1
|
Bentaberry-Rosa A, Nicaise Y, Delmas C, Gouazé-Andersson V, Cohen-Jonathan-Moyal E, Seva C. Overexpression of Growth Differentiation Factor 15 in Glioblastoma Stem Cells Promotes Their Radioresistance. Cancers (Basel) 2023; 16:27. [PMID: 38201456 PMCID: PMC10778311 DOI: 10.3390/cancers16010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
GSCs play an important role in GBM recurrence. Understanding the resistance mechanisms in these cells is therefore crucial for radiation therapy optimization. In this study, using patient-derived GSCs, we demonstrate that GDF15, a cytokine belonging to the TGF-β superfamily, is regulated by irradiation (IR) and the transcription factor WWTR1/TAZ. Blocking WWTR1/TAZ using specific siRNAs significantly reduces GDF15 basal expression and reverses the upregulation of this cytokine induced by IR. Furthermore, we demonstrate that GDF15 plays an important role in GSC radioresistance. Targeting GDF15 expression by siRNA in GSCs expressing high levels of GDF15 sensitizes the cells to IR. In addition, we also found that GDF15 expression is critical for GSC spheroid formation, as GDF15 knockdown significantly reduces the number of GSC neurospheres. This study suggests that GDF15 targeting in combination with radiotherapy may be a feasible approach in patients with GBM.
Collapse
Affiliation(s)
- Alexandre Bentaberry-Rosa
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31062 Toulouse, France; (A.B.-R.); (Y.N.); (C.D.); (V.G.-A.); (E.C.-J.-M.)
- IUCT-Oncopole, 31100 Toulouse, France
| | - Yvan Nicaise
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31062 Toulouse, France; (A.B.-R.); (Y.N.); (C.D.); (V.G.-A.); (E.C.-J.-M.)
| | - Caroline Delmas
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31062 Toulouse, France; (A.B.-R.); (Y.N.); (C.D.); (V.G.-A.); (E.C.-J.-M.)
- IUCT-Oncopole, 31100 Toulouse, France
| | - Valérie Gouazé-Andersson
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31062 Toulouse, France; (A.B.-R.); (Y.N.); (C.D.); (V.G.-A.); (E.C.-J.-M.)
- IUCT-Oncopole, 31100 Toulouse, France
| | - Elizabeth Cohen-Jonathan-Moyal
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31062 Toulouse, France; (A.B.-R.); (Y.N.); (C.D.); (V.G.-A.); (E.C.-J.-M.)
- IUCT-Oncopole, 31100 Toulouse, France
| | - Catherine Seva
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31062 Toulouse, France; (A.B.-R.); (Y.N.); (C.D.); (V.G.-A.); (E.C.-J.-M.)
| |
Collapse
|
2
|
Chang TY, Lan KC, Wu CH, Sheu ML, Yang RS, Liu SH. Nε-(1-Carboxymethyl)-L-lysine, an advanced glycation end product, exerts malignancy on chondrosarcoma via the activation of cancer stemness. Arch Toxicol 2023; 97:2231-2244. [PMID: 37314482 DOI: 10.1007/s00204-023-03539-8] [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: 01/04/2023] [Accepted: 06/06/2023] [Indexed: 06/15/2023]
Abstract
Despite epidemiological evidence that suggests diabetes mellitus is a risk factor for cancer, the link between diabetes mellitus and primary bone cancer is rarely discussed. Chondrosarcomas are primary malignant cartilage tumors with poor prognosis and high metastatic potential. It remains unclear whether hyperglycemia affects the stemness and malignancy of chondrosarcoma cells. Nε-(1-Carboxymethyl)-L-lysine (CML), an advanced glycation end product (AGE), is a major immunological epitope detected in the tissue proteins of diabetic patients. We hypothesized that CML could enhance cancer stemness in chondrosarcoma cells. CML enhanced tumor-sphere formation and the expression of cancer stem cell markers in human chondrosarcoma cell lines. Migration and invasion ability and the epithelial-mesenchymal transition (EMT) process were also induced by CML treatment. Moreover, CML increased the protein expression levels of the receptor for AGE (RAGE), phosphorylated NFκB-p65, and decreased the phosphorylation of AKT and GSK-3. We also found that hyperglycemia with high CML levels facilitated tumor metastasis, whereas tumor growth was not affected in the streptozotocin (STZ)-induced diabetic NOD/SCID tumor xenograft mouse models. Our results indicate that CML enhances chondrosarcoma stemness and metastasis, which may reveal the relationship between AGE and bone cancer metastasis.
Collapse
Affiliation(s)
- Ting-Yu Chang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Hung Wu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Rong-Sen Yang
- Department of Orthopedics, National Taiwan University Hospital, Taipei, Taiwan
| | - Shing-Hwa Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
- Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei, Taiwan.
| |
Collapse
|
3
|
Ganjoo S, Puebla-Osorio N, Nanez S, Hsu E, Voss T, Barsoumian H, Duong LK, Welsh JW, Cortez MA. Bone morphogenetic proteins, activins, and growth and differentiation factors in tumor immunology and immunotherapy resistance. Front Immunol 2022; 13:1033642. [PMID: 36353620 PMCID: PMC9638036 DOI: 10.3389/fimmu.2022.1033642] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2024] Open
Abstract
The TGF-β superfamily is a group of secreted polypeptides with key roles in exerting and regulating a variety of physiologic effects, especially those related to cell signaling, growth, development, and differentiation. Although its central member, TGF-β, has been extensively reviewed, other members of the family-namely bone morphogenetic proteins (BMPs), activins, and growth and differentiation factors (GDFs)-have not been as thoroughly investigated. Moreover, although the specific roles of TGF-β signaling in cancer immunology and immunotherapy resistance have been extensively reported, little is known of the roles of BMPs, activins, and GDFs in these domains. This review focuses on how these superfamily members influence key immune cells in cancer progression and resistance to treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria Angelica Cortez
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
4
|
Pathophysiological role of growth differentiation factor 15 (GDF15) in obesity, cancer, and cachexia. Cytokine Growth Factor Rev 2021; 64:71-83. [PMID: 34836750 DOI: 10.1016/j.cytogfr.2021.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 02/08/2023]
Abstract
Growth differentiation factor 15 or macrophage inhibitory cytokine-1 (GDF15/MIC-1) is a divergent member of the transforming growth factor β superfamily and has a diverse pathophysiological roles in cancers, cardiometabolic disorders, and other diseases. GDF15 controls hematopoietic growth, energy homeostasis, adipose tissue metabolism, body growth, bone remodeling, and response to stress signals. The role of GDF15 in cancer development and progression is complicated and depends on the specific cancer type, stage, and tumor microenvironment. Recently, research on GDF15 and GDF15-associated signaling has accelerated due to the identification of the GDF15 receptor: glial cell line-derived neurotrophic factor (GDNF) family receptor α-like (GFRAL). Therapeutic interventions to target GDF15 and/or GFRAL revealed the mechanisms that drive its activity and might improve overall outcomes of patients with metabolic disorders and cancer. This review highlights the structure and functions of GDF15 and its receptor, emphasizing the pleiotropic role of GDF15 in obesity, tumorigenesis, metastasis, immunomodulation, and cachexia.
Collapse
|
5
|
Buchholz K, Antosik P, Grzanka D, Gagat M, Smolińska M, Grzanka A, Gzil A, Kasperska A, Klimaszewska-Wiśniewska A. Expression of the Body-Weight Signaling Players: GDF15, GFRAL and RET and their clinical relevance in Gastric Cancer. J Cancer 2021; 12:4698-4709. [PMID: 34149933 PMCID: PMC8210553 DOI: 10.7150/jca.55511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
The existence, the functional role and clinical relevance of GDF15 and its signaling through a GFRAL/RET-dependent complex in gastric cancer (GC) and other human tumors remain to be elucidated, despite the widespread recognition of obesity as an important cancer-predisposing factor. Therefore, we aimed to analyze the expression levels of GDF15, GFRAL and RET in GC tissues in relation to each other and clinicopathological features, including patient survival, in order to establish a potential implication of the body-weight signaling pathway in the pathology and clinical outcome of GC. Protein expression was examined by immunohistochemistry on tissue microarrays containing 104 and 30 consecutive GC and normal gastric mucosa samples, whereas gene expression data for The Cancer Genome Atlas cohort of 413 GC patients were obtained from public sources. We found that the protein expression of GDF15, GFRAL and RET was significantly elevated and positively correlated in our set of GC tissues, which was reflected in their tendency to be overexpressed in low-grade and intermediate-grade tumors rather than high-grade ones. No other relationships between the expression status of the examined proteins and clinicopathological characteristics of GC patients were found. Through in silico data analysis, we showed that high GDF15 expression was associated with better overall survival (OS) of GC patients, whereas the opposite was true for high levels of GFRAL or RET. Specifically, GFRAL and RET emerged as independent prognostic factors associated with poor OS. Furthermore, high combined expression of the three markers: GDF15+GFRAL+RET was significantly associated with reduced OS, and it was an independent prognostic factor of borderline significance in terms of OS, when adjusted for covariates. If validated in large-scale studies, the individual and combined expression of GDF15, GFRAL and RET may provide significant clinical implications for the prognosis prediction of GC patients.
Collapse
Affiliation(s)
- Karolina Buchholz
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland.,Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Paulina Antosik
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Maciej Gagat
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Marta Smolińska
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Alina Grzanka
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Arkadiusz Gzil
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Anna Kasperska
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| | - Anna Klimaszewska-Wiśniewska
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
6
|
5-Aza-2'-Deoxycytidine and Valproic Acid in Combination with CHIR99021 and A83-01 Induce Pluripotency Genes Expression in Human Adult Somatic Cells. Molecules 2021; 26:molecules26071909. [PMID: 33805347 PMCID: PMC8036574 DOI: 10.3390/molecules26071909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/04/2021] [Accepted: 03/20/2021] [Indexed: 12/17/2022] Open
Abstract
A generation of induced pluripotent stem cells (iPSC) by ectopic expression of OCT4, SOX2, KLF4, and c-MYC has established promising opportunities for stem cell research, drug discovery, and disease modeling. While this forced genetic expression represents an advantage, there will always be an issue with genomic instability and transient pluripotency genes reactivation that might preclude their clinical application. During the reprogramming process, a somatic cell must undergo several epigenetic modifications to induce groups of genes capable of reactivating the endogenous pluripotency core. Here, looking to increase the reprograming efficiency in somatic cells, we evaluated the effect of epigenetic molecules 5-aza-2'-deoxycytidine (5AZ) and valproic acid (VPA) and two small molecules reported as reprogramming enhancers, CHIR99021 and A83-01, on the expression of pluripotency genes and the methylation profile of the OCT4 promoter in a human dermal fibroblasts cell strain. The addition of this cocktail to culture medium increased the expression of OCT4, SOX2, and KLF4 expression by 2.1-fold, 8.5-fold, and 2-fold, respectively, with respect to controls; concomitantly, a reduction in methylated CpG sites in OCT4 promoter region was observed. The epigenetic cocktail also induced the expression of the metastasis-associated gene S100A4. However, the epigenetic cocktail did not induce the morphological changes characteristic of the reprogramming process. In summary, 5AZ, VPA, CHIR99021, and A83-01 induced the expression of OCT4 and SOX2, two critical genes for iPSC. Future studies will allow us to precise the mechanisms by which these compounds exert their reprogramming effects.
Collapse
|
7
|
Takenaga K, Ochiya T, Endo H. Inhibition of the invasion and metastasis of mammary carcinoma cells by NBD peptide targeting S100A4 via the suppression of the Sp1/MMP‑14 axis. Int J Oncol 2021; 58:397-408. [PMID: 33650647 PMCID: PMC7864152 DOI: 10.3892/ijo.2021.5173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/12/2020] [Indexed: 12/30/2022] Open
Abstract
A synthetic peptide that blocks the interaction between the metastasis‑enhancing calcium‑binding protein, S100A4, and its effector protein, methionine aminopeptidase 2 (MetAP2) (the NBD peptide), was previously demonstrated to inhibit the angiogenesis of endothelial cells, leading to the regression of human prostate cancer in a xenograft model. However, the effects of the NBD peptide on the malignant properties of cancer cells that express S100A4 remain to be elucidated. The present study demonstrates that the NBD peptide inhibits the invasiveness and metastasis of highly metastatic human mammary carcinoma cells. The introduction of the peptide into MDA‑MB‑231 variant cells resulted in the suppression of matrix degradation in a gelatin invadopodia assay and invasiveness in a Matrigel invasion assay. In line with these results, the peptide significantly downregulated the expression of matrix metalloproteinase (MMP)‑14 (MT1‑MMP). Mechanistic analysis of the downregulation of MMP‑14 revealed the suppression of the expression of the transcription factor, specificity protein 1 (Sp1), but not that of nuclear factor (NF)‑κB, early growth response 1 (EGR1) or ELK3, all of which were reported to be involved in transcriptional regulation of the MMP‑14 gene. At the same time, evidence suggested that the NBD peptide also suppressed Sp1 and MMP‑14 expression levels in MDA‑MB‑468 cells. Importantly, the intravenous administration of the NBD peptide encapsulated in liposomes inhibited pulmonary metastasis from mammary gland tumors in mice with xenograft tumors. These results indicate that the NBD peptide can suppress malignant tumor growth through the suppression of the Sp1/MMP‑14 axis. Taken together, these results reveal that the NBD peptide acts on not only endothelial cells, but also on tumor cells in an integrated manner, suggesting that the peptide may prove to be a promising cancer therapeutic peptide drug.
Collapse
Affiliation(s)
- Keizo Takenaga
- Department of Life Science, Faculty of Medicine, Shimane University, Shimane 690-0823
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo 160-0023
| | - Hideya Endo
- Division of Cellular and Molecular Biology, Department of Cancer Biology
- Division of Molecular Pathology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
8
|
GDF15 promotes glioma stem cell-like phenotype via regulation of ERK1/2-c-Fos-LIF signaling. Cell Death Discov 2021; 7:3. [PMID: 33431816 PMCID: PMC7801449 DOI: 10.1038/s41420-020-00395-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/05/2020] [Accepted: 11/19/2020] [Indexed: 01/01/2023] Open
Abstract
Growth differentiation factor 15 (GDF15), a member of the transforming growth factor β family, is associated with tumor progression, metastasis, and cell apoptosis. However, controversy persists regarding the role of GDF15 in different tumor types, and its function in glioma stem cells (GSCs) remains unknown. Here, we report that GDF15 promotes the GSC-like phenotype in GSC-like cells (GSCLCs) through the activation of leukemia inhibitor factor (LIF)–STAT3 signaling. Mechanistically, GDF15 was found to upregulate expression of the transcription factor c-Fos, which binds to the LIF promoter, leading to enhanced transcription of LIF in GSCLCs. Furthermore, GDF15 may activate the ERK1/2 signaling pathway in GSCLCs, and the upregulation of LIF expression and the GSC-like phenotype was dependent on ERK1/2 signaling. In addition, the small immunomodulator imiquimod induced GDF15 expression, which in turn activated the LIF–STAT3 pathway and subsequently promoted the GSC-like phenotype in GSCLCs. Thus, our results demonstrate that GDF15 can act as a proliferative and pro-stemness factor for GSCs, and therefore, it may represent a potential therapeutic target in glioma treatment.
Collapse
|
9
|
Bunwanna A, Damrongrungruang T, Puasiri S, Kantrong N, Chailertvanitkul P. Preservation of the viability and gene expression of human periodontal ligament cells by Thai propolis extract. Dent Traumatol 2020; 37:123-130. [PMID: 33185962 DOI: 10.1111/edt.12612] [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: 06/02/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND/AIM Success of tooth replantation depends on the quality and quantity of periodontal ligament (PDL) cells. The aims of this study were to evaluate Thai propolis extract as a storage medium for maintaining PDL cell viability and preserving gene expressions in PDL tissues. MATERIALS AND METHODS PDL cells from human premolars were tested for cytotoxicity of the extract by PrestoBlue assay to determine a non-toxic concentration. Subsequently, 96 freshly extracted premolars were allocated into different treatment groups. Control groups were freshly extracted premolars or they had been stored dry for 12 hours. Experimental avulsed teeth were created by leaving them air-dried for 30 minutes immediately after extraction, then they were immersed in Thai propolis extract, HBSS or milk for 3, 6 and 12 hours. After tooth storage, the remaining PDL cells were determined for their cell viability. RNA isolated from PDL tissues of three premolars treated similarly was analysed for periostin and S100A4 expressions using RT-qPCR. RESULTS Thai propolis extract at 0.625 mg mL-1 promoted the greatest PDL cell viability. Tooth storage in 0.625 mg mL-1 Thai propolis extract, HBSS or milk showed no difference in maintaining cell viability. Periostin mRNA level was preserved by Thai propolis extract. Expression of S100A4 mRNA in PDL tissues stored in all tested media was dampened. CONCLUSIONS PDL cells from mock avulsed teeth stored in 0.625 mg mL-1 Thai propolis extract for 3, 6 and 12 hours remained viable and the expression of periostin was preserved. This study suggests this extract as an alternative for a tooth storage medium for up to 12 hours. However, transporting an avulsed tooth in a storage medium for extended extra-oral time might affect the PDL cell phenotypes.
Collapse
Affiliation(s)
- Atittaya Bunwanna
- Department of Restorative Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | | | - Subin Puasiri
- Department of Preventive Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Nutthapong Kantrong
- Department of Restorative Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Pattama Chailertvanitkul
- Department of Restorative Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
10
|
GDF15 Repression Contributes to 5-Fluorouracil Resistance in Human Colon Cancer by Regulating Epithelial-Mesenchymal Transition and Apoptosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2826010. [PMID: 33062674 PMCID: PMC7542494 DOI: 10.1155/2020/2826010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Chemotherapy based on 5-fluorouracil (5-FU) is the standard approach for colon cancer treatment, and resistance to 5-FU is a significant obstacle in the clinical treatment of colon cancer. However, the mechanisms underlying 5-FU resistance in colon cancer cells remain largely unknown. This study aimed at determining whether 5-FU-resistant colon cancer cells undergo epithelial-mesenchymal transition (EMT) and apoptosis and the role of GDF15—a member of the transforming growth factor β/bone morphogenetic protein super family and a protein known to be involved in cancer progression—in the regulation of EMT and apoptosis of these cells, along with the underlying mechanisms. In vitro apoptosis detection assay, growth inhibition assay, transwell, and wound healing experiments revealed that 5-FU-resistant colon cancer cells possessed enhanced EMT and antiapoptotic ability. These cells also showed a stronger tendency to proliferate and metastasize in vivo. Quantitative reverse transcription-PCR and western blotting revealed that 5-FU-resistant colon cancer cells expressed lower levels of growth differentiation factor 15 (GDF15) than did 5-FU-sensitive colon cancer cells. Moreover, the transient GDF15 overexpression resensitized 5-FU-resistant colon cells to 5-FU. Collectively, these findings indicate the mechanism underlying the 5-FU resistance of colon cancer cells and provide new therapeutic targets for improving the prognosis of colon cancer patients.
Collapse
|
11
|
Chen L, Qiao L, Bian Y, Sun X. GDF15 knockdown promotes erastin-induced ferroptosis by decreasing SLC7A11 expression. Biochem Biophys Res Commun 2020; 526:293-299. [DOI: 10.1016/j.bbrc.2020.03.079] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/13/2020] [Indexed: 12/12/2022]
|
12
|
Shen W, Tan X, Hao F. S100A4 expression is associated with poor prognosis in patients with resectable gastrointestinal stromal tumor. Libyan J Med 2020; 14:1659669. [PMID: 31474194 PMCID: PMC6735325 DOI: 10.1080/19932820.2019.1659669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
S100A4 is particularly associated with the progression and metastasis of numerous human malignancies. This study was designed to examine the clinicopathologic significance of S100A4 in gastrointestinal stromal tumor (GISTs). The level of OPNS100A4 expression in a large cohort of resectable GISTs was evaluated with immunohistochemistry. Its correlation with the clinicopathologic parameters of patients with resectable GISTs was analyzed. A survival analysis was performed to evaluate the prognostic significance of S100A4 expression using Kaplan-Meier method. Results: In 108 patients with resectable GISTs, the most high-risk GISTs had a strong level of S100A4 expression. Strong S100A4 expression was significantly associated with tumor size, mitosis, and recurrence, but not gender and age. Patients with weak S100A4 expression had a relatively longer disease-free survival compared to patients with strong S100A4 expression.Therefore, S100A4 expression is a putative marker for tumor progression and an adverse prognosis in GISTs.
Collapse
Affiliation(s)
- Wenlong Shen
- Department of Anorectal, Qilu Hospital of Shandong University , Qingdao , Shandong , PR China
| | - Xiaojie Tan
- Department of General surgery, Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Fengyun Hao
- Department of Pathology, Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| |
Collapse
|
13
|
Li Y, Wang J, Song K, Liu S, Zhang H, Wang F, Ni C, Zhai W, Liang J, Qin Z, Zhang J. S100A4 promotes hepatocellular carcinogenesis by intensifying fibrosis-associated cancer cell stemness. Oncoimmunology 2020; 9:1725355. [PMID: 32117590 PMCID: PMC7028350 DOI: 10.1080/2162402x.2020.1725355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/03/2019] [Accepted: 11/25/2019] [Indexed: 02/07/2023] Open
Abstract
A cancer-promoting role of fibrogenesis in the liver has long been speculated; however, the molecular mechanisms regarding this phenomenon are largely unknown. We demonstrated in our previous study that macrophage-derived S100A4 promotes liver fibrosis via activation of hepatic stellate cells; however, whether and how S100A4 directly contributes to the development of fibrosis-associated liver cancer remains elusive. High expression of S100A4 in the fibrotic region was observed in human liver tumor tissues which associated with advanced disease severity. Through an established hepatocarcinogenesis model involving apparent liver fibrogenesis, we found that S100A4-deficient mice developed significantly less and smaller liver tumor nodules, with no change in the liver inflammation but decreased liver fibrosis and expression of stem cell markers in hepatocellular carcinoma (HCC) tissues. Mechanistically, S100A4 directly promoted stem cell-associated genes signatures in a way synergistic with its interacting protein, extracellular matrix component collagen I. This process is dependent on the receptor of advanced glycation end products (RAGE) and β-catenin signaling. Furthermore, the liver tumor sphere formation in vitro and tumor growth in vivo were greatly enhanced only when the cancer cells were pretreated with both S100A4 and collagen I. Our work firstly demonstrated a key role of S100A4 in synergy with extracellular matrix in the promotion of hepatocellular carcinoma by affecting the stemness of cancer cells.
Collapse
Affiliation(s)
- Yanan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jun Wang
- Department of Immunobiology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kun Song
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shuangqing Liu
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Huilei Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Fei Wang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chen Ni
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenlong Zhai
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jialu Liang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of Protein and Peptide Pharmaceuticals, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
14
|
Modi A, Dwivedi S, Roy D, Khokhar M, Purohit P, Vishnoi J, Pareek P, Sharma S, Sharma P, Misra S. Growth differentiation factor 15 and its role in carcinogenesis: an update. Growth Factors 2019; 37:190-207. [PMID: 31693861 DOI: 10.1080/08977194.2019.1685988] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Growth differentiation factor-15 (GDF-15) is a novel cytokine secreted by a variety of cells like macrophages, adipocytes, normally expressed in high amounts by placenta. It is also highly expressed in multiple carcinomas like Colon, Breast, Pancreas, Liver, and Ovarian. Several reports on serum GDF-15 as a potential biomarker for diagnosis and prognosis of cancer are hampered by the lack of robust data, with large sample size and critical patient recruitment. However, experimental accounts on cancer tumors, cell lines, and animal models suggest GDF-15's role in cancer progression via endothelial mesenchymal transition, angiogenesis, metastasis, drug resistance and even stemness of various cancers. GDF-15 could be the point of amalgamation for the various hallmarks of cancer and can prove a useful therapeutic target in cancer. The current review was conceptualized with a thought of critically appraising the existing information of GDF-15 in carcinogenesis.
Collapse
Affiliation(s)
- Anupama Modi
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | | | - Dipayan Roy
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Manoj Khokhar
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Purvi Purohit
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | | | - Puneet Pareek
- Department of Radiotherapy, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Shailja Sharma
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Praveen Sharma
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Sanjeev Misra
- Department of Oncosurgery, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
15
|
Fang L, Li F, Gu C. GDF-15: A Multifunctional Modulator and Potential Therapeutic Target in Cancer. Curr Pharm Des 2019; 25:654-662. [PMID: 30947652 DOI: 10.2174/1381612825666190402101143] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022]
Abstract
Various pathological processes are associated with the aberrant expression and function of cytokines, especially those belonging to the transforming growth factor-β (TGF-β) family. Nevertheless, the functions of members of the TGF-β family in cancer progression and therapy are still uncertain. Growth differentiation factor- 15, which exists in intracellular and extracellular forms, is classified as a divergent member of the TGF-β superfamily. It has been indicated that GDF-15 is also connected to the evolution of cancer both positively and negatively depending upon the cellular state and environment. Under normal physiological conditions, GDF-15 inhibits early tumour promotion. However, its abnormal expression in advanced cancers causes proliferation, invasion, metastasis, cancer stem cell formation, immune escape and a reduced response to therapy. As a clinical indicator, GDF-15 can be used as a tool for the diagnosis and therapy of an extensive scope of cancers. Although some basic functions of GDF-15 are noncontroversial, their mechanisms remain unclear and complicated at the molecular level. Therefore, GDF-15 needs to be further explored and reviewed.
Collapse
Affiliation(s)
- Lei Fang
- Department of Thoracic surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, China
| | - Fengzhou Li
- Department of Thoracic surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, China
| | - Chundong Gu
- Department of Thoracic surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, China
| |
Collapse
|
16
|
Liu J, Dong X, Lu J, Zhang Y, Liu K, Liu L, E Q, Lu X, Yin J, Shen Y. Utility of GDF-15 as a diagnostic biomarker in gastric cancer: an investigation combining GEO, TCGA and meta-analysis. FEBS Open Bio 2019; 9:35-42. [PMID: 30652072 PMCID: PMC6325603 DOI: 10.1002/2211-5463.12537] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/20/2018] [Accepted: 09/27/2018] [Indexed: 12/17/2022] Open
Abstract
It was recently suggested that growth differentiation factor-15 (GDF-15) is associated with gastric cancer (GC) carcinogenesis. However, the diagnostic potential of GDF-15 for GC remains unclear. To address this issue, we obtained RNA sequencing and microarray data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, and searched PubMed, Google Scholar and Web of Science for relevant literature. We then used STATA to perform a meta-analysis. In total, reports of 253 GC patients and 112 healthy controls who contributed peripheral blood samples were taken from the four literature sources, while information on 754 GC tumor and 263 gastric normal tissues was drawn from TCGA and seven GEO datasets. The expression level of GDF-15 mRNA was significantly higher in tumor tissues than in normal tissues, with a standard mean difference (SMD) of 0.79% and a 95% confidence interval (95% CI) of 0.63-0.95. Consistently, the GDF-15 protein in blood was significantly increased in GC patients as compared to controls (SMD = 3.74, 95% CI = 1.81-5.68). In addition, based on information from TCGA and GEO datasets, the expression level of GDF-15 mRNA may be of use for the diagnosis of GC, with a combined sensitivity, specificity and odds ratio of 0.69 (95% CI = 0.58-0.79), 0.90 (95% CI = 0.84-0.93) and 6.32 (95% CI = 4.22-9.49), respectively. The summary receiver operating characteristic curve demonstrated that the area under the curve was 0.90 (95% CI = 0.87-0.93). The results suggest higher levels of GDF-15 may be associated with GC tumorigenesis and may have the potential to be a diagnostic biomarker of GC.
Collapse
Affiliation(s)
- Jie‐yu Liu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Xing‐xuan Dong
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Jia‐nan Lu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Kai‐fan Liu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Ling‐feng Liu
- School of Basic MedicineMedical College of Soochow UniversitySuzhouChina
| | - Qing‐zhi E
- School of Basic MedicineMedical College of Soochow UniversitySuzhouChina
| | - Xiao‐jing Lu
- School of Basic MedicineMedical College of Soochow UniversitySuzhouChina
| | - Jie‐yun Yin
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| | - Yue‐ping Shen
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSchool of Public HealthMedical College of Soochow UniversitySuzhouChina
| |
Collapse
|
17
|
Bian Y, Guo J, Qiao L, Sun X. miR-3189-3p Mimics Enhance the Effects of S100A4 siRNA on the Inhibition of Proliferation and Migration of Gastric Cancer Cells by Targeting CFL2. Int J Mol Sci 2018; 19:ijms19010236. [PMID: 29342841 PMCID: PMC5796184 DOI: 10.3390/ijms19010236] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/27/2017] [Accepted: 01/08/2018] [Indexed: 12/17/2022] Open
Abstract
GDF15 is a downstream gene of S100A4. miR-3189 is embedded in the intron of GDF15—and coexpressed with it. miR-3189-3p functions to inhibit the proliferation and migration of glioblastoma cells. We speculated that S100A4 might regulate miR-3189-3p to affect its function in gastric cancer cells. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) showed that miR-3189-3p expression was significantly downregulated in MGC803 cells after S100A4 knockdown. Overexpression of miR-3189-3p significantly inhibited the proliferation and migration of the cells. Moreover, miR-3189-3p mimics enhanced the effects of an S100A4 siRNA on the inhibition of cell proliferation and migration. Dual luciferase reporter assays, qRT-PCR, and Western blotting verified that CFL2 is a direct target of miR-3189-3p. CFL2 mediates the regulation of miR-3189-3p on the proliferation and migration of MGC803 cells. Data mining based on Kaplan–Meier plots showed that high CFL2 expression is associated with poor overall survival and first progression in gastric cancer. These data suggested that miR-3189-3p mimics enhanced the effects of the S100A4 siRNA on the inhibition of gastric cancer cell proliferation and migration by targeting CFL2. The findings suggested that when targeting S100A4 to treat gastric cancer, consideration and correction for counteracting factors should obtain a satisfactory effect.
Collapse
Affiliation(s)
- Yue Bian
- Department of Medical Genetics, China Medical University, Shenyang 110122, China.
| | - Junfu Guo
- Department of Medical Genetics, China Medical University, Shenyang 110122, China.
- Teaching and Experiment Center, Liaoning University of Traditional Chinese Medicine, Shenyang110847, China.
| | - Linlin Qiao
- Department of Medical Genetics, China Medical University, Shenyang 110122, China.
| | - Xiuju Sun
- Department of Medical Genetics, China Medical University, Shenyang 110122, China.
| |
Collapse
|
18
|
Jiao J, González Á, Stevenson HL, Gagea M, Sugimoto H, Kalluri R, Beretta L. Depletion of S100A4 + stromal cells does not prevent HCC development but reduces the stem cell-like phenotype of the tumors. Exp Mol Med 2018; 50:e422. [PMID: 29303514 PMCID: PMC5992984 DOI: 10.1038/emm.2017.175] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023] Open
Abstract
There is a pressing need for the development of novel approaches to treat and prevent hepatocellular carcinoma (HCC). The S100 calcium-binding protein S100A4 is associated with poor prognosis and metastasis in several human cancers. In addition, a role for S100A4 in modulating cancer-initiating cells stemness properties was recently proposed in head and neck and gastric cancers. Whether S100A4+ stromal cells contribute to tumor onset remains, however, an unanswered question. To address that question, we generated a new mouse model allowing for the depletion of S100A4+ cells in a mouse model of HCC with stemness properties, by crossing mice with hepatic deletion of phosphatase and tensin homolog (PTEN) with mice expressing viral thymidine kinase under the control of S100A4 promoter. Depletion of S100A4+ cells by ganciclovir injection did not prevent the development of HCC but reduced the stemness phenotype of the tumor as measured by the expression of progenitor cell, biliary cell and hepatocyte markers. The results were further confirmed by histology analysis showing reduction of cholangiolar tumor components and degree of oval cell hyperplasia in the adjacent liver. Depletion of S100A4+ cells had also some beneficial effect on the underlying liver disease with a reduction of NAS score, largely due to the reduction of inflammation. In conclusion, this study demonstrated that S100A4+ cells do not contribute to HCC onset but maintain the stemness phenotype of the tumor. This study also suggests for the first time a crosstalk between inflammation and stemness.
Collapse
Affiliation(s)
- Jingjing Jiao
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Álvaro González
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather L Stevenson
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mihai Gagea
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
19
|
Guo J, Bian Y, Wang Y, Chen L, Yu A, Sun X. FAM107Bis regulated byS100A4and mediates the effect ofS100A4on the proliferation and migration of MGC803 gastric cancer cells. Cell Biol Int 2017; 41:1103-1109. [DOI: 10.1002/cbin.10816] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 07/01/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Junfu Guo
- Department of Medical Genetics; China Medical University; Shenyang Liaoning 110122 China
- Teaching and Experiment Center; Liaoning University of Traditional Chinese Medicine; Shenyang Liaoning 110847 China
| | - Yue Bian
- Department of Medical Genetics; China Medical University; Shenyang Liaoning 110122 China
| | - Yu Wang
- Department of Medical Genetics; China Medical University; Shenyang Liaoning 110122 China
| | - Lisha Chen
- Department of Medical Genetics; China Medical University; Shenyang Liaoning 110122 China
| | - Aiwen Yu
- Department of Medical Genetics; China Medical University; Shenyang Liaoning 110122 China
- Department of Rehabilitation; The First Affiliated Hospital of China Medical University; Shenyang Liaoning 110001 China
| | - Xiuju Sun
- Department of Medical Genetics; China Medical University; Shenyang Liaoning 110122 China
| |
Collapse
|
20
|
Fei F, Qu J, Zhang M, Li Y, Zhang S. S100A4 in cancer progression and metastasis: A systematic review. Oncotarget 2017; 8:73219-73239. [PMID: 29069865 PMCID: PMC5641208 DOI: 10.18632/oncotarget.18016] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/08/2017] [Indexed: 12/11/2022] Open
Abstract
Metastasis is the leading cause of cancer-related death and directly associates with cancer progression, resistance to anticancer therapy, and poor patient survival. Current efforts focusing on the underlying molecular mechanisms of cancer metastasis attract a special attention to cancer researchers. The epithelial-mesenchymal transition is a complex of molecular program during embryogenesis, inflammation, tissue fibrosis, and cancer progression and metastasis. S100A4, an important member of S100 family proteins, functions to increase the tumor progression and metastasis. The molecular mechanisms of S100A4 involving in the progression and metastasis are diverse in various malignant tumors. Detection of S100A4 expression becomes a promising candidate biomarker in cancer early diagnosis and prediction of cancer metastasis and therefore, S100A4 may be a therapeutic target. This review summarized up to date advancement on the role of S100A4 in human cancer development, progression, and metastasis and the underlying molecular events and then strategies to target S100A4 expression experimentally.
Collapse
Affiliation(s)
- Fei Fei
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R.China.,Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Jie Qu
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R.China.,Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Mingqing Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| |
Collapse
|