarkers for disease follow-up and effective therapies. Aiming to fill this gap, we performed a bioinformatics assessment on our data and two additional GEO microarray profiles, followed by a deep analysis of the 40 differentially expressed genes identified. PPI network analysis and MCODE plug-in pointed out nine upregulated hub genes coding for proteins from the collagen family (COL12A1, COL5A2, and COL10A1) or involved in the assembly (BGN) or degradation of collagens (CTHRC1), and also associated with cell adhesion (THBS2 and SPP1) and extracellular matrix degradation (FAP, SULF1). Those genes were highly upregulated at the mRNA and protein level, the increase being correlated with pathological T stages. The high expression of BGN (p = 8 × 10−12), THBS2 (p = 1.2 × 10−6), CTHRC1 (p = 1.1 × 10−4), SULF1 (p = 3.8 × 10−4), COL5A1 (p = 1.3 × 10−4), COL10A1 (p = 5.7 × 10−4), COL12A1 (p = 2 × 10−3) correlated with poor overall survival and an immune infiltrate based especially on immunosuppressive M2 macrophages (p-value range 4.82 × 10−7–1.63 × 10−13). Our results emphasize that these genes could be candidate biomarkers for GC progression and prognosis and new therapeutic targets.
Collapse
Affiliation(s)
- Mihaela Chivu-Economescu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.G.N.); (L.M.); (D.D.); (C.B.); (C.C.D.)
- Correspondence: or ; Tel.: +40-21-324-2592
| | - Laura G. Necula
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.G.N.); (L.M.); (D.D.); (C.B.); (C.C.D.)
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania; (V.H.); (I.P.)
| | - Lilia Matei
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.G.N.); (L.M.); (D.D.); (C.B.); (C.C.D.)
| | - Denisa Dragu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.G.N.); (L.M.); (D.D.); (C.B.); (C.C.D.)
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.G.N.); (L.M.); (D.D.); (C.B.); (C.C.D.)
| | - Andrei Sorop
- Center of Excellence for Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.S.); (S.D.)
| | - Vlad Herlea
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania; (V.H.); (I.P.)
- Department of Pathology, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Simona Dima
- Center of Excellence for Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania; (A.S.); (S.D.)
- Center of General Surgery and Liver Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Irinel Popescu
- Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania; (V.H.); (I.P.)
- Center of General Surgery and Liver Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Carmen C. Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (L.G.N.); (L.M.); (D.D.); (C.B.); (C.C.D.)
| |
Collapse
|