1
|
Xie Q, Shen Y, Yang Y, Liang J, Wu T, Hu C, Wang Y, Tao H. Identification of XD23 as a potent inhibitor of osteosarcoma via downregulation of DKK1 and activation of the WNT/β-catenin pathway. iScience 2024; 27:110758. [PMID: 39280613 PMCID: PMC11402217 DOI: 10.1016/j.isci.2024.110758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/13/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Osteosarcoma, the most prevalent malignant bone tumor, is notorious for its aggressive growth and invasiveness. The highly mutable genome of osteosarcoma has made identifying a key oncogene challenging, hindering the development of targeted treatments. Our study validates the effectiveness of XD23, an anti-cancer agent we previously identified, in curbing osteosarcoma proliferation, metastasis, EMT differentiation, and bone destruction and promoting osteosarcoma apoptosis. It further elucidated that XD23 thwarts osteosarcoma by suppressing DKK1 expression, which in turn activates the WNT-β/Catenin pathway. This research presents the concrete evidence of DKK1's involvement in osteosarcoma development, offering a foundation for the development of DKK1 inhibitors as novel treatments for this disease.
Collapse
Affiliation(s)
- Qian Xie
- Department of Orthopedics, Shenzhen University General Hospital, Shenzhen 518055, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yanni Shen
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yipei Yang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jianhui Liang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Tailin Wu
- Department of Orthopedics, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Chun Hu
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yan Wang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huiren Tao
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
2
|
Niedra H, Peculis R, Saksis R, Mandrika I, Vilisova S, Nazarovs J, Breiksa A, Gerina A, Earl J, Ruz-Caracuel I, Rosas MG, Pukitis A, Senterjakova N, Rovite V. Tumor and α-SMA-expressing stromal cells in pancreatic neuroendocrine tumors have a distinct RNA profile depending on tumor grade. Mol Oncol 2024. [PMID: 39245631 DOI: 10.1002/1878-0261.13727] [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: 09/07/2023] [Revised: 07/12/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Alpha-smooth muscle actin (α-SMA) expression in the stroma is linked to the presence of cancer-associated fibroblasts and is known to correlate with worse outcomes in various tumors. In this study, using a GeoMx digital spatial profiling approach, we characterized the gene expression of the tumor and α-SMA-expressing stromal cell compartments in pancreatic neuroendocrine tumors (PanNETs). The profiling was performed on tissues from eight retrospective cases (three grade 1, four grade 2, and one grade 3). Selected regions of interest were segmented geometrically based on tissue morphology and fluorescent signals from synaptophysin and α-SMA markers. The α-SMA-expressing stromal-cell-associated genes were involved in pathways of extracellular matrix modification, whereas, in tumor cells, the gene expression profiles were associated with pathways involved in cell proliferation. The comparison of gene expression profiles across all three PanNET grades revealed that the differences between grades are not only present at the level of the tumor but also in the α-SMA-expressing stromal cells. Furthermore, the tumor cells from regions with a rich presence of adjacent α-SMA-expressing stromal cells revealed an upregulation of matrix metalloproteinase-9 (MMP9) expression in grade 3 tumors. This study provides an in-depth characterization of gene expression profiles in α-SMA-expressing stromal and tumor cells, and outlines potential crosstalk mechanisms.
Collapse
Affiliation(s)
- Helvijs Niedra
- Department of Molecular and Functional Genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Raitis Peculis
- Department of Molecular and Functional Genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Rihards Saksis
- Department of Molecular and Functional Genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ilona Mandrika
- Department of Molecular and Functional Genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Sofija Vilisova
- Oncology clinic, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Jurijs Nazarovs
- Institute of Pathology, Pauls Stradins Clinical University Hospital, Riga, Latvia
- Department of Pathology, Riga Stradins University, Latvia
| | - Austra Breiksa
- Institute of Pathology, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Aija Gerina
- Oncology clinic, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Julie Earl
- Ramón y Cajal Health Research Institute (IRYCIS), Ramón y Cajal University Hospital. Ctra. Colmenar Viejo, CIBERONC, Madrid, Spain
| | - Ignacio Ruz-Caracuel
- Ramón y Cajal Health Research Institute (IRYCIS), Ramón y Cajal University Hospital. Ctra. Colmenar Viejo, CIBERONC, Madrid, Spain
- Department of Pathology, Ramón y Cajal University Hospital. Ctra, Colmenar Viejo, Madrid, Spain
| | - Marta Gabriela Rosas
- Department of Pathology, Ramón y Cajal University Hospital. Ctra, Colmenar Viejo, Madrid, Spain
| | - Aldis Pukitis
- Centre of Gastroenterology, Hepatology and Nutrition Therapy, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Natalja Senterjakova
- Centre of Gastroenterology, Hepatology and Nutrition Therapy, Pauls Stradins Clinical University Hospital, Riga, Latvia
| | - Vita Rovite
- Department of Molecular and Functional Genomics, Latvian Biomedical Research and Study Centre, Riga, Latvia
| |
Collapse
|
3
|
Gál E, Parvaneh S, Miklós V, Hegyi P, Kemény L, Veréb Z, Venglovecz V. Investigating the influence of taurochenodeoxycholic acid (TCDCA) on pancreatic cancer cell behavior: An RNA sequencing approach. J Biotechnol 2024; 391:20-32. [PMID: 38815810 DOI: 10.1016/j.jbiotec.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Pancreatic cancer (PC) poses a substantial global health challenge, ranking as the fourth leading cause of cancer-related deaths due to its high mortality rate. Late-stage diagnoses are common due to the absence of specific symptoms. Pancreatic ductal adenocarcinoma (PDAC) accounts for the majority of PC cases. Recent research has suggested a potential link between elevated serum levels of bile acids (BAs) and tumorigenesis of PDAC. This study aims to understand how taurochenodeoxycholic acid (TCDCA), a secondary BA, influences PDAC using RNA sequencing techniques on the Capan-1 cell line. We identified 2,950 differentially expressed genes (DEGs) following TCDCA treatment, with 1,597 upregulated and 1,353 downregulated genes. These DEGs were associated with critical PDAC pathways, including coagulation, angiogenesis, cell migration, and signaling regulation. Furthermore, we reviewed relevant literature highlighting genes like DKK-1, KRT80, UPLA, and SerpinB2, known for their roles in PDAC tumorigenesis and metastasis. Our study sheds light on the complex relationship between BAs and PDAC, offering insights into potential diagnostic markers and therapeutic targets. Further research is needed to unravel these findings' precise mechanisms and clinical implications, potentially improving PDAC diagnosis and treatment.
Collapse
Affiliation(s)
- Eleonóra Gál
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Shahram Parvaneh
- Regenerative Medicine and Cellular Pharmacology Research Laboratory, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Vanda Miklós
- University Biobank, University of Szeged, Szeged, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary; Translational Pancreatology Research Group, Interdisciplinary Center of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary; Centre for Translational Medicine, Semmelweis University, Budapest, Hungary; Institute for Pancreatic Disorders, Semmelweis University, Budapest, Hungary
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Research Laboratory, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary; Interdisciplinary Research Development and Innovation, Center of Excellence, University of Szeged, Szeged, Hungary; HCEMM-USZ Skin Research Group, HCEMM, Szeged, Hungary
| | - Zoltán Veréb
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary.
| | - Viktória Venglovecz
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary; Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary; Translational Pancreatology Research Group, Interdisciplinary Center of Excellence for Research Development and Innovation, University of Szeged, Szeged, Hungary
| |
Collapse
|
4
|
Zhao C, Liu L, He Q, Li Y, Feng J, Chen Y, Li Y, Xu X, Zhu S, Ye Y, Wen Y, Zeng Z, Zhou D, Jin F. Dickkopf-1 is an immune infiltration-related prognostic biomarker of head and neck squamous cell carcinoma. Aging (Albany NY) 2024; 16:3837-3855. [PMID: 38376441 PMCID: PMC10929804 DOI: 10.18632/aging.205563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/11/2024] [Indexed: 02/21/2024]
Abstract
Immunotherapy is currently one of the most viable therapies for head and neck squamous cell carcinoma (HNSCC), characterized by high immune cell infiltration. The Wnt-signaling inhibitor and immune activation mediator, Dickkopf-1 (DKK1), has a strong correlation with tumor growth, tumor microenvironment, and, consequently, disease prognosis. Nevertheless, it is still unclear how DKK1 expression, HNSCC prognosis, and tumor-infiltrating lymphocytes are related. To better understand these associations, we examined how DKK1 expression varies across different tumor and normal tissues. In our study, we investigated the association between DKK1 mRNA expression and clinical outcomes. Next, we assessed the link between DKK1 expression and tumor immune cell infiltration. Additionally, using immunohistochemistry, we evaluated the expression of DKK1 in 15 healthy head and neck tissue samples, and the expression of CD3, CD4, and DKK1 in 27 HNSCC samples. We also explored aberrant DKK1 expression during tumorigenesis. DKK1 expression was remarkably higher in HNSCC tissues than in healthy tissues, and was shown to be associated with tumor stage, grade, lymph node metastasis, histology, and a dismal clinical prognosis in HNSCC. DKK1 expression in HNSCC tissues was inversely correlated with CD3+ (P < 0.0001) and CD4+ (P < 0.0001) immune cell infiltration, while that in immune cells was inversely associated with HNSCC prognosis. These findings offer a bioinformatics perspective on the function of DKK1 in HNSCC immunotherapy and provide justification for clinical research on DKK1-targeted HNSCC treatments. DKK1 is a central target for improving the efficacy of HNSCC immunotherapy.
Collapse
Affiliation(s)
- Chaofen Zhao
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Lina Liu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Qianyong He
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Yuanyuan Li
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Jianglong Feng
- Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
| | - Yue Chen
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Yuxin Li
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Xinyu Xu
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Shaoyuan Zhu
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Yuanmei Ye
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Yajing Wen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Zhirui Zeng
- Department of Physiology, The School of Basic Medicine, Guizhou Medical University, Guiyang, P.R. China
| | - Ding’an Zhou
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Feng Jin
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, P.R. China
- Department of Oncology, The School of Clinical Medicine, Guizhou Medical University, Guiyang, P.R. China
| |
Collapse
|
5
|
Mishima T, Takano S, Takayashiki T, Kuboki S, Suzuki D, Sakai N, Hosokawa I, Konishi T, Nishino H, Nakada S, Kouchi Y, Kishimoto T, Ohtsuka M. Serum elastase-1 predicts malignancy in intraductal papillary mucinous neoplasm of the pancreas. Pancreatology 2024; 24:93-99. [PMID: 38102054 DOI: 10.1016/j.pan.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND The indication for surgical resection of intraductal papillary mucinous neoplasms (IPMNs) is defined by imaging features, such as mural nodules. Although carbohydrate antigen (CA) 19-9 was selected as a parameter for worrisome features, no serum biomarkers were considered when deciding on surgical indications in the latest international consensus guideline. In this study, we assessed whether clinical factors, imaging findings, and serum biomarkers are useful in predicting malignant IPMNs. METHODS A total of 234 resected IPMN cases in Chiba University Hospital from July 2005 to December 2021 were retrospectively analyzed. RESULTS Among the 234 patients with resected IPMNs diagnosed by preoperative imaging, 117 were diagnosed with malignant pathologies (high-grade dysplasia and invasive IPMNs) according to the histological classification. In the multivariate analysis, cyst diameter ≥30 mm; p = 0.035), enhancing mural nodules on multidetector computed tomography (≥5 mm; p = 0.018), and high serum elastase-1 (≥230 ng/dl; p = 0.0007) were identified as independent malignant predictors, while CA19-9 was not. Furthermore, based on the receiver operator characteristic curve analyses, elastase-1 was superior to CA19-9 for predicting malignant IPMNs. Additionally, high serum elastase-1 levels (≥230 ng/dl; p = 0.0093) were identified as independent predictors of malignant IPMNs in patients without mural nodules on multidetector computed tomography (MDCT) in multivariate analysis. CONCLUSION The serum elastase-1 level was found to be a potentially useful biomarker for predicting malignant IPMNs.
Collapse
Affiliation(s)
- Takashi Mishima
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Shigetsugu Takano
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Tsukasa Takayashiki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Satoshi Kuboki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Daisuke Suzuki
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Nozomu Sakai
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Isamu Hosokawa
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Takanori Konishi
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Hitoe Nishino
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Shinichiro Nakada
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Yusuke Kouchi
- Department of Molecular Pathology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Takashi Kishimoto
- Department of Molecular Pathology, Chiba University, Graduate School of Medicine, Chiba, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Chiba University, Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
6
|
Chen SJ, Ren LK, Fei XB, Liu P, Wang X, Zhu CH, Pan YZ. A study on the role of Taxifolin in inducing apoptosis of pancreatic cancer cells: screening results using weighted gene co-expression network analysis. Aging (Albany NY) 2024; 16:2617-2637. [PMID: 38305809 PMCID: PMC10911370 DOI: 10.18632/aging.205500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024]
Abstract
Pancreatic adenocarcinoma (PAAD) is a frequent malignant tumor in the pancreas. The incomplete understanding of cancer etiology and pathogenesis, as well as the limitations in early detection and diagnostic methods, have created an urgent need for the discovery of new therapeutic targets and drugs to control this disease. As a result, the current therapeutic options are limited. In this study, the weighted gene co-expression network analysis (WGCNA) method was employed to identify key genes associated with the progression and prognosis of pancreatic adenocarcinoma (PAAD) patients in the Gene Expression Profiling Interactive Analysis (GEPIA) database. To identify small molecule drugs with potential in the treatment of pancreatic adenocarcinoma (PAAD), we compared key genes to the reference dataset in the CMAP database. First, we analyzed the antitumor properties of small molecule drugs using cell counting kit-8 (CCK-8), AO/EB and Transwell assays. Subsequently, we integrated network pharmacology with molecular docking to explore the potential mechanisms of the identified molecules' anti-tumor effects. Our findings indicated that the progression and prognosis of PAAD patients in pancreatic cancer were associated with 11 genes, namely, DKK1, S100A2, CDA, KRT6A, ITGA3, GPR87, IL20RB, ZBED2, PMEPA1, CST6, and MUC16. These genes were filtered based on their therapeutic potential through comparing them with the reference dataset in the CMAP database. Taxifolin, a natural small molecule drug with the potential for treating PAAD, was screened by comparing it with the reference dataset in the CMAP database. Cell-based experiments have validated the potential of Taxifolin to facilitate apoptosis in pancreatic cancer cells while restraining their invasion and metastasis. This outcome is believed to be achieved via the HIF-1 signaling pathway. In conclusion, this study provided a theoretical basis for screening genes related to the progression of pancreatic cancer and discovered potentially active small molecule drugs. The experimental results confirm that Taxifolin has the ability to promote apoptosis in pancreatic cancer cells.
Collapse
Affiliation(s)
- Shao-Jie Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Li-Kun Ren
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Xiao-Bin Fei
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Peng Liu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xing Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
- Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Chang-Hao Zhu
- Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Yao-Zhen Pan
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
- Department of Hepatobiliary Surgery, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
7
|
Seo SH, Cho KJ, Park HJ, Lee HW, Kim BK, Park JY, Kim DY, Ahn SH, Cheon JH, Yook JI, Kim MD, Joo DJ, Kim SU. Inhibition of Dickkopf-1 enhances the anti-tumor efficacy of sorafenib via inhibition of the PI3K/Akt and Wnt/β-catenin pathways in hepatocellular carcinoma. Cell Commun Signal 2023; 21:339. [PMID: 38012711 PMCID: PMC10680194 DOI: 10.1186/s12964-023-01355-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/14/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Sorafenib improves the overall survival in patients with advanced hepatocellular carcinoma (HCC). Dickkopf-1 (DKK1) is commonly overexpressed in HCC. In this study, we investigated whether the inhibition of DKK1 enhances the anti-tumor efficacy of sorafenib in HCC. METHODS HCC cells were treated with sorafenib and WAY-262611, which is an inhibitor of DKK1. Transgenic mouse models were also developed using hydrodynamic tail vein injection. Mice were orally administered with sorafenib (32 mg/kg), WAY-262611 (16 mg/kg), or sorafenib + WAY-262611 for 10 days. Mechanisms of sorafenib and WAY-262611 were explored via western blotting, immunostaining, and RNA sequencing. RESULTS DKK1 was significantly overexpressed in patients with HCC than in the healthy controls and patients with liver diseases except HCC (all P < 0.05). Compared with sorafenib alone, sorafenib + WAY-262611 significantly inhibited the cell viability, invasion, migration, and colony formation by promoting apoptosis and altering the cell cycles in HCC cells (all P < 0.05). Moreover, sorafenib + WAY-262611 decreased the p110α, phospho-Akt (all P < 0.05), active β-catenin (all P < 0.05) and phospho-GSK-3β (Ser9) expression levels, while increasing the phospho-GSK-3β (Tyr216) expression levels compared with those in the sorafenib alone in vitro and in vivo. In addition, sorafenib + WAY-262611 inhibited tumor progression by regulating cell proliferation and apoptosis, significantly better than sorafenib alone in mouse models. CONCLUSIONS Our results indicate that DKK1 inhibition significantly enhances the anti-tumor efficacy of sorafenib by inhibiting the PI3K/Akt and Wnt/β-catenin pathways via regulation of GSK3β activity, suggesting a novel therapeutic strategy for HCC. Video Abstract.
Collapse
Affiliation(s)
- Sang Hyun Seo
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Kyung Joo Cho
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Hye Jung Park
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Hye Won Lee
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | - Beom Kyung Kim
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | - Jun Yong Park
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | - Do Young Kim
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | - Sang Hoon Ahn
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong In Yook
- Department of Oral Pathology, Yonsei University College of Dentistry, Seoul, Korea
| | - Man-Deuk Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Jin Joo
- Department of Surgery, Yonsei University of College of Medicine, Seoul, Korea
| | - Seung Up Kim
- Yonsei Liver Center, Severance Hospital, Seoul, Korea.
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea.
| |
Collapse
|
8
|
Wang Y, Yu T, Chen J, Zhao R, Diao M, Mei P, He S, Qiu W, Ye G, Jiang L, Xiao H, Liao Y. Immune characteristics analysis and construction of a four-gene prognostic signature for lung adenocarcinoma based on estrogen reactivity. BMC Cancer 2023; 23:1047. [PMID: 37907850 PMCID: PMC10619241 DOI: 10.1186/s12885-023-11415-y] [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: 03/03/2023] [Accepted: 09/17/2023] [Indexed: 11/02/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is a common type of malignant tumor with poor prognosis and high mortality. In our previous studies, we found that estrogen is an important risk factor for LUAD, and different estrogen statuses can predict different prognoses. Therefore, in this study, we constructed a prognostic signature related to estrogen reactivity to determine the relationship between different estrogen reactivities and prognosis. We downloaded the LUAD dataset from The Cancer Genome Atlas (TCGA) database, calculated the estrogen reactivity of each sample, and divided them into a high-estrogen reactivity group and a low-estrogen reactivity group. The difference in overall survival between the groups was significant. We also analyzed the status of immune cell infiltration and immune checkpoint expression between the groups. We analyzed the differential gene expression between the groups and screened four key prognostic factors by the least absolute shrinkage and selection operator (LASSO) regression and univariable and multivariable Cox regression. Based on the four genes, a risk signature was established. To a certain extent, the receiver operating characteristic (ROC) curve showed the predictive ability of the risk signature, which was further verified using the GSE31210 dataset. We also determined the role of estrogen in LUAD using an orthotopic mouse model. Additionally, we developed a predictive nomogram combining the risk signature with other clinical characteristics. In conclusion, our four-gene prognostic signature based on estrogen reactivity had prognostic value and can provide new insights into the development of treatment strategies for LUAD.
Collapse
Affiliation(s)
- Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Yu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaping Chen
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxin Diao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peiyuan Mei
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwen He
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenlin Qiu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanchao Ye
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijuan Jiang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Xiao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Fard D, Giraudo E, Tamagnone L. Mind the (guidance) signals! Translational relevance of semaphorins, plexins, and neuropilins in pancreatic cancer. Trends Mol Med 2023; 29:817-829. [PMID: 37598000 DOI: 10.1016/j.molmed.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/21/2023]
Abstract
Pancreatic cancer is a major cause of demise worldwide. Although key associated genetic changes have been discovered, disease progression is sustained by pathogenic mechanisms that are poorly understood at the molecular level. In particular, the tissue microenvironment of pancreatic adenocarcinoma (PDAC) is usually characterized by high stromal content, scarce recruitment of immune cells, and the presence of neuronal fibers. Semaphorins and their receptors, plexins and neuropilins, comprise a wide family of regulatory signals that control neurons, endothelial and immune cells, embryo development, and normal tissue homeostasis, as well as the microenvironment of human tumors. We focus on the role of these molecular signals in pancreatic cancer progression, as revealed by experimental research and clinical studies, including novel approaches for cancer treatment.
Collapse
Affiliation(s)
- Damon Fard
- Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy
| | - Enrico Giraudo
- Department of Science and Drug Technology, University of Turin, Turin, Italy; Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, Italy
| | - Luca Tamagnone
- Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy; Fondazione Policlinico Gemelli, IRCCS, Rome, Italy.
| |
Collapse
|
10
|
Iliesiu A, Toma RV, Ciongariu AM, Costea R, Zarnescu N, Bîlteanu L. A pancreatic adenocarcinoma mimicking hepatoid carcinoma of uncertain histogenesis: A case report and literature review. Oncol Lett 2023; 26:442. [PMID: 37720666 PMCID: PMC10502951 DOI: 10.3892/ol.2023.14029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 06/19/2023] [Indexed: 09/19/2023] Open
Abstract
In rare cases, metastatic adenocarcinomas of different origin may exhibit the features of hepatoid carcinoma (HC), a rare malignant epithelial tumor, most commonly occurring in the ovaries and stomach, as well as in the pancreas and biliary ducts. A case of a 72-year-old female patient who developed a highly aggressive, poorly differentiated pancreatic ductal adenocarcinoma with peritoneal carcinomatosis, demonstrating hepatoid differentiation upon conventional hematoxylin and eosin staining is reported in the present study. The patient presented with severe abdominal pain, and the radiological investigations performed revealed ovarian and hepatic tumor masses and peritoneal lesions, which were surgically removed. The gross examination of the peritoneum and omentum revealed multiple solid, firm, grey-white nodules, diffusely infiltrating the adipose tissue. The microscopic examination revealed a malignant epithelial proliferation, composed of polygonal cells with abundant eosinophilic cytoplasm and irregular, pleomorphic nuclei. Certain cells presented with intracytoplasmic mucus inclusions, raising suspicion of a HC with an uncertain histogenesis. Immunohistochemical staining was performed, and the tumor cells were found to be positive for cytokeratin (CK)7, CK18 and mucin 5AC, whereas negative staining for CK20, caudal-type homeobox transcription factor 2, α-fetoprotein, paired box gene 8, GATA-binding protein 3 and Wilms tumor 1 were documented. Thus, the diagnosis of metastatic pancreatic adenocarcinoma was established. The main aim of the present study was to provide further knowledge concerning poorly differentiated metastatic adenocarcinoma resembling HC, emphasizing the histopathological and immunohistochemical features of these malignant lesions and raising awareness of the diagnostic difficulties that may arise, as well as the importance of the use immunohistochemistry in differentiating carcinomas of uncertain histogenesis.
Collapse
Affiliation(s)
- Andreea Iliesiu
- Department of Pathology, University Emergency Hospital of Bucharest, Bucharest 014461, Romania
- Faculty of General Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Radu-Valeriu Toma
- Faculty of General Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
- Oncological Institute ‘Alexandru Trestioreanu’, Bucharest 022328, Romania
| | - Ana Maria Ciongariu
- Department of Pathology, University Emergency Hospital of Bucharest, Bucharest 014461, Romania
- Faculty of General Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Radu Costea
- Faculty of General Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
- Second Department of Surgery, University Emergency Hospital of Bucharest, Bucharest 050098, Romania
| | - Narcis Zarnescu
- Faculty of General Medicine, ‘Carol Davila’ University of Medicine and Pharmacy, Bucharest 050474, Romania
- Second Department of Surgery, University Emergency Hospital of Bucharest, Bucharest 050098, Romania
| | - Liviu Bîlteanu
- Oncological Institute ‘Alexandru Trestioreanu’, Bucharest 022328, Romania
- Department of Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine, Bucharest 050097, Romania
| |
Collapse
|
11
|
Ungkulpasvich U, Hatakeyama H, Hirotsu T, di Luccio E. Pancreatic Cancer and Detection Methods. Biomedicines 2023; 11:2557. [PMID: 37760999 PMCID: PMC10526344 DOI: 10.3390/biomedicines11092557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The pancreas is a vital organ with exocrine and endocrine functions. Pancreatitis is an inflammation of the pancreas caused by alcohol consumption and gallstones. This condition can heighten the risk of pancreatic cancer (PC), a challenging disease with a high mortality rate. Genetic and epigenetic factors contribute significantly to PC development, along with other risk factors. Early detection is crucial for improving PC outcomes. Diagnostic methods, including imagining modalities and tissue biopsy, aid in the detection and analysis of PC. In contrast, liquid biopsy (LB) shows promise in early tumor detection by assessing biomarkers in bodily fluids. Understanding the function of the pancreas, associated diseases, risk factors, and available diagnostic methods is essential for effective management and early PC detection. The current clinical examination of PC is challenging due to its asymptomatic early stages and limitations of highly precise diagnostics. Screening is recommended for high-risk populations and individuals with potential benign tumors. Among various PC screening methods, the N-NOSE plus pancreas test stands out with its high AUC of 0.865. Compared to other commercial products, the N-NOSE plus pancreas test offers a cost-effective solution for early detection. However, additional diagnostic tests are required for confirmation. Further research, validation, and the development of non-invasive screening methods and standardized scoring systems are crucial to enhance PC detection and improve patient outcomes. This review outlines the context of pancreatic cancer and the challenges for early detection.
Collapse
Affiliation(s)
| | | | | | - Eric di Luccio
- Hirotsu Bioscience Inc., 22F The New Otani Garden Court, 4-1 Kioi-cho, Chiyoda-ku, Tokyo 102-0094, Japan; (U.U.); (H.H.); (T.H.)
| |
Collapse
|
12
|
Liu L, Yang X, Zhang J, Jiang W, Hou T, Zong Y, Bai H, Yang K, Yang X. Long non-coding RNA SNHG11 regulates the Wnt/β-catenin signaling pathway through rho/ROCK in trabecular meshwork cells. FASEB J 2023; 37:e22873. [PMID: 36929360 DOI: 10.1096/fj.202201733rrr] [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: 10/22/2022] [Revised: 02/16/2023] [Accepted: 03/01/2023] [Indexed: 03/18/2023]
Abstract
Trabecular meshwork (TM) cell dysfunction is the leading cause of elevated intraocular pressure (IOP) and glaucoma. The long non-coding RNA (lncRNA) small nucleolar RNA host gene 11 (SNHG11) is associated with cell proliferation and apoptosis, but its biological functions and role in glaucoma pathogenesis remain unclear. In the present study, we investigated the role of SNHG11 in TM cells using immortalized human TM and glaucomatous human TM (GTM3 ) cells and an acute ocular hypertension mouse model. SNHG11 expression was depleted using siRNA targeting SNHG11. Transwell assays, quantitative real-time PCR analysis (qRT-PCR), western blotting, and CCK-8 assay were used to evaluate cell migration, apoptosis, autophagy, and proliferation. Wnt/β-catenin pathway activity was inferred from qRT-PCR, western blotting, immunofluorescence, and luciferase reporter and TOPFlash reporter assays. The expression of Rho kinases (ROCKs) was detected using qRT-PCR and western blotting. SNHG11 was downregulated in GTM3 cells and mice with acute ocular hypertension. In TM cells, SNHG11 knockdown inhibited cell proliferation and migration, activated autophagy, and apoptosis, repressing the Wnt/β-catenin signaling pathway, and activated Rho/ROCK. Wnt/β-catenin signaling pathway activity increased in TM cells treated with ROCK inhibitor. SNHG11 regulated Wnt/β-catenin signaling through Rho/ROCK by increasing GSK-3β expression and β-catenin phosphorylation at Ser33/37/Thr41 while decreasing β-catenin phosphorylation at Ser675. We demonstrate that the lncRNA SNHG11 regulates Wnt/β-catenin signaling through Rho/ROCK via β-catenin phosphorylation at Ser675 or GSK-3β-mediated phosphorylation at Ser33/37/Thr41, affecting cell proliferation, migration, apoptosis, and autophagy. Through its effects on Wnt/β-catenin signaling, SNHG11 is implicated in glaucoma pathogenesis and is a potential therapeutic target.
Collapse
Affiliation(s)
- Lu Liu
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Xuejiao Yang
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Jingjing Zhang
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Wenlan Jiang
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Tianyu Hou
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yao Zong
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Haiqing Bai
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Kun Yang
- Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Xian Yang
- Department of Ophthalmology, Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| |
Collapse
|
13
|
Ledinek Ž, Sobočan M, Sisinger D, Hojnik M, Büdefeld T, Potočnik U, Knez J. The association of Wnt-signalling and EMT markers with clinical characteristics in women with endometrial cancer. Front Oncol 2023; 13:1013463. [PMID: 36969079 PMCID: PMC10031053 DOI: 10.3389/fonc.2023.1013463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Endometrial cancer is the most common gynecologic malignancy in the developed world. Risk stratification and treatment approaches are changing due to better understanding of tumor biology. Upregulated Wnt signaling plays an important role in cancer initiation and progression with promising potential for development of specific Wnt inhibitor therapy. One of the ways in which Wnt signaling contributes to progression of cancer, is by activating epithelial-to-mesenchymal transition (EMT) in tumor cells, causing the expression of mesenchymal markers, and enabling tumor cells to dissociate and migrate. This study analyzed the expression of Wnt signaling and EMT markers in endometrial cancer. Wnt signaling and EMT markers were significantly correlated with hormone receptors status in EC, but not with other clinico-pathological characteristics. Expression of Wnt antagonist, Dkk1 was significantly different between the ESGO-ESTRO-ESP patient risk assessment categories using integrated molecular risk assessment.
Collapse
Affiliation(s)
- Živa Ledinek
- Department of Pathology, University Medical Centre Maribor, Maribor, Slovenia
| | - Monika Sobočan
- Divison for Gynaecology and Perinatology, University Medical Centre Maribor, Maribor, Slovenia
- Department of Gynaecology and Obstetrics, Faculty of Medicine University of Maribor, Maribor, Slovenia
| | - Damjan Sisinger
- Department of Pathology, University Medical Centre Maribor, Maribor, Slovenia
| | - Marko Hojnik
- Department of Pathology, University Medical Centre Maribor, Maribor, Slovenia
| | - Tomaž Büdefeld
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Uroš Potočnik
- Center for Human Molecular Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Laboratory for Biochemistry, Molecular Biology and Genomics, University of Maribor, Maribor, Slovenia
- Department for Science and Research, University Medical Centre Maribor, Maribor, Slovenia
| | - Jure Knez
- Divison for Gynaecology and Perinatology, University Medical Centre Maribor, Maribor, Slovenia
- Department of Gynaecology and Obstetrics, Faculty of Medicine University of Maribor, Maribor, Slovenia
- *Correspondence: Jure Knez,
| |
Collapse
|
14
|
Liang T, Wu X, Wang L, Song T, Wu P, Niu Y, Huang H. Correlation of NNMT and DKK1 Protein Expression With Clinicopathological Characteristics and Prognosis of Breast Cancer. Clin Med Insights Oncol 2023; 17:11795549231168073. [PMID: 37114075 PMCID: PMC10126688 DOI: 10.1177/11795549231168073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/15/2023] [Indexed: 04/29/2023] Open
Abstract
Background Nicotinamide N-methyltransferase (NNMT) and Dickkopf-1 (DKK1) play an important role in the development of breast cancer, and the purpose of this study was designed to examine the clinical and prognostic significance of NNMT and DKK1 in breast cancer. Methods The GEPIA2 database was used to evaluate the expression and survival of NNMT mRNA and DKK1 mRNA of breast cancer. Then an immunohistochemical study was carried out on 374 cases of breast tissue to identify the protein expression and significance of NNMT and DKK1. Next, the prognostic significance of DKK1 in breast cancer was explored by COX and Kaplan-Meier models. Results Protein NNMT expression was correlated with lymph node metastasis and histological grade (P < .05) while protein DKK1 expression was related to tumor size, pT stage, histological grade, and Ki-67 (P < .05). Protein DKK1 was related to disease-specific survival (DSS), and low DKK1 expression indicated a poor prognosis of breast cancer patients (P < .05). Combined expression of protein NNMT and protein DKK1 predicted different prognosis of DSS (P < .05). Conclusions Nicotinamide N-methyltransferase and DKK1 were linked to breast cancer malignancy and invasion. Breast cancer patients with low DKK1 expression had a worse prognosis. Oncotypes of NNMT and DKK1 expression predicted patient outcomes.
Collapse
Affiliation(s)
- Tairong Liang
- Department of Pathology, The Second
Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xiuqian Wu
- The Affiliated Cancer Hospital of
Shantou University Medical College, Shantou, China
| | - Lan Wang
- Department of Pathology, The Second
Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Tiantian Song
- Department of Pharmacology, Shantou
University Medical College, Shantou, China
- Department of Preventive Medicine,
Shantou University Medical College, Shantou, China
| | - Peishan Wu
- Department of Pathology, The Second
Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yongdong Niu
- Department of Pharmacology, Shantou
University Medical College, Shantou, China
| | - Haihua Huang
- Department of Pathology, The Second
Affiliated Hospital of Shantou University Medical College, Shantou, China
- Haihua Huang, Department of Pathology, The
Second Affiliated Hospital of Shantou University Medical College, Shantou
515000, China.
| |
Collapse
|
15
|
Du Y, Guo Z. Recent progress in ferroptosis: inducers and inhibitors. Cell Death Dis 2022; 8:501. [PMID: 36581640 PMCID: PMC9800531 DOI: 10.1038/s41420-022-01297-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022]
Abstract
Ferroptosis is a new iron-dependent form of programmed cell death characterized by iron accumulation and lipid peroxidation. In recent years, ferroptosis has garnered enormous interest in disease treatment research communities in pursuit to reveal the mechanism and key targets of ferroptosis because ferroptosis is closely related to the pathophysiological processes of many diseases. Recent studies have shown some key targets, such as glutathione peroxidase 4 (GPX4) and System Xc-, and several inducers and inhibitors have been developed to regulate these key targets. With the emergence of new ferroptosis targets, studies on inducers and inhibitors have made new developments. The selection and use of inducers and inhibitors are very important for related work. This paper briefly introduces important regulatory targets in the ferroptosis metabolic pathway, lists and categorizes commonly used and recently developed inducers and inhibitors, and discusses their medical application. The paper ends of with potential future research direction for ferroptosis.
Collapse
Affiliation(s)
- Yunxi Du
- grid.20513.350000 0004 1789 9964Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China
| | - Zhong Guo
- grid.20513.350000 0004 1789 9964Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China ,grid.20513.350000 0004 1789 9964Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
16
|
Luo M, Chen YJ, Xie Y, Wang QR, Xiang YN, Long NY, Yang WX, Zhao Y, Zhou JJ. Dickkopf-related protein 1/cytoskeleton-associated protein 4 signaling activation by Helicobacter pylori-induced activator protein-1 promotes gastric tumorigenesis via the PI3K/AKT/mTOR pathway. World J Gastroenterol 2022; 28:6769-6787. [PMID: 36620343 PMCID: PMC9813938 DOI: 10.3748/wjg.v28.i47.6769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/05/2022] [Accepted: 11/30/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common malignant tumor with high incidence and mortality rates globally, especially in East Asian countries. Helicobacter pylori (H. pylori) infection is a significant and independent risk factor for GC. However, its underlying mechanism of action is not fully understood. Dickkopf-related protein (DKK) 1 is a Wnt signaling antagonist, and cytoskeleton-associated protein (CKAP) 4 is a newly identified DKK1 receptor. Recent studies found that the binding of DKK1 to CAKP4 mediated the procancer signaling of DKK1 inde-pendent of Wnt signaling. We hypothesize that H. pylori-induced activation of DKK1/CKAP4 signaling contributes to the initiation and progression of GC.
AIM To investigate the interaction of H. pylori infection, DKK1 and CAKP4 in GC, as well as the underlying molecular mechanisms.
METHODS RNA sequencing was used to identify differentially expressed genes (DEGs) between H. pylori-infected and uninfected primary GC cells. Gain- and loss-of-function experiments were performed to verify the H. pylori-induced upregulation of activator protein-1 (AP-1) in GC cells. A dual-luciferase reporter assay and co-immunoprecipitation were used to determine the binding of AP-1 to the DKK1 promoter and DKK1 to CKAP4. Western blotting and immunohistochemistry detected the expression of DKK1, CKAP4, and phos-phatidylinositol 3-kinase (PI3K) pathway-related proteins in GC cells and tissues. Functional experiments and tumorigenicity in nude mice detected malignant behavior of GC cells in vitro and in vivo.
RESULTS We identified 32 DEGs between primary GC cells with and without H. pylori infection, including JUN, fos-like antigen-1 (FOSL1), and DKK1, and confirmed that the three proteins and CKAP4 were highly expressed in H. pylori-infected GC cells, H. pylori-infected gerbil gastric tissues, and human GC tissues. JUN and FOSL1 form AP-1 to transcriptionally activate DKK1 expression by binding to the DKK1 promoter. Activated DKK1 bound to CKAP4, but not the most common Wnt coreceptor low-density lipoprotein receptor-related protein 5/6, to promote GC cell growth, colony formation, migration, invasion, and xenograft tumor growth in nude mice. All these effects were driven by activation of the PI3K/AKT/mammalian target of rapamycin (mTOR) pathway. Targeting the PI3K signaling pathway by LY294002 inhibited DKK1-mediated CKAP4/PI3K signaling activity and the malignant behavior of GC cells.
CONCLUSION H. pylori induces JUN and FOSL1 expression to form AP-1, which transcriptionally activates DKK1. Binding of DKK1 to KAKP4 contributes to gastric tumorigenesis via the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Mei Luo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yuan-Jia Chen
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yuan Xie
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Qin-Rong Wang
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yi-Ning Xiang
- Department of Pathology of Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Ni-Ya Long
- Department of Neurology of Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Wen-Xiu Yang
- Department of Pathology of Affiliated Hospital, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yan Zhao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Jian-Jiang Zhou
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & Key Laboratory of Medical Molecular Biology of Guizhou Province, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| |
Collapse
|
17
|
Geleta B, Tout FS, Lim SC, Sahni S, Jansson PJ, Apte MV, Richardson DR, Kovačević Ž. Targeting Wnt/tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. J Biol Chem 2022; 298:101608. [PMID: 35065073 PMCID: PMC8881656 DOI: 10.1016/j.jbc.2022.101608] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
A major barrier to successful pancreatic cancer (PC) treatment is the surrounding stroma, which secretes growth factors/cytokines that promote PC progression. Wnt and tenascin C (TnC) are key ligands secreted by stromal pancreatic stellate cells (PSCs) that then act on PC cells in a paracrine manner to activate the oncogenic β-catenin and YAP/TAZ signaling pathways. Therefore, therapies targeting oncogenic Wnt/TnC cross talk between PC cells and PSCs constitute a promising new therapeutic approach for PC treatment. The metastasis suppressor N-myc downstream-regulated gene-1 (NDRG1) inhibits tumor progression and metastasis in numerous cancers, including PC. We demonstrate herein that targeting NDRG1 using the clinically trialed anticancer agent di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) inhibited Wnt/TnC-mediated interactions between PC cells and the surrounding PSCs. Mechanistically, NDRG1 and DpC markedly inhibit secretion of Wnt3a and TnC by PSCs, while also attenuating Wnt/β-catenin and YAP/TAZ activation and downstream signaling in PC cells. This antioncogenic activity was mediated by direct inhibition of β-catenin and YAP/TAZ nuclear localization and by increasing the Wnt inhibitor, DKK1. Expression of NDRG1 also inhibited transforming growth factor (TGF)-β secretion by PC cells, a key mechanism by which PC cells activate PSCs. Using an in vivo orthotopic PC mouse model, we show DpC downregulated β-catenin, TnC, and YAP/TAZ, while potently increasing NDRG1 expression in PC tumors. We conclude that NDRG1 and DpC inhibit Wnt/TnC-mediated interactions between PC cells and PSCs. These results further illuminate the antioncogenic mechanism of NDRG1 and the potential of targeting this metastasis suppressor to overcome the oncogenic effects of the PC-PSC interaction.
Collapse
Affiliation(s)
- Bekesho Geleta
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Faten S Tout
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Department of Medical Laboratory Science, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Syer Choon Lim
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia; Cancer Drug Resistance & Stem Cell Program, Faculty of Medicine and Health, School of Medical Science, University of Sydney, Sydney, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, UNSW Sydney, Sydney, New South Wales, Australia; Pancreatic Research Group, Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Žaklina Kovačević
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
18
|
Luu TT. Review of Immunohistochemistry Biomarkers in Pancreatic Cancer Diagnosis. Front Oncol 2022; 11:799025. [PMID: 34988027 PMCID: PMC8720928 DOI: 10.3389/fonc.2021.799025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is one of the cancer types with poor prognosis and high rate of mortality. Diagnostic modalities for early detection of pancreatic cancer have been among the academic concerns. On account of the potential role of immunohistochemistry (IHC) biomarkers in overcoming certain limitations of imaging diagnostic tools in discriminating pancreatic cancer tissues from benign ones, a growing scholarly attention has been given to the diagnostic efficacy of IHC biomarkers for pancreatic cancer. This review will analyze and synthesize published articles to provide an insight into potential IHC biomarkers for pancreatic cancer diagnosis.
Collapse
Affiliation(s)
- Tuan Trong Luu
- Management & Marketing Department, Swinburne University of Technology, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Zhang S, Wei H, Ha X, Zhang Y, Guo Y. NK4 Regulates Laryngeal Squamous Cell Carcinoma Cell Properties and Inhibits Tumorigenicity by Modulating the DKK1/Wnt/β-Catenin Axis. Front Oncol 2022; 11:783575. [PMID: 34970492 PMCID: PMC8712930 DOI: 10.3389/fonc.2021.783575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/24/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the effects of NK4 gene on the properties and tumorigenicity in laryngeal squamous cell carcinoma cell. Methods Here, we used the attenuated Salmonella carrying the NK4 gene to transfect the AMC-HN-8 cells and detected the expression of NK4 by the real-time quantitative polymerase chain reaction (q RT-PCR). The properties of NK4 gene was determined by MTT method, cell scratch test, and flow cytometry. A nude mouse tumorigenesis model was used to evaluate the effect of NK4 gene on the growth of AMC-HN-8 cells in vivo. While a western blot assay was used to assess the expression of DKK1, Wnt1 and β-Catenin in nude mouse tumors. Results qRT-PCR showed that the expression of NK4 in the transfection group was significantly higher than that in the control group (P<0.01), and the expression increased with the time of transfection. MTT results showed NK4 overexpression inhibited the proliferation of AMC-HN-8 cells, and the inhibitory activity no longer increased with increasing dose when 30% expression supernatant was added (P<0.01). Scratch experiment showed that NK4 overexpression decreased the cell migration ability (P<0.01). Annexin V/PI double staining experiment showed that NK4 gene induced AMC-HN-8 cell apoptosis (P<0.01), and cell cycle arrest in S phase (P<0.01). NK4 overexpression inhibited tumor formation ability of AMC-HN-8 cells in vivo (P <0.05). WB detection showed that the expression of DKK1 increased, Wnt1 and β-Catenin protein decreased after the high expression of NK4. Conclusions NK4 gene inhibit cell proliferation and migration, while promote cell apoptosis, and induce cell cycle arrest in S phase of laryngeal carcinoma AMC-HN-8 cells. NK4 overexpression inhibit the tumorigenesis ability of AMC-HN-8 cells, which may be related to the regulation of DKK1/Wnt1/β-Catenin signal axis.
Collapse
Affiliation(s)
- Shoukai Zhang
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Otolaryngology-Head Neck Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Hulai Wei
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiaoqin Ha
- Laboratory, People's Liberation Army Joint Logistics Support Force 940th Hospital, Lanzhou, China
| | - Yueyu Zhang
- Otolaryngology-Head Neck Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Yufen Guo
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Otolaryngology-Head Neck Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
20
|
Tang P, Qu W, Wu D, Chen S, Liu M, Chen W, Ai Q, Tang H, Zhou H. Identifying and Validating an Acidosis-Related Signature Associated with Prognosis and Tumor Immune Infiltration Characteristics in Pancreatic Carcinoma. J Immunol Res 2021; 2021:3821055. [PMID: 34993253 PMCID: PMC8727107 DOI: 10.1155/2021/3821055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Acidosis in the tumor microenvironment (TME) is involved in tumor immune dysfunction and tumor progression. We attempted to develop an acidosis-related index (ARI) signature to improve the prognostic prediction of pancreatic carcinoma (PC). METHODS Differential gene expression analyses of two public datasets (GSE152345 and GSE62452) from the Gene Expression Omnibus database were performed to identify the acidosis-related genes. The Cancer Genome Atlas-pancreatic carcinoma (TCGA-PAAD) cohort in the TCGA database was set as the discovery dataset. Univariate Cox regression and the Kaplan-Meier method were applied to screen for prognostic genes. The least absolute shrinkage and selection operator (LASSO) Cox regression was used to establish the optimal model. The tumor immune infiltrating pattern was characterized by the single-sample gene set enrichment analysis (ssGSEA) method, and the prediction of immunotherapy responsiveness was conducted using the tumor immune dysfunction and exclusion (TIDE) algorithm. RESULTS We identified 133 acidosis-related genes, of which 37 were identified as prognostic genes by univariate Cox analysis in combination with the Kaplan-Meier method (p values of both methods < 0.05). An acidosis-related signature involving seven genes (ARNTL2, DKK1, CEP55, CTSV, MYEOV, DSG2, and GBP2) was developed in TCGA-PAAD and further validated in GSE62452. Patients in the acidosis-related high-risk group consistently showed poorer survival outcomes than those in the low-risk group. The 5-year AUCs (areas under the curve) for survival prediction were 0.738 for TCGA-PAAD and 0.889 for GSE62452, suggesting excellent performance. The low-risk group in TCGA-PAAD showed a higher abundance of CD8+ T cells and activated natural killer cells and was predicted to possess an elevated proportion of immunotherapeutic responders compared with the high-risk counterpart. CONCLUSIONS We developed a reliable acidosis-related signature that showed excellent performance in prognostic prediction and correlated with tumor immune infiltration, providing a new direction for prognostic evaluation and immunotherapy management in PC.
Collapse
Affiliation(s)
- Pingfei Tang
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Weiming Qu
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Dajun Wu
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Shihua Chen
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Minji Liu
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Weishun Chen
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Qiongjia Ai
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Haijuan Tang
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| | - Hongbing Zhou
- Department of Digestive Diseases, Zhuzhou Central Hospital, The Affiliated Zhuzhou Hospital of Xiangya Medical College of Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
21
|
Gao S, Jin Y, Zhang H. Pan-Cancer Analyses Reveal Oncogenic and Immunological Role of Dickkopf-1 (DKK1). Front Genet 2021; 12:757897. [PMID: 34899842 PMCID: PMC8654726 DOI: 10.3389/fgene.2021.757897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/05/2021] [Indexed: 12/15/2022] Open
Abstract
WNT signaling pathway inhibitor Dickkopf-1 (DKK1) is related to cancer progression; however, its diagnostic and prognostic potential have not been investigated in a pan-cancer perspective. In this study, multiple bioinformatic analyses were conducted to evaluate therapeutic value of DKK1 in human cancers. The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) project served as data resources. The Wilcoxon rank test was performed to evaluate the expression difference of DKK1 between cancer tissues and normal tissues. A Kaplan-Meier curve and Cox regression were used for prognosis evaluation. Single-sample gene set enrichment analysis (ssGSEA) was used to evaluate the association of DKK1 expression with the immune cell infiltration. The potential function of DKK1 was explored by STRING and clusterProfiler. We found that the expression level of DKK1 is significantly different in different cancer types. Importantly, we demonstrated that DKK1 is an independent risk factor in ESCA, LUAD, MESO, and STAD. Further analysis revealed that DKK1 had a large effect on the immune cell infiltration and markers of certain immune cells, such as Th1 and Th2 cells. PPI network analysis and further pathway enrichment analysis indicated that DKK1 was mainly involved in the WNT signaling pathway. Our findings suggested that DKK1 might serve as a marker of prognosis for certain cancers by affecting the WNT signaling pathway and tumor immune microenvironment.
Collapse
Affiliation(s)
- Shuang Gao
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Ye Jin
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Hongmei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China.,School of Clinical Medicine, North China University of Science and Technology, Tangshan, China.,Hebei Province Key Laboratory of Occupational Health and Safety for Coal Industry, School of Public Health, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
22
|
Jin H, Cui M. Recognition of potential therapeutic role of 2-hydroxy-3-methylanthraquinones in the treatment of gallbladder carcinoma: A proteomics analysis. Fundam Clin Pharmacol 2021; 36:350-362. [PMID: 34850442 DOI: 10.1111/fcp.12740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/31/2021] [Accepted: 11/23/2021] [Indexed: 12/01/2022]
Abstract
Gallbladder carcinoma (GBC), with early metastasis and high recurrence rates, is an enormous threat to health. As an anthraquinones monomer of traditional Chinese medicine Hedyotis diffusa, 2-hydroxy-3-methylanthraquinone (HMA) has been reported to inhibit the growth of several cancers. But in our preliminary study, HMA could only weakly induce GBC cell apoptosis. To explore other possible mechanism underlying the inhibition effect of HMA on GBC, this proteomics analysis was performed. A proteomics analysis was performed on one GBC cell line bought from the China Life Science Cell Bank. Several computational techniques were merged to develop analysis for those differently expressed proteins. A comparative protein-protein interaction network analysis was carried out among the differently expressed proteins to identify the proteins potentially inhibiting GBC. Thus, a GO and KEGG analysis was performed to identify the signaling pathways underlying a potential therapeutic role for HMA. A total of 285 proteins were affected by HMA, including 187 upregulated and 98 downregulated. The subcellular localization of differently expressed proteins were identified, including 142 in nuclear, 67 in cytoplasm, 67 in extracellular matrix, 46 in plasma membrane, 13 in mitochondrion, 3 in lysosome, and 1 in cytoskeleton. HMA could regulate EGFR, FN1, PLG, PLAUR, LAMA3, HRG, THBS1, PLAT, KNG1, ENAM, SERPINE1, ECM1, interleukin-8, and trypsin in GBC. Most of the regulated proteins involve in cell migration. Pathways including PI3K-Akt, Wnt, HIF-1, focal adhesion, microRNAs were regulated by HMA. HMA was shown to be an inhibition agent for GBC development, and this analysis would contribute to the development of new anti-GBC drugs.
Collapse
Affiliation(s)
- Hao Jin
- The Second Department of General Surgery, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| | - Min Cui
- Deputy Secretary of Party Committee, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, China
| |
Collapse
|
23
|
O'Neill RS, Stoita A. Biomarkers in the diagnosis of pancreatic cancer: Are we closer to finding the golden ticket? World J Gastroenterol 2021; 27:4045-4087. [PMID: 34326612 PMCID: PMC8311531 DOI: 10.3748/wjg.v27.i26.4045] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/24/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is a leading cause of cancer related mortality on a global scale. The disease itself is associated with a dismal prognosis, partly due to its silent nature resulting in patients presenting with advanced disease at the time of diagnosis. To combat this, there has been an explosion in the last decade of potential candidate biomarkers in the research setting in the hope that a diagnostic biomarker may provide a glimmer of hope in what is otherwise quite a substantial clinical dilemma. Currently, serum carbohydrate antigen 19-9 is utilized in the diagnostic work-up of patients diagnosed with PC however this biomarker lacks the sensitivity and specificity associated with a gold-standard marker. In the search for a biomarker that is both sensitive and specific for the diagnosis of PC, there has been a paradigm shift towards a focus on liquid biopsy and the use of diagnostic panels which has subsequently proved to have efficacy in the diagnosis of PC. Currently, promising developments in the field of early detection on PC using diagnostic biomarkers include the detection of microRNA (miRNA) in serum and circulating tumour cells. Both these modalities, although in their infancy and yet to be widely accepted into routine clinical practice, possess merit in the early detection of PC. We reviewed over 300 biomarkers with the aim to provide an in-depth summary of the current state-of-play regarding diagnostic biomarkers in PC (serum, urinary, salivary, faecal, pancreatic juice and biliary fluid).
Collapse
Affiliation(s)
- Robert S O'Neill
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St George and Sutherland Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2010, Australia
| | - Alina Stoita
- Department of Gastroenterology, St Vincent's Hospital Sydney, Sydney 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney 2010, Australia
| |
Collapse
|
24
|
Wang S, Zheng Y, Yang F, Zhu L, Zhu XQ, Wang ZF, Wu XL, Zhou CH, Yan JY, Hu BY, Kong B, Fu DL, Bruns C, Zhao Y, Qin LX, Dong QZ. The molecular biology of pancreatic adenocarcinoma: translational challenges and clinical perspectives. Signal Transduct Target Ther 2021; 6:249. [PMID: 34219130 PMCID: PMC8255319 DOI: 10.1038/s41392-021-00659-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an increasingly common cause of cancer mortality with a tight correspondence between disease mortality and incidence. Furthermore, it is usually diagnosed at an advanced stage with a very dismal prognosis. Due to the high heterogeneity, metabolic reprogramming, and dense stromal environment associated with pancreatic cancer, patients benefit little from current conventional therapy. Recent insight into the biology and genetics of pancreatic cancer has supported its molecular classification, thus expanding clinical therapeutic options. In this review, we summarize how the biological features of pancreatic cancer and its metabolic reprogramming as well as the tumor microenvironment regulate its development and progression. We further discuss potential biomarkers for pancreatic cancer diagnosis, prediction, and surveillance based on novel liquid biopsies. We also outline recent advances in defining pancreatic cancer subtypes and subtype-specific therapeutic responses and current preclinical therapeutic models. Finally, we discuss prospects and challenges in the clinical development of pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Feng Yang
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Xiao-Qiang Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhe-Fang Wang
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Xiao-Lin Wu
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Cheng-Hui Zhou
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Jia-Yan Yan
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - De-Liang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Christiane Bruns
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany
| | - Yue Zhao
- General, Visceral and Cancer Surgery, University Hospital of Cologne, Cologne, Germany.
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, Shanghai, China.
- Key laboratory of whole-period monitoring and precise intervention of digestive cancer, Shanghai Municipal Health Commission (SMHC), Shanghai, China.
| |
Collapse
|
25
|
Liu W, Fu X, Li R. CNN1 regulates the DKK1/Wnt/β-catenin/c-myc signaling pathway by activating TIMP2 to inhibit the invasion, migration and EMT of lung squamous cell carcinoma cells. Exp Ther Med 2021; 22:855. [PMID: 34178128 PMCID: PMC8220635 DOI: 10.3892/etm.2021.10287] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
The present study aimed to investigate the effect of calponin 1 (CNN1) on the invasion and migration of lung squamous cell carcinoma (LUSC) cells and the associations between CNN1, tissue inhibitor of metalloproteinases 2 (TIMP2), Dickkopf-1 (DKK1) and the Wnt/β-catenin/c-myc signaling pathway. The expression levels of CNN1 and TIMP2 in LUSC cells and the association between CNN1 and TIMP2 were predicted using the GEPIA database. The cells were transiently transfected to overexpress CNN1, which resulted in inhibition of DKK1 and TIMP2 expression levels. Wound healing and Transwell assays were used to detect the invasive and migratory abilities of LUSC cells. Reverse transcription-quantitative PCR and western blotting were used to investigate the expression levels of CNN1, MMP2, MMP9, E-cadherin, N-cadherin (N-cad), SLUG, DKK1, β-catenin and c-myc. The expression levels of N-cad were detected using immunofluorescence staining. The results indicated that overexpression of CNN1 inhibited the invasion and migration of NCI-H2170 cells. Inhibition of DKK1 reversed this change and the expression levels of β-catenin and c-myc were upregulated, whereas the expression levels of DKK1 were downregulated with a concomitant inhibition of TIMP2. In summary, these results demonstrated that CNN1 regulated the DKK1/Wnt/β-catenin/c-myc signaling pathway by activating TIMP2 to inhibit the invasion, migration and epithelial-to-mesenchymal transition of LUSC cells.
Collapse
Affiliation(s)
- Wusheng Liu
- Department of Respiratory Medicine, The Affiliated Xinyu Hospital of Nanchang University, Xinyu, Jiangxi 338000, P.R. China
| | - Xiaogang Fu
- Department of Respiratory Medicine, Xinyu People's Hospital, Xinyu, Jiangxi 338000, P.R. China
| | - Rumei Li
- Department of Endocrinology, Xinyu People's Hospital, Xinyu, Jiangxi 338000, P.R. China
| |
Collapse
|
26
|
Chu HY, Chen Z, Wang L, Zhang ZK, Tan X, Liu S, Zhang BT, Lu A, Yu Y, Zhang G. Dickkopf-1: A Promising Target for Cancer Immunotherapy. Front Immunol 2021; 12:658097. [PMID: 34093545 PMCID: PMC8174842 DOI: 10.3389/fimmu.2021.658097] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/04/2021] [Indexed: 01/15/2023] Open
Abstract
Clinical studies in a range of cancers have detected elevated levels of the Wnt antagonist Dickkopf-1 (DKK1) in the serum or tumors of patients, and this was frequently associated with a poor prognosis. Our analysis of DKK1 gene profile using data from TCGA also proves the high expression of DKK1 in 14 types of cancers. Numerous preclinical studies have demonstrated the cancer-promoting effects of DKK1 in both in vitro cell models and in vivo animal models. Furthermore, DKK1 showed the ability to modulate immune cell activities as well as the immunosuppressive cancer microenvironment. Expression level of DKK1 is positively correlated with infiltrating levels of myeloid-derived suppressor cells (MDSCs) in 20 types of cancers, while negatively associated with CD8+ T cells in 4 of these 20 cancer types. Emerging experimental evidence indicates that DKK1 has been involved in T cell differentiation and induction of cancer evasion of immune surveillance by accumulating MDSCs. Consequently, DKK1 has become a promising target for cancer immunotherapy, and the mechanisms of DKK1 affecting cancers and immune cells have received great attention. This review introduces the rapidly growing body of literature revealing the cancer-promoting and immune regulatory activities of DKK1. In addition, this review also predicts that by understanding the interaction between different domains of DKK1 through computational modeling and functional studies, the underlying functional mechanism of DKK1 could be further elucidated, thus facilitating the development of anti-DKK1 drugs with more promising efficacy in cancer immunotherapy.
Collapse
Affiliation(s)
- Hang Yin Chu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Zihao Chen
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China.,School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Luyao Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Zong-Kang Zhang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China.,School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xinhuan Tan
- Department of Microsurgery (II), Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Wendeng, China
| | - Shuangshuang Liu
- Department of Microsurgery (II), Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Wendeng, China
| | - Bao-Ting Zhang
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China.,School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-based Translational Medicine and Drug Discovery, Hong Kong, China
| |
Collapse
|
27
|
Zhang C, Huang L, Xiong J, Xie L, Ying S, Jia Y, Yao Y, Song X, Zeng Z, Yuan J. Isoalantolactone inhibits pancreatic cancer proliferation by regulation of PI3K and Wnt signal pathway. PLoS One 2021; 16:e0247752. [PMID: 33661942 PMCID: PMC7932101 DOI: 10.1371/journal.pone.0247752] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND/AIMS Isoalantolactone (IATL) is one of multiple isomeric sesquiterpene lactones and is isolated from inula helenium. IATL has multiple functions such as antibacterial, antihelminthic and antiproliferative activities. IATL also inhibits pancreatic cancer proliferation and induces apoptosis by increasing ROS production. However, the detailed mechanism of IATL-mediated pancreatic cancer apoptosis remains largely unknown. METHODS In current study, pancreatic carcinoma cell lines (PANC-1, AsPC-1, BxPC-3) and a mouse xenograft model were used to determine the mechanism of IATL-mediated toxic effects. RESULTS IATL (20μM) inhibited pancreatic adenocarcinoma cell lines proliferation in a time-dependent way; while scratch assay showed that IATL significantly inhibited PANC-1 scratch closure (P<0.05); Invasion assays indicated that IATL significantly attenuated pancreatic adenocarcinoma cell lines invasion on matrigel. Signal analysis showed that IATL inhibited pancreatic adenocarcinoma cell proliferation by blocking EGF-PI3K-Skp2-Akt signal axis. Moreover, IATL induced pancreatic adenocarcinoma cell apoptosis by increasing cytosolic Caspase3 and Box expression. This apoptosis was mediated by inhibition of canonical wnt signal pathway. Finally, xenograft studies showed that IATL also significantly inhibited pancreatic adenocarcinoma cell proliferation and induced pancreatic adenocarcinoma cell apoptosis in vivo. CONCLUSIONS IATL inhibits pancreatic cancer proliferation and induces apoptosis on cellular and in vivo models. Signal pathway studies reveal that EGF-PI3K-Skp2-Akt signal axis and canonical wnt pathway are involved in IATL-mediated cellular proliferation inhibition and apoptosis. These studies indicate that IATL may provide a future potential therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Chaoxiong Zhang
- Research Center for Occupational Respiratory Disease, West China Fourth Hospital, Sichuan University, Chengdu, China
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
- Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Lei Huang
- Department of Gastroenterology, Chengdu First People’s Hospital, Chengdu, China
| | - Jingyuan Xiong
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
| | - Linshen Xie
- Research Center for Occupational Respiratory Disease, West China Fourth Hospital, Sichuan University, Chengdu, China
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
| | - Shi Ying
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
| | - You Jia
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Research Center for Occupational Respiratory Disease, West China Fourth Hospital, Sichuan University, Chengdu, China
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
| | - Xuejiao Song
- Healthy Food Evaluation Center, West China School of Public Health, Sichuan University, Chengdu, China
| | - Zhenguo Zeng
- Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialing Yuan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
28
|
Hu Y, Liu M, Xu S, Li S, Yang M, Su T, Yuan Z, Peng H. The Clinical Significance of Dickkopf Wnt Signaling Pathway Inhibitor Gene Family in Head and Neck Squamous Cell Carcinoma. Med Sci Monit 2020; 26:e927368. [PMID: 33281184 PMCID: PMC7706141 DOI: 10.12659/msm.927368] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/15/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Dickkopf Wnt signaling pathway inhibitor (DKK) gene family, which is known to inhibit the Wnt regulation process, is widely found in cancers. However, the roles and functions of specific family members in head and neck squamous cell carcinoma (HNSCC) are still unclear. MATERIAL AND METHODS Online bioinformatics tools (Oncomine, UALCAN, Kaplan-Meier plotter, GEPIA, Metascape, and STRING) were used to analyze the relationships between distinct DKKs and HNSCC. The transcriptome expression, clinical association, functions, pathways, and protein-protein interaction networks of DKKs in HNSCC were explored. RESULTS The mRNA expression of DKK1, DKK3, and Dickkopf-like acrosomal protein 1 (DKKL1) in HNSCC was significantly higher than in normal tissues, while that of DKK4 was lower. The mRNA expression of DKK1, DKK3, and DKKL1 was elevated in higher-grade HNSCC. The mRNA expression of DKK1 and DKK3 was elevated in human papillomavirus (HPV)-negative HNSCC, while DKKL1 had a higher mRNA expression in HPV-positive HNSCC. In addition, DKK1 was significantly associated with unfavorable overall survival in HNSCC patients. DKK3 was more likely to be a negative factor for the 5-year survival rate, while DKK4 was the opposite. DKK1 function was mainly enriched in GTPase-mediated signal transduction. Porcupine O-acyltransferase, a key regulator of the Wnt signaling pathway, was also associated with DKK1 in the protein-protein interaction network. CONCLUSIONS With regard to improving the therapeutic strategies of HNSCC in the future, DKK1 could be an unfavorable prognostic biomarker. DKK3, DKK4, and DKKL1 might be potential biomarkers for HNSCC.
Collapse
|
29
|
Jiang X, Hui F, Qin X, Wu Y, Liu H, Gao J, Li X, Xu Y, Zhang Y. Diagnosis Accuracy and Prognostic Significance of the Dickkopf-1 Protein in Gastrointestinal Carcinomas: Systematic Review and Network Meta-analysis. J Cancer 2020; 11:7091-7100. [PMID: 33193872 PMCID: PMC7646173 DOI: 10.7150/jca.49970] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/06/2020] [Indexed: 12/17/2022] Open
Abstract
Objective: To evaluate the diagnosis accuracy and prognostic significance of bio-marker dickkopf-1(DKK-1) protein in GIC, and also sub-type of hepatocellular carcinoma (HCC), pancreas carcinomas (PC), oesophageal carcinoma (EPC) and Adenocarcinoma of esophago-gastric junction (AEGJ), etc. Methods: Electronic databases were searched from inception to May 2020. Patients were diagnosed with gastrointestinal carcinomas, and provided data on the correlation between high and low DKK-1 expression and diagnosis or prognosis. Results: Forty-three publications involving 9318 participants were included in the network meta-analysis, with 31 of them providing data for diagnosis value and 18 records were eligible for providing prognosis value of DKK-1. DKK-1 has a moderate diagnostic value for overall GIC, HCC and PC. In addition, for the combined diagnosis value of DKK-1 +AFP, high diagnostic accuracy value could be determined in HCC and early HCC group, respectively. Whereas, diagnosis efficiency of DKK-1+CA19-9 was also better than that of DKK-1 alone with AUC value is above 0.95. For the prognosis meta-analysis of histopathological stratification, we found that EPC and AEGJ ranked the best for the histopathological stratification of prognosis from network meta-analysis. This systematic review protocol was registered with the PROSPERO registry (No.CRD42020167910). Conclusion: DKK-1 has good diagnostic accuracy, especially combination of DKK-1+AFP in HCC and DKK-1+CA19-9 in PC, whereas modest prognostic significant in GIC. Future head-to-head researches are warranted for DKK-1 expression in HCC and PC tissue.
Collapse
Affiliation(s)
- Xiaowen Jiang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Fuhai Hui
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiaochun Qin
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuting Wu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Haihan Liu
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jing Gao
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiang Li
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yali Xu
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yingshi Zhang
- Department of Life Science and Biochemistry, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
30
|
Bu F, Nie H, Zhu X, Wu T, Lin K, Zhao J, Huang J. A signature of 18 immune-related gene pairs to predict the prognosis of pancreatic cancer patients. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:713-726. [PMID: 33128857 PMCID: PMC7654420 DOI: 10.1002/iid3.363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer is one of the most lethal malignancies. With the promising prospects conveyed by immunotherapy in cancers, we aimed to construct an immune‐related gene pairs (IRGPs) signature to predict the prognosis of pancreatic cancer patients. We downloaded clinical and transcriptional data of pancreatic cancer patients from The Cancer Genome Atlas data set as the training group and GSE57495 data set as the verification group. We filtered immune‐related transcriptional data by IMMPORT. With the assistance of lasso penalized Cox regression, we constructed our prognostic IRGPs signature and divided all samples into high‐/low‐risk groups by receiver operating characteristic curve for further comparisons. The comparisons between high‐ and low‐risk groups including survival rate, multivariate, and univariate Cox proportional‐hazards analysis, infiltration of immune cells, and Gene Set Enrichment Analysis (GSEA). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) are facilitated to analyze the proceedings in which our IRGPs signature may involve in. The results revealed that 18 IRGPs were defined as our prognostic signature. The prognostic value of this IRGPs signature was verified from the GSE57495 data set. We further demonstrated the independent prognostic value of this IRGPs signature. The contents of six immune cells between high‐/low‐risk groups were different, which was associated with the progression of diverse cancers. Results from GO, KEGG, and GSEA revealed that this IRGPs signature was involved in extracellular space, immune response, cancer pathways, cation channel, and gated channel activities. Evidently, this IRGPs signature will provide remarkable value for the therapy of pancreatic cancer patients.
Collapse
Affiliation(s)
- Fanqin Bu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Han Nie
- Department of Vascular Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- Zhongshan School of Medicine, Research Center of the Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Ting Wu
- Infection Department of Guixi Traditional Chinese Medicine Hospital, Guixi, Jiangxi, China
| | - Kang Lin
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiefeng Zhao
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jun Huang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
31
|
Identification of the prognostic value of immune gene signature and infiltrating immune cells for esophageal cancer patients. Int Immunopharmacol 2020; 87:106795. [PMID: 32707495 DOI: 10.1016/j.intimp.2020.106795] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Esophageal cancer (ESCA) is one of the deadliest solid malignancies with worse survival rate worldwide. Here, we aimed to establish an immune-gene prognostic signature for predicting patients' survival and providing accurate targets for personalized therapy or immunotherapy. METHODS Gene expression profile of patients with ESCA were download from The Cancer Genome Atlas (TCGA) database (dataset 1: n = 159) and immune-related genes from the ImmPORT database. Dataset 1 was subdivided into two groups (dataset 2: n = 80; dataset 3: n = 79). Kaplan-Meier and receiver operating characteristic (ROC) curves were plotted to validate the predictive effect of the prognostic signature on the three datasets. TIMER and CIBERSORT analysis were used to evaluate the correlation between the prognostic signature and infiltrating immune cells. RESULTS We constructed a prognostic signature composed of six immune genes (HSPA6, S100A12, FABP3, DKK1, OSM and NR2F2). Kaplan-Meier curves validated the good predictive ability of the prognostic signature in datasets 1, 2 and 3 (P = 0.0034, P = 0.0081, and P = 0.0363, respectively). The area under the curve (AUC) of the ROC curves validated the predictive accuracy of the immune signature (AUCs = 0.757, 0.800, and 0.701, respectively). We also revealed the good prognostic value of the immune cells, including activated memory CD4 T cells, T follicular helper cells and monocytes. Potential target drugs, including Olopatadine and Amlexanox, were identified for clinical therapies to improve patients' survival outcomes. CONCLUSION Our study indicated that the immune-related prognostic signature could serve as a novel biomarker for predicting patients' prognosis and providing new immunotherapy targets in ESCA.
Collapse
|
32
|
Gan LL, Hii LW, Wong SF, Leong CO, Mai CW. Molecular Mechanisms and Potential Therapeutic Reversal of Pancreatic Cancer-Induced Immune Evasion. Cancers (Basel) 2020; 12:cancers12071872. [PMID: 32664564 PMCID: PMC7408947 DOI: 10.3390/cancers12071872] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/06/2020] [Accepted: 07/09/2020] [Indexed: 02/05/2023] Open
Abstract
Pancreatic cancer ranks high among the causes of cancer-related mortality. The prognosis of this grim condition has not improved significantly over the past 50 years, despite advancement in imaging techniques, cancer genetics and treatment modalities. Due to the relative difficulty in the early detection of pancreatic tumors, as low as 20% of patients are eligible for potentially curative surgery; moreover, chemotherapy and radiotherapy (RT) do not confer a great benefit in the overall survival of the patients. Currently, emerging developments in immunotherapy have yet to bring a significant clinical advantage among pancreatic cancer patients. In fact, pancreatic tumor-driven immune evasion possesses one of the greatest challenges leading to immunotherapeutic resistance. Most of the immune escape pathways are innate, while poor priming of hosts' immune response and immunoediting constitute the adaptive immunosuppressive machinery. In this review, we extensively discuss the pathway perturbations undermining the anti-tumor immunity specific to pancreatic cancer. We also explore feasible up-and-coming therapeutic strategies that may restore immunity and address therapeutic resistance, bringing hope to eliminate the status quo in pancreatic cancer prognosis.
Collapse
Affiliation(s)
- Li-Lian Gan
- School of Postgraduate Study, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (L.-L.G.); (L.-W.H.)
| | - Ling-Wei Hii
- School of Postgraduate Study, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (L.-L.G.); (L.-W.H.)
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Shew-Fung Wong
- School of Medicine, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Environmental and Population Health, Institute for Research, Development and Innovation (IRDI), International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Chee-Onn Leong
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development and Innovation (IRDI), International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Chun-Wai Mai
- School of Pharmacy, International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- Centre for Cancer and Stem Cells Research, Institute for Research, Development and Innovation (IRDI), International Medical University, 126, Jalan Jalil Perkasa 19, Bukit Jalil, Kuala Lumpur 57000, Malaysia
- Correspondence: ; Tel.: +60-3-2731-7596
| |
Collapse
|
33
|
The RBP1-CKAP4 axis activates oncogenic autophagy and promotes cancer progression in oral squamous cell carcinoma. Cell Death Dis 2020; 11:488. [PMID: 32587255 PMCID: PMC7316825 DOI: 10.1038/s41419-020-2693-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/02/2022]
Abstract
Retinol-binding protein 1 (RBP1) is involved in several physiological functions, including the regulation of the metabolism and retinol transport. Studies have shown that it plays an important role in the pathogenesis of several types of cancer. However, the role of RBP1 and its correlation with autophagy in oral squamous cell carcinoma (OSCC) pathogenesis remain unknown. In this study, RBP1 was identified as the most significantly upregulated DEPs with a >2-fold change in OSCC samples when compared to normal tissues through iTRAQ-based proteomics analysis coupled with 2D LC–MS/MS. RBP1 overexpression was significantly associated with malignant phenotypes (differentiation, TNM stage, and lymphatic metastasis) of OSCC. In vitro experiments demonstrated that RBP1 was significantly increased in OSCC tissues and cell lines compared with control group. RBP1 overexpression promoted cell growth, migration, and invasion of OSCC cells. Silencing of RBP1 suppressed tumor formation in xenografted mice. We further demonstrated that the RBP1–CKAP4 axis was a critical regulator of the autophagic machinery in OSCC, inactivation of autophagy rescued the RBP1–CKAP4-mediated malignant biological behaviors of OSCC cells. Overall, a mechanistic link was provided by RBP1–CKAP4 between primary oncogenic features and the induction of autophagy, which may provide a potential therapeutic target that warrants further investigation for treatment of OSCC.
Collapse
|
34
|
Li J, Gao Y, Yue W. The Clinical Diagnostic and Prognostic Value of Dickkopf-1 in Cancer. Cancer Manag Res 2020; 12:4253-4260. [PMID: 32606922 PMCID: PMC7292247 DOI: 10.2147/cmar.s254596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/19/2020] [Indexed: 12/25/2022] Open
Abstract
The Wnt signaling pathway extensively participates in diverse processes such as embryonic development, maintenance of homeostasis and tumor pathogenesis. Dickkopf-1 (DKK1), a Wnt inhibitor, plays a vital role for over the past decades regarding its role in the regulation of several types of cancers. However, studies have shown that DKK1 is expressed differently in cancer and plays a role as a cancer-promoting factor or a tumor suppressor, which is worthy of further exploration. We herein study whether DKK1 is highly expressed in all cancers and plays a crucial role in promoting cancer. Furthermore, we discussed as to which stages of cancer development it plays in. Finally, the present detection methods were introduced and indicated the clinical application of DKK1 in tumor development.
Collapse
Affiliation(s)
- Jie Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Yan Gao
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| | - Wentao Yue
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, People's Republic of China
| |
Collapse
|
35
|
DKK1 is epigenetically downregulated by promoter methylation and inhibits bile acid-induced gastric intestinal metaplasia. Biochem Biophys Res Commun 2020; 523:780-786. [PMID: 31952791 DOI: 10.1016/j.bbrc.2019.12.109] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
Dickkopf-related protein 1 (DKK1) is essential to gastric cancer as an inhibitor of Wnt signaling. Gastric intestinal metaplasia (GIM) is an important precancerous lesion of gastric cancer that can be activated by bile acid reflux and chronic inflammation. However, the exact mechanism of DKK1 in bile acid-induced GIM has not been completely elucidated. We aimed to explore the epigenetic alterations and biological functions of DKK1 in the development of GIM. In the present study, bile acid was found to induce the expression of intestinal markers in gastric epithelial cells, whereas DKK1 was downregulated in response to bile acid stimulation. The mRNA and protein expression levels of DKK1 were decreased in GIM tissues as evidenced by qRT-PCR and immunohistochemical staining. Surprisingly, the methylation of the DKK1 promoter increased in GIM tissues, and we discovered 28 differential methylation sites of the DKK1 promoter in GIM tissues. Bile acid was able to induce the partial methylation of the DKK1 promoter, while 5-aza could increase DKK1 expression as well as decrease intestinal markers expression in gastric epithelial cells. In conclusion, the promoter methylation and downregulation of DKK1 might play important roles in the development of GIM, especially bile acid-induced GIM.
Collapse
|