51
|
Bad phosphorylation as a target of inhibition in oncology. Cancer Lett 2017; 415:177-186. [PMID: 29175460 DOI: 10.1016/j.canlet.2017.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022]
Abstract
Bcl-2 agonist of cell death (BAD) is a BH3-only member of the Bcl-2 family which possesses important regulatory function in apoptosis. BAD has also been shown to possess many non-apoptotic functions closely linked to cancer including regulation of glycolysis, autophagy, cell cycle progression and immune system development. Interestingly, BAD can be either pro-apoptotic or pro-survival depending on the phosphorylation state of three specific serine residues (human S75, S99 and S118). Expression of BAD and BAD phosphorylation patterns have been shown to influence tumor initiation and progression and play a predictive role in disease prognosis, drug response and chemosensitivity in various cancers. This review aims to summarize the current evidence on the functional role of BAD phosphorylation in human cancer and evaluate the potential utility of modulating BAD phosphorylation in cancer.
Collapse
|
52
|
Liu J, Jin S, Wang R. MicroRNA‑139 suppressed tumor cell proliferation, migration and invasion by directly targeting HDGF in epithelial ovarian cancer. Mol Med Rep 2017; 16:3379-3386. [PMID: 28713954 DOI: 10.3892/mmr.2017.6956] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 04/05/2017] [Indexed: 11/05/2022] Open
Abstract
The current study investigated the expression and functional roles of microRNA‑139 (miR‑139) on human epithelial ovarian cancer (EOC). Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was performed to measure miR‑139 expression in EOC tissues and cell lines. The effects of miR‑139 on EOC cell proliferation, migration and invasion were assessed using MTT, cell migration and invasion assays, respectively. Subsequently, the molecular mechanism underlying the tumor suppressive roles of miR‑139 on EOC was determined by bioinformatics analysis, RT‑qPCR, western blotting and the luciferase reporter assay. According to the results, it was identified that miR‑139 was significantly downregulated in EOC tissues and cell lines. In addition, restoration of miR‑139 suppressed tumor cell proliferation, migration and invasion in EOC. Furthermore, hepatoma‑derived growth factor (HDGF) was identified as a target of miR‑139 in EOC. Upregulation of HDGF could rescue the inhibitory effects exerted by miR‑139 overexpression on EOC cell proliferation, migration and invasion. Collectively, the results indicated that miR‑139 was downregulated in EOC, and acted as a tumor suppressor by directly targeting HDGF. To the best of our knowledge, this was the first study to identify that miR‑139 contributes to the growth and metastasis of EOC.
Collapse
Affiliation(s)
- Junli Liu
- Department of Gynaecology and Obstetrics, Changzhi Peace Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Shuangling Jin
- Department of Gynaecology and Obstetrics, Changzhi Peace Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Rui Wang
- Department of Gynaecology and Obstetrics, Changzhi Peace Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| |
Collapse
|
53
|
Liu X, Chang X, Liu R, Yu X, Chen L, Aihara K. Quantifying critical states of complex diseases using single-sample dynamic network biomarkers. PLoS Comput Biol 2017; 13:e1005633. [PMID: 28678795 PMCID: PMC5517040 DOI: 10.1371/journal.pcbi.1005633] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 07/19/2017] [Accepted: 06/19/2017] [Indexed: 02/04/2023] Open
Abstract
Dynamic network biomarkers (DNB) can identify the critical state or tipping point of a disease, thereby predicting rather than diagnosing the disease. However, it is difficult to apply the DNB theory to clinical practice because evaluating DNB at the critical state required the data of multiple samples on each individual, which are generally not available, and thus limit the applicability of DNB. In this study, we developed a novel method, i.e., single-sample DNB (sDNB), to detect early-warning signals or critical states of diseases in individual patients with only a single sample for each patient, thus opening a new way to predict diseases in a personalized way. In contrast to the information of differential expressions used in traditional biomarkers to “diagnose disease”, sDNB is based on the information of differential associations, thereby having the ability to “predict disease” or “diagnose near-future disease”. Applying this method to datasets for influenza virus infection and cancer metastasis led to accurate identification of the critical states or correct prediction of the immediate diseases based on individual samples. We successfully identified the critical states or tipping points just before the appearance of disease symptoms for influenza virus infection and the onset of distant metastasis for individual patients with cancer, thereby demonstrating the effectiveness and efficiency of our method for quantifying critical states at the single-sample level. The concept of dynamic network biomarkers (DNB) was proposed for detecting the critical state or tipping point of a complex disease (a pre-disease state immediately preceding the disease state), and has been applied to study the mechanism of cell fate decision and immune checkpoint blockade. But DNB cannot be used to identify the critical state or tipping point for a single patient because evaluating DNB for critical state required the data of multiple samples. The proposed method can identify the critical state of a complex disease for a single patient by implementing the concept of DNB. This method not only can be applied to detect the critical state or tipping point of a single sample, but also can be used to study the mechanism of complex disease at a single sample level. The ability of accurately and efficiently identifying the critical state for a single sample can benefit the development of personalized medicine.
Collapse
Affiliation(s)
- Xiaoping Liu
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, Anhui Province, China
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Mathematics and Statistics, Shandong University at Weihai, Weihai, China
| | - Xiao Chang
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- College of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, Anhui Province, China
| | - Rui Liu
- School of Mathematics, South China University of Technology, Guangzhou, China
| | - Xiangtian Yu
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Luonan Chen
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- * E-mail: (LC); (KA)
| | - Kazuyuki Aihara
- Institute of Industrial Science, the University of Tokyo, Tokyo, Japan
- * E-mail: (LC); (KA)
| |
Collapse
|
54
|
Song R, Cong L, Ni G, Chen M, Sun H, Sun Y, Chen M. MicroRNA-195 inhibits the behavior of cervical cancer tumors by directly targeting HDGF. Oncol Lett 2017; 14:767-775. [PMID: 28693232 PMCID: PMC5494760 DOI: 10.3892/ol.2017.6210] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 01/31/2017] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are a class of conserved non-coding endogenous small regulatory RNAs that regulate target gene expression by binding to the 3'-untranslated region of target mRNAs in a base-pairing manner, resulting in repression of transcription or degradation of target mRNAs. It has been demonstrated previously that the abnormal expression of miRNAs is involved in the carcinogenesis and progression of cervical cancer. The aim of the present study was to investigate the expression, biological functions and underlying molecular mechanisms of miR-195 in cervical cancer. The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression level of miR-195 in cervical cancer tissues and cell lines. Following transfection, an MTT assay, cell migration and invasion assays, western blot analysis and a dual-luciferase reporter assay were performed in human cervical cancer cells. In the present study, it was identified that miR-195 was downregulated in cervical cancer tissues and cell lines. Additionally, upregulation of miR-195 and knockdown of hepatoma-derived growth factor (HDGF) inhibited proliferation, migration and invasion of cervical cancer cells. Furthermore, a dual-luciferase reporter assay identified that HDGF was a direct target gene of miR-195. RT-qPCR and western blot analysis demonstrated that miR-195 mimic inhibited HDGF expression at the mRNA and protein levels, whereas miR-195 inhibitor enhanced HDGF expression at the mRNA and protein levels. These results indicated that miR-195 targeted HDGF to inhibit the behavior of tumors in cervical cancer. These results also suggested that miR-195 was a potential therapeutic biomarker of cervical cancer.
Collapse
Affiliation(s)
- Rongrong Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
- Prenatal Diagnosis Center, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
- Department of Obstetrics and Gynecology, The People's Hospital of Xuancheng, Xuancheng, Anhui 242000, P.R. China
| | - Lin Cong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
- Prenatal Diagnosis Center, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Guantai Ni
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Min Chen
- Department of Obstetrics and Gynecology, The People's Hospital of Xuancheng, Xuancheng, Anhui 242000, P.R. China
| | - Honmei Sun
- Department of Obstetrics and Gynecology, The People's Hospital of Xuancheng, Xuancheng, Anhui 242000, P.R. China
| | - Yunxia Sun
- Department of Obstetrics and Gynecology, The People's Hospital of Xuancheng, Xuancheng, Anhui 242000, P.R. China
| | - Meiling Chen
- Department of Obstetrics and Gynecology, The People's Hospital of Xuancheng, Xuancheng, Anhui 242000, P.R. China
| |
Collapse
|
55
|
TRPV4 plays a role in breast cancer cell migration via Ca 2+-dependent activation of AKT and downregulation of E-cadherin cell cortex protein. Oncogenesis 2017; 6:e338. [PMID: 28530703 PMCID: PMC5523072 DOI: 10.1038/oncsis.2017.39] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 03/26/2017] [Accepted: 04/05/2017] [Indexed: 12/31/2022] Open
Abstract
TRPV4 belongs to the 'Transient Receptor Potential' (TRP) superfamily. It has been identified to profoundly affect a variety of physiological processes, including nociception, heat sensation and inflammation. Unlike other TRP superfamily channels, its role in cancers are unknown until recently when we reported TRPV4 to be required for cancer cell softness that may promote breast cancer cell extravasation and metastasis. Here, we elucidated the molecular mechanisms mediated by TRPV4 in the metastatic breast cancer cells. TRPV4-mediated signaling was demonstrated to involve Ca2+-dependent activation of AKT and downregulation of E-cadherin expression, which was abolished upon TRPV4 silencing. Functionally, TRPV4-enhanced breast caner cell transendothelial migration requires AKT activity while a combination of transcriptional and post-translational regulation contributed to the TRPV4-mediated E-cadherin downregulation. Finally, mass spectrometry analysis revealed that TRPV4 is required for the expression of a network of secreted proteins involved in extracellular matrix remodeling. In conclusion, TRPV4 may regulate breast cancer metastasis by regulating cell softness through the Ca2+-dependent AKT-E-cadherin signaling axis and regulation of the expression of extracellular proteins.
Collapse
|
56
|
Mukherjee S, Patra CR. Therapeutic application of anti-angiogenic nanomaterials in cancers. NANOSCALE 2016; 8:12444-12470. [PMID: 27067119 DOI: 10.1039/c5nr07887c] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Angiogenesis, the formation of new blood vessels from pre-existing vasculature, plays a vital role in physiological and pathological processes (embryonic development, wound healing, tumor growth and metastasis). The overall balance of angiogenesis inside the human body is maintained by pro- and anti-angiogenic signals. The processes by which drugs inhibit angiogenesis as well as tumor growth are called the anti-angiogenesis technique, a most promising cancer treatment strategy. Over the last couple of decades, scientists have been developing angiogenesis inhibitors for the treatment of cancers. However, conventional anti-angiogenic therapy has several limitations including drug resistance that can create problems for a successful therapeutic strategy. Therefore, a new comprehensive treatment strategy using antiangiogenic agents for the treatment of cancer is urgently needed. Recently researchers have been developing and designing several nanoparticles that show anti-angiogenic properties. These nanomedicines could be useful as an alternative strategy for the treatment of various cancers using anti-angiogenic therapy. In this review article, we critically focus on the potential application of anti-angiogenic nanomaterial and nanoparticle based drug/siRNA/peptide delivery systems in cancer therapeutics. We also discuss the basic and clinical perspectives of anti-angiogenesis therapy, highlighting its importance in tumor angiogenesis, current status and future prospects and challenges.
Collapse
Affiliation(s)
- Sudip Mukherjee
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad - 500007, Telangana, India.
| | | |
Collapse
|
57
|
Progranulin and granulin-like protein as novel VEGF-independent angiogenic factors derived from human mesothelioma cells. Oncogene 2016; 36:714-722. [PMID: 27345409 DOI: 10.1038/onc.2016.226] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/24/2022]
Abstract
Malignant mesothelioma is an aggressive tumor arising from the mesothelial cells of serous membranes and is associated with tumor angiogenesis, which is a prerequisite for tumor progression. Vascular endothelial growth factors (VEGFs) including VEGF-A have a crucial role in tumor angiogenesis. However, bevacizumab, a monoclonal antibody to VEGF-A, has recently been reported not to improve the progression-free survival of patients with malignant mesothelioma. Cell culture supernatant contains extracellular components such as serum, which can mask the existence of unknown cell-derived factors in the supernatant and make it difficult to detect the factors by subsequent protein analysis. We tried using serum-free culture for human mesothelioma cell lines, NCI-H28, NCI-H2452 and NCI-H2052, and only NCI-H2052 cells adapted to serum-free culture. We found that serum-free culture supernatant derived from NCI-H2052 cells induces the formation of capillary-like tube structures (tube formation) in three-dimensional culture, in which endothelial cells sandwiched between two layers of collagen or embedded in collagen are incubated with various angiogenic inducers. However, neither neutralization of VEGF-A nor RNA interference of VEGF receptor 2 (VEGFR2) suppressed the supernatant-induced tube formation. Using mass spectrometry, we identified a total of 399 proteins in the supernatant, among which interleukin-8 (IL-8), growth-regulated α-protein, midkine, IL-18, IL-6, hepatoma-derived growth factor, clusterin and granulin (GRN), also known as progranulin (PGRN), were included as a candidate protein inducing angiogenesis. Neutralizing assays and RNA interference showed that PGRN, but not the above seven candidate proteins, caused the supernatant-induced tube formation. We also found that NCI-H28 and NCI-H2452 cells express PGRN. Furthermore, we demonstrate that not only PGRN but also GRN-like protein have an important role in the supernatant-induced tube formation. Thus, mesothelioma-derived GRNs induce VEGF-independent angiogenesis.
Collapse
|
58
|
Kozlov SV, Waardenberg AJ, Engholm-Keller K, Arthur JW, Graham ME, Lavin M. Reactive Oxygen Species (ROS)-Activated ATM-Dependent Phosphorylation of Cytoplasmic Substrates Identified by Large-Scale Phosphoproteomics Screen. Mol Cell Proteomics 2016; 15:1032-47. [PMID: 26699800 PMCID: PMC4813686 DOI: 10.1074/mcp.m115.055723] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/08/2015] [Indexed: 01/06/2023] Open
Abstract
Ataxia-telangiectasia, mutated (ATM) protein plays a central role in phosphorylating a network of proteins in response to DNA damage. These proteins function in signaling pathways designed to maintain the stability of the genome and minimize the risk of disease by controlling cell cycle checkpoints, initiating DNA repair, and regulating gene expression. ATM kinase can be activated by a variety of stimuli, including oxidative stress. Here, we confirmed activation of cytoplasmic ATM by autophosphorylation at multiple sites. Then we employed a global quantitative phosphoproteomics approach to identify cytoplasmic proteins altered in their phosphorylation state in control and ataxia-telangiectasia (A-T) cells in response to oxidative damage. We demonstrated that ATM was activated by oxidative damage in the cytoplasm as well as in the nucleus and identified a total of 9,833 phosphorylation sites, including 6,686 high-confidence sites mapping to 2,536 unique proteins. A total of 62 differentially phosphorylated peptides were identified; of these, 43 were phosphorylated in control but not in A-T cells, and 19 varied in their level of phosphorylation. Motif enrichment analysis of phosphopeptides revealed that consensus ATM serine glutamine sites were overrepresented. When considering phosphorylation events, only observed in control cells (not observed in A-T cells), with predicted ATM sites phosphoSerine/phosphoThreonine glutamine, we narrowed this list to 11 candidate ATM-dependent cytoplasmic proteins. Two of these 11 were previously described as ATM substrates (HMGA1 and UIMCI/RAP80), another five were identified in a whole cell extract phosphoproteomic screens, and the remaining four proteins had not been identified previously in DNA damage response screens. We validated the phosphorylation of three of these proteins (oxidative stress responsive 1 (OSR1), HDGF, and ccdc82) as ATM dependent after H2O2 exposure, and another protein (S100A11) demonstrated ATM-dependence for translocation from the cytoplasm to the nucleus. These data provide new insights into the activation of ATM by oxidative stress through identification of novel substrates for ATM in the cytoplasm.
Collapse
Affiliation(s)
- Sergei V Kozlov
- From the ‡University of Queensland Centre for Clinical Research, University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston, Brisbane, QLD 4029 Australia
| | - Ashley J Waardenberg
- §Bioinformatics Unit, Children's Medical Research Institute, University of Sydney, 214 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Kasper Engholm-Keller
- ¶Synapse Proteomics Group, Children's Medical Research Institute, University of Sydney, 214 Hawkesbury Road, Westmead, NSW, 2145, Australia; ‖Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Jonathan W Arthur
- §Bioinformatics Unit, Children's Medical Research Institute, University of Sydney, 214 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Mark E Graham
- ¶Synapse Proteomics Group, Children's Medical Research Institute, University of Sydney, 214 Hawkesbury Road, Westmead, NSW, 2145, Australia
| | - Martin Lavin
- From the ‡University of Queensland Centre for Clinical Research, University of Queensland, Royal Brisbane & Women's Hospital Campus, Herston, Brisbane, QLD 4029 Australia;
| |
Collapse
|
59
|
Bao CH, Liu K, Wang XT, Ma W, Wang JB, Wang C, Jia YB, Wang NN, Tan BX, Song QX, Cheng YF. Prognostic role of hepatoma-derived growth factor in solid tumors of Eastern Asia: a systematic review and meta- analysis. Asian Pac J Cancer Prev 2016; 16:1803-11. [PMID: 25773828 DOI: 10.7314/apjcp.2015.16.5.1803] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Hepatoma-derived growth factor (HDGF) is a novel jack-of-all-trades in cancer. Here we quantify the prognostic impact of this biomarker and assess how consistent is its expression in solid tumors. A comprehensive search strategy was used to search relevant literature updated on October 3, 2014 in PubMed, EMBASE and WEB of Science. Correlations between HDGF expression and clinicopathological features or cancer prognosis was analyzed. All pooled HRs or ORs were derived from random-effects models. Twenty-six studies, primarily in Eastern Asia, covering 2,803 patients were included in the analysis, all of them published during the past decade. We found that HDGF overexpression was significantly associated with overall survival (OS) (HROS=2.35, 95%CI=2.04-2.71, p<0.001) and disease free survival (DFS) (HRDFS=2.25, 95%CI =1.81-2.79, p<0.001) in solid tumors, especially in non-small cell lung cancer, hepatocellular carcinoma and cholangiocarcinoma (CCA). Moreover, multivariate survival analysis showed that HDGF overexpression was an independent predictor of poor prognosis (HROS=2.41, 95%CI: 2.02-2.81, p<0.001; HRDFS=2.39, 95%CI: 1.77-3.24, p<0.001). In addition, HDGF overexpression was significantly associated with tumor category (T3-4 versus T1-2, OR=2.12, 95%CI: 1.17-3.83, p=0.013) and lymph node status (N+ versus N-, OR=2.37, 95%CI: 1.31-4.29, p=0.03) in CCA. This study provides a comprehensive examination of the literature available on the association of HDGF overexpression with OS, DFS and some clinicopathological features in solid tumors. Meta-analysis results provide evidence that HDGF may be a new indicator of poor cancer prognosis. Considering the limitations of the eligible studies, other large-scale prospective trials must be conducted to clarify the prognostic value of HDGF in predicting cancer survival.
Collapse
Affiliation(s)
- Ci-Hang Bao
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Shandong University, Jinan, China E-mail :
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Embryonic Lethality Due to Arrested Cardiac Development in Psip1/Hdgfrp2 Double-Deficient Mice. PLoS One 2015; 10:e0137797. [PMID: 26367869 PMCID: PMC4569352 DOI: 10.1371/journal.pone.0137797] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 08/20/2015] [Indexed: 12/28/2022] Open
Abstract
Hepatoma-derived growth factor (HDGF) related protein 2 (HRP2) and lens epithelium-derived growth factor (LEDGF)/p75 are closely related members of the HRP2 protein family. LEDGF/p75 has been implicated in numerous human pathologies including cancer, autoimmunity, and infectious disease. Knockout of the Psip1 gene, which encodes for LEDGF/p75 and the shorter LEDGF/p52 isoform, was previously shown to cause perinatal lethality in mice. The function of HRP2 was by contrast largely unknown. To learn about the role of HRP2 in development, we knocked out the Hdgfrp2 gene, which encodes for HRP2, in both normal and Psip1 knockout mice. Hdgfrp2 knockout mice developed normally and were fertile. By contrast, the double deficient mice died at approximate embryonic day (E) 13.5. Histological examination revealed ventricular septal defect (VSD) associated with E14.5 double knockout embryos. To investigate the underlying molecular mechanism(s), RNA recovered from ventricular tissue was subjected to RNA-sequencing on the Illumina platform. Bioinformatic analysis revealed several genes and biological pathways that were significantly deregulated by the Psip1 knockout and/or Psip1/Hdgfrp2 double knockout. Among the dozen genes known to encode for LEDGF/p75 binding factors, only the expression of Nova1, which encodes an RNA splicing factor, was significantly deregulated by the knockouts. However the expression of other RNA splicing factors, including the LEDGF/p52-interacting protein ASF/SF2, was not significantly altered, indicating that deregulation of global RNA splicing was not a driving factor in the pathology of the VSD. Tumor growth factor (Tgf) β-signaling, which plays a key role in cardiac morphogenesis during development, was the only pathway significantly deregulated by the double knockout as compared to control and Psip1 knockout samples. We accordingly speculate that deregulated Tgf-β signaling was a contributing factor to the VSD and prenatal lethality of Psip1/Hdgfrp2 double-deficient mice.
Collapse
|
61
|
Abstract
AIM To investigate the expression of hepatoma-derived growth factor (HDGF) in osteosarcoma (OS) and the correlation with clinicopathologic factors, prognosis, and tumor progression. METHOD HDGF expression in OS tissues was detected by immunohistochemistry. The correlation between HDGF and clinicopathologic factors was analyzed by chi-square test, and the association between HDGF expression and the overall survival rates was evaluated by univariate analysis using Kaplan-Meier method. HDGF concentration in cell medium or cell lysates was detected by enzyme-linked immunosorbent assay method. The effect of extrinsic and intrinsic HDGF on OS cell proliferation was detected by MTT assay after recombinant HDGF stimulation or HDGF knockdown, respectively. RESULTS Proportion of HDGF high expression was 18.69% (20/107) in OS. HDGF high expression was significantly associated with larger tumor size (P=0.004). With experiments in vitro, we demonstrated that human recombinant HDGF could activate AKT and MAPK signaling pathway, resulting in OS cell proliferation. By knocking down HDGF expression, we proved that intrinsic HDGF was required in OS proliferation. CONCLUSION High HDGF expression was significantly associated with larger OS tumor size and could promote OS cell proliferation, indicating that HDGF could be an effective biomarker and a potential drug target in OS treatment.
Collapse
Affiliation(s)
- Zhiguo Chen
- Department of Orthopedics, Linyi People's Hospital, Linyi City, Shandong Province, People's Republic of China
| | - Shenghai Qiu
- Department of Orthopedics, People's Hospital of Taiyuan, Taiyuan City, Shanxi Province, People's Republic of China
| | - Xiaofei Lu
- Department of General Surgery, Jinan Central Hospital affiliated to Shandong University, Jinan City, Shandong Province, People's Republic of China
| |
Collapse
|
62
|
Liu XJ, Liu WL, Yang FM, Yang XQ, Lu XF. Hepatoma-derived growth factor predicts unfavorable prognosis of epithelial ovarian cancer. Onco Targets Ther 2015; 8:2101-9. [PMID: 26316779 PMCID: PMC4540117 DOI: 10.2147/ott.s85660] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aim To evaluate the expression and clinical significance of hepatoma-derived growth factor (HDGF) in epithelial ovarian cancer (EOC). Background Recent studies have demonstrated that HDGF overexpression correlates to the progression and poor prognosis in several kinds of cancers. However, the clinical significance and prognostic value of HDGF in EOC have not been investigated. Methods Expression of HDGF was visualized by immunohistology and then the cohort was divided into higher- and lower-expression groups. The correlation between HDGF and clinicopathologic factors was analyzed by χ2 test. The prognostic value of HDGF was assessed by univariate analysis with Kaplan–Meier method, and by multivariate analysis with Cox-regression model. With experiments in vitro, HDGF expression in ovarian cancer cell lines was detected by immunoblotting. Results Higher HDGF expression rate was 52.76% in EOC. HDGF expression was significantly associated with lymphatic metastasis (P=0.006). Higher HDGF expression was closely correlated to poorer 5-year overall survival rate with univariate analysis (P=0.003), and was identified as an independent prognostic factor with multivariate analysis (P=0.007). With experiments in vitro, HDGF was proved to exist in all ovarian cancer cell lines with different expression levels. Conclusion HDGF expression correlates to unfavorable prognosis and can be considered as an independent prognostic factor, indicating that HDGF may be a promising potential molecular drug target.
Collapse
Affiliation(s)
- Xue-Jun Liu
- Department of Obstetrics, Linyi Hospital Affiliated to Shandong University, Linyi City, People's Republic of China
| | - Wen-Lian Liu
- Department of Obstetrics, Linyi Hospital Affiliated to Shandong University, Linyi City, People's Republic of China
| | - Fang-Mei Yang
- Department of Obstetrics, Linyi Hospital Affiliated to Shandong University, Linyi City, People's Republic of China
| | - Xiao-Qing Yang
- Department of Pathology, Qianfoshan Hospital Affiliated to Shandong University, Jinan City, People's Republic of China
| | - Xiao-Fei Lu
- Department of General Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan City, People's Republic of China
| |
Collapse
|
63
|
Enomoto H, Nakamura H, Liu W, Nishiguchi S. Hepatoma-Derived Growth Factor: Its Possible Involvement in the Progression of Hepatocellular Carcinoma. Int J Mol Sci 2015; 16:14086-97. [PMID: 26101867 PMCID: PMC4490540 DOI: 10.3390/ijms160614086] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/11/2015] [Accepted: 06/17/2015] [Indexed: 02/05/2023] Open
Abstract
The development of hepatocellular carcinoma (HCC) is an important complication of viral infection induced by hepatitis virus C, and our major research theme is to identify a new growth factor related to the progression of HCC. HDGF (hepatoma-derived growth factor) is a novel growth factor that belongs to a new gene family. HDGF was initially purified from the conditioned medium of a hepatoma cell line. HDGF promotes cellular proliferation as a DNA binding nuclear factor and a secreted protein acting via a receptor-mediated pathway. HDGF is a unique multi-functional protein that can function as a growth factor, angiogenic factor and anti-apoptotic factor and it participates in the development and progression of various malignant diseases. The expression level of HDGF may be an independent prognostic factor for predicting the disease-free and overall survival in patients with various malignancies, including HCC. Furthermore, the overexpression of HDGF promotes the proliferation of HCC cells, while a reduction in the HDGF expression inhibits the proliferation of HCC cells. This article provides an overview of the characteristics of HDGF and describes the potential role of HDGF as a growth-promoting factor for HCC.
Collapse
Affiliation(s)
- Hirayuki Enomoto
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo 663-8501, Japan.
| | - Hideji Nakamura
- Department of Gastroenterology and Hepatology, Nissay Hospital, Itachibori 6-3-8, Nishi-ku, Osaka 550-0012, Japan.
| | - Weidong Liu
- Department of Hepatology and Infectious Diseases, the Second Affiliated Hospital, Shantou University Medical College, No. 69, Dongxiabei, Jinping, Shantou 515041, China.
| | - Shuhei Nishiguchi
- Division of Hepatobiliary and Pancreatic Disease, Department of Internal Medicine, Hyogo College of Medicine, Mukogawa-cho 1-1, Nishinomiya, Hyogo 663-8501, Japan.
| |
Collapse
|
64
|
Bao CH, Wang XT, Ma W, Wang NN, Un Nesa E, Wang JB, Wang C, Jia YB, Wang K, Tian H, Cheng YF. Irradiated fibroblasts promote epithelial-mesenchymal transition and HDGF expression of esophageal squamous cell carcinoma. Biochem Biophys Res Commun 2015; 458:441-7. [PMID: 25677618 DOI: 10.1016/j.bbrc.2015.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/01/2015] [Indexed: 12/31/2022]
Abstract
Recent evidence suggested that nonirradiated cancer-associated fibroblasts (CAFs) promoted aggressive phenotypes of cancer cells through epithelial-mesenchymal transition (EMT). Hepatoma-derived growth factor (HDGF) is a radiosensitive gene of esophageal squamous cell carcinoma (ESCC). This study aimed to investigate the effect of irradiated fibroblasts on EMT and HDGF expression of ESCC. Our study demonstrated that coculture with nonirradiated fibroblasts significantly increased the invasive ability of ESCC cells and the increased invasiveness was further accelerated when they were cocultured with irradiated fibroblasts. Scattering of ESCC cells was also accelerated by the supernatant from irradiated fibroblasts. Exposure of ESCC cells to supernatant from irradiated fibroblasts resulted in decreased E-cadherin, increased vimentin in vitro and β-catenin was demonstrated to localize to the nucleus in tumor cells with irradiated fibroblasts in vivo models. The expression of HDGF and β-catenin were increased in both fibroblasts and ESCC cells of irradiated group in vitro and in vivo models. Interestingly, the tumor cells adjoining the stromal fibroblasts displayed strong nuclear HDGF immunoreactivity, which suggested the occurrence of a paracrine effect of fibroblasts on HDGF expression. These data suggested that irradiated fibroblasts promoted invasion, growth, EMT and HDGF expression of ESCC.
Collapse
Affiliation(s)
- Ci-Hang Bao
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xin-Tong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Wei Ma
- Department of Radiation Oncology, Cancer Hospital, Genaral Hospital of Ningxia Medical University, Yinchuan 750000, China
| | - Na-Na Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Effat Un Nesa
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Jian-Bo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yi-Bin Jia
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Kai Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yu-Feng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan 250012, China.
| |
Collapse
|