1
|
Hou C, Huo J, Yan S, Sun F, Yang X. Identification of fibrosis-associated biomarkers in heart failure and human cancers. J Transl Med 2024; 22:1042. [PMID: 39563337 PMCID: PMC11575019 DOI: 10.1186/s12967-024-05759-7] [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: 03/23/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Heart failure (HF) and cancer share common risk factors and pathophysiological mechanisms, including fibrosis. Identifying biomarkers and therapeutic targets for both conditions is crucial. MATERIALS AND METHODS RNA sequencing data from HF patients were analyzed to identify 12 genes associated with myocardial fibrosis. Validation was performed using public datasets, and functional enrichment analyses were conducted. Gene expression patterns and prognostic value in various cancers were assessed. RESULTS Fibromodulin (FMOD), Periostin (POSTN), Latent Transforming Growth Factor Beta Binding Protein 2 (LTBP2), Collagen Type I Alpha 1 Chain (COL1A1), Collagen Type VIII Alpha 1 Chain (COL8A1), Asporin (ASPN), and Hemoglobin Subunit Beta (HBB) showed significant dysregulation in heart failure tissues and were implicated in multiple cancer types. Pan-cancer analysis revealed associations between these genes and prognosis. Correlations with cancer-associated fibroblasts were also observed. CONCLUSION FMOD, POSTN, LTBP2, COL1A1, COL8A1, ASPN, and HBB are potential biomarkers for HF and cancer with fibrotic microenvironments. Targeting fibrosis may offer novel therapeutic approaches. Further validation and mechanistic studies are needed. This study contributes to understanding HF and cancer at the molecular level and suggests personalized treatment strategies.
Collapse
Affiliation(s)
- Can Hou
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213000, China
| | - Junyu Huo
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213000, China
| | - Si Yan
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213000, China
| | - Fei Sun
- Department of Radiotherapy, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, 213000, China.
| | - Xiaoyu Yang
- Department of Cardiovascular Medicine, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, 213000, China.
| |
Collapse
|
2
|
Lall SP, Alsafwani ZW, Batra SK, Seshacharyulu P. ASPORIN: A root of the matter in tumors and their host environment. Biochim Biophys Acta Rev Cancer 2024; 1879:189029. [PMID: 38008263 PMCID: PMC10872503 DOI: 10.1016/j.bbcan.2023.189029] [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: 09/10/2023] [Revised: 11/16/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
Asporin (ASPN) has been identified as one of the members of the class I small leucine-rich proteoglycans (SLRPs) family in the extracellular matrix (ECM). It is involved in classic ensigns of cancers such as self-dependent growth, resistance to growth inhibitors, restricting apoptosis, cancer metastasis, and bone-related disorders. ASPN is different from other members of SLRPs, such as decorin (DCN) and biglycan (BGN), in a way that it contains a distinctive length of aspartate (D) residues in the amino (N) -terminal region. These D-repeats residues possess germline polymorphisms and are identified to be linked with cancer progression and osteoarthritis (OA). The polyaspartate stretch in the N-terminal region of the protein and its resemblance to DCN are the reasons it is called asporin. In this review, we comprehensively summarized and updated the dual role of ASPN in various malignancies, its structure in mice and humans, variants, mutations, cancer-associated signalings and functions, the relationship between ASPN and cancer-epithelial, stromal fibroblast crosstalk, immune cells and immunosuppression in cancer and other diseases. In cancer and other bone-related diseases, ASPN is identified to be regulating various signaling pathways such as TGFβ, Wnt/β-catenin, notch, hedgehog, EGFR, HER2, and CD44-mediated Rac1. These pathways promote cancer cell invasion, proliferation, and migration by mediating the epithelial-to-mesenchymal transition (EMT) process. Finally, we discussed mouse models mimicking ASPN in vivo function in cancers and the probability of therapeutic targeting of ASPN in cancer cells, fibrosis, and other bone-related diseases.
Collapse
Affiliation(s)
- Shobhit P Lall
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Zahraa W Alsafwani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| |
Collapse
|
3
|
The Expression of ARMCX1 in Gastric Cancer Contributes to Prognosis and Influences Chemotherapy. J Immunol Res 2023; 2023:2623317. [PMID: 36726491 PMCID: PMC9886469 DOI: 10.1155/2023/2623317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/26/2022] [Indexed: 01/24/2023] Open
Abstract
The altered expression of ARMCX1 in patients with gastric cancer has been reported frequently, yet its correlation to prognosis and chemotherapy needs to be unveiled. In combination of the gene expression data retrieved from TCGA database and bioinformatic analysis, this study discovered 590 differentially expressed genes in the cancerous biopsies isolated from gastric patients, compared with controls. Among which, ARMCX1 exhibited great potential to serve as a prognostic biomarker for gastric patients; furthermore, patients with low expression of ARMCX1 could be more sensitive to these 9 chemotherapeutic agents: A-770041, AMG-706, ATRA, BEZ235, bortezomib, CGP60474, dasatinib, HG-64-1, and pazopanib, rather than the other chemotherapeutic agents. This study helps the improvement of evaluating the prognosis of gastric cancer patients, and would help optimize chemotherapeutic strategies in consideration of the expression of ARMCX1.
Collapse
|
4
|
Li H, Zhu D, Yang Y, Ma Y, Chen Y, Xue P, Chen J, Qin M, Xu D, Cai C, Cheng H. Determinants of DNMT2/TRDMT1 preference for substrates tRNA and DNA during the evolution. RNA Biol 2023; 20:875-892. [PMID: 37966982 PMCID: PMC10653749 DOI: 10.1080/15476286.2023.2272473] [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] [Accepted: 10/12/2023] [Indexed: 11/17/2023] Open
Abstract
RNA methyltransferase DNMT2/TRDMT1 is the most conserved member of the DNMT family from bacteria to plants and mammals. In previous studies, we found some determinants for tRNA recognition of DNMT2/TRDMT1, but the preference mechanism of this enzyme for substrates tRNA and DNA remains to be explored. In the present study, CFT-containing target recognition domain (TRD) and target recognition extension domain (TRED) in DNMT2/TRDMT1 play a crucial role in the substrate DNA and RNA selection during the evolution. Moreover, the classical substrate tRNA for DNMT2/TRDMT1 had a characteristic sequence CUXXCAC in the anticodon loop. Position 35 was occupied by U, making cytosine-38 (C38) twist into the loop, whereas C, G or A was located at position 35, keeping the C38-flipping state. Hence, the substrate preference could be modulated by the easily flipped state of target cytosine in tRNA, as well as TRD and TRED. Additionally, DNMT2/TRDMT1 cancer mutant activity was collectively mediated by five enzymatic characteristics, which might impact gene expressions. Importantly, G155C, G155V and G155S mutations reduced enzymatic activities and showed significant associations with diseases using seven prediction methods. Altogether, these findings will assist in illustrating the substrate preference mechanism of DNMT2/TRDMT1 and provide a promising therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Huari Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Daiyun Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yapeng Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yunfei Ma
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Yong Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Pingfang Xue
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Juan Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Mian Qin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Dandan Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Chao Cai
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Hongjing Cheng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
5
|
Fan R, Yan X, Zhang W. Relationship between asporin and extracellular matrix behavior: A literature review. Medicine (Baltimore) 2022; 101:e32490. [PMID: 36595867 PMCID: PMC9794316 DOI: 10.1097/md.0000000000032490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Asporin (ASPN), as a member of the small leucine-rich repeat proteoglycan family, is a type of protein that is found in the extracellular matrix. Collagen deposition or transformation is involved in a variety of pathological processes. ASPN is identified in cancerous tissue, pathological cardiac tissue, articular cartilage, keloid, and fibrotic lung tissue, and it has a role in the development of cancer, cardiovascular, bone and joint, keloid, and pulmonary fibrosis by interfering with collagen metabolism. This review article summarizes the data on ASPN expressions in mouse and human and highlights that overexpress of ASPN might play a role in a variety of diseases. Although our knowledge of ASPN is currently limited, these instances may help us better understand how it interacts with diseases.
Collapse
Affiliation(s)
- Rui Fan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Shandong, China
| | - Xiaoyan Yan
- Department of Geriatrics, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, China
- * Correspondence: Wei Zhang, Department of Respiratory and Critical Care Medicine, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong 250014, China (e-mail: )
| |
Collapse
|
6
|
Karagiorgou Z, Fountas PN, Manou D, Knutsen E, Theocharis AD. Proteoglycans Determine the Dynamic Landscape of EMT and Cancer Cell Stemness. Cancers (Basel) 2022; 14:5328. [PMID: 36358747 PMCID: PMC9653992 DOI: 10.3390/cancers14215328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 03/15/2024] Open
Abstract
Proteoglycans (PGs) are pivotal components of extracellular matrices, involved in a variety of processes such as migration, invasion, morphogenesis, differentiation, drug resistance, and epithelial-to-mesenchymal transition (EMT). Cellular plasticity is a crucial intermediate phenotypic state acquired by cancer cells, which can modulate EMT and the generation of cancer stem cells (CSCs). PGs affect cell plasticity, stemness, and EMT, altering the cellular shape and functions. PGs control these functions, either by direct activation of signaling cascades, acting as co-receptors, or through regulation of the availability of biological compounds such as growth factors and cytokines. Differential expression of microRNAs is also associated with the expression of PGs and their interplay is implicated in the fine tuning of cancer cell phenotype and potential. This review summarizes the involvement of PGs in the regulation of EMT and stemness of cancer cells and highlights the molecular mechanisms.
Collapse
Affiliation(s)
- Zoi Karagiorgou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Panagiotis N. Fountas
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9010 Tromsø, Norway
- Centre for Clinical Research and Education, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
7
|
Tanaka M. Crosstalk of tumor stromal cells orchestrates invasion and spreading of gastric cancer. Pathol Int 2022; 72:219-233. [PMID: 35112770 DOI: 10.1111/pin.13211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/18/2022] [Indexed: 01/22/2023]
Abstract
Tumors contain various stromal cells that support cancer progression. Some types of cancer, such as scirrhous gastric cancer, are characterized by large areas of fibrosis accompanied by cancer-associated fibroblasts (CAFs). Asporin (ASPN) is a small leucine-rich proteoglycan highly expressed in CAFs of various tumors. ASPN accelerates CAF migration and invasion, resulting in CAF-led cancer cell invasion. In addition, ASPN further upregulated the expression of genes specific to a characteristic subgroup of fibroblasts in tumors. These cells were preferentially located at the tumor periphery and could be generated by a unique mechanism involving the CAF-mediated education of normal fibroblasts (CEFs). In this review, we at first describe recent findings regarding the function of ASPN in the tumor microenvironment, as well as the mechanism involved in the generation of CEFs. CAFs are derived from heterogeneous origins besides resident normal fibroblasts. Among them, CAFs derived from mesothelial cells (mesothelial cell-derived CAF [MC-CAFs]) play pivotal roles in peritoneal carcinomatosis. We observed that MC-CAFs on the surfaces of organs also participate in tumor formation by infiltrating into the parenchyma, promoting local invasion by gastric cancers. This review also highlights the potential functions of macrophages in the formation of MC-CAFs in gastric cancers, by transfer the contents of cancer cell-derived extracellular vesicles.
Collapse
Affiliation(s)
- Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
8
|
Guo Y, Ning B, Zhang Q, Ma J, Zhao L, Lu Q, Zhang D. Identification of Hub Diagnostic Biomarkers and Candidate Therapeutic Drugs in Heart Failure. Int J Gen Med 2022; 15:623-635. [PMID: 35058712 PMCID: PMC8765546 DOI: 10.2147/ijgm.s349235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/31/2021] [Indexed: 01/08/2023] Open
Abstract
Purpose The objective of this study was to identify the potential regulatory mechanisms, diagnostic biomarkers, and therapeutic drugs for heart failure (HF). Methods Differentially expressed genes (DEGs) between HF and non-failing donors were screened from the GSE57345, GSE5406, and GSE3586 datasets. Database for Annotation Visualization and Integrated Discovery and Metascape were used for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses respectively. The GSE57345 dataset was used for weighted gene co-expression network analysis (WGCNA). The intersecting hub genes from the DEGs and WGCNA were identified and verified with the GSE5406 and GSE3586 datasets. The diagnostic value of the hub genes was calculated through receiver operating characteristic analysis and net reclassification index (NRI). Gene set enrichment analysis (GSEA) was used to filter out the signaling pathways associated with the hub genes. SYBYL 2.1 was used for molecular docking of hub targets and potential HF drugs obtained from the connection map. Results Functional annotation of the DEGs showed enrichment of negative regulation of angiogenesis, endoplasmic reticulum stress response, and heart development. PTN, LUM, ISLR, and ASPN were identified as the hub genes of HF. GSEA showed that the key genes were related to the transforming growth factor-β (TGF-β) and Wnt signaling pathways. Sirolimus, LY-294002, and wortmannin have been confirmed as potential drugs for HF. Conclusion We identified new hub genes and candidate therapeutic drugs for HF, which are potential diagnostic, therapeutic and prognostic targets and warrant further investigation.
Collapse
Affiliation(s)
- Yang Guo
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Department of Eco-Environmental Engineering, Qinghai University, Xining, 810016, People's Republic of China
| | - Bobin Ning
- Department of Medicine, The General Hospital of the People's Liberation Army, Beijing, 100038, People's Republic of China
| | - Qunhui Zhang
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Department of Eco-Environmental Engineering, Qinghai University, Xining, 810016, People's Republic of China
| | - Jing Ma
- Department of Eco-Environmental Engineering, Qinghai University, Xining, 810016, People's Republic of China
| | - Linlin Zhao
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Department of Eco-Environmental Engineering, Qinghai University, Xining, 810016, People's Republic of China
| | - QiQin Lu
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Qinghai-Utah Joint Research Key Lab for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Department of Eco-Environmental Engineering, Qinghai University, Xining, 810016, People's Republic of China
| | - Dejun Zhang
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, 810001, People's Republic of China.,Department of Eco-Environmental Engineering, Qinghai University, Xining, 810016, People's Republic of China
| |
Collapse
|
9
|
Basak D, Jamal Z, Ghosh A, Mondal PK, Dey Talukdar P, Ghosh S, Ghosh Roy B, Ghosh R, Halder A, Chowdhury A, Dhali GK, Chattopadhyay BK, Saha ML, Basu A, Roy S, Mukherjee C, Biswas NK, Chatterji U, Datta S. Reciprocal interplay between asporin and decorin: Implications in gastric cancer prognosis. PLoS One 2021; 16:e0255915. [PMID: 34379688 PMCID: PMC8357146 DOI: 10.1371/journal.pone.0255915] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
Effective patient prognosis necessitates identification of novel tumor promoting drivers of gastric cancer (GC) which contribute to worsened conditions by analysing TCGA-gastric adenocarcinoma dataset. Small leucine-rich proteoglycans, asporin (ASPN) and decorin (DCN), play overlapping roles in development and diseases; however, the mechanisms underlying their interplay remain elusive. Here, we investigated the complex interplay of asporin, decorin and their interaction with TGFβ in GC tumor and corresponding normal tissues. The mRNA levels, protein expressions and cellular localizations of ASPN and DCN were analyzed using real-time PCR, western blot and immunohistochemistry, respectively. The protein-protein interaction was predicted by in-silico interaction analysis and validated by co-immunoprecipitation assay. The correlations between ASPN and EMT proteins, VEGF and collagen were achieved using western blot analysis. A significant increase in expression of ASPN in tumor tissue vs. normal tissue was observed in both TCGA and our patient cohort. DCN, an effective inhibitor of the TGFβ pathway, was negatively correlated with stages of GC. Co-immunoprecipitation demonstrated that DCN binds with TGFβ, in normal gastric epithelium, whereas in GC, ASPN preferentially binds TGFβ. Possible activation of the canonical TGFβ pathway by phosphorylation of SMAD2 in tumor tissues suggests its role as an intracellular tumor promoter. Furthermore, tissues expressing ASPN showed unregulated EMT signalling. Our study uncovers ASPN as a GC-promoting gene and DCN as tumor suppressor, suggesting that ASPN can act as a prognostic marker in GC. For the first time, we describe the physical interaction of TGFβ with ASPN in GC and DCN with TGFβ in GC and normal gastric epithelium respectively. This study suggests that prevention of ASPN-TGFβ interaction or overexpression of DCN could serve as promising therapeutic strategies for GC patients.
Collapse
Affiliation(s)
- Dipjit Basak
- Human Genetics Unit, Indian Statistical Institute, Kolkata, India
| | - Zarqua Jamal
- Cancer Research Lab, Department of Zoology, University of Calcutta, Kolkata, India
| | - Arnab Ghosh
- National Institute of Biomedical Genomics, Kalyani, India
| | | | | | - Semanti Ghosh
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India
| | | | - Ranajoy Ghosh
- The School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Aniket Halder
- The School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Abhijit Chowdhury
- The School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Gopal Krishna Dhali
- The School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | | | - Makhan Lal Saha
- Department of Surgery, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Abhimanyu Basu
- Department of Surgery, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Sukanta Roy
- The School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | | | | | - Urmi Chatterji
- Cancer Research Lab, Department of Zoology, University of Calcutta, Kolkata, India
| | - Shalini Datta
- Human Genetics Unit, Indian Statistical Institute, Kolkata, India
- * E-mail:
| |
Collapse
|
10
|
Itoh G, Takagane K, Fukushi Y, Kuriyama S, Umakoshi M, Goto A, Yanagihara K, Yashiro M, Tanaka M. Cancer-associated fibroblasts educate normal fibroblasts to facilitate cancer cell spreading and T cell suppression. Mol Oncol 2021; 16:166-187. [PMID: 34379869 PMCID: PMC8732346 DOI: 10.1002/1878-0261.13077] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 11/11/2022] Open
Abstract
In some tumors, a small number of cancer cells are scattered in a large fibrotic stroma. Here, we demonstrate a novel mechanism for expansion of pro‐tumor fibroblasts via cancer‐associated fibroblast (CAF)‐mediated education of normal fibroblasts (NFs). When NFs were incubated with conditioned medium from CAFs, the resulting CAF‐educated fibroblasts (CEFs) generated reactive oxygen species, which induced NF‐κB‐mediated expression of inflammatory cytokines and the extracellular matrix protein asporin (ASPN), while expression of a common CAF marker gene, α‐SMA, was not increased. ASPN further increased CEF expression of downstream molecules, including indoleamine 2,3‐dioxygenase 1 (IDO‐1), kynureninase (KYNU), and pregnancy‐associated plasma protein‐A (PAPP‐A). These CEFs induce cytocidal effects against CD8+ T cells and IGF‐I activation in cancer cells. CEFs were generated without cancer cells by the direct mixture of NFs and CAFs in mouse xenografts, and once CEFs were generated, they sequentially educated NFs, leading to continuous generation of CEFs. In diffuse‐type gastric cancers, ASPNhigh/IDO‐1high/KYNUhigh/α‐SMA− CEFs were located at the distal invading front. These CEFs expanded in the fibrotic stroma and caused dissemination of cancer cells. ASPN may therefore be a key molecule in facilitating tumor spreading and T‐cell suppression.
Collapse
Affiliation(s)
- Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Kurara Takagane
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Yuma Fukushi
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.,Department of Life Science, Faculty and Graduate School of Engineering and Resource Science, Akita University, 1-1 Tegata Gakuenmachi, Akita, 010-8502, Japan
| | - Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Michinobu Umakoshi
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8545, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| |
Collapse
|
11
|
Liu L, Yu H, Long Y, You Z, Ogawa R, Du Y, Huang C. Asporin inhibits collagen matrix-mediated intercellular mechanocommunications between fibroblasts during keloid progression. FASEB J 2021; 35:e21705. [PMID: 34105826 DOI: 10.1096/fj.202100111r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 05/17/2021] [Indexed: 01/03/2023]
Abstract
Keloids are fibrotic lesions that grow unceasingly and invasively and are driven by local mechanical stimuli. Unlike other fibrotic diseases and normal wound healing, keloids exhibit little transformation of dermal fibroblasts into α-SMA+ myofibroblasts. This study showed that asporin is the most strongly expressed gene in keloids and its gene-ontology terms relate strongly to ECM metabolism/organization. Experiments with human dermal cells (HDFs) showed that asporin overexpression/treatment abrogated the HDF ability to adopt a perpendicular orientation when subjected to stretching tension. It also induced calcification of the surrounding 3D collagen matrix. Asporin overexpression/treatment also prevented the HDFs from remodeling the surrounding 3D collagen matrix, leading to a disorganized network of thick, wavy collagen fibers that resembled keloid collagen architecture. This in turn impaired the ability of the HDFs to contract the collagen matrix. Asporin treatment also made the fibroblasts impervious to the fibrous collagen contraction of α-SMA+ myofibroblasts, which normally activates fibroblasts. Thus, by calcifying collagen, asporin prevents fibroblasts from linearly rearranging the surrounding collagen; this reduces both their mechanosensitivity and mechanosignaling to each other through the collagen network. This blocks fibroblast activation and differentiation into the mature myofibroblasts that efficiently remodel the extracellular matrix. Consequently, the fibroblasts remain immature, highly proliferative, and continue laying down abundant extracellular matrix, causing keloid growth and invasion. Notably, dermal injection of asporin-overexpressing HDFs into murine wounds recapitulated keloid collagen histopathological characteristics. Thus, disrupted interfibroblast mechanocommunication may promote keloid progression. Asporin may be a new diagnostic biomarker and therapeutic target for keloids.
Collapse
Affiliation(s)
- Longwei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yi Long
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Chenyu Huang
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Yashiro M, Hasegawa T, Yamamoto Y, Tsujio G, Nishimura S, Sera T, Sugimoto A, Kushiyama S, Kasashima H, Fukuoka T, Sakurai K, Toyokawa T, Kubo N, Ohira M. Asporin Expression on Stromal Cells and/or Cancer Cells Might Be A Useful Prognostic Marker in Patients with Diffuse-Type Gastric Cancer. Eur Surg Res 2021; 62:53-60. [PMID: 33882483 DOI: 10.1159/000515458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/22/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Asporin (ASPN), a member of the proteoglycan family, has been shown to have a close correlation with cancer progression. It is not known whether ASPN is an oncogenic driver or a tumor suppressor in human gastric cancer. We sought herein to determine the relationship between ASPN expression and clinicopathological features of gastric cancer. PATIENTS AND METHODS A total of 296 gastric cancer patients (diffuse type, n = 144; intestinal type, n = 152) were enrolled. The ASPN expression level in each case was analyzed by immunohistochemistry. RESULTS ASPN was mainly found on stromal cells, especially on fibroblasts in tumor stroma, i.e., cancer-associated fibroblasts. The ASPN expression on either cancer cells or stromal cells was significantly high in macroscopic scirrhous-type tumors (p < 0.001) and histologically abundant stroma-type tumors (p < 0.001). Interestingly, a Kaplan-Meier survival curve of the 144 cases of diffuse-type gastric cancer revealed a significantly poorer prognosis in patients with ASPN-positive expression (p = 0.043; log rank) compared to those with ASPN-negative expression, but the prognoses were not significantly different in these subgroups of the 152 cases of intestinal-type gastric cancer. A multivariate analysis with respect to overall survival showed that ASPN expression on stromal cells and/or cancer cells was significantly correlated with overall survival in patients with diffuse-type gastric cancer (p = 0.041). CONCLUSION In gastric cancer, ASPN was expressed mainly on stromal cells and partially on cancer cells. ASPN expression on stromal cells and/or cancer cells might be a useful prognostic marker in patients with diffuse-type gastric cancer.
Collapse
Affiliation(s)
- Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Tsuyoshi Hasegawa
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Yurie Yamamoto
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Gen Tsujio
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Sadaaki Nishimura
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Tomohiro Sera
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Atsushi Sugimoto
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Shuhei Kushiyama
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Hiroaki Kasashima
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Tatsunari Fukuoka
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan.,Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Katsunobu Sakurai
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Takahiro Toyokawa
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Naoshi Kubo
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| | - Masaichi Ohira
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka City, Japan
| |
Collapse
|
13
|
Mickova A, Kharaishvili G, Kurfurstova D, Gachechiladze M, Kral M, Vacek O, Pokryvkova B, Mistrik M, Soucek K, Bouchal J. Skp2 and Slug Are Coexpressed in Aggressive Prostate Cancer and Inhibited by Neddylation Blockade. Int J Mol Sci 2021; 22:ijms22062844. [PMID: 33799604 PMCID: PMC8000894 DOI: 10.3390/ijms22062844] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related deaths in men in Western countries, and there is still an urgent need for a better understanding of PCa progression to inspire new treatment strategies. Skp2 is a substrate-recruiting component of the E3 ubiquitin ligase complex, whose activity is regulated through neddylation. Slug is a transcriptional repressor involved in the epithelial-to-mesenchymal transition, which may contribute to therapy resistance. Although Skp2 has previously been associated with a mesenchymal phenotype and prostate cancer progression, the relationship with Slug deserves further elucidation. We have previously shown that a high Gleason score (≥8) is associated with higher Skp2 and lower E-cadherin expression. In this study, significantly increased expression of Skp2, AR, and Slug, along with E-cadherin downregulation, was observed in primary prostate cancer in patients who already had lymph node metastases. Skp2 was slightly correlated with Slug and AR in the whole cohort (Rs 0.32 and 0.37, respectively), which was enhanced for both proteins in patients with high Gleason scores (Rs 0.56 and 0.53, respectively) and, in the case of Slug, also in patients with metastasis to lymph nodes (Rs 0.56). Coexpression of Skp2 and Slug was confirmed in prostate cancer tissues by multiplex immunohistochemistry and confocal microscopy. The same relationship between these two proteins was observed in three sets of prostate epithelial cell lines (PC3, DU145, and E2) and their mesenchymal counterparts. Chemical inhibition of Skp2, but not RNA interference, modestly decreased Slug protein in PC3 and its docetaxel-resistant subline PC3 DR12. Importantly, chemical inhibition of Skp2 by MLN4924 upregulated p27 and decreased Slug expression in PC3, PC3 DR12, and LAPC4 cells. Novel treatment strategies targeting Skp2 and Slug by the neddylation blockade may be promising in advanced prostate cancer, as recently documented for other aggressive solid tumors.
Collapse
Affiliation(s)
- Alena Mickova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, 779 00 Olomouc, Czech Republic; (A.M.); (D.K.); (M.G.)
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic;
| | - Gvantsa Kharaishvili
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, 779 00 Olomouc, Czech Republic; (A.M.); (D.K.); (M.G.)
- Correspondence: (G.K.); (J.B.)
| | - Daniela Kurfurstova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, 779 00 Olomouc, Czech Republic; (A.M.); (D.K.); (M.G.)
| | - Mariam Gachechiladze
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, 779 00 Olomouc, Czech Republic; (A.M.); (D.K.); (M.G.)
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic;
| | - Milan Kral
- Department of Urology, University Hospital, 779 00 Olomouc, Czech Republic;
| | - Ondrej Vacek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 65 Brno, Czech Republic; (O.V.); (K.S.)
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne’s University Hospital in Brno, 602 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Barbora Pokryvkova
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, 252 50 Vestec, Czech Republic;
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic;
| | - Karel Soucek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 612 65 Brno, Czech Republic; (O.V.); (K.S.)
- International Clinical Research Center, Center for Biomolecular and Cellular Engineering, St. Anne’s University Hospital in Brno, 602 00 Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, 779 00 Olomouc, Czech Republic; (A.M.); (D.K.); (M.G.)
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic;
- Correspondence: (G.K.); (J.B.)
| |
Collapse
|
14
|
Sasaki Y, Takagane K, Konno T, Itoh G, Kuriyama S, Yanagihara K, Yashiro M, Yamada S, Murakami S, Tanaka M. Expression of asporin reprograms cancer cells to acquire resistance to oxidative stress. Cancer Sci 2021; 112:1251-1261. [PMID: 33393151 PMCID: PMC7935789 DOI: 10.1111/cas.14794] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022] Open
Abstract
Asporin (ASPN), a small leucine‐rich proteoglycan expressed predominantly by cancer associated fibroblasts (CAFs), plays a pivotal role in tumor progression. ASPN is also expressed by some cancer cells, but its biological significance is unclear. Here, we investigated the effects of ASPN expression in gastric cancer cells. Overexpression of ASPN in 2 gastric cancer cell lines, HSC‐43 and 44As3, led to increased migration and invasion capacity, accompanied by induction of CD44 expression and activation of Rac1 and MMP9. ASPN expression increased resistance of HSC‐43 cells to oxidative stress by reducing the amount of mitochondrial reactive oxygen species. ASPN induced expression of the transcription factor HIF1α and upregulated lactate dehydrogenase A (LDHA) and PDH‐E1α, suggesting that ASPN reprograms HSC‐43 cells to undergo anaerobic glycolysis and suppresses ROS generation in mitochondria, which has been observed in another cell line HSC‐44PE. By contrast, 44As3 cells expressed high levels of HIF1α in response to oxidant stress and escaped apoptosis regardless of ASPN expression. Examination of xenografts in the gastric wall of ASPN–/– mice revealed that growth of HSC‐43 tumors with increased micro blood vessel density was significantly accelerated by ASPN; however, ASPN increased the invasion depth of both HSC‐43 and 44As3 tumors. These results suggest that ASPN has 2 distinct effects on cancer cells: HIF1α‐mediated resistance to oxidative stress via reprogramming of glucose metabolism, and activation of CD44‐Rac1 and MMP9 to promote cell migration and invasion. Therefore, ASPN may be a new therapeutic target in tumor fibroblasts and cancer cells in some gastric carcinomas.
Collapse
Affiliation(s)
- Yuto Sasaki
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan.,Department of Life Science, Faculty and Graduate School of Engineering and Resource Science, Akita University, Akita, Japan
| | - Kurara Takagane
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Takumi Konno
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan.,Department of Life Science, Faculty and Graduate School of Engineering and Resource Science, Akita University, Akita, Japan
| | - Go Itoh
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Sei Kuriyama
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuyoshi Yanagihara
- Division of Biomarker Discovery, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Masakazu Yashiro
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Satoru Yamada
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Department of Periodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Shinya Murakami
- Department of Periodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masamitsu Tanaka
- Department of Molecular Medicine and Biochemistry, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
15
|
The Tumor Microenvironment as a Driving Force of Breast Cancer Stem Cell Plasticity. Cancers (Basel) 2020; 12:cancers12123863. [PMID: 33371274 PMCID: PMC7766255 DOI: 10.3390/cancers12123863] [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: 11/29/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Breast cancer stem cells are a subset of transformed cells that sustain tumor growth and can metastasize to secondary organs. Since metastasis accounts for most cancer deaths, it is of paramount importance to understand the cellular and molecular mechanisms that regulate this subgroup of cells. The tumor microenvironment (TME) is the habitat in which transformed cells evolve, and it is composed by many different cell types and the extracellular matrix (ECM). A body of evidence strongly indicates that microenvironmental cues modulate stemness in breast cancer, and that the coevolution of the TME and cancer stem cells determine the fate of breast tumors. In this review, we summarize the studies providing links between the TME and the breast cancer stem cell phenotype and we discuss their specific interactions with immune cell subsets, stromal cells, and the ECM. Abstract Tumor progression involves the co-evolution of transformed cells and the milieu in which they live and expand. Breast cancer stem cells (BCSCs) are a specialized subset of cells that sustain tumor growth and drive metastatic colonization. However, the cellular hierarchy in breast tumors is rather plastic, and the capacity to transition from one cell state to another depends not only on the intrinsic properties of transformed cells, but also on the interplay with their niches. It has become evident that the tumor microenvironment (TME) is a major player in regulating the BCSC phenotype and metastasis. The complexity of the TME is reflected in its number of players and in the interactions that they establish with each other. Multiple types of immune cells, stromal cells, and the extracellular matrix (ECM) form an intricate communication network with cancer cells, exert a highly selective pressure on the tumor, and provide supportive niches for BCSC expansion. A better understanding of the mechanisms regulating these interactions is crucial to develop strategies aimed at interfering with key BCSC niche factors, which may help reducing tumor heterogeneity and impair metastasis.
Collapse
|
16
|
Hummitzsch K, Hatzirodos N, Macpherson AM, Schwartz J, Rodgers RJ, Irving-Rodgers HF. Transcriptome analyses of ovarian stroma: tunica albuginea, interstitium and theca interna. Reproduction 2020; 157:545-565. [PMID: 30925461 DOI: 10.1530/rep-18-0323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 03/29/2019] [Indexed: 01/15/2023]
Abstract
The ovary has specialised stromal compartments, including the tunica albuginea, interstitial stroma and theca interna, which develops concurrently with the follicular antrum. To characterise the molecular determinants of these compartments, stroma adjacent to preantral follicles (pre-theca), interstitium and tunica albuginea were laser microdissected (n = 4 per group) and theca interna was dissected from bovine antral follicles (n = 6). RNA microarray analysis showed minimal differences between interstitial stroma and pre-theca, and these were combined for some analyses and referred to as stroma. Genes significantly upregulated in theca interna compared to stroma included INSL3, LHCGR, HSD3B1, CYP17A1, ALDH1A1, OGN, POSTN and ASPN. Quantitative RT-PCR showed significantly greater expression of OGN and LGALS1 in interstitial stroma and theca interna versus tunica and greater expression of ACD in tunica compared to theca interna. PLN was significantly higher in interstitial stroma compared to tunica and theca. Ingenuity pathway, network and upstream regulator analyses were undertaken. Cell survival was also upregulated in theca interna. The tunica albuginea was associated with GPCR and cAMP signalling, suggesting tunica contractility. It was also associated with TGF-β signalling and increased fibrous matrix. Western immunoblotting was positive for OGN, LGALS1, ALDH1A1, ACD and PLN with PLN and OGN highly expressed in tunica and interstitial stroma (each n = 6), but not in theca interna from antral follicles (n = 24). Immunohistochemistry localised LGALS1 and POSTN to extracellular matrix and PLN to smooth muscle cells. These results have identified novel differences between the ovarian stromal compartments.
Collapse
Affiliation(s)
- Katja Hummitzsch
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas Hatzirodos
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anne M Macpherson
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jeff Schwartz
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| | - Raymond J Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Helen F Irving-Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia.,School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
17
|
Zhan S, Li J, Ge W. Multifaceted Roles of Asporin in Cancer: Current Understanding. Front Oncol 2019; 9:948. [PMID: 31608236 PMCID: PMC6771297 DOI: 10.3389/fonc.2019.00948] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
The small leucine-rich proteoglycan (SLRP) family consists of 18 members categorized into five distinct classes, the traditional classes I–III, and the non-canonical classes IV–V. Unlike the other class I SLRPs (decorin and biglycan), asporin contains a unique and conserved stretch of aspartate (D) residues in its N terminus, and germline polymorphisms in the D-repeat-length are associated with osteoarthritis and prostate cancer progression. Since the first discovery of asporin in 2001, previous studies have focused mainly on its roles in bone and joint diseases, including osteoarthritis, intervertebral disc degeneration and periodontal ligament mineralization. Recently, asporin gene expression was also reported to be dysregulated in tumor tissues of different types of cancer, and to act as oncogene in pancreatic, colorectal, gastric, and prostate cancers, and some types of breast cancer, though it is also reported to function as a tumor suppressor gene in triple-negative breast cancer. Furthermore, asporin is also positively or negatively correlated with tumor proliferation, migration, invasion, and patient prognosis through its regulation of different signaling pathways, including the TGF-β, EGFR, and CD44 pathways. In this review, we seek to elucidate the signaling pathways and functions regulated by asporin in different types of cancer and to highlight some important issues that require investigation in future research.
Collapse
Affiliation(s)
- Shaohua Zhan
- National Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China.,National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China.,Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
18
|
Hughes RM, Simons BW, Khan H, Miller R, Kugler V, Torquato S, Theodros D, Haffner MC, Lotan T, Huang J, Davicioni E, An SS, Riddle RC, Thorek DLJ, Garraway IP, Fertig EJ, Isaacs JT, Brennen WN, Park BH, Hurley PJ. Asporin Restricts Mesenchymal Stromal Cell Differentiation, Alters the Tumor Microenvironment, and Drives Metastatic Progression. Cancer Res 2019; 79:3636-3650. [PMID: 31123087 PMCID: PMC6734938 DOI: 10.1158/0008-5472.can-18-2931] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 04/17/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
Tumor progression to metastasis is not cancer cell autonomous, but rather involves the interplay of multiple cell types within the tumor microenvironment. Here we identify asporin (ASPN) as a novel, secreted mesenchymal stromal cell (MSC) factor in the tumor microenvironment that regulates metastatic development. MSCs expressed high levels of ASPN, which decreased following lineage differentiation. ASPN loss impaired MSC self-renewal and promoted terminal cell differentiation. Mechanistically, secreted ASPN bound to BMP-4 and restricted BMP-4-induced MSC differentiation prior to lineage commitment. ASPN expression was distinctly conserved between MSC and cancer-associated fibroblasts (CAF). ASPN expression in the tumor microenvironment broadly impacted multiple cell types. Prostate tumor allografts in ASPN-null mice had a reduced number of tumor-associated MSCs, fewer cancer stem cells, decreased tumor vasculature, and an increased percentage of infiltrating CD8+ T cells. ASPN-null mice also demonstrated a significant reduction in lung metastases compared with wild-type mice. These data establish a role for ASPN as a critical MSC factor that extensively affects the tumor microenvironment and induces metastatic progression. SIGNIFICANCE: These findings show that asporin regulates key properties of mesenchymal stromal cells, including self-renewal and multipotency, and asporin expression by reactive stromal cells alters the tumor microenvironment and promotes metastatic progression.
Collapse
Affiliation(s)
- Robert M Hughes
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Brian W Simons
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Hamda Khan
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rebecca Miller
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Valentina Kugler
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Samantha Torquato
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Debebe Theodros
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michael C Haffner
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Tamara Lotan
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jessie Huang
- The Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Elai Davicioni
- Genome Dx Biosciences, Inc., Vancouver, British Columbia, Canada
| | - Steven S An
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- The Whiting School of Engineering, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Ryan C Riddle
- The Department of Orthopedic Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Daniel L J Thorek
- The Department of Radiology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Isla P Garraway
- The Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Elana J Fertig
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John T Isaacs
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - W Nathaniel Brennen
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ben H Park
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Whiting School of Engineering, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Paula J Hurley
- The James Buchanan Brady Urological Institute, Department of Urology, Johns Hopkins School of Medicine, Baltimore, Maryland.
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Li H, Zhang Z, Chen L, Sun X, Zhao Y, Guo Q, Zhu S, Li P, Min L, Zhang S. Cytoplasmic Asporin promotes cell migration by regulating TGF-β/Smad2/3 pathway and indicates a poor prognosis in colorectal cancer. Cell Death Dis 2019; 10:109. [PMID: 30728352 PMCID: PMC6365561 DOI: 10.1038/s41419-019-1376-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/29/2018] [Accepted: 01/17/2019] [Indexed: 01/10/2023]
Abstract
Previous studies revealed that Asporin (ASPN) is a potential mediator in the development of various types of cancer as a secreted stroma protein, but the function of ASPN inside the cancer cells remains largely unknown. Here, we demonstrated a higher expression level of ASPN in colorectal cancer (CRC) than matched normal tissues, and 25% (2/8) CRC showed copy number variation (CNV) gain/amplification in ASPN gene. Both higher ASPN expression levels and ASPN CNV gain/amplification indicated a worse prognosis in CRC patients. ASPN can promote proliferation, migration, and invasion of CRC cells, and inhibit apoptosis by activating Akt/Erk and TGF-β/Smad2/3 signalings. Further investigations revealed that ASPN interacts with Smad2/3, facilitates its translocation into nucleus, and up-regulates the expression of Epithelial-mesenchymal transition (EMT) related genes. Rescue assays confirmed that TGF-β signaling is essential for the effects of ASPN on promoting CRC cell migration and invasion. In conclusion, ASPN promotes the migration and invasion of CRC cells via TGF-β/Smad2/3 pathway and could serve as a potential prognostic biomarker in CRC patients.
Collapse
Affiliation(s)
- Hengcun Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Zheng Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Lei Chen
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Xiujing Sun
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Yu Zhao
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Qingdong Guo
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Peng Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China.
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, 100050, Beijing, P. R. China.
| |
Collapse
|
20
|
Maccarana M, Svensson RB, Knutsson A, Giannopoulos A, Pelkonen M, Weis M, Eyre D, Warman M, Kalamajski S. Asporin-deficient mice have tougher skin and altered skin glycosaminoglycan content and structure. PLoS One 2017; 12:e0184028. [PMID: 28859141 PMCID: PMC5578652 DOI: 10.1371/journal.pone.0184028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/16/2017] [Indexed: 11/24/2022] Open
Abstract
The main structural component of connective tissues is fibrillar, cross-linked collagen whose fibrillogenesis can be modulated by Small Leucine-Rich Proteins/Proteoglycans (SLRPs). Not all SLRPs’ effects on collagen and extracellular matrix in vivo have been elucidated; one of the less investigated SLRPs is asporin. Here we describe the successful generation of an Aspn-/- mouse model and the investigation of the Aspn-/- skin phenotype. Functionally, Aspn-/- mice had an increased skin mechanical toughness, although there were no structural changes present on histology or immunohistochemistry. Electron microscopy analyses showed 7% thinner collagen fibrils in Aspn-/- mice (not statistically significant). Several matrix genes were upregulated, including collagens (Col1a1, Col1a2, Col3a1), matrix metalloproteinases (Mmp2, Mmp3) and lysyl oxidases (Lox, Loxl2), while lysyl hydroxylase (Plod2) was downregulated. Intriguingly no differences were observed in collagen protein content or in collagen cross-linking-related lysine oxidation or hydroxylation. The glycosaminoglycan content and structure in Aspn-/- skin was profoundly altered: chondroitin/dermatan sulfate was more than doubled and had an altered composition, while heparan sulfate was halved and had a decreased sulfation. Also, decorin and biglycan were doubled in Aspn-/- skin. Overall, asporin deficiency changes skin glycosaminoglycan composition, and decorin and biglycan content, which may explain the changes in skin mechanical properties.
Collapse
Affiliation(s)
- Marco Maccarana
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - René B. Svensson
- Institute of Sports Medicine, Bispebjerg Hospital, and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Anki Knutsson
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Antonis Giannopoulos
- Institute of Sports Medicine, Bispebjerg Hospital, and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Mea Pelkonen
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - MaryAnn Weis
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - David Eyre
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington, United States of America
| | - Matthew Warman
- Children’s Hospital Boston, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sebastian Kalamajski
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
21
|
Wang L, Wu H, Wang L, Zhang H, Lu J, Liang Z, Liu T. Asporin promotes pancreatic cancer cell invasion and migration by regulating the epithelial-to-mesenchymal transition (EMT) through both autocrine and paracrine mechanisms. Cancer Lett 2017; 398:24-36. [PMID: 28400334 DOI: 10.1016/j.canlet.2017.04.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/13/2017] [Accepted: 04/01/2017] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer is histopathologically characterized by excessive desmoplasia induced by pancreatic stellate cells (PSCs). Asporin, an extracellular matrix (ECM) protein, is highly expressed in cancer-associated fibroblasts (CAFs). Asporin expression in PSCs and its roles in PSC-pancreatic cancer cell (PCC) interaction remain unclear. The present study firstly showed that Asporin is highly expressed in activated PSCs and is involved in PSC-mediated invasion and migration of PCCs. Exogenous Asporin interacted with the transmembrane receptor CD44 on PCCs to activate NF-κB/p65 and promoted the epithelial-mesenchymal transition (EMT) in PCCs. Furthermore, AKT and ERK pathways participated in Asporin/CD44-induced NF-κB/p65 activation in pancreatic cancer. Asporin had similar effects on PCCs via an autocrine mechanism. Consistent with our in vitro experiments, we showed that Asporin in peritumoral stroma of pancreatic cancer tissues was associated with poor clinical outcome. In conclusion, this is the first study to show that Asporin promotes EMT, invasion, and migration of PCCs by activating CD44-AKT/ERK-NF-κB pathway in paracrine and autocrine manners. Moreover, our results indicate that Asporin may be a prognostic marker and suggest that targeting the tumor microenvironment represents a promising therapeutic strategy in pancreatic cancer.
Collapse
Affiliation(s)
- Lili Wang
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Huanwen Wu
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Li Wang
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hui Zhang
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Junliang Lu
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhiyong Liang
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Tonghua Liu
- Molecular Pathology Research Center, Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
22
|
Kahounová Z, Kurfürstová D, Bouchal J, Kharaishvili G, Navrátil J, Remšík J, Šimečková Š, Študent V, Kozubík A, Souček K. The fibroblast surface markers FAP, anti-fibroblast, and FSP are expressed by cells of epithelial origin and may be altered during epithelial-to-mesenchymal transition. Cytometry A 2017; 93:941-951. [PMID: 28383825 DOI: 10.1002/cyto.a.23101] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/13/2017] [Accepted: 03/07/2017] [Indexed: 12/15/2022]
Abstract
The identification of fibroblasts and cancer-associated fibroblasts from human cancer tissue using surface markers is difficult, especially because the markers used currently are usually not expressed solely by fibroblasts, and the identification of fibroblast-specific surface molecules is still under investigation. It was aimed to compare three commercially available antibodies in the detection of different surface epitopes of fibroblasts (anti-fibroblast, fibroblast activation protein α, and fibroblast surface protein). The specificity of their expression, employing fibroblast cell lines and tumor-derived fibroblasts from breast and prostate tissues was investigated. Both the established fibroblast cell line HFF-1 and ex vivo primary fibroblasts isolated from breast and prostate cancer tissues expressed the tested surface markers to different degrees. Surprisingly, those markers were expressed also by permanent cell lines of epithelial origin, both benign and cancer-derived (breast-cell lines MCF 10A, HMLE and prostate-cell lines BPH-1, DU 145, and PC-3). The expression of fibroblast activation protein α increased on the surface of previously described models of epithelial cells undergoing epithelial-to-mesenchymal transition in response to treatment with TGF-β1. To prove the co-expression of the fibroblast markers on cells of epithelial origin, we used freshly dissociated human prostate and breast cancer tissues. The results confirmed the co-expression of anti-fibroblast and fibroblast surface protein on CD31/CD45-negative/EpCAM-positive epithelial cells. In summary, our data support the findings that the tested fibroblast markers are not fibroblast specific and may be expressed also by cells of epithelial origin (e.g., cells undergoing EMT). Therefore, the expression of these markers should be interpreted with caution, and the combination of several epitopes for both positive (anti-fibroblast or fibroblast activation protein α) and negative (EpCAM) identification of fibroblasts from breast and prostate tumor tissues is advised. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Zuzana Kahounová
- Department of Cytokinetics, Institute of Biophysics of the CAS, v.v.i, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Daniela Kurfürstová
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Gvantsa Kharaishvili
- Department of Clinical and Molecular Pathology, Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jiří Navrátil
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Ján Remšík
- Department of Cytokinetics, Institute of Biophysics of the CAS, v.v.i, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Šárka Šimečková
- Department of Cytokinetics, Institute of Biophysics of the CAS, v.v.i, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Vladimír Študent
- Department of Urology, University Hospital Olomouc, Olomouc, Czech Republic
| | - Alois Kozubík
- Department of Cytokinetics, Institute of Biophysics of the CAS, v.v.i, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karel Souček
- Department of Cytokinetics, Institute of Biophysics of the CAS, v.v.i, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
23
|
Rochette A, Boufaied N, Scarlata E, Hamel L, Brimo F, Whitaker HC, Ramos-Montoya A, Neal DE, Dragomir A, Aprikian A, Chevalier S, Thomson AA. Asporin is a stromally expressed marker associated with prostate cancer progression. Br J Cancer 2017; 116:775-784. [PMID: 28152543 PMCID: PMC5355923 DOI: 10.1038/bjc.2017.15] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Prostate cancer shows considerable heterogeneity in disease progression and we propose that markers expressed in tumour stroma may be reliable predictors of aggressive tumour subtypes. METHODS We have used Kaplan-Meier, univariate and multivariate analysis to correlate the expression of Asporin (ASPN) mRNA and protein with prostate cancer progression in independent cohorts. We used immunohistochemistry and H scoring to document stromal localisation of ASPN in a tissue microarray and mouse prostate cancer model, and correlated expression with reactive stroma, defined using Masson Trichrome staining. We used cell cultures of primary prostate cancer fibroblasts treated with serum-free conditioned media from prostate cancer cell lines to examine regulation of ASPN mRNA in tumour stromal cells. RESULTS We observed increased expression of ASPN mRNA in a data set derived from benign vs tumour microdissected tissue, and a correlation with biochemical recurrence using Kaplan-Meier and Cox proportional hazard analysis. ASPN protein localised to tumour stroma and elevated expression of ASPN was correlated with decreased time to biochemical recurrence, in a cohort of 326 patients with a median follow up of 9.6 years. Univariate and multivariate analysis demonstrated that ASPN was correlated with progression, as were Gleason score, and clinical stage. Additionally, ASPN expression correlated with the presence of reactive stroma, suggesting that it may be a stromal marker expressed in response to the presence of tumour cells and particularly with aggressive tumour subtypes. We observed expression of ASPN in the stroma of tumours induced by p53 inhibition in a mouse model of prostate cancer, and correlation with neuroendocrine marker expression. Finally, we demonstrated that ASPN transcript expression in normal and cancer fibroblasts was regulated by conditioned media derived from the PC3, but not LNCaP, prostate cancer cell lines. CONCLUSIONS Our results suggest that ASPN is a stromally expressed biomarker that correlates with disease progression, and is observed in reactive stroma. ASPN expression in stroma may be part of a stromal response to aggressive tumour subtypes.
Collapse
Affiliation(s)
- Annie Rochette
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Nadia Boufaied
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Eleonora Scarlata
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Lucie Hamel
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Fadi Brimo
- Department of Pathology, Division of Urology, McGill University and The McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Hayley C Whitaker
- Department of Oncology, University of Cambridge, Box 279, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Antonio Ramos-Montoya
- Department of Oncology, University of Cambridge, Box 279, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - David E Neal
- Department of Oncology, University of Cambridge, Box 279, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Alice Dragomir
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Armen Aprikian
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Simone Chevalier
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Axel A Thomson
- Department of Surgery, Division of Urology, McGill University and the Cancer Research Program of the Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| |
Collapse
|
24
|
Simkova D, Kharaishvili G, Slabakova E, Murray PG, Bouchal J. Glycoprotein asporin as a novel player in tumour microenvironment and cancer progression. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160:467-473. [PMID: 27605398 DOI: 10.5507/bp.2016.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/08/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Small leucine rich proteoglycans (SLRPs), major non-collagen components of the extracellular matrix (ECM), have multiple biological roles with diverse effects. Asporin, a member of the SLRPs class I, competes with other molecules in binding to collagen and affects its mineralization. Its role in cancer is only now being elucidated. METHODS The PubMed online database was used to search relevant reviews and original articles. Furthermore, altered asporin expression was analysed in publicly available genome-wide expression data at the Gene Expression Omnibus database. RESULTS Polymorphisms in the N-terminal polyaspartate domain, which binds calcium, are associated with osteoarthritis and prostate cancer. Asporin also promotes the progression of scirrhous gastric cancer where it is required for coordinated invasion by cancer associated fibroblasts and cancer cells. Besides the enhanced expression of asporin observed in multiple cancer types, such as breast, prostate, gastric, pancreas and colon cancer, tumour suppressive effects of asporin were described in triple-negative breast cancer. We also discuss a number of factors modulating asporin expression in different cell types relevant for alterations toing the tumour microenvironment. CONCLUSION The apparent contradicting tumour promoting and suppressive effects of asporin require further investigation. Deciphering the role of asporin and other SLRPs in tumour-stroma interactions is needed for a better understanding of cancer progression and potentially also for novel tumour microenvironment based therapies.
Collapse
Affiliation(s)
- Dana Simkova
- Department of Clinical and Molecular Pathology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Gvantsa Kharaishvili
- Department of Clinical and Molecular Pathology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Eva Slabakova
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i., Brno, Czech Republic
| | - Paul G Murray
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Jan Bouchal
- Department of Clinical and Molecular Pathology and Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|