1
|
Pan D, Li X, Qiao X, Wang Q. Immunosuppressive tumor microenvironment in pancreatic cancer: mechanisms and therapeutic targets. Front Immunol 2025; 16:1582305. [PMID: 40443678 PMCID: PMC12119487 DOI: 10.3389/fimmu.2025.1582305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/17/2025] [Indexed: 06/02/2025] Open
Abstract
Pancreatic cancer is projected to become the second leading cause of cancer-related death by 2030. Conventional interventions including surgery, radiotherapy, and chemotherapy provide only modest survival benefits, underscoring an urgent need for more effective therapies. Although immunotherapy has revolutionized the management of several solid tumors, its clinical benefit in pancreatic cancer has so far been disappointing. Mounting evidence indicates that a highly immunosuppressive tumor microenvironment (TME), dominated by tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), drives immune evasion, tumor progression, metastasis, and chemoresistance through complex cytokine and chemokine networks. This review summarizes current knowledge of these immunosuppressive mechanisms and provides emerging strategies aimed at re-educating or depleting these cellular constituents to enhance the efficacy of immunotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Da Pan
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Xinyue Li
- First College for Clinical Medicine, Xuzhou Medical University, Jiangsu, Xuzhou, China
| | - Xiao Qiao
- Department of Gastroenterology, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| | - Qiqi Wang
- Department of Gastroenterology, Wenzhou Central Hospital, Wenzhou, China
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
2
|
Zajkowska M, Orywal K, Gryko M. Potential Utility of A Proliferation-Inducing Ligand (APRIL) in Colorectal Cancer. Int J Mol Sci 2024; 25:12496. [PMID: 39684206 DOI: 10.3390/ijms252312496] [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: 10/14/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
APRIL (A proliferation-inducing ligand) is a member of the tumor necrosis factor superfamily that is overexpressed in a variety of malignant tumors, including colorectal cancer (CRC). Its key physiological roles include inducing the immunoglobulin switch and ensuring plasmocyte survival. In terms of pathological roles, APRIL antagonism has been identified as a key target in autoimmune diseases and immunoglobulin disorders. As previously demonstrated, several inflammatory processes occur at the site of neoplastic initial stages, and their local symptoms are difficult to detect, particularly in the early stages. That is why we chose to study the current literature on APRIL's role in the development of colorectal cancer. The main objective of our research was to investigate the role of APRIL in cancer initiation and its usefulness in the detection and therapy of CRC. Interestingly, the findings conducted so far show that the selected protein has a significant potential as a CRC biomarker and treatment target. Importantly, based on its concentration, it is possible to identify CRC patients, but whether the lesion has a benign or malignant nature, indicating the possibility of rapid detection of an ongoing disease process.
Collapse
Affiliation(s)
- Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok Clinical Hospital, 15-269 Białystok, Poland
| | - Karolina Orywal
- Department of Biochemical Diagnostics, Medical University of Bialystok Clinical Hospital, 15-269 Białystok, Poland
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Białystok, Poland
| | - Mariusz Gryko
- Department of Surgical Nursing, Medical University of Białystok, 15-274 Białystok, Poland
- 1st Clinical Department of General and Endocrine Surgery, Medical University of Bialystok Clinical Hospital, 15-276 Białystok, Poland
| |
Collapse
|
3
|
Abukwaik R, Vera-Siguenza E, Tennant D, Spill F. p53 Orchestrates Cancer Metabolism: Unveiling Strategies to Reverse the Warburg Effect. Bull Math Biol 2024; 86:124. [PMID: 39207627 PMCID: PMC11362376 DOI: 10.1007/s11538-024-01346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Cancer cells exhibit significant alterations in their metabolism, characterised by a reduction in oxidative phosphorylation (OXPHOS) and an increased reliance on glycolysis, even in the presence of oxygen. This metabolic shift, known as the Warburg effect, is pivotal in fuelling cancer's uncontrolled growth, invasion, and therapeutic resistance. While dysregulation of many genes contributes to this metabolic shift, the tumour suppressor gene p53 emerges as a master player. Yet, the molecular mechanisms remain elusive. This study introduces a comprehensive mathematical model, integrating essential p53 targets, offering insights into how p53 orchestrates its targets to redirect cancer metabolism towards an OXPHOS-dominant state. Simulation outcomes align closely with experimental data comparing glucose metabolism in colon cancer cells with wild-type and mutated p53. Additionally, our findings reveal the dynamic capability of elevated p53 activation to fully reverse the Warburg effect, highlighting the significance of its activity levels not just in triggering apoptosis (programmed cell death) post-chemotherapy but also in modifying the metabolic pathways implicated in treatment resistance. In scenarios of p53 mutations, our analysis suggests targeting glycolysis-instigating signalling pathways as an alternative strategy, whereas targeting solely synthesis of cytochrome c oxidase 2 (SCO2) does support mitochondrial respiration but may not effectively suppress the glycolysis pathway, potentially boosting the energy production and cancer cell viability.
Collapse
Affiliation(s)
- Roba Abukwaik
- Mathematics Department, King Abdulaziz University, Rabigh, Saudi Arabia.
- School of Mathematics, University of Birmingham, Birmingham, B15 2TS, UK.
| | - Elias Vera-Siguenza
- School of Mathematics, University of Birmingham, Birmingham, B15 2TS, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniel Tennant
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Fabian Spill
- School of Mathematics, University of Birmingham, Birmingham, B15 2TS, UK.
| |
Collapse
|
4
|
Khosroshahi EM, Maghsoudloo M, Fahimi H, Mokhtari K, Entezari M, Peymani M, Hashemi M, Wan R. Determining expression changes of ANO7 and SLC38A4 membrane transporters in colorectal cancer. Heliyon 2024; 10:e34464. [PMID: 39114022 PMCID: PMC11305260 DOI: 10.1016/j.heliyon.2024.e34464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/21/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Membrane transporters are proteins responsible for facilitating the movement of molecules within biological membranes. They play a vital role in maintaining cellular homeostasis by regulating the transport of nutrients, ions, and other molecules into and out of cells. Our aim is to identify biomarkers in colorectal cancer using membrane transporter proteins. We utilized COAD TCGA data for this purpose. Subsequently, we conducted differential gene analysis and feature selection using membrane transporter proteins. Furthermore, we identified two potential genes, including ANO7 and SLC38A4. To validate the expression profiles of ANO7 and SLC38A4, key genes in this context, RT-qPCR was employed on colorectal cancer samples and adjacent normal tissues. Additionally, utilizing GEPIA2, Kaplan-Meier survival analysis, and cBioPortal, we assessed the status of these genes in various cancers, examining their methylation and mutation patterns. In conclusion, we suggest that ANO7 and SLC38A4 serve as prognostic biomarkers in colorectal cancer.
Collapse
Affiliation(s)
- Elaheh Mohandesi Khosroshahi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Hossein Fahimi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
5
|
An S, Kim SK, Kwon HY, Kim CS, Bang HJ, Do H, Kim B, Kim K, Kim Y. Expression of Immune-Related and Inflammatory Markers and Their Prognostic Impact in Colorectal Cancer Patients. Int J Mol Sci 2023; 24:11579. [PMID: 37511338 PMCID: PMC10380610 DOI: 10.3390/ijms241411579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The tumor microenvironment of colorectal cancer (CRC) is heterogenous; thus, it is likely that multiple immune-related and inflammatory markers are simultaneously expressed in the tumor. The aim of this study was to identify immune-related and inflammatory markers expressed in freshly frozen CRC tissues and to investigate whether they are related to the clinicopathological features and prognosis of CRC. Seventy patients with CRC who underwent curative surgical resection between December 2014 and January 2017 were included in this study. Tissue samples were obtained from tumor and non-tumor areas in the patients' colons. The concentrations of immune-related markers (APRIL/TNFSF13, BAFF, LAG-3, PD-1, PD-L1, and CTLA-4) and inflammatory markers (CHIT, MMP-3, osteocalcin, pentraxin-3, sTNF-R1, and sTNF-R2) in the samples were measured using the Bio-plex Multiplex Immunoassay system. The concentrations of APRIL/TNFSF13, BAFF, and MMP-3 in the samples were significantly high; thus, we conducted analyses based on the cut-off values for these three markers. The high-APRIL/TNFSH13-expression group showed a significantly higher rate of metastatic lesions than the low-expression group, whereas the high-MMP-3-expression group had higher CEA levels, more lymph node metastases, and more advanced disease stages than the low-expression group. The five-year disease-free survival of the high-MMP-3-expression group was significantly shorter than that of the low-expression group (65.1% vs. 90.2%, p = 0.033). This study provides evidence that the APRIL/TNFSF13, BAFF, and MMP-3 pathway is overexpressed in CRC tissues and is associated with unfavorable clinicopathological features and poor prognosis in CRC patients. These markers could serve as diagnostic or prognostic biomarkers for CRC.
Collapse
Affiliation(s)
- Sanghyun An
- Department of Colorectal Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Wonju Surgical Research Collaboration, Wonju 26465, Republic of Korea
| | - Soo-Ki Kim
- Department of Microbiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Hye Youn Kwon
- Department of Colorectal Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Wonju Surgical Research Collaboration, Wonju 26465, Republic of Korea
| | - Cheol Su Kim
- Department of Microbiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Hui-Jae Bang
- Wonju Surgical Research Collaboration, Wonju 26465, Republic of Korea
- Department of Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Republic of Korea
| | - Hyejin Do
- Department of Anesthesiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - BoRa Kim
- Department of Internal Medicine, Division of Gastroenterology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Kwangmin Kim
- Department of Colorectal Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Wonju Surgical Research Collaboration, Wonju 26465, Republic of Korea
| | - Youngwan Kim
- Department of Colorectal Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Wonju Surgical Research Collaboration, Wonju 26465, Republic of Korea
| |
Collapse
|
6
|
Arévalo B, Blázquez-García M, Valverde A, Serafín V, Montero-Calle A, Solís-Fernández G, Barderas R, Campuzano S, Yáñez-Sedeño P, Pingarrón JM. Binary MoS2 nanostructures as nanocarriers for amplification in multiplexed electrochemical immunosensing: simultaneous determination of B cell activation factor and proliferation-induced signal immunity-related cytokines. Mikrochim Acta 2022; 189:143. [PMID: 35286499 PMCID: PMC8921152 DOI: 10.1007/s00604-022-05250-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022]
Abstract
A dual immunosensor is reported for the simultaneous determination of two important immunity-related cytokines: BAFF (B cell activation factor) and APRIL (a proliferation-induced signal). Sandwich-type immunoassays with specific antibodies (cAbs) and a strategy for signal amplification based on labelling the detection antibodies (dAbs) with binary MoS2/MWCNTs nanostructures and using horseradish peroxidase (HRP) were implemented. Amperometric detection was carried out at screen-printed dual carbon electrodes (SPdCEs) through the hydroquinone HQ/H2O2 system. The developed dual immunosensor provided limit of detection (LOD) of 0.08 and 0.06 ng mL−1 for BAFF and APRIL, respectively, and proved to be useful for the determination of both cytokines in cancer cell lysates and serum samples from patients diagnosed with autoimmune diseases and cancer. The obtained results agreed with those found using ELISA methodologies.
Collapse
Affiliation(s)
- Beatriz Arévalo
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain
| | - Marina Blázquez-García
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain
| | - Alejandro Valverde
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain
| | - Verónica Serafín
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, 28220, MajadahondaMadrid, Spain
| | | | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, 28220, MajadahondaMadrid, Spain
| | - Susana Campuzano
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Paloma Yáñez-Sedeño
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain.
| | - José M Pingarrón
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040, Madrid, Spain
| |
Collapse
|
7
|
Zhang Q, Ni Y, Zhi X, Wang J, Li Z, Tang J, Wang L, Wang W, Xu Z. Involvement of APRIL in Helicobacter pylori-related gastric cancer. J Cancer Res Clin Oncol 2021; 147:1685-1697. [PMID: 33738544 DOI: 10.1007/s00432-021-03574-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/17/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND/AIMS A proliferation-inducing ligand (APRIL, also known as TNFSF13, CD256) is a member of the tumor necrosis factor (TNF) superfamily and involved in a diverse set of diseases. In this work, we explored the potential associations and underlying mechanism in patients suffered from gastric cancer between the expression of APRIL and H. pylori infection. METHODS We analyzed APRIL expression levels in 200 GC tissue samples by immunohistochemistry staining. H. pylori infection was detected by modified Giemsa staining. The biological effects of APRIL on human GC cells in vitro and in vivo were tested by CCK-8 assay, colony formation, flow cytometry detection, transwell migration assay, matrigel invasion assay, and tumor xenograft assay in animals. RESULTS APRIL reactivity was positively correlated with H. pylori infection in vitro and vivo. It turned out that the decrease of miR-145 expression was dose-dependent and time-dependent on H. pylori infection and in consistent with APRIL expression. MiR-145 significantly attenuated the effect of H. pylori infection on APRIL gene expression in SGC7901 and BGC823 cell lines. Furthermore, APRIL overexpression promoted the proliferation, migration, invasion, and transfer of GC cells and decreased apoptosis, while APRIL knockdown suppressed these effects. We confirmed that APRIL activated the canonical NF-κB pathway through phosphorylation of AKT. CONCLUSION The expression of APRIL, which promoted the proliferation, migration, invasion, viability, and metastasis of GC cells, was upregulated in human H. pylori-infected GC through miR-145. Besides, APRIL-induced gastric tumorigenicity via activating NF-κB pathway. These results may provide a framework for the deeper analysis of APRIL in GC risk and prognosis.
Collapse
Affiliation(s)
- Qun Zhang
- Department of Oncological Surgery, Minhang Branch of Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ying Ni
- Department of Hematology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaofei Zhi
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jiwei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Tang
- Department of Pediatric Surgery, Nanjing Children's Hospital, Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linjun Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weizhi Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Vijaya Kumar A, Brézillon S, Untereiner V, Sockalingum GD, Kumar Katakam S, Mohamed HT, Kemper B, Greve B, Mohr B, Ibrahim SA, Goycoolea FM, Kiesel L, Pavão MSG, Motta JM, Götte M. HS2ST1-dependent signaling pathways determine breast cancer cell viability, matrix interactions, and invasive behavior. Cancer Sci 2020; 111:2907-2922. [PMID: 32573871 PMCID: PMC7419026 DOI: 10.1111/cas.14539] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) act as signaling co‐receptors by interaction of their sulfated glycosaminoglycan chains with numerous signaling molecules. In breast cancer, the function of heparan sulfate 2‐O‐sulfotransferase (HS2ST1), the enzyme mediating 2‐O‐sulfation of HS, is largely unknown. Hence, a comparative study on the functional consequences of HS2ST1 overexpression and siRNA knockdown was performed in the breast cancer cell lines MCF‐7 and MDA‐MB‐231. HS2ST1 overexpression inhibited Matrigel invasion, while its knockdown reversed the phenotype. Likewise, cell motility and adhesion to fibronectin and laminin were affected by altered HS2ST1 expression. Phosphokinase array screening revealed a general decrease in signaling via multiple pathways. Fluorescent ligand binding studies revealed altered binding of fibroblast growth factor 2 (FGF‐2) to HS2ST1‐expressing cells compared with control cells. HS2ST1‐overexpressing cells showed reduced MAPK signaling responses to FGF‐2, and altered expression of epidermal growth factor receptor (EGFR), E‐cadherin, Wnt‐7a, and Tcf4. The increased viability of HS2ST1‐depleted cells was reduced to control levels by pharmacological MAPK pathway inhibition. Moreover, MAPK inhibitors generated a phenocopy of the HS2ST1‐dependent delay in scratch wound repair. In conclusion, HS2ST1 modulation of breast cancer cell invasiveness is a compound effect of altered E‐cadherin and EGFR expression, leading to altered signaling via MAPK and additional pathways.
Collapse
Affiliation(s)
- Archana Vijaya Kumar
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Stéphane Brézillon
- CNRS, MEDyC UMR 7369, UFR de Médecine, Université de Reims Champagne-Ardenne, Reims, France
| | | | | | - Sampath Kumar Katakam
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Hossam Taha Mohamed
- CNRS, MEDyC UMR 7369, UFR de Médecine, Université de Reims Champagne-Ardenne, Reims, France.,Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.,Faculty of Biotechnology, October University for Modern Sciences and Arts, Giza, Egypt
| | - Björn Kemper
- Biomedical Technology Center of the Medical Faculty, University of Münster, Münster, Germany
| | - Burkhard Greve
- Department of Radiotherapy - Radiooncology, University Hospital Münster, Münster, Germany
| | - Benedikt Mohr
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | | | | | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - Mauro S G Pavão
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana M Motta
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| |
Collapse
|
9
|
Retraction: APRIL Induces Tumorigenesis and Metastasis of Colorectal Cancer Cells via Activation of the PI3K/Akt Pathway. PLoS One 2019; 14:e0222525. [PMID: 31513667 PMCID: PMC6742464 DOI: 10.1371/journal.pone.0222525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
10
|
Yang B, Pan CS, Li Q, Yang Z, Long FX, Fan JY, Wang CS, Han JY, Tang DX. Inhibitory effects of Chanling Gao on the proliferation and liver metastasis of transplanted colorectal cancer in nude mice. PLoS One 2019; 14:e0201504. [PMID: 30789971 PMCID: PMC6383928 DOI: 10.1371/journal.pone.0201504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/31/2019] [Indexed: 12/20/2022] Open
Abstract
This study aimed to explore the efficacy and mechanism of Chanling Gao (CLG), a compound Chinese medicine, on colorectal cancer (CRC). A model of transplanted CRC was established in nude mice. The mice were treated 7 days after CRC transplantation with either Capecitabine or CLG for 3 weeks. On the 28th day after the operation, CRC growth and liver metastasis were assessed by morphology, the changes in the expression of HIF-1α (hypoxia inducible factor-1α), stromal cell-derived factor-1 alpha (SDF-1α), CXCR4 (C-X-C chemokine receptor type 4), PI3K, and Akt in the transplanted tumor and SDF-1α and CXCR4 in the liver were detected by Western blot and immunohistochemistry. The protein contents of vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-2, and collagen IV in the serum and transplanted tumor and SDF-1α and CXCR4 in liver tissues were detected by enzyme-linked immunosorbent assay. In the Capecitabine and high dose CLG groups, the growth and liver metastasis of CRC were significantly inhibited, the protein levels of HIF-1α, SDF-1α, CXCR4, MMP-2, VEGF, PI3K, Akt, P-PI3K and P-Akt in the transplanted tumor were lower, while the content of collagen IV in the transplanted tumor was higher, than in Model group. A high dose of CLG inhibited the growth of transplanted tumor and liver metastasis of CRC in nude mice, probably by inhibiting the HIF-1α/SDF-1α-CXCR4/PI3K-Akt signaling pathway reducing the synthesis and release of VEGF and degradation of collagen IV.
Collapse
Affiliation(s)
- Bing Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Zhu Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Feng-Xi Long
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Chuan-She Wang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing-Yan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dong-Xin Tang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Department of Oncology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
11
|
APRIL is Involved in the Proliferation and Metastasis of Acute Lymphoblastic Leukemia Cells. J Pediatr Hematol Oncol 2018; 40:588-593. [PMID: 29697578 DOI: 10.1097/mph.0000000000001198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Our previous work showed that a proliferation-inducing ligand (APRIL) was involved in the development of acute lymphoblastic leukemia (ALL) in children. However, the precise role of APRIL in ALL remains unknown. To investigate this issue, we silenced and overexpressed APRIL in Nalm-6 ALL cells using short hairpin RNA targeting the APRIL gene and recombinant human APRIL, respectively, and evaluated the effects on cell proliferation, apoptosis, and migration. APRIL mRNA and APRIL and matrix metalloproteinase-2 protein levels were evaluated by semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) and western blott, respectively. We found that APRIL expression was reduced by shRNA-mediated knockdown in Nalm-6 cells; this was associated with a decrease in cell proliferation (P<0.05). APRIL knockdown increased apoptosis (P<0.01) but suppressed cell migration along with matrix metalloproteinase-2 protein level. Overexpressing recombinant human APRIL had the opposite effects in each case (P<0.05). These results demonstrate a link between APRIL expression and ALL development and suggest that APRIL is a potential therapeutic target for ALL treatment.
Collapse
|
12
|
Prognostic Significance of Serum BAFF, APRIL, TACI and BCMA Levels in Chronic Lymphocytic Leukemia. Indian J Hematol Blood Transfus 2018; 35:265-271. [PMID: 30988562 DOI: 10.1007/s12288-018-1029-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/08/2018] [Indexed: 01/12/2023] Open
Abstract
As chronic lymphocytic leukemia (CLL) has a variable disease course, novel prognostic markers and risk assessment models are being developed in order to identify high-risk patients who may need early treatment. The two tumor necrosis factor family proteins BAFF and APRIL and their receptors BAFF-R, TACI and BCMA are considered to play a critical role in the survival of normal B cells. In order to highlight the pathophysiological role of this complicated biological network, we aimed to analyze the potential prognostic effects of BAFF, APRIL, TACI and BCMA in CLL patients. We investigated the prognostic impact of serum BCMA, TACI, BAFF and APRIL levels in 129 newly diagnosed CLL patients [median age: 64 (39-88) years; male/female: 85/44]. Serum BAFF, TACI and BCMA levels were significantly lower in the patient group compared to the control group (p < 0.001), while serum APRIL level did not differ significantly between two groups (p > 0.05). Serum BCMA [(p = 0.029; r = 0.208)] and TACI levels [(p = 0.011; r = 0.241)] were positively correlated with serum free light chain ratio. Serum BAFF [(p = 0.008; r = - 0.236)] and BCMA [(p = 0.042; r = - 0.183)] levels were negatively correlated with Rai stage. Overall survival (OS) was relatively better in patients with low serum BAFF levels [60 (1-187) months vs 39.5 (0-256) months; p = 0.063]. Probability of OS was higher in patients with low BAFF levels when compared to patients with normal levels, without statistical significance (53.6% vs 23.6%; p > 0.05). Large prospective studies are needed to validate the prognostic role of this essential biological pathway in CLL.
Collapse
|
13
|
Tai YT, Lin L, Xing L, Cho SF, Yu T, Acharya C, Wen K, Hsieh PA, Dulos J, van Elsas A, Munshi N, Richardson P, Anderson KC. APRIL signaling via TACI mediates immunosuppression by T regulatory cells in multiple myeloma: therapeutic implications. Leukemia 2018; 33:426-438. [PMID: 30135465 PMCID: PMC6367000 DOI: 10.1038/s41375-018-0242-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/16/2018] [Accepted: 07/23/2018] [Indexed: 01/08/2023]
Abstract
We here investigate how APRIL impacts immune regulatory T cells and directly contributes to the immunosuppressive multiple myeloma (MM) bone marrow (BM) microenvironment. First, APRIL receptor TACI expression is significantly higher in regulatory T cells (Tregs) than conventional T cells (Tcons) from the same patient, confirmed by upregulated Treg markers, i.e., Foxp3, CTLA-4. APRIL significantly stimulates proliferation and survival of Tregs, whereas neutralizing anti-APRIL monoclonal antibodies (mAbs) inhibit theses effects. Besides TACI-dependent induction of cell cycle progression and anti-apoptosis genes, APRIL specifically augments Foxp3, IL-10, TGFβ1, and PD-L1 in Tregs to further enhance Treg-inhibited Tcon proliferation. APRIL further increases MM cell-driven Treg (iTreg) via TACI-dependent proliferation associated with upregulated IL-10, TGFβ1, and CD15s in iTreg, which further inhibits Tcons. Osteoclasts producing APRIL and PD-L1 significantly block Tcon expansion by iTreg generation, which is overcome by combined treatment with anti-APRIL and -PD1/PD-L1 mAbs. Finally, APRIL increases IL-10-producing B regulatory cells (Bregs) via TACI on BM Bregs of MM patients. Taken together, these results define novel APRIL actions via TACI on Tregs and Bregs to promote MM cell survival, providing the rationale for targeting APRIL/TACI system to alleviate the immunosuppressive BM milieu and improve patient outcome in MM.
Collapse
Affiliation(s)
- Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Liang Lin
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Lijie Xing
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Hematology, Shandong Provincial Hospital affiliated to Shandong University, No. 324, Jingwu Road, Jinan, Shandong, 250021, People's Republic of China
| | - Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tengteng Yu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chirag Acharya
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Phillip A Hsieh
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - John Dulos
- Aduro Biotech Europe, Oss, The Netherlands
| | | | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Paul Richardson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
14
|
Wang S, Zhang C, Zhang Z, Qian W, Sun Y, Ji B, Zhang Y, Zhu C, Ji D, Wang Q, Sun Y. Transcriptome analysis in primary colorectal cancer tissues from patients with and without liver metastases using next-generation sequencing. Cancer Med 2017; 6:1976-1987. [PMID: 28745433 PMCID: PMC5548889 DOI: 10.1002/cam4.1147] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/06/2017] [Accepted: 06/25/2017] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide and liver metastases are the leading cause of death in patients with CRC. In this study, we performed next-generation sequencing profiling on primary colorectal tumor tissues obtained from three CRC patients with liver metastases and three CRC patients without liver metastases to identify differentially expressed genes (DEGs) that might be responsible for the metastases process. After filtering 2690 DEGs, comprising 996 upregulated and 1694 downregulated RNAs, 22 upregulated and 73 downregulated DEGs were identified. Gene ontology (GO) and pathway analyses were performed to determine the underlying mechanisms. Single-organism process (biological process), cell (cellular component), and binding (molecular function) were the most related terms in the GO analysis. We selected the top 13 upregulated and top 12 downregulated genes by fold change to verify their differential expression using quantitative real-time reverse transcription PCR (qRT-PCR) and immunohistochemistry (IHC). The validation showed that three most significantly upregulated DEGs were HOXD10, UGT2A3, and SLC13A2, whereas the five most significantly downregulated DEGs were SPP1, CXCL8, MMP3, OSM, and CXCL6, respectively. These aberrantly expressed genes may play pivotal roles in promoting or inhibiting metastases. Further studies are required to determine the functions of DEGs to promote the diagnosis of metastases and provide novel chemotherapy targets.
Collapse
Affiliation(s)
- Sen Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chuan Zhang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Zhiyuan Zhang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Wenwei Qian
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Ye Sun
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Bing Ji
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yue Zhang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chunyan Zhu
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Dongjian Ji
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Qingyuan Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yueming Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| |
Collapse
|
15
|
Wang H, Liu X, Long M, Huang Y, Zhang L, Zhang R, Zheng Y, Liao X, Wang Y, Liao Q, Li W, Tang Z, Tong Q, Wang X, Fang F, Rojo de la Vega M, Ouyang Q, Zhang DD, Yu S, Zheng H. NRF2 activation by antioxidant antidiabetic agents accelerates tumor metastasis. Sci Transl Med 2016; 8:334ra51. [PMID: 27075625 DOI: 10.1126/scitranslmed.aad6095] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/18/2016] [Indexed: 12/18/2022]
Abstract
Cancer is a common comorbidity of diabetic patients; however, little is known about the effects that antidiabetic drugs have on tumors. We discovered that common classes of drugs used in type 2 diabetes mellitus, the hypoglycemic dipeptidyl peptidase-4 inhibitors (DPP-4i) saxagliptin and sitagliptin, as well as the antineuropathic α-lipoic acid (ALA), do not increase tumor incidence but increase the risk of metastasis of existing tumors. Specifically, these drugs induce prolonged activation of the nuclear factor E2-related factor 2 (NRF2)-mediated antioxidant response through inhibition of KEAP1-C151-dependent ubiquitination and subsequent degradation of NRF2, resulting in up-regulated expression of metastasis-associated proteins, increased cancer cell migration, and promotion of metastasis in xenograft mouse models. Accordingly, knockdown of NRF2 attenuated naturally occurring and DPP-4i-induced tumor metastasis, whereas NRF2 activation accelerated metastasis. Furthermore, in human liver cancer tissue samples, increased NRF2 expression correlated with metastasis. Our findings suggest that antioxidants that activate NRF2 signaling may need to be administered with caution in cancer patients, such as diabetic patients with cancer. Moreover, NRF2 may be a potential biomarker and therapeutic target for tumor metastasis.
Collapse
Affiliation(s)
- Hui Wang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xiufei Liu
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Min Long
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yi Huang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Immunity and Infectious Diseases, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Linlin Zhang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Rui Zhang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yi Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xiaoyu Liao
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yuren Wang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Qian Liao
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Wenjie Li
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Zili Tang
- Molecular and Translational Radiation Oncology, Heidelberg Ion Therapy Center, Heidelberg Institute of Radiation Oncology, University of Heidelberg Medical School, National Center for Cancer Diseases, German Cancer Research Center, Heidelberg 69120, Germany
| | - Qiang Tong
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xiaocui Wang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Fang Fang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Montserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Qin Ouyang
- College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA.
| | - Shicang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
16
|
Jiang X, Mao W, Yang Z, Zeng J, Zhang Y, Song Y, Kong Y, Ren S, Zuo Y. Silencing P2X7 receptor downregulates the expression of TCP-1 involved in lymphoma lymphatic metastasis. Oncotarget 2016; 6:42105-17. [PMID: 26556873 PMCID: PMC4747213 DOI: 10.18632/oncotarget.5870] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 10/27/2015] [Indexed: 01/26/2023] Open
Abstract
P2X7R is an ATP-gated cation channel that participates in cell proliferation and apoptosis. TCP-1 assists with the protein folding. According to our previous research, the P2X7R has a potential role in P388D1 lymphoid neoplasm cells dissemination to peripheral lymph nodes. In order to make a further exploration about the probable mechanism, the lymph nodes which metastasized by P2X7R-silenced P388D1 cells or non-silenced cells were analyzed by 2DE and a MALDI-TOF-based proteomics approach. In the 64 proteins which were differentially expressed between two groups, TCP-1 was found to be significantly decreased in P2X7R shRNA group compared to controls. This correlation was also found in subsequent experiments in vivo and in vitro. The positive correlation between P2X7R and TCP-1 was also proved in both lymphoma and benign lymphadenopathy tissues from patients. It indicates that TCP-1 may be a crucial downstream molecular of P2X7R and plays a novel role in lymphoid neoplasm metastasis.
Collapse
Affiliation(s)
- Xudong Jiang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China
| | - Wenjuan Mao
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China
| | - Ziyi Yang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China
| | - Jia Zeng
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China
| | - Yi Zhang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China.,Department of Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yang Song
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China.,Department of Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Ying Kong
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Shuangyi Ren
- Department of Surgery, the Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Yunfei Zuo
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
17
|
Hou W, Meng X, Wang Y, Mo W, Wu Y, Yu M. Characterization and high-yield production of non- N-glycosylated recombinant human BCMA-Fc in Pichia pastoris. Eng Life Sci 2016; 17:96-106. [PMID: 32624756 DOI: 10.1002/elsc.201600039] [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: 01/29/2016] [Revised: 05/17/2016] [Accepted: 06/09/2016] [Indexed: 11/11/2022] Open
Abstract
B-cell maturation antigen (BCMA) fused at the C-terminus to the Fc portion of human IgG1 (BCMA-Fc) blocks B-cell activating factor (BAFF) and proliferation-inducing ligand (APRIL)-mediated B-cell activation, leading to immune disorders. The fusion protein has been cloned and produced by several engineering cell lines. To reduce cost and enhance production, we attempted to express recombinant human BCMA-Fc (rhBCMA-Fc) in Pichia pastoris under the control of the AOX1 methanol-inducible promoter. To produce the target protein with uniform molecular weight and reduced immunogenicity, we mutated two predicted N-linked glycosylation sites. The secretory yield was improved by codon optimization of the target gene sequence. After fed-batch fermentation under optimized conditions, the highest yield (207 mg/L) of rhBCMA-Fc was obtained with high productivity (3.45 mg/L/h). The purified functional rhBCMA-Fc possessed high-binding affinity to APRIL and dose-dependent inhibition of APRIL-induced proliferative activity in vitro through three-step purification. Thus, this yeast-derived expression method could be a low-cost and effective alternative to the production of rhBCMA-Fc in mammalian cell lines.
Collapse
Affiliation(s)
- Weihua Hou
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Xianchao Meng
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Yuxiong Wang
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Wei Mo
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Yi Wu
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| | - Min Yu
- Ministry of Education and Department of Biochemistry and Molecular Biology School of Basic Medicine Fudan University Shanghai China
| |
Collapse
|
18
|
Jujuboside A Protects H9C2 Cells from Isoproterenol-Induced Injury via Activating PI3K/Akt/mTOR Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:9593716. [PMID: 27293469 PMCID: PMC4884826 DOI: 10.1155/2016/9593716] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/26/2016] [Indexed: 11/17/2022]
Abstract
Jujuboside A is a kind of the saponins isolated from the seeds of Ziziphus jujuba, which possesses multiple biological effects, such as antianxiety, antioxidant, and anti-inflammatory effects; however, its mediatory effect on isoproterenol-stimulated cardiomyocytes has not been investigated yet. In this study, we tried to detect the protective effect and potential mechanism of JUA on ISO-induced cardiomyocytes injury. H9C2 cells were treated with ISO to induce cell damage. Cells were pretreated with JUA to investigate the effects on the cell viability, morphological changes, light chain 3 conversion, and the activation of PI3K/Akt/mTOR signaling pathway. Results showed that ISO significantly inhibited the cell viability in a time- and dose-dependent manner. JUA pretreatment could reverse the reduction of cell viability and better the injury of H9C2 cells induced by ISO. Western blot analysis showed that JUA could accelerate the phosphorylation of PI3K, Akt, and mTOR. Results also indicated that JUA could significantly decrease the ratio of microtubule-associated protein LC3-II/I in H9C2 cells. Taken together, our research showed that JUA could notably reduce the damage cause by ISO via promoting the phosphorylation of PI3K, Akt, and mTOR and inhibiting LC3 conversion, which may be a potential choice for the treatment of heart diseases.
Collapse
|
19
|
YU YANG, YU XIAOFENG, MA JIANXIA, TONG YILI, YAO JIANFENG. Effects of NVP-BEZ235 on the proliferation, migration, apoptosis and autophagy in HT-29 human colorectal adenocarcinoma cells. Int J Oncol 2016; 49:285-93. [DOI: 10.3892/ijo.2016.3507] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/21/2016] [Indexed: 11/05/2022] Open
|
20
|
Xiang T, Yu F, Fei R, Qian J, Chen W. CHRNA7 inhibits cell invasion and metastasis of LoVo human colorectal cancer cells through PI3K/Akt signaling. Oncol Rep 2015; 35:999-1005. [PMID: 26719016 DOI: 10.3892/or.2015.4462] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/23/2015] [Indexed: 11/06/2022] Open
Abstract
The α7 neuronal nicotinic receptor gene (CHRNA7) is widely expressed in both the brain and periphery whereas its encoding protein of α7 neuronal acetylcholine receptor (α7nAChR) belongs to the nicotinic acetylcholine receptor family. Considerable evidence suggests that α7nAChR plays an important role in chronic inflammatory and neuropathic pain signaling and thus has been proposed as a potential target for treating cognitive deficits in patients with schizophrenia, attention deficit hyperactivity disorder (ADHD) and Alzheimer's disease. The aim of the present study was to determine the role of endogenous α7nAChR signaling in human colorectal cancer growth and metastasis. pLVX‑CHRNA7 encoding the full length of CHRNA7 was constructed and transfected into LoVo human colorectal cancer cells. Cell proliferation was measured by Cell Counting Kit‑8 (CCK‑8), and cell migration and invasion were detected by Transwell chamber assays. Expression and activity of metastasis‑related metalloproteinases (MMPs) were analyzed by western blotting and gelatin zymography, respectively. Activation of metastasis-related signaling molecules was detected by western blotting. LY294002 was used to specifically block the phosphatidylinositol 3‑kinase/v‑akt murine thymoma viral oncogene homologue (PI3K/Akt) pathway. We showed that concomitantly with an increase in α7nAChR expression after transfection, LoVo cells presented reduced abilities for migration and invasion, which was accompanied by reduced expression levels of MMP‑1 and MMP‑9 as well as activation of the PI3K/Akt signaling pathway. The application of LY294002 restored the migration and invasion abilities of the LoVo cells bearing CHRNA7. Collectively, we conclude that overexpression of CHRNA7 negatively controls colorectal cancer LoVo cell invasion and metastasis via PI3K/Akt pathway activation and may serve as either a diagnostic marker or a therapeutic target for colorectal cancer metastasis.
Collapse
Affiliation(s)
- Tao Xiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Feng Yu
- Anorectal Department, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310012, P.R. China
| | - Rushan Fei
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Jing Qian
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Research Center of Infection and Immunity, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Wenbin Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
21
|
Tumor necrosis factor superfamily member 13 is a novel biomarker for diagnosis and prognosis and promotes cancer cell proliferation in laryngeal squamous cell carcinoma. Tumour Biol 2015; 37:2635-45. [PMID: 26395262 DOI: 10.1007/s13277-015-4016-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/28/2015] [Indexed: 12/27/2022] Open
Abstract
Tumor necrosis factor superfamily member 13 (TNFSF13) modulates cell proliferation and apoptosis and participates in the pathogenesis of solid tumors, but its role in laryngeal cancer development is not clearly defined. In order to investigate whether TNFSF13 can be used as a biomarker for diagnosis and prognosis in laryngeal squamous cell carcinoma (LSCC) and the role of TNFSF13 in laryngeal cancer carcinogenesis, we conducted immunohistochemistry and ELISA assays to evaluate the expression level of TNFSF13 in laryngeal cancer patients and the contrast. We also conducted experiments on the functional study of TNFSF13 in vitro. We found that the expression levels of TNFSF13, ki-67, and NF-κB p65 in LSCC tumor tissues were higher than those in vocal polyp and para-carcinoma tissues. The Spearman rank correlation analysis showed that the expression of TNFSF13 had a positive correlation with the expression of ki-67 and NF-κB p65. Cox regression analysis and Kaplan-Meier plots confirmed the expression level of TNFSF13 was a prognostic factor for LSCC. Moreover, the serum TNFSF13 level was significantly higher in LSCC patients than in the controls, and the serum expression level of TNFSF13 can distinguish LSCC from healthy people, precancerosis, or laryngeal benign tumor. In addition, functional study of TNFSF13 in vitro revealed that knockdown of TNFSF13 inhibited cell proliferation by inducing G1 phase cell cycle arrest in Hep-2 cells. In conclusion, TNFSF13 may be a potential novel molecular target for diagnosis and prognosis in human LSCC, and therapies that target TNFSF13 may have clinical significance for the treatment of LSCC.
Collapse
|
22
|
miR-383 inhibits hepatocellular carcinoma cell proliferation via targeting APRIL. Tumour Biol 2015; 37:2497-507. [DOI: 10.1007/s13277-015-4071-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/13/2015] [Indexed: 01/26/2023] Open
|
23
|
Lee C, Park JW, Suh JH, Moon KC. High expression of APRIL correlates with poor prognosis in clear cell renal cell carcinoma. Pathol Res Pract 2015; 211:824-8. [PMID: 26296917 DOI: 10.1016/j.prp.2015.07.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/16/2015] [Indexed: 12/26/2022]
Abstract
INTRODUCTION The members of the Tumor Necrosis Factor Superfamily (TNFSF), including A PRoliferation Inducing Ligand (APRIL), have been studied in RCC and other tumors. In this study, we investigated the expression of APRIL in resected clear cell renal cell carcinoma (CCRCC) samples by immunohistochemistry (IHC) and analyzed its association with the clinicopathologic characteristics and prognosis of the patients. METHODS We examined 206 CCRCC samples from patients who underwent radical or partial nephrectomy at Seoul National University Hospital between 1999 and 2002. Tissue microarray (TMA) blocks were made, and immunohistochemical staining for APRIL expression was performed. RESULTS We classified the IHC results as high expression or low expression. Of the 206 cases, 89 cases (43.2%) were classified as showing high expression, and 117 cases (56.8%) showed low expression. The high expression of APRIL was significantly correlated with higher Fuhrman nuclear grade and higher pathologic stage (p=0.000 and 0.004), and we observed that the high expression of APRIL was significantly correlated with the overall survival of the patients (p=0.045) and cancer-specific survival (p=0.020), but was not correlated with disease-free survival (p=0.106). In multivariate analysis adjusted for Fuhrman nuclear grade and pathologic stage, the high expression of APRIL was not an independent prognostic factor for CCRCC, as determined by overall survival (p=0.830) and cancer-specific survival (p=0.792). CONCLUSIONS We found that the high expression of APRIL in CCRCCs was correlated with high Fuhrman nuclear grade, high pathologic stage, and poor overall and cancer-specific survival of the patients. However, it did not correlate with disease-free survival and was not an independent prognostic factor.
Collapse
Affiliation(s)
- Cheol Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Whan Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ja Hee Suh
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea; Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
24
|
He XQ, Guan J, Liu F, Li J, He MR. Identification of the sAPRIL binding peptide and its growth inhibition effects in the colorectal cancer cells. PLoS One 2015; 10:e0120564. [PMID: 25826583 PMCID: PMC4380366 DOI: 10.1371/journal.pone.0120564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 02/05/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A proliferation-inducing ligand (APRIL) is a member of the tumor necrosis factor (TNF) super family. It binds to its specific receptors and is involved in multiple processes during tumorigenesis and tumor cells proliferation. High levels of APRIL expression are closely correlated to the growth, metastasis, and 5-FU drug resistance of colorectal cancer. The aim of this study was to identify a specific APRIL binding peptide (BP) able to block APRIL activity that could be used as a potential treatment for colorectal cancer. METHODS A phage display library was used to identify peptides that bound selectively to soluble recombinant human APRIL (sAPRIL). The peptides with the highest binding affinity for sAPRIL were identified using ELISA. The effects of sAPRIL-BP on cell proliferation and cell cycle/apoptosis in vitro were evaluated using the CCK-8 assay and flow cytometry, respectively. An in vivo mouse model of colorectal cancer was used to determine the anti-tumor efficacy of the sAPRIL-BP. RESULTS Three candidate peptides were characterized from eight phage clones with high binding affinity for sAPRIL. The peptide with the highest affinity was selected for further characterization. The identified sAPRIL-BP suppressed tumor cell proliferation and cell cycle progression in LOVO cells in a dose-dependent manner. In vivo in a mouse colorectal challenge model, the sAPRIL-BP reduced the growth of tumor xenografts in nude mice by inhibiting proliferation and inducing apoptosis intratumorally. Moreover, in an in vivo metastasis model, sAPRIL-BP reduced liver metastasis of colorectal cancer cells. CONCLUSIONS sAPRIL-BP significantly suppressed tumor growth in vitro and in vivo and might be a candidate for treating colorectal cancers that express high levels of APRIL.
Collapse
Affiliation(s)
- Xiao-qing He
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Digestive Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Oncology Department, Wuzhou Red Cross Hospital, Wuzhou 543002, Guangxi Province, China
| | - Jing Guan
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Digestive Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- Army Reserve Anti-aircraft Artillery Hospital, Zhengzhou 450002, Henan Province, China
| | - Fang Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Digestive Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Jing Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Digestive Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Mei-rong He
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Digestive Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
- * E-mail:
| |
Collapse
|
25
|
Gao W, Cai L, Xu X, Fan J, Xue X, Yan X, Qu Q, Wang X, Zhang C, Wu G. Anti-CTGF single-chain variable fragment dimers inhibit human airway smooth muscle (ASM) cell proliferation by down-regulating p-Akt and p-mTOR levels. PLoS One 2014; 9:e113980. [PMID: 25478966 PMCID: PMC4257608 DOI: 10.1371/journal.pone.0113980] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 11/02/2014] [Indexed: 11/19/2022] Open
Abstract
Connective tissue growth factor (CTGF) contributes to airway smooth muscle (ASM) cell hyperplasia in asthma. Humanized single-chain variable fragment antibody (scFv) was well characterized as a CTGF antagonist in the differentiation of fibroblast into myofibroblast and pulmonary fibrosis in our previous studies. To further improve the bioactivity of scFv, we constructed a plasmid to express scFv-linker-matrilin-6×His fusion proteins that could self-assemble into the scFv dimers by disulfide bonds in matrilin under non-reducing conditions. An immunoreactivity assay demonstrated that the scFv dimer could highly bind to CTGF in a concentration-dependent manner. The MTT and EdU assay results revealed that CTGF (≥10 ng/mL) promoted the proliferation of ASM cells, and this effect was inhibited when the cells were treated with anti-CTGF scFv dimer. The western blot analysis results showed that increased phosphorylation of Akt and mTOR induced by CTGF could be suppressed by this scFv dimer. Based on these findings, anti-CTGF scFv dimer may be a potential agent for the prevention of airway remodeling in asthma.
Collapse
Affiliation(s)
- Wei Gao
- Medical School, Southeast University, Nanjing 210009, China
| | - Liting Cai
- Medical School, Southeast University, Nanjing 210009, China
| | - Xudong Xu
- Department of Biological engineering, Southeast University, Nanjing 210009, China
| | - Juxiang Fan
- Medical School, Southeast University, Nanjing 210009, China
| | - Xiulei Xue
- Medical School, Southeast University, Nanjing 210009, China
| | - Xuejiao Yan
- Medical School, Southeast University, Nanjing 210009, China
| | - Qinrong Qu
- Medical School, Southeast University, Nanjing 210009, China
| | - Xihua Wang
- Department of Respiration, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Chen Zhang
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Guoqiu Wu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing 210009, China
| |
Collapse
|
26
|
Kwak TK, Sohn EJ, Kim S, Won G, Choi JU, Jeong K, Jeong M, Kwon OS, Kim SH. Inhibitory effect of ethanol extract of Ocimum sanctum on osteopontin mediated metastasis of NCI-H460 non-small cell lung cancer cells. Altern Ther Health Med 2014; 14:419. [PMID: 25345853 PMCID: PMC4219006 DOI: 10.1186/1472-6882-14-419] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 06/27/2014] [Indexed: 01/10/2023]
Abstract
Background Osteopontin (OPN) is one of important molecular targets in cancer progression, metastasis as a calcium-binding, extracellular-matrix-associated protein of the small integrin-binding ligand and, N-linked glycoprotein. In the present study, anti-metastatic mechanism of ethanol extracts of Ocimum sanctum (EEOS) was elucidated on OPN enhanced metastasis in NCI-H460 non- small cell lung cancer cells. Methods Cell viability was measured by MTT assay. Adhesion and invasion assays were carried out to see that EEOS inhibited cell adhesion and invasion in OPN treated and non-treated NCI-H 460 cells. RT-PCR was used to determine the mRNA levels of uPA, uPAR, and EGFR. Results EEOS significantly inhibited cell adhesion and invasion in OPN treated and non treated NCI-H460 cells, though EEOS did not show any toxicity up to 200 μg/ml. EEOS effectively attenuated the expression of OPN and CD44 and also OPN activated the expression of CD44 in NCI-H460 cells. In addition, EEOS effectively suppressed the expression of phosphatidylinositide 3-kinases (PI3K) and cyclooxygenase 2 (COX-2) and the phosphorylation of Akt at protein level in OPN treated NCI-H460 cells. Also, EEOS significantly attenuated the expression of urokinase plasminogen activator (uPA), its receptor (uPAR) and epidermal growth factor receptor (EGFR) at mRNA level and reduced vascular endothelial growth factor (VEGF) production and MMP-9 activity in OPN treated NCI-H460 cells. Furthermore, PI3K/Akt inhibitor LY294002 enhanced anti-metastatic potential of EEOS to attenuate the expression of uPA and MMP-9 in OPN treated NCI-H 460 cells. Conclusion Overall, our findings suggest that anti-metastatic mechanism of EEOS is mediated by inhibition of PI3K/Akt in OPN treated NCI-H460 non-small cell lung cancer cells.
Collapse
|
27
|
Amelioration of colorectal cancer using negative lipidoid nanoparticles to encapsulate siRNA against APRIL by enema delivery mode. Pathol Oncol Res 2014; 20:953-64. [PMID: 24771268 DOI: 10.1007/s12253-014-9779-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 04/09/2014] [Indexed: 01/24/2023]
Abstract
A proliferation-inducing ligand (APRIL) is a key cell proliferation-regulatory molecule and have been investigated well enough in immunity regulation and a few of immune diseases. APRIL can stimulate tumor cell growth and is up-expressed in cancer tissues, especially in CRC (colorectal cancer). However, whether inhibition of APRIL can regulate tumor-relative genes expression in vivo and subsequently ameliorate the pathological progress of CRC remains obscure. To address this question, we developed a novel negative lipidoid nanoparticles (NLNs) encapsulating small interference RNA (siRNA) for selectively silencing APRIL in the parenchyma of CRC focus in vivo, which uptake proceeded through a lipid raft endocytotic pathway. Local enema delivery of APRIL-NLNs silenced APRIL in CRC cells and animal models, and then ameliorated experimentally the progress of CRC by suppressing CRC cell proliferation, metastasis, and apoptosis-related cytokine expression and did not affect the function of liver and kidneys and not trigger the immune response of CRC models. This study reveals APRIL to be a potential anti-CRC target by in vivo experiments, and suggests that the application of similar modes of siRNA delivery may be feasible in other therapeutic settings.
Collapse
|
28
|
Malinowsky K, Nitsche U, Janssen KP, Bader FG, Späth C, Drecoll E, Keller G, Höfler H, Slotta-Huspenina J, Becker KF. Activation of the PI3K/AKT pathway correlates with prognosis in stage II colon cancer. Br J Cancer 2014; 110:2081-9. [PMID: 24619078 PMCID: PMC3992486 DOI: 10.1038/bjc.2014.100] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 11/05/2013] [Accepted: 01/28/2014] [Indexed: 12/27/2022] Open
Abstract
Background: Patients with UICC/AJCC stage II colon cancer have a high 5-year overall survival rate after surgery. Nevertheless, a significant subgroup of patients develops tumour recurrence. Currently, there are no clinically established biomarkers available to identify this patient group. We applied reverse-phase protein arrays (RPPA) for phosphatidylinositide-3-kinase pathway activation mapping to stratify patients according to their risk of tumour recurrence after surgery. Methods: Full-length proteins were extracted from formalin-fixed, paraffin-embedded tissue samples of 118 patients who underwent curative resection. RPPA technology was used to analyse expression and/or phosphorylation levels of six major factors of the phosphatidylinositide-3-kinase pathway. Oncogenic mutations of KRAS and BRAF, and DNA microsatellite status, currently discussed as prognostic markers, were analysed in parallel. Results: Expression of phospho-AKT (HR=3.52; P=0.032), S6RP (HR=6.3; P=0.044), and phospho-4E-BP1 (HR=4.12; P=0.011) were prognostic factors for disease-free survival. None of the molecular genetic alterations were significantly associated with prognosis. Conclusions: Our data indicate that activation of the PI3K/AKT pathway evidenced on the protein level might be a valuable prognostic marker to stratify patients for their risk of tumour recurrence. Beside adjuvant chemotherapy targeting of upregulated PI3K/AKT signalling may be an attractive strategy for treatment of high-risk patients.
Collapse
Affiliation(s)
- K Malinowsky
- Department of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - U Nitsche
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - K-P Janssen
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - F G Bader
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - C Späth
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - E Drecoll
- Department of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - G Keller
- Department of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - H Höfler
- 1] Department of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany [2] Department of Pathology, Helmholtz-Centre Munich, Ingolstädter Landstrasse 1, 85764 Munich, Germany
| | - J Slotta-Huspenina
- Department of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| | - K-F Becker
- Department of Pathology, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany
| |
Collapse
|
29
|
Zhang ZJ, Yang YK, Wu WZ. Bufalin attenuates the stage and metastatic potential of hepatocellular carcinoma in nude mice. J Transl Med 2014; 12:57. [PMID: 24581171 PMCID: PMC4015709 DOI: 10.1186/1479-5876-12-57] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 02/05/2014] [Indexed: 11/10/2022] Open
Abstract
Background Advanced hepatocellular carcinoma (HCC) patients undergo significant tumor growth and metastasis. Here, we investigated bufalin for treating HCC, which exhibits anti-tumor activities in many tumor cell lines. Method In our experiment, HCCLM3-R cells were injected into nude mice to form subcutaneous human HCC tumors that were implanted into the liver to establish orthotopic transplantation tumor models. Bufalin was injected intraperitoneally at 1 or 1.5 mg/kg. LY294002 (100 mg/kg), a potent inhibitor of Akt which reduced the levels of pAkt in HCCLM3 cell lines, was injected intraperitoneally into one group thrice weekly. The control was injected with an equal volume of saline. Morphological alterations were evaluated in the liver and lung by stereomicroscopy, the apoptotic rate was measured by TUNEL staining, and expression of AKT/GSK3β/β-catenin/E-cadherin signaling pathway-related proteins was detected by immunohistochemistry (IHC) and western blot analysis. Results These results suggested that the sizes and qualities of orthotopic transplanted tumors as well as pulmonary metastasis decreased markedly at the highest bufalin dose compared with that in the control. Orthotopic transplanted tumor tissues were necrotic in bufalin-treated groups and the apoptotic cell number was markedly higher at the highest bufalin dose compared with that in the control. Certain changes of expression of AKT/GSK3β/β-catenin/E-cadherin signaling pathway-related proteins were in tumor tissues, which were related to the bufalin dose. Similar results were observed in the LY294002-treated group. Conclusion Based on the above, one can draw conclusions that bufalin has significant anti-tumor activities and reduces the metastatic potential in an orthotopic transplantation tumor model of human HCC. Inhibition of AKT/GSK3β/β-catenin/E-cadherin signaling pathways by bufalin may show therapeutic effects in advanced HCC patients.
Collapse
Affiliation(s)
| | - Yun-Ke Yang
- Department of Traditional Chinese Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, China.
| | | |
Collapse
|
30
|
Chen Y, Xin X, Li J, Xu J, Yu X, Li T, Mo Z, Hu Y. RTK/ERK pathway under natural selection associated with prostate cancer. PLoS One 2013; 8:e78254. [PMID: 24223781 PMCID: PMC3817240 DOI: 10.1371/journal.pone.0078254] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 09/10/2013] [Indexed: 12/28/2022] Open
Abstract
Prostate cancer (PCa) is a global disease causing large numbers of deaths every year. Recent studies have indicated the RTK/ERK pathway might be a key pathway in the development of PCa. However, the exact association and evolution-based mechanism remain unclear. This study was conducted by combining genotypic and phenotypic data from the Chinese Consortium for Prostate Cancer Genetics (ChinaPCa) with related databases such as the HapMap Project and Genevar. In this analysis, expression of quantitative trait loci (eQTLs) analysis, natural selection and gene-based pathway analysis were involved. The pathway analysis confirmed the positive relationship between PCa risk and several key genes. In addition, combined with the natural selection, it seems that 4 genes (EGFR, ERBB2, PTK2, and RAF1) with five SNPs (rs11238349, rs17172438, rs984654, rs11773818, and rs17172432) especially rs17172432, might be pivotal factors in the development of PCa. The results indicate that the RTK/ERK pathway under natural selection is a key link in PCa risk. The joint effect of the genes and loci with positive selection might be one reason for the development of PCa. Dealing with all the factors simultaneously might give insight into prevention and aid in predicting the success of potential therapies for PCa.
Collapse
Affiliation(s)
- Yang Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xianxiang Xin
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Medical Research Center, Guangxi Medical University, Nanning, Guangxi, China
| | - Jie Li
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Research Center for Guangxi Reproductive Medicine, First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, China
| | - Jianfeng Xu
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
- Fudan Center for Genetic Epidemiology, School of Life Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Xiaoxiang Yu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Urology, the 303rd Hospital of Chinese People's Liberation Army, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Tianyu Li
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Department of Urology and Nephrology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yanling Hu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- Medical Research Center, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
31
|
Fu M, Fan W, Pu X, Ni H, Zhang W, Chang F, Gong L, Xiong L, Wang J, Gu X. Elevated expression of SHIP2 correlates with poor prognosis in non-small cell lung cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:2185-2191. [PMID: 24133597 PMCID: PMC3796241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 08/17/2013] [Indexed: 06/02/2023]
Abstract
SH2-containing inositol 5'-phosphatase 2 (SHIP2) is a vital regulator of phosphoinositide pools in metabolic pathways and is considered to downregulate phosphatidylinositol 3'-kinase signaling, which underlies the development of several kinds of human cancers. However, SHIP2 expression in non-small cell lung cancer (NSCLC) and its relationship with the clinical characteristics of NSCLC remain poorly understood. In this study, one-step quantitative reverse transcription-polymerase chain reaction and immunohistochemistry analysis with tissue microarray was used to evaluate SHIP2 expression in NSCLC and to investigate the relationship of this expression to NSCLC prognosis. Results showed that the expression of SHIP2 messenger RNA and protein was significantly higher in NSCLC than in corresponding non-cancerous tissues (both p < 0.05). SHIP2 protein expression in NSCLC was related to lymph node metastasis (p = 0.042), TNM stage (p = 0.036), and 5-year survival rate (p = 0.046). The Kaplan-Meier method and log-rank test suggested that high SHIP2 expression, tobacco consumption, and advanced tumor stage were significantly associated with low survival of NSCLC patients. The results of this research suggested that SHIP2 expression was correlated with malignant phenotypes of NSCLC and may thus serve as a poor prognostic factor and valuable oncogene for NSCLC.
Collapse
Affiliation(s)
- Maoying Fu
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Weifei Fan
- Department of Hematology and Oncology, Jiangsu Province Geriatric InstituteNanjing 210029, China
| | - Xiaolin Pu
- Department of Hematology and Oncology, Jiangsu Province Geriatric InstituteNanjing 210029, China
| | - Huihui Ni
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Wei Zhang
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Feng Chang
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Li Gong
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| | - Lin Xiong
- Department of Pathology, The Second Affiliated Hospital of Nanjing Medical UniversityNanjing 210011, China
| | - Jun Wang
- Department of Hematology and Oncology, Jiangsu Province Geriatric InstituteNanjing 210029, China
| | - Xuefeng Gu
- Department of Infectious Diseases, The First People’s Hospital of Kunshan Affiliated with Jiangsu UniversitySuzhou 215000, China
| |
Collapse
|
32
|
APRIL depletion induces cell cycle arrest and apoptosis through blocking TGF-β1/ERK signaling pathway in human colorectal cancer cells. Mol Cell Biochem 2013; 383:179-89. [DOI: 10.1007/s11010-013-1766-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/10/2013] [Indexed: 02/06/2023]
|
33
|
Ding W, Wang J, Wang F, Wang G, Wu Q, Ju S, Cong H, Wang H. Serum sAPRIL: a potential tumor-associated biomarker to colorectal cancer. Clin Biochem 2013; 46:1590-4. [PMID: 23792260 DOI: 10.1016/j.clinbiochem.2013.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/28/2013] [Accepted: 06/11/2013] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The purpose of our study was to investigate the serum levels of soluble a-proliferation-inducing ligand (sAPRIL) in patients with colorectal cancer (CRC), benign intestinal disease and healthy volunteers and explore the potential possibility of sAPRIL severing as a CRC biomarker. METHODS Our investigation was conducted on 35 blood samples obtained from CRC patients, 32 blood samples from patients with benign intestinal diseases and 31 blood samples from healthy volunteers. The sAPRIL concentrations were examined by an enzyme-linked immunosorbent assay (ELISA) and data were analyzed with non-parametric Mann-Whitney U-test and X²-test. The correlation relationship between sAPRIL and CEA, as well as sAPRIL and CA19-9 was assessed by non-parametric Spearmen's correlation test, respectively. RESULTS The median value of sAPRIL in the malignant group was 10.43 ng/mL, compared with those of the benign group (4.89 ng/mL) and control group (3.30 ng/mL), respectively, which had an obvious significance (P<0.0003). Area under the receiver-operating characteristic (ROC) curve for sAPRIL was 0.854 (95% CI, 0.776-0.933). The optimal cut-off level of sAPRIL was 5.49 ng/mL. Serum sAPRIL had a positive correlation with CEA (r=0.637, P=0.000) and CA19-9 (r=0.357, P=0.008) in 35 patients with colorectal cancer. sAPRIL showed higher sensitivity (82.9%) than those of CEA (74.3%) and CA19-9 (65.7%) in CRC, respectively. CONCLUSION The results indicated that serum sAPRIL, as a potential biomarker, had a positive diagnostic value for colorectal cancer.
Collapse
Affiliation(s)
- Weifeng Ding
- Medical Laboratory Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China; School of Public Health, Nantong University, Nantong, Jiangsu Province, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhen Y, Ye Y, Yu X, Mai C, Zhou Y, Chen Y, Yang H, Lyu X, Song Y, Wu Q, Fu Q, Zhao M, Hua S, Wang H, Liu Z, Zhang Y, Fang W. Reduced CTGF expression promotes cell growth, migration, and invasion in nasopharyngeal carcinoma. PLoS One 2013; 8:e64976. [PMID: 23755163 PMCID: PMC3670884 DOI: 10.1371/journal.pone.0064976] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/19/2013] [Indexed: 01/11/2023] Open
Abstract
Background The role of CTGF varies in different types of cancer. The purpose of this study is to investigate the involvement of CTGF in tumor progression and prognosis of human nasopharyngeal carcinoma (NPC). Experimental design CTGF expression levels were examined in NPC tissues and cells, nasopharynx (NP) tissues, and NP69 cells. The effects and molecular mechanisms of CTGF expression on cell proliferation, migration, invasion, and cell cycle were also explored. Results NPC cells exhibited decreased mRNA expression of CTGF compared to immortalized human nasopharyngeal epithelial cell line NP69. Similarly, CTGF was observed to be downregulated in NPC compared to normal tissues at mRNA and protein levels. Furthermore, reduced CTGF was negatively associated with the progression of NPC. Knocking down CTGF expression enhanced the colony formation, cell migration, invasion, and G1/S cell cycle transition. Mechanistic analysis revealed that CTGF suppression activated FAK/PI3K/AKT and its downstream signals regulating the cell cycle, epithelial-mesenchymal transition (EMT) and MMPs. Finally, DNA methylation microarray revealed a lack of hypermethylation at the CTGF promoter, suggesting other mechanisms are associated with suppression of CTGF in NPC. Conclusion Our study demonstrates that reduced expression of CTGF promoted cell proliferation, migration, invasion and cell cycle progression through FAK/PI3K/AKT, EMT and MMP pathways in NPC.
Collapse
Affiliation(s)
- Yan Zhen
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Yanfen Ye
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Xiaoli Yu
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Chunping Mai
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Ying Zhou
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Yan Chen
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Huiling Yang
- School of Pharmacy, Guangdong Medical College, Dongguan, PR China
| | - Xiaoming Lyu
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Qiangyun Wu
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Qiaofen Fu
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Mengyang Zhao
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Shengni Hua
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Hao Wang
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
| | - Zhen Liu
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
- Department of Pathology of Basic School, Medical University of Guangzhou, Guangzhou, PR China
- * E-mail: (ZL); (YZ); (WF)
| | - Yajie Zhang
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
- Department of Pathology of Basic School, Medical University of Guangzhou, Guangzhou, PR China
- * E-mail: (ZL); (YZ); (WF)
| | - Weiyi Fang
- Cancer Research Institute of Basic Medicine School, Southern Medical University, Guangzhou, PR China
- * E-mail: (ZL); (YZ); (WF)
| |
Collapse
|