51
|
Fu Y, Wang Y. SRSF7 knockdown promotes apoptosis of colon and lung cancer cells. Oncol Lett 2018; 15:5545-5552. [PMID: 29556298 PMCID: PMC5844074 DOI: 10.3892/ol.2018.8072] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 01/10/2018] [Indexed: 02/04/2023] Open
Abstract
Serine/arginine-rich (SR) proteins are a family of important splicing factors, which are involved in multiple aspects of RNA processing, including splicing, mRNA nuclear export, mRNA stability and translation. Previous studies have identified a number of SR proteins that exhibit abnormal expression in various tumor types. In the present study, the expression and function of serine/arginine-rich splicing factor 7 (SRSF7) were investigated in colon and lung cancer. Using tissue immunohistochemistry, it was observed that SRSF7 was overexpressed in colon and lung cancer tissues. As the role of SRSF7 in cancer remains to be fully elucidated, the expression of SRSF7 was knocked down in the present study by transfecting SRSF7-specific small interfering RNAs (siRNAs) into the HCT116 colon cancer cell line and A549 lung cancer cell line, which exhibited elevated expression of SRSF7. MTS assays, western blot analysis, flow cytometry and spectrofluorometer analyses were performed to assess the effects of SRSF7 knockdown on the proliferation and apoptosis of cells. The results demonstrated that the expression of SRSF7 was efficiently knocked down by SRSF7 siRNA, and that SRSF7 knockdown inhibited proliferation and enhanced apoptosis of HCT116 and A549 cells. Further experiments involving BEAS-2B cells stably overexpressing SRSF7, and A549 cells with stable knockdown of SRSF7 revealed that SRSF7 regulated the splicing of the apoptosis regulator Fas. Collectively, these data indicated that SRSF7 is critical for the survival of colon and lung cancer cells, and may be a potential therapeutic target for the treatment of colon and lung cancer.
Collapse
Affiliation(s)
- Yu Fu
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang, Hebei 050018, P.R. China
| | - Yingze Wang
- College of Bioscience and Bioengineering, Hebei University of Science and Technology, Shijiazhuang, Hebei 050018, P.R. China
| |
Collapse
|
52
|
Zeng D, Lin J, He H, Tan G, Lan Y, Jiang F, Sheng S. Therapeutic effect of targeted Fas-expressing adenoviruses method combining γδ T cells in a mouse model of human ovarian carcinoma. Oncol Lett 2018; 15:2555-2561. [PMID: 29434973 DOI: 10.3892/ol.2017.7599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/07/2017] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to investigate the therapeutic effect and safety of targeted use of Fas-expressing adenoviruses combined with γδ T cell-mediated killing to treat human ovarian cancer xenografts in BALB/c mice. Shuttle plasmids containing control elements of human telomerase reverse transcriptase promoter and two-step transcriptional amplification system were constructed and packaged into adenovirus-5 vectors to generate expression of an exogenous Fas gene. A mouse xenograft model of human ovarian carcinoma was constructed. A total of 35 BALB/c mice were randomly divided into five groups, which were injected with PBS, γδ T cells, Fas-expressing adenoviruses, taxol, or Fas-expressing adenovirus and γδ T cells. The weight and volume of tumors in mice in each group was monitored. Tissue sections of the various tissues of mice in the Fas-expressing adenovirus and γδ T cells group was compared with those in the PBS group to evaluate the safety of Fas-expressing adenovirus and γδ T cells in the treatment of human ovarian cancer xenograft tumors. The results of the present study indicated that mice in all treatment groups were alive at the end of the treatment course. Tumor weight and volume was the highest in the PBS group, followed successively by the adenovirus group, the γδ T cell group, the adenovirus and γδ T cell group, and the taxol group. The weight and volume inhibition rate in adenovirus and γδ T cell group were significantly higher compared with in the PBS group (P<0.05). Pathological observation of tissue samples revealed that none of vital organs in the adenovirus and γδ T cell group developed any evident morphological changes during treatment, when compared with healthy controls. In conclusion, the combined therapy with Fas-expressing adenoviruses and γδ T cells is efficient and safe for the treatment of mouse human ovarian carcinoma xenografts.
Collapse
Affiliation(s)
- Dingyuan Zeng
- Department of Oncology, Maternity and Children's Hospital Affiliated to the Guangxi University of Science and Technology, Liuzhou, Guangxi 545002, P.R. China
| | - Jiajing Lin
- Department of Gynecology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Nanning 545000, P.R. China
| | - Hongying He
- Department of Gynecology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Nanning 545000, P.R. China
| | - Guangping Tan
- Department of Gynecology, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Nanning 545000, P.R. China
| | - Ying Lan
- Department of Gynecology, The First Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi 545002, P.R. China
| | - Fuyan Jiang
- Liuzhou Tumor Hospital, Liuzhou, Guangxi 545005, P.R. China
| | - Shuting Sheng
- Liuzhou Hospital of Traditional Chinese Medicine, Liuzhou, Guangxi 545001, P.R. China
| |
Collapse
|
53
|
Deregulated FADD expression and phosphorylation in T-cell lymphoblastic lymphoma. Oncotarget 2018; 7:61485-61499. [PMID: 27556297 PMCID: PMC5308666 DOI: 10.18632/oncotarget.11370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/11/2016] [Indexed: 12/14/2022] Open
Abstract
In the present work, we show that T-cell lymphoblastic lymphoma cells exhibit a reduction of FADD availability in the cytoplasm, which may contribute to impaired apoptosis. In addition, we observe a reduction of FADD phosphorylation that inversely correlates with the proliferation capacity and tumor aggressiveness. The resultant balance between FADD-dependent apoptotic and non-apoptotic abilities may define the outcome of the tumor. Thus, we propose that FADD expression and phosphorylation can be reliable biomarkers with prognostic value for T-LBL stratification.
Collapse
|
54
|
Wang M, Su P. The role of the Fas/FasL signaling pathway in environmental toxicant-induced testicular cell apoptosis: An update. Syst Biol Reprod Med 2018; 64:93-102. [DOI: 10.1080/19396368.2017.1422046] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mei Wang
- Family Planning Research Institute of Tongji Medical College, Huazhong University of Science and Technology, Hubei, P.R. China
| | - Ping Su
- Family Planning Research Institute of Tongji Medical College, Huazhong University of Science and Technology, Hubei, P.R. China
| |
Collapse
|
55
|
Muraki M. Development of expression systems for the production of recombinant human Fas ligand extracellular domain derivatives using <em>Pichia pastoris</em> and preparation of the conjugates by site-specific chemical modifications: A review. AIMS BIOENGINEERING 2018. [DOI: 10.3934/bioeng.2018.1.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
56
|
Ye C, Yu X, Zeng J, Dai M, Zhang B. Effects of baicalein on proliferation, apoptosis, migration and invasion of Ewing's sarcoma cells. Int J Oncol 2017; 51:1785-1792. [PMID: 29039470 DOI: 10.3892/ijo.2017.4148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 09/25/2017] [Indexed: 11/06/2022] Open
Abstract
Ewing's sarcoma (ES) is a rare tumor that is more frequent in pediatric and adolescent age groups. In the past few decades, long-term survival in affected patients has improved due to the success of multimodal therapy. However, long-term survival is inevitably restricted by the late side-effects of chemotherapy. Besides, early metastasis also contributes to the poor prognosis of ES. Recently, traditional Chinese medicines (TCMs) have increasingly attracted interest due to the promising clinical results and fewer side-effects for the treatment of cancers. Among the various TCMs, the root of Scutellaria baicalensis exerts anti-inflammatory properties as a well-known herb in traditional Chinese medicine. Baicalein (5,6,7-trihydroxyflavone) derived from the root of Scutellaria baicalensis is a bioactive compound, which possesses a powerful pro-apoptotic activity in various cancers such as hepatocellular carcinoma and myeloma. However, the effects of baicalein on ES cells remain still unknown. We anticipated that baicalein also has apoptotic activity in ES. The aim of the present study was to investigate the effects of baicalein on viability, apoptosis, migration and invasion of ES cells, and further to elaborate the molecular mechanism of baicalein-induced ES cell apoptosis. We found that baicalein markedly inhibited ES cells viability in a time- and dose-dependent manner, especially SK-ES-1 cells and could promote the apoptosis of ES cells. Additionally, baicalein was capable of upregulating the expression of the pro-apoptotic proteins Bax and cytochrome c, reducing the expression of the anti-apoptotic protein Bcl-2, elevating the ratio of Bax/Bcl-2, and triggering the mitochondrial apoptotic pathway, which led to caspase-3 and caspase-9 activation and PARP cleavage. Meanwhile, the activation of caspase-8 and the death receptor pathway was also observed. Besides, baicalein could reduce ES migration and invasion in vitro, which showed its potential to inhibit ES metastasis, besides contributing to the decrease in the expression of matrix metalloproteinases (MMP)-2 and MMP-9. In conclusion, baicalein has a potent tumor-suppressor activity by inducing cell apoptosis through the mitochondrial apoptotic pathway and the death receptor pathway in ES cells, thus it may serve as a novel and effective candidate agent for ES treatment.
Collapse
Affiliation(s)
- Conglin Ye
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaolong Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Jin Zeng
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
57
|
Tian H, Yao ST, Yang NN, Ren J, Jiao P, Zhang X, Li DX, Zhang GA, Xia ZF, Qin SC. D4F alleviates macrophage-derived foam cell apoptosis by inhibiting the NF-κB-dependent Fas/FasL pathway. Sci Rep 2017; 7:7333. [PMID: 28779128 PMCID: PMC5544683 DOI: 10.1038/s41598-017-07656-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/28/2017] [Indexed: 01/08/2023] Open
Abstract
This study was designed to explore the protective effect of D4F, an apolipoprotein A-I mimetic peptide, on nuclear factor-κB (NF-κB)-dependent Fas/Fas ligand (FasL) pathway-mediated apoptosis in macrophages induced by oxidized low-density lipoprotein (ox-LDL). Our results showed that ox-LDL induced apoptosis, NF-κB P65 nuclear translocation and the upregulation of Fas/FasL pathway-related proteins, including Fas, FasL, Fas-associated death domain proteins (FADD), caspase-8 and caspase-3 in RAW264.7 macrophages, whereas silencing of Fas blocked ox-LDL-induced macrophage apoptosis. Furthermore, silencing of P65 attenuated macrophage apoptosis and the upregulation of Fas caused by ox-LDL, whereas P65 expression was not significantly affected by treatment with Fas siRNA. D4F attenuated the reduction of cell viability and the increase in lactate dehydrogenase leakage and apoptosis. Additionally, D4F inhibited ox-LDL-induced P65 nuclear translocation and upregulation of Fas/FasL pathway-related proteins in RAW264.7 cells and in atherosclerotic lesions of apoE-/- mice. However, Jo2, a Fas-activating monoclonal antibody, reversed the inhibitory effect of D4F on ox-LDL-induced cell apoptosis and upregulation of Fas, FasL and FADD. These data indicate that NF-κB mediates Fas/FasL pathway activation and apoptosis in macrophages induced by ox-LDL and that D4F protects macrophages from ox-LDL-induced apoptosis by suppressing the activation of NF-κB and the Fas/FasL pathway.
Collapse
Affiliation(s)
- Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China
| | - Shu-Tong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China. .,College of Basic Medical Sciences, Taishan Medical University, Taian, 271000, China.
| | - Na-Na Yang
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China
| | - Jie Ren
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, 250022, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China
| | - Xiangjian Zhang
- Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease and Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, 050000, China
| | - Dong-Xuan Li
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China
| | - Gong-An Zhang
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China
| | - Zhen-Fang Xia
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China
| | - Shu-Cun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, 271000, China.
| |
Collapse
|
58
|
Mion F, Tonon S, Valeri V, Pucillo CE. Message in a bottle from the tumor microenvironment: tumor-educated DCs instruct B cells to participate in immunosuppression. Cell Mol Immunol 2017; 14:730-732. [PMID: 28757609 DOI: 10.1038/cmi.2017.63] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 06/10/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- Francesca Mion
- Department of Medicine, University of Udine, Udine, Italy
| | - Silvia Tonon
- Department of Medicine, University of Udine, Udine, Italy
| | - Viviana Valeri
- Department of Medicine, University of Udine, Udine, Italy
| | | |
Collapse
|
59
|
Radisky ES, Raeeszadeh-Sarmazdeh M, Radisky DC. Therapeutic Potential of Matrix Metalloproteinase Inhibition in Breast Cancer. J Cell Biochem 2017; 118:3531-3548. [PMID: 28585723 PMCID: PMC5621753 DOI: 10.1002/jcb.26185] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc endopeptidases that cleave nearly all components of the extracellular matrix as well as many other soluble and cell-associated proteins. MMPs have been implicated in normal physiological processes, including development, and in the acquisition and progression of the malignant phenotype. Disappointing results from a series of clinical trials testing small molecule, broad spectrum MMP inhibitors as cancer therapeutics led to a re-evaluation of how MMPs function in the tumor microenvironment, and ongoing research continues to reveal that these proteins play complex roles in cancer development and progression. It is now clear that effective targeting of MMPs for therapeutic benefit will require selective inhibition of specific MMPs. Here, we provide an overview of the MMP family and its biological regulators, the tissue inhibitors of metalloproteinases (TIMPs). We then summarize recent research from model systems that elucidate how specific MMPs drive the malignant phenotype of breast cancer cells, including acquisition of cancer stem cell features and induction of the epithelial-mesenchymal transition, and we also outline clinical studies that implicate specific MMPs in breast cancer outcomes. We conclude by discussing ongoing strategies for development of inhibitors with therapeutic potential that are capable of selectively targeting the MMPs most responsible for tumor promotion, with special consideration of the potential of biologics including antibodies and engineered proteins based on the TIMP scaffold. J. Cell. Biochem. 118: 3531-3548, 2017. © 2017 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville 32224, Florida
| | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville 32224, Florida
| |
Collapse
|
60
|
Muraki M, Hirota K. Site-specific chemical conjugation of human Fas ligand extracellular domain using trans-cyclooctene - methyltetrazine reactions. BMC Biotechnol 2017; 17:56. [PMID: 28673349 PMCID: PMC5496246 DOI: 10.1186/s12896-017-0381-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/27/2017] [Indexed: 11/13/2022] Open
Abstract
Background Fas ligand plays a key role in the human immune system as a major cell death inducing protein. The extracellular domain of human Fas ligand (hFasLECD) triggers apoptosis of malignant cells, and therefore is expected to have substantial potentials in medical biotechnology. However, the current application of this protein to clinical medicine is hampered by a shortage of the benefits relative to the drawbacks including the side-effects in systemic administration. Effective procedures for the engineering of the protein by attaching useful additional functions are required to overcome the problem. Results A procedure for the site-specific chemical conjugation of hFasLECD with a fluorochrome and functional proteins was devised using an inverse-electron-demand Diels-Alder reaction between trans-cyclooctene group and methyltetrazine group. The conjugations in the present study were attained by using much less molar excess amounts of the compounds to be attached as compared with the conventional chemical modification reactions using maleimide derivatives in the previous study. The isolated conjugates of hFasLECD with sulfo-Cy3, avidin and rabbit IgG Fab’ domain presented the functional and the structural integrities of the attached molecules without impairing the specific binding activity toward human Fas receptor extracellular domain. Conclusions The present study provided a new fundamental strategy for the production of the engineered hFasLECDs with additional beneficial functions, which will lead to the developments of the improved diagnostic systems and the effective treatment methods of serious diseases by using this protein as a component of novel molecular tools. Electronic supplementary material The online version of this article (doi:10.1186/s12896-017-0381-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michiro Muraki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan.
| | - Kiyonori Hirota
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki, 305-8566, Japan
| |
Collapse
|
61
|
El-Mesery M, El-Mowafy M, Elgaml A, Youssef LF, Abed SY. Correlation of Serum Soluble Fibrinogen-Like Protein 2 with Soluble FAS Ligand and Interferon Gamma in Egyptian Hepatitis C Virus-Infected Patients and Hepatocellular Carcinoma Patients. J Interferon Cytokine Res 2017; 37:342-347. [PMID: 28609212 DOI: 10.1089/jir.2016.0128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Infection with hepatitis C virus (HCV) remains one of the serious human diseases worldwide, especially in Egypt, which can lead to cirrhosis or hepatocellular carcinoma (HCC). However, the exact molecular mechanism of HCC progress in HCV-infected patients remains unclear. Soluble fibrinogen-like protein 2 (sFGL2) is a modulator of the immune response that is secreted by T cells and inhibits maturation of dendritic cells and T cell proliferation. In the current study, serum sFGL2 levels were analyzed by enzyme-linked immunosorbent assay (ELISA) technique in 30 chronic HCV-infected patients (HCV group), 30 chronic HCV-infected patients with HCC (HCC group), and 12 healthy individuals (control group). Moreover, serum levels of soluble FAS ligand (sFASL) and interferon gamma (IFN-γ) were analyzed and correlated with sFGL2 levels. According to our results, serum sFGL2 levels were significantly elevated in all patients with chronic HCV infection. However, HCC patients showed lower sFGL2 levels than HCV-infected patients without HCC incidence. In addition, serum sFASL levels were significantly elevated in both HCV and HCC groups, whereas serum IFN-γ levels were only elevated in the HCC group. Interestingly, sFGL2 correlated positively with serum total bilirubin level and negatively with serum levels of sFASL, IFN-γ, and albumin in HCV and HCC groups. Thus, conclusively, sFGL2 level increases in Egyptian HCV-infected and HCC patients. Taken together, the current work may open future possibility of designing new treatment strategies for HCV infection targeting sFGL2 and its immunosuppressive effect.
Collapse
Affiliation(s)
- Mohamed El-Mesery
- 1 Department of Biochemistry, Faculty of Pharmacy, Mansoura University , Mansoura, Egypt
| | - Mohammed El-Mowafy
- 2 Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University , Mansoura, Egypt
| | - Abdelaziz Elgaml
- 2 Department of Microbiology and Immunology, Faculty of Pharmacy, Mansoura University , Mansoura, Egypt
| | - Laila F Youssef
- 1 Department of Biochemistry, Faculty of Pharmacy, Mansoura University , Mansoura, Egypt
| | - Sally Y Abed
- 3 Department of Tropical Medicine, Faculty of Medicine, Mansoura University , Mansoura, Egypt
| |
Collapse
|
62
|
Khalifa RH, Shahin RMH, Aboukhalil REE, Marie MA, El-Deeb AM, Riad NM. Digging more in the genetic risk prediction of hepatitis C virus epidemic in Egypt: Apoptosis genes polymorphisms in the susceptibility of hepatitis C virus and association with viral load. Virus Res 2017; 238:35-39. [PMID: 28587866 DOI: 10.1016/j.virusres.2017.05.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 02/07/2023]
Abstract
Egypt is confronted with the highest hepatitis C virus (HCV) epidemic. Apoptosis and cellular immune responses are crucial to the clearance or persistence of viral infections. This case-control study was carried out to detect whether apoptosis genes single nucleotide polymorphisms (SNPs) confer risk to HCV in a cohort of Egyptian patients and to explore their association with viral load. One hundred and ninety six blood samples were withdrawn from 96 HCV patients and 100 controls. The Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) -1525G>A and FasL-844T>C SNPs were genotyped using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Hepatitis C viral load was measured using Real time PCR. Results Genotypes distributions of TRAIL -1525G>A and FasL-844 T>C polymorphisms in controls were in accordance with Hardy-Weinberg equilibrium (p>0.05). The study showed a statistically significant difference in the distribution of the TRAIL -1525G>A polymorphism genotypes and the FasL-844 T>C polymorphism genotypes between the HCV patients and the controls (p=0.001 and 0.02 respectively), with association of the -1525GA genotype and -844 TT genotype with increased risk of HCV infection (OR=2.68, 1.942 respectively, 95% CI=1.482-4.846, 1.1-3.43, respectively). No significant association was detected between TRAIL, FasL and the viral load. Our results suggest that the FasL -844T>C SNP is implicated in the susceptibility to HCV in Egyptian patients and firstly report the involvement of TRAIL gene polymorphism in the risk of the disease. Therefore we recommend national programs to delineate genetic factors that may put individuals at risk for contracting HCV.
Collapse
Affiliation(s)
- Rania H Khalifa
- Department of Clinical & Chemical Pathology, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt.
| | | | - Reham Emad Eldin Aboukhalil
- Department of Clinical & Chemical Pathology, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt; Department of BMS at UCM, Qassim University, Saudi Arabia.
| | - Mohamed A Marie
- Department of Internal medicine, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt.
| | - Amr M El-Deeb
- Department of Tropical Medicine, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt.
| | - Nermine Magdi Riad
- Department of Clinical & Chemical Pathology, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt.
| |
Collapse
|
63
|
Wu J, Chen K, Zhang F, Jin J, Zhang N, Li D, Ying L, Chen W, Yu H, Mao W, Su D. Overcoming Linsitinib intrinsic resistance through inhibition of nuclear factor-κB signaling in esophageal squamous cell carcinoma. Cancer Med 2017; 6:1353-1361. [PMID: 28440057 PMCID: PMC5463077 DOI: 10.1002/cam4.1068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 02/20/2017] [Accepted: 03/04/2017] [Indexed: 12/30/2022] Open
Abstract
The aim of this study is to evaluate the efficacy of insulin-like growth factor 1 receptor (IGF-1R) inhibitor Linsitinib, in esophageal squamous cell carcinoma (ESCC), and to characterize special biomarker to screen Linsitinib-sensitive patients as well as explore the molecular-resistant mechanism to Linsitinib in ESCC. Our study evaluated the sensitivity of insulin-like growth factor 1 receptor (IGF-1R) inhibitor, Linsitinib in ESCC cells with MTT assay. After Linsitinib treatment, the expressions of downstream signaling molecules and apoptosis pathways were measured by western blot. And the antitumor effect of Linsitinib and JSH-23, an inhibitor of nuclear factor-κB transcriptional activity, was analyzed both as single agent and in combination in ESCC. Apoptosis, cell viability, and clonogenic survival analysis were also investigated. The sensitivity of Linsitinib was relatively variable in patient-derived primary ESCC cells as well as in human commercial cell lines. And the downstream AKT/mTOR and ERK signaling pathways were inhibited by Linsitinib, while phosphorylation level of NF-κB p65 was obviously activated to reduce apoptosis effect in Linsitinib-resistant cell lines. Most importantly, blockage of NF-κB activity by JSH-23 could sensitize resistant cells to Linsitinib treatment. Results from this study demonstrated that the intrinsic resistance to Linsitinib was predominantly mediated by NF-κB activation in ESCC. Moreover, combination of Linsitinib and JSH-23 as therapy provides a novel strategy to overcome resistance to Linsitinib in ESCC.
Collapse
Affiliation(s)
- Junzhou Wu
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Kaiyan Chen
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China.,Department of Oncology, The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fanrong Zhang
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Jiaoyue Jin
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Nan Zhang
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Dan Li
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China.,Department of Oncology, The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lisha Ying
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Wei Chen
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Herbert Yu
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Weimin Mao
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| | - Dan Su
- Cancer Research Institute, Zhejiang Cancer Hospital & Key Laboratory Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang Province, Hangzhou, China
| |
Collapse
|
64
|
Rani I, Sharma B, Kumar S, Kaur S, Agnihotri N. Apoptosis mediated chemosensitization of tumor cells to 5-fluorouracil on supplementation of fish oil in experimental colon carcinoma. Tumour Biol 2017; 39:1010428317695019. [PMID: 28349837 DOI: 10.1177/1010428317695019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
5-Fluorouracil has been considered as a cornerstone therapy for colorectal cancer; however, it suffers from low therapeutic response rate and severe side effects. Therefore, there is an urgent need to increase the clinical efficacy of 5-fluorouracil. Recently, fish oil rich in n-3 polyunsaturated fatty acids has been reported to chemosensitize tumor cells to anti-cancer drugs. This study is designed to understand the underlying mechanisms of synergistic effect of fish oil and 5-fluorouracil by evaluation of tumor cell-associated markers such as apoptosis and DNA damage. The colon cancer was developed by administration of N,N-dimethylhydrazine dihydrochloride and dextran sulfate sodium salt. Further these animals were treated with 5-fluorouracil, fish oil, or a combination of both. In carcinogen-treated animals, a decrease in DNA damage and apoptotic index was observed. There was also a decrease in the expression of Fas, FasL, caspase 8, and Bax, and an increase in Bcl-2. In contrast, administration of 5-fluorouracil and fish oil as an adjuvant increased both DNA damage and apoptotic index by activation of both extrinsic and intrinsic apoptotic pathways as compared to the other groups. The increased pro-apoptotic effect by synergism of 5-fluorouracil and fish oil may be attributed to the incorporation of n-3 polyunsaturated fatty acids in membrane, which alters membrane fluidity in cancer cells. In conclusion, this study highlights that the induction of apoptotic pathway by fish oil may increase the susceptibility of tumors to chemotherapeutic regimens.
Collapse
Affiliation(s)
- Isha Rani
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Bhoomika Sharma
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Sandeep Kumar
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Satinder Kaur
- Department of Biochemistry, Panjab University, Chandigarh, India
| | | |
Collapse
|
65
|
Kang Y, Wang ZJ, Xie D, Sun X, Yang W, Zhao X, Xu N. Characterization and Potential Antitumor Activity of Polysaccharide from Gracilariopsis lemaneiformis. Mar Drugs 2017; 15:md15040100. [PMID: 28353631 PMCID: PMC5408246 DOI: 10.3390/md15040100] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 11/06/2022] Open
Abstract
Substances with valuable antitumor properties have been identified in many marine algae, including an edible polysaccharide from the marine alga Gracilariopsis lemaneiformis (PGL). We previously reported transcriptome profiling data showing that PGL induced transcriptional alterations generate anti-lung cancer activity. To identify how PGL is detrimental to tumors, we purified PGL to characterize its chemical composition, molecular weight, and sugar and protein content and investigated its antitumor activity. We demonstrated that PGL exerted its antitumor activities by modulating cell viability, morphology, apoptosis, and the apoptosis-related Fas/FasL signaling pathway in the human lung cancer cell line A549, the gastric cancer cell line MKN28, and the mouse melanoma cell line B16. Our data provide the first evidence that PGL inhibits cell proliferation by inducing apoptosis, which is largely mediated by Fas/FasL in cancer cells, suggesting that PGL might be a novel therapeutic agent against cancer.
Collapse
Affiliation(s)
- Yani Kang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo 315211, Zhejiang, China.
- School of Biomedical Engineering, Bio-ID Research Center, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhi-Jiang Wang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo 315100, China.
| | - Dongsheng Xie
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xue Sun
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo 315211, Zhejiang, China.
| | - Wenge Yang
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo 315211, Zhejiang, China.
| | - Xiaodong Zhao
- School of Biomedical Engineering, Bio-ID Research Center, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Nianjun Xu
- Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
66
|
Genetic disruption of oncogenic Kras sensitizes lung cancer cells to Fas receptor-mediated apoptosis. Proc Natl Acad Sci U S A 2017; 114:3648-3653. [PMID: 28320962 DOI: 10.1073/pnas.1620861114] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Genetic lesions that activate KRAS account for ∼30% of the 1.6 million annual cases of lung cancer. Despite clinical need, KRAS is still undruggable using traditional small-molecule drugs/inhibitors. When oncogenic Kras is suppressed by RNA interference, tumors initially regress but eventually recur and proliferate despite suppression of Kras Here, we show that tumor cells can survive knockout of oncogenic Kras, indicating the existence of Kras-independent survival pathways. Thus, even if clinical KRAS inhibitors were available, resistance would remain an obstacle to treatment. Kras-independent cancer cells exhibit decreased colony formation in vitro but retain the ability to form tumors in mice. Comparing the transcriptomes of oncogenic Kras cells and Kras knockout cells, we identified 603 genes that were specifically up-regulated in Kras knockout cells, including the Fas gene, which encodes a cell surface death receptor involved in physiological regulation of apoptosis. Antibodies recognizing Fas receptor efficiently induced apoptosis of Kras knockout cells but not oncogenic Kras-expressing cells. Increased Fas expression in Kras knockout cells was attributed to decreased association of repressive epigenetic marks at the Fas promoter. Concordant with this observation, treating oncogenic Kras cells with histone deacetylase inhibitor and Fas-activating antibody efficiently induced apoptosis, thus bypassing the need to inhibit Kras. Our results suggest that activation of Fas could be exploited as an Achilles' heel in tumors initiated by oncogenic Kras.
Collapse
|
67
|
Chien MH, Chang WM, Lee WJ, Chang YC, Lai TC, Chan DV, Sharma R, Lin YF, Hsiao M. A Fas Ligand (FasL)-Fused Humanized Antibody Against Tumor-Associated Glycoprotein 72 Selectively Exhibits the Cytotoxic Effect Against Oral Cancer Cells with a Low FasL/Fas Ratio. Mol Cancer Ther 2017; 16:1102-1113. [PMID: 28292939 DOI: 10.1158/1535-7163.mct-16-0314] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/27/2016] [Accepted: 03/07/2017] [Indexed: 12/16/2022]
Abstract
Altered expression of the Fas ligand (FasL)/Fas ratio exhibits a direct impact on the prognosis of cancer patients, and its impairment in cancer cells may lead to apoptosis resistance. Thus, the development of effective therapies targeting the FasL/Fas system may play an important role in the fight against cancer. In this study, we evaluated whether a fusion protein (hcc49scFv-FasL) comprising of the cytotoxicity domain of the FasL fused to a humanized antibody (CC49) against tumor-associated glycoprotein 72, which is expressed on oral squamous cell carcinoma (OSCC), can selectively kill OSCC cells with different FasL/Fas ratios. In clinical samples, the significantly low FasL and high Fas transcripts were observed in tumors compared with normal tissues. A lower FasL/Fas ratio was correlated with a worse prognosis of OSCC patients and higher proliferative and invasive abilities of OSCC cells. The hcc49scFv-FasL showed a selective cytotoxic effect on OSCC cells (Cal-27 and SAS) but not on normal oral keratinocytes cells (HOK) through apoptosis induction. Moreover, SAS cells harboring a lower FasL/Fas ratio than Cal-27 were more sensitive to the cytotoxic effect of hcc49scFv-FasL. Unlike wild-type FasL, hcc49scFv-FasL was not cleaved by matrix metalloproteinases and did not induce nonapoptotic signaling in SAS cells. In vivo, we found that hcc49scFv-FasL drastically reduced the formation of lymph node metastasis and decreased primary tumor growth in SAS orthotopic and subcutaneous xenograft tumor models. Collectively, our data indicate that a tumor-targeting antibody fused to the FasL can be a powerful tool for OSCC treatment, especially in populations with a low FasL/Fas ratio. Mol Cancer Ther; 16(6); 1102-13. ©2017 AACR.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody-Dependent Cell Cytotoxicity/immunology
- Antigens, Neoplasm/immunology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/immunology
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Survival
- Cell Transformation, Neoplastic/metabolism
- Disease Models, Animal
- Fas Ligand Protein/genetics
- Fas Ligand Protein/metabolism
- Glycoproteins/antagonists & inhibitors
- Glycoproteins/immunology
- Humans
- Mice
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/immunology
- Mouth Neoplasms/metabolism
- Mouth Neoplasms/pathology
- Prognosis
- Protein Binding
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/pharmacology
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
- fas Receptor/metabolism
Collapse
Affiliation(s)
- Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wei-Min Chang
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Jiunn Lee
- Department of Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chan Chang
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Tsung-Ching Lai
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Derek V Chan
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Rahul Sharma
- Center for Immunity, Inflammation, and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Michael Hsiao
- The Genomics Research Center, Academia Sinica, Taipei, Taiwan.
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- The PhD Program for Translational Medicine, College of Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
68
|
Ozdemirkiran FG, Nalbantoglu S, Gokgoz Z, Payzin BK, Vural F, Cagirgan S, Berdeli A. FAS/FASL gene polymorphisms in Turkish patients with chronic myeloproliferative disorders. Arch Med Sci 2017; 13:426-432. [PMID: 28261298 PMCID: PMC5332439 DOI: 10.5114/aoms.2015.53963] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/18/2015] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Chronic myeloproliferative disorders (CMPD) are chronic myeloid hematological disorders, characterized by increased myeloid cell proliferation and fibrosis. Impaired apoptotic mechanisms, increased cell proliferation, uncontrolled hematopoietic cell proliferation and myeloaccumulation may contribute to the pathogenesis of CMPD. The aim of our study was to show the possible role of FAS/FASL gene polymorphisms in CMPD pathogenesis and investigate the association with clinical parameters and susceptibility to disease. MATERIAL AND METHODS We included 101 (34 polycythemia vera (PV), 23 primary myelofibrosis (PMF), 44 essential thrombocythemia (ET)) CMPD patients diagnosed according to the WHO classification criteria and 95 healthy controls in this study. All the patients and the controls were investigated for FAS/FASL gene expression, allele frequencies and phenotype features, and also FAS mRNA levels were analyzed. RESULTS Chronic myeloproliferative disorders patients showed increased FAS-670AG + GG genotype distribution compared with the control group (p < 0.05). While the A allele was more frequent in both groups, AG genotype was more frequent in CMPD patients. There was no association between FAS-670A>G gene polymorphism and some clinical parameters such as splenomegaly and thrombosis (p > 0.05). No statistically significant difference in FASL+843C>T genotype or allele frequency was found between groups (p > 0.05). Moreover, no statistically significant difference was detected in FASL and JAK2V617F mutations (p > 0.05). FAS mRNA expression was 1.5-fold reduced in patients compared to healthy subjects. CONCLUSIONS According to our findings, FAS/FASL gene expression may contribute to the molecular and immunological pathogenesis of CMPD. More investigations are needed to support these data.
Collapse
Affiliation(s)
- Fusun Gediz Ozdemirkiran
- Department of Hematology, Atatürk Research and Education Hospital, Izmir Katip Çelebi University, Izmir, Turkey
| | - Sinem Nalbantoglu
- Department of Molecular Genetics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Zafer Gokgoz
- Department of Hematology, Baskent University, Adana, Turkey
| | - Bahriye Kadriye Payzin
- Department of Hematology, Atatürk Research and Education Hospital, Izmir Katip Çelebi University, Izmir, Turkey
| | - Filiz Vural
- Department of Hematology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Seckin Cagirgan
- Department of Hematology, Izmir Medical Park Hospital, Izmir, Turkey
| | - Afig Berdeli
- Department of Molecular Genetics, Faculty of Medicine, Ege University, Izmir, Turkey
| |
Collapse
|
69
|
Ke B, Fan C, Yang L, Fang X. Matrix Metalloproteinases-7 and Kidney Fibrosis. Front Physiol 2017; 8:21. [PMID: 28239354 PMCID: PMC5301013 DOI: 10.3389/fphys.2017.00021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/10/2017] [Indexed: 12/18/2022] Open
Abstract
Matrix metalloproteinase-7 (MMP-7) is a secreted zinc- and calcium-dependent endopeptidase that degrades a broad range of extracellular matrix substrates and additional substrates. MMP-7 playsa crucial role in a diverse array of cellular processes and appears to be a key regulator of fibrosis in several diseases, including pulmonary fibrosis, liver fibrosis, and cystic fibrosis. In particular, the relationship between MMP-7 and kidney fibrosis has attracted significant attention in recent years. Growing evidence indicates that MMP-7 plays an important role in the pathogenesis of kidney fibrosis. Here, we summarize the recent progress in the understanding of the role of MMP-7 in kidney fibrosis. In particular, we discuss how MMP-7 contributes to kidney fibrotic lesions via the following three pathways: epithelial-mesenchymal transition (EMT), transforming growth factor-beta (TGF-β) signaling, and extracellular matrix (ECM) deposition. Further dissection of the crosstalk among and regulation of these pathways will help clinicians and researchers develop effective therapeutic approaches for treating chronic kidney disease.
Collapse
Affiliation(s)
- Ben Ke
- The Third Hospital of Nanchang Nanchang, China
| | - Chuqiao Fan
- Nanchang University School of Medicine Nanchang, China
| | - Liping Yang
- Department of Nephrology, The Second Affiliated Hospital to Nanchang University Nanchang, China
| | - Xiangdong Fang
- Department of Breast Surgery, Jiangxi Cancer Hospital Nanchang, China
| |
Collapse
|
70
|
Kodigepalli KM, Li M, Liu SL, Wu L. Exogenous expression of SAMHD1 inhibits proliferation and induces apoptosis in cutaneous T-cell lymphoma-derived HuT78 cells. Cell Cycle 2016; 16:179-188. [PMID: 27929746 DOI: 10.1080/15384101.2016.1261226] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Sterile α motif and HD domain-containing protein 1 (SAMHD1) is a mammalian dNTP hydrolase (dNTPase) that regulates intracellular dNTP balance. We have previously reported that SAMHD1 mRNA and protein levels are significantly downregulated in CD4+ T-cells of patients with cutaneous T-cell lymphoma (CTCL), a disease characterized by infiltration of neoplastic CD4+ T-lymphocytes into the skin. However, functional significance of SAMHD1 in CTCL development and progression remains unknown. Here we investigate the mechanism by which SAMHD1 induces apoptosis in CTCL-derived CD4+ T-cells. We stably expressed exogenous SAMHD1 in the CTCL-derived HuT78 T-cell line containing a very low level of endogenous SAMHD1 protein. We found that low-level exogenous expression of SAMHD1 led to a significant reduction in HuT78 cell growth, proliferation, and colony formation. Exogenous SAMHD1 expression in HuT78 cells also resulted in increased spontaneous and Fas ligand (Fas-L)-induced apoptosis levels via activation of the extrinsic pathway, including caspase-8, -3 and -7. Additionally, increased SAMHD1 significantly reduced the protein and mRNA expression of the short isoform of cFLIP (cFLIPS), an important negative regulator of Fas-L-mediated apoptotic signaling. Our results indicate that exogenous SAMHD1 expression inhibits HuT78 cell growth and proliferation in part by increasing apoptosis. These findings implicate that SAMHD1 acts as an inhibitor in CTCL cell growth, suggesting that downregulation of SAMHD1 expression in neoplastic T-cells can facilitate uncontrolled cell proliferation.
Collapse
Affiliation(s)
- Karthik M Kodigepalli
- a Center of Retrovirus Research, Department of Veterinary Biosciences ; The Ohio State University , Columbus , OH , USA
| | - Minghua Li
- a Center of Retrovirus Research, Department of Veterinary Biosciences ; The Ohio State University , Columbus , OH , USA
| | - Shan-Lu Liu
- a Center of Retrovirus Research, Department of Veterinary Biosciences ; The Ohio State University , Columbus , OH , USA
| | - Li Wu
- a Center of Retrovirus Research, Department of Veterinary Biosciences ; The Ohio State University , Columbus , OH , USA.,b Comprehensive Cancer Center, The Ohio State University , Columbus , OH , USA.,c Department of Microbial Infection and Immunity , The Ohio State University , Columbus , OH , USA
| |
Collapse
|
71
|
Abdullahi A, Jeschke MG. White Adipose Tissue Browning: A Double-edged Sword. Trends Endocrinol Metab 2016; 27:542-552. [PMID: 27397607 PMCID: PMC5234861 DOI: 10.1016/j.tem.2016.06.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/09/2016] [Accepted: 06/13/2016] [Indexed: 12/16/2022]
Abstract
The study of white adipose tissue (WAT) 'browning' has become a 'hot topic' in various acute and chronic metabolic conditions, based on the idea that WAT browning might be able to facilitate weight loss and improve metabolic health. However, this view cannot be translated into all areas of medicine. Recent studies identified effects of browning associated with adverse outcomes, and as more studies are being conducted, a very different picture has emerged about WAT browning and its detrimental effect in acute and chronic hypermetabolic conditions. Therefore, the notion that browning is supposedly beneficial may be inadequate. In this review we analyze how and why browning in chronic hypermetabolic associated diseases can be detrimental and lead to adverse outcomes.
Collapse
Affiliation(s)
- Abdikarim Abdullahi
- Institute of Medical Science, University of Toronto, Canada; Sunnybrook Research Institute, Totonto, Canada
| | - Marc G Jeschke
- Institute of Medical Science, University of Toronto, Canada; Sunnybrook Research Institute, Totonto, Canada; Ross Tilley Burn Centre, Sunnybrook Health Sciences Centre, University of Toronto, Canada; Department of Surgery, Division of Plastic Surgery, Department of Immunology, University of Toronto, Canada.
| |
Collapse
|
72
|
Muraki M. Preparation of a functional fluorescent human Fas ligand extracellular domain derivative using a three-dimensional structure guided site-specific fluorochrome conjugation. SPRINGERPLUS 2016; 5:997. [PMID: 27398274 PMCID: PMC4936993 DOI: 10.1186/s40064-016-2673-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/24/2016] [Indexed: 11/04/2022]
Abstract
Background Human Fas ligand extracellular domain has been investigated as an important target protein in the field of medical biotechnology. In a recent study, the author developed an effective method to produce biologically active human Fas ligand extracellular domain derivatives using site-specific chemical modifications. Findings A human Fas ligand extracellular domain derivative containing a reactive cysteine residue within its N-terminal tag sequence, which locates not proximal to the binding interface between the ligand and the receptor in terms of the three-dimensional structure, was modified by Fluorescein-5-Maleimide without impairing the specific binding activity toward human Fas receptor extracellular domain. The purified protein sample free of low molecular-weight contaminants showed a characteristic fluorescence spectrum derived from the attached Fluorescein moieties, and formed a stable binding complex with human Fas receptor extracellular domain—human IgG1 Fc domain fusion protein in solution. The conjugation number of the fluorochrome was estimated to be 2.5 per a single human Fas ligand extracellular domain trimer from the ratio of the absorbance value at 280 nm to that at 495 nm. Conclusions A functional fluorescent human Fas ligand extracellular domain derivative was prepared via a site-specific conjugation of fluorochrome, which was guided by the three-dimensional structure information on the ligand-receptor complex. Fluorescent derivatives created by this method may contribute to the development of an improved diagnosis system for the diseases related to Fas receptor. Electronic supplementary material The online version of this article (doi:10.1186/s40064-016-2673-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michiro Muraki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, 305-8566 Japan
| |
Collapse
|
73
|
LIU XUEZHONG, ZHANG YIRAN, WANG YI, YAN YUAN, WANG JIAJING, GU JIANHONG, CHUN BIANJIAN, LIU ZONGPING. Investigation of cadmium-induced apoptosis and the protective effect of N-acetylcysteine in BRL 3A cells. Mol Med Rep 2016; 14:373-9. [DOI: 10.3892/mmr.2016.5218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 12/30/2015] [Indexed: 11/06/2022] Open
|
74
|
Synergistic Effect and Molecular Mechanisms of Traditional Chinese Medicine on Regulating Tumor Microenvironment and Cancer Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1490738. [PMID: 27042656 PMCID: PMC4793102 DOI: 10.1155/2016/1490738] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 01/26/2016] [Indexed: 12/23/2022]
Abstract
The interaction of tumor cells with the microenvironment is like a relationship between the “seeds” and “soil,” which is a hotspot in recent cancer research. Targeting at tumor microenvironment as well as tumor cells has become a new strategy for cancer treatment. Conventional cancer treatments mostly focused on single targets or single mechanism (the seeds or part of the soil); few researches intervened in the whole tumor microenvironment and achieved ideal therapeutic effect as expected. Traditional Chinese medicine displays a broad range of biological effects, and increasing evidence has shown that it may relate with synergistic effect on regulating tumor microenvironment and cancer cells. Based on literature review and our previous studies, we summarize the synergistic effect and the molecular mechanisms of traditional Chinese medicine on regulating tumor microenvironment and cancer cells.
Collapse
|
75
|
Khalifa RH, Bahgat DMR, Darwish HAH, Shahin RMH. Significant association between FasL gene -844T/C polymorphism and risk to hepatocellular carcinoma in Egyptian patients. Immunol Lett 2016; 172:84-8. [PMID: 26891954 DOI: 10.1016/j.imlet.2016.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/12/2016] [Accepted: 02/12/2016] [Indexed: 01/14/2023]
Abstract
Fas/Fas ligand (FasL) system is the most critical apoptotic signaling entity in the extrinsic apoptotic pathway; hence mutations affecting this pathway may prevent the immune system from the removal of newly-formed tumor cells, and thus lead to tumor formation. The present study investigated the association between the FasL -844T/C polymorphism and the risk of hepatocellular carcinoma (HCC) in a cohort of Egyptian patients and explored the relationship of various clinical and pathological parameters with this single nucleotide polymorphism (SNP). Blood samples were withdrawn from hundred HCC patients and 100 age-, sex- and ethnically matched controls. The FasL -844T/C (rs763110) gene polymorphism was typed from genomic DNA using polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) assay. Genotype distributions and allelic frequencies between patients and control subjects showed that the TT homozygous patients were two times more likely to develop HCC (p=0.011). Also, the T allele was found to be a significant risk factor for the disease (OR 1.970, 95% CI 1.250-3.105, p=0.003). No association was detected between different parameters of the disease and the SNP. For the first time, our results suggest that the -844T/C polymorphism in the FasL gene confers risk to HCC. The alarming increase in the incidence of HCC in Egypt encourages further studies to document our results in a larger sample, and recommends more genetic studies hoping to define a genomic risk prediction specific to this cancer in our population.
Collapse
Affiliation(s)
- Rania H Khalifa
- Department of Clinical & Chemical Pathology, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt.
| | - Dina M Rasheed Bahgat
- Department of Clinical & Chemical Pathology, Kasr Al-Ainy, School of Medicine, Cairo University, Egypt
| | | | | |
Collapse
|
76
|
Regan D, Dow S. Manipulation of Innate Immunity for Cancer Therapy in Dogs. Vet Sci 2015; 2:423-439. [PMID: 29061951 PMCID: PMC5644648 DOI: 10.3390/vetsci2040423] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/20/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
Over the last one to two decades, the field of cancer immunotherapy has rapidly progressed from early preclinical studies to a successful clinical reality and fourth major pillar of human cancer therapy. While current excitement in the field of immunotherapy is being driven by several major breakthroughs including immune checkpoint inhibitors and adoptive cell therapies, these advances stem from a foundation of pivotal studies demonstrating the immune systems role in tumor control and eradication. The following will be a succinct review on veterinary cancer immunotherapy as it pertains to manipulation of the innate immune system to control tumor growth and metastasis. In addition, we will provide an update on recent progress in our understanding of the innate immune system in veterinary tumor immunology, and how these gains may lead to novel therapies for the treatment of cancer in companion animals.
Collapse
Affiliation(s)
- Daniel Regan
- Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
- The Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
| | - Steven Dow
- Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
- The Center for Immune and Regenerative Medicine, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80525, USA.
| |
Collapse
|
77
|
Gravina GL, Mancini A, Sanita P, Vitale F, Marampon F, Ventura L, Landesman Y, McCauley D, Kauffman M, Shacham S, Festuccia C. KPT-330, a potent and selective exportin-1 (XPO-1) inhibitor, shows antitumor effects modulating the expression of cyclin D1 and survivin [corrected] in prostate cancer models. BMC Cancer 2015; 15:941. [PMID: 26620414 PMCID: PMC4666032 DOI: 10.1186/s12885-015-1936-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND AIMS Increased expression of Chromosome Region Maintenance (CRM-1)/exportin-1 (XPO-1) has been correlated with poor prognosis in several aggressive tumors, making it an interesting therapeutic target. Selective Inhibitor of Nuclear Export (SINE) compounds bind to XPO-1 and block its ability to export cargo proteins. Here, we investigated the effects of a new class of SINE compounds in models of prostate cancer. MATERIAL AND METHODS We evaluated the expression of XPO-1 in human prostate cancer tissues and cell lines. Next, six SINE (KPT-127, KPT-185, KPT-205, KPT-225, KPT-251 and KPT-330) compounds having different potency with broad-spectrum, tumor-selective cytotoxicity, tolerability and pharmacokinetic profiles were tested in a panel of prostate cancer cells representing distinct differentiation/progression states of disease and genotypes. Two SINE candidates for clinical trials (KPT-251 and KPT-330) were also tested in vivo in three cell models of aggressive prostate cancer engrafted in male nude mice. RESULTS AND CONCLUSIONS XPO-1 is overexpressed in prostate cancer compared to normal or hyperplastic tissues. Increased XPO-1 expression, mainly in the nuclear compartment, was associated with increased Gleason score and bone metastatic potential supporting the use of SINEs in advanced prostate cancer. SINE compounds inhibited proliferation and promoted apoptosis of tumor cells, but did not affect immortalized non-transformed prostate epithelial cells. Nuclei from SINE treated cells showed increased protein localization of XPO-1, survivin and cyclin D1 followed by degradation of these proteins leading to cell cycle arrest and apoptosis. Oral administration of KPT-251 and KPT-330 in PC3, DU145 and 22rv1 tumor-bearing nude mice reduced tumor cell proliferation, angiogenesis and induced apoptosis. Our results provide supportive evidence for the therapeutic use of SINE compounds in advanced/castration resistant prostate cancers and warrants further clinical investigation.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Patrizia Sanita
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Flora Vitale
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| | - Luca Ventura
- Pathology Division, San Salvatore Hospital, L'Aquila, Italy.
| | | | | | | | | | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
78
|
Zhao S, Wu D, Wu P, Wang Z, Huang J. Serum IL-10 Predicts Worse Outcome in Cancer Patients: A Meta-Analysis. PLoS One 2015; 10:e0139598. [PMID: 26440936 PMCID: PMC4595202 DOI: 10.1371/journal.pone.0139598] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 09/14/2015] [Indexed: 12/16/2022] Open
Abstract
Background IL–10 is an important immunosuppressive cytokine which is frequently elevated in tumor microenvironment. Some studies have reported that overexpression of serous IL–10 is correlated with worse outcome in patients with malignant tumor. Here, we conducted a meta-analysis to assess the prognostic impact of serous IL–10 expression in cancer patients. Methods We searched PubMed and EBSCO for studies in evaluating the association of IL–10 expression—in serum and clinical outcome in cancer patients. Overall survival (OS) was the primary prognostic indicator and disease-free survival (DFS) was the secondary indicator. Extracted data were computed into odds ratios (ORs) and 95% confidence interval (CI) or a P value for survival at 1, 3 and 5 years. Pooled data were weighted using the Mantel–Haenszel Fixed-effect model. All statistical tests were two-sided. Results A total of 1788 patients with cancer from 21 published studies were incorporated into this meta-analysis. High level of serum IL–10 was significantly associated with worse OS at 1-year (OR = 3.70, 95% CI = 2.81 to 4.87, P < 0.00001), 3-year (OR = 3.33, 95% CI = 2.53 to 4.39, P < 0.0001) and 5-year (OR = 2.80, 95% CI = 1.90 to 4.10, P < 0.0001) of cancer. Subgroup analysis showed that the correlation between serous IL–10 expression and outcome of patients with solid tumors and hematological malignancies are consistent. The association of IL–10 with worse DFS at 1-year (OR = 3.34, 95% CI = 1.40 to 7.94, P = 0.006) and 2-year (OR = 3.91, 95% CI = 1.79 to 8.53, P = 0.0006) was also identified. Conclusions High expression of serous IL–10 leads to an adverse survival in most types of cancer. IL–10 is a valuable biomarker for prognostic prediction and targeting IL–10 treatment options for both solid tumors and hematological malignancies.
Collapse
Affiliation(s)
- Shuai Zhao
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Provincial Key Laboratory of Molecular Biology in Medical Sciences), Zhejiang University, Hangzhou, China
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Dang Wu
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Provincial Key Laboratory of Molecular Biology in Medical Sciences), Zhejiang University, Hangzhou, China
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Pin Wu
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Provincial Key Laboratory of Molecular Biology in Medical Sciences), Zhejiang University, Hangzhou, China
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhen Wang
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Provincial Key Laboratory of Molecular Biology in Medical Sciences), Zhejiang University, Hangzhou, China
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Huang
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Provincial Key Laboratory of Molecular Biology in Medical Sciences), Zhejiang University, Hangzhou, China
- Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- * E-mail:
| |
Collapse
|
79
|
Yu X, Zhou X, Fu C, Wang Q, Nie T, Zou F, Guo R, Liu H, Zhang B, Dai M. Celastrol induces apoptosis of human osteosarcoma cells via the mitochondrial apoptotic pathway. Oncol Rep 2015; 34:1129-36. [PMID: 26165547 PMCID: PMC4530898 DOI: 10.3892/or.2015.4124] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/15/2015] [Indexed: 12/28/2022] Open
Abstract
Celastrol is an active compound extracted from the root bark of Triptergium wilfordii Hook F., also known as 'Thunder of God Vine'. It is a well-known Chinese medicinal herb that was found to inhibit tumor cell growth and promote apoptosis in several tumor cell lines. However, research into its effects on osteosarcoma cell apoptosis is still extremely limited. The present study was undertaken to determine the effect of celastrol on viability and apoptosis of osteosarcoma cells and furthermore, to illuminate the molecular mechanism of celastrol-induced osteosarcoma cell apoptosis. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay was used to evaluate the viability of the cells following treatment with celastrol. The effect of celastrol on the apoptotic rate of the cells was evaluated by flow cytometry using Annexin V-PE/7-AAD staining assay. Fluorescence microscopy was used to detect the morphological changes in the human osteosarcoma U-2OS cell lines. The expression of Bcl-2 family proteins, caspase-3, caspase-8, caspase-9, cytochrome c and PARP was measured by western blotting. We found that celastrol significantly inhibited the growth of osteosarcoma cells in a dose-dependent manner, particularly U-2OS cells. Furthermore, we observed that celastrol upregulated the expression of the pro-apoptotic proteins Bax and cytochrome c and altered the ratio of Bax/Bcl-2, and triggered the mitochondrial apoptotic pathway, resulting in caspase-3 and -9 activation and PARP cleavage. To conclude, the results indicate that celastrol inhibits the proliferation of human osteosarcoma cancer cells by inducing apoptosis via the mitochondrial-dependent pathway.
Collapse
Affiliation(s)
- Xiaolong Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xin Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changlin Fu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiang Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tao Nie
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fan Zou
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Runsheng Guo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hucheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
80
|
Effect and Molecular Mechanisms of Traditional Chinese Medicine on Regulating Tumor Immunosuppressive Microenvironment. BIOMED RESEARCH INTERNATIONAL 2015; 2015:261620. [PMID: 26161392 PMCID: PMC4486742 DOI: 10.1155/2015/261620] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/16/2014] [Indexed: 01/07/2023]
Abstract
Traditional Chinese medicine (TCM) is an important complementary strategy for treating cancer in China. The mechanism is related to regulating the internal environment and remodeling the tumor immunosuppressive microenvironment (TIM). Herein we illustrate how TIM is reformed and its protumor activity on promoting tumor cell proliferation, angiogenesis and lymphangiogenesis, tumor invasion, and the oncogenicity of cancer stem cells. Furthermore we summarize the effects and mechanism of TCM on regulating TIM via enhancing antitumor immune responses (e.g., regulating the expression of MHC molecules and Fas/FasL, attenuating cancerigenic ability of cancer stem cells) and remolding immunosuppressive cells (e.g., reversing immune phenotypes of T lymphocytes and tumor associated macrophages, promoting dendritic cells mature, restraining myeloid derived suppressor cells function, and regulating Th1/Th2 factors). We also reveal the bidirectional and multitargeting functions of TCM on regulating TIM. Hopefully, it provides new theoretical basis for TCM clinical practice in cancer treatment and prevention.
Collapse
|
81
|
Radisky ES, Radisky DC. Matrix metalloproteinases as breast cancer drivers and therapeutic targets. Front Biosci (Landmark Ed) 2015; 20:1144-63. [PMID: 25961550 DOI: 10.2741/4364] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Members of the matrix metalloproteinase (MMP) family have been identified as poor prognosis markers for breast cancer patients and as drivers of many facets of the tumor phenotype in experimental models. Early enthusiasm for MMPs as therapeutic targets was tempered following disappointing clinical trials that utilized broad spectrum, small molecule catalytic site inhibitors. However, subsequent research has continued to define key roles for MMPs as breast cancer promoters, to elucidate the complex roles that that these proteins play in breast cancer development and progression, and to identify how these roles are linked to specific and unique biochemical features of individual members of the MMP family. Here, we provide an overview of the structural features of the MMPs, then discuss clinical studies identifying which MMP family members are linked with breast cancer development and new experimental studies that reveal how these specific MMPs may play unique roles in the breast cancer microenvironment. We conclude with a discussion of the most promising avenues for development of therapeutic agents capable of targeting the tumor-promoting properties of MMPs.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida 32224,
| | | |
Collapse
|
82
|
Pei Q, Pan J, Ding X, Wang J, Zou X, Lv Y. Gemcitabine sensitizes pancreatic cancer cells to the CTLs antitumor response induced by BCG-stimulated dendritic cells via a Fas-dependent pathway. Pancreatology 2015; 15:233-9. [PMID: 25937078 DOI: 10.1016/j.pan.2015.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 04/01/2015] [Accepted: 04/02/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES There are increasing evidences suggesting that chemotherapeutic agents can enhance the cytotoxic T lymphocytes (CTLs) antitumor effect, but the precise mechanism is not fully explained. This study aims to investigate whether gemcitabine (GEM) can sensitize pancreatic cancer cells to the CTLs antitumor response, and explore the potential mechanism. METHODS Cell counting kit-8 assays (CCK-8) were performed to determine the tumor cell proliferation. Flow cytometric analysis was conducted to analyze maturation of DCs and the expression of Fas. An Annexin V FITC Apoptosis Detection Kit was performed to detect tumor cell apoptosis. CytoTox 96 Nonradioactive Cytotoxicity assays were used to determine T cell-mediated tumor cell lysis. RESULTS First, it was demonstrated that Bacillus Calmette Guérin (BCG) could be used to induce effective CTLs antitumor response. Then, GEM inhibited the growth of SW1990 cells, induced apoptosis and upregulated the Fas expression even at a low concentration. When antagonistic anti-Fas mAb ZB4 was preincubated with GEM-treated SW1990 cells, the lysis induced by CTLs was reduced. Moreover, agonistic anti-Fas mAb CH11 induced more apoptosis of GEM-treated SW1990 cells. CONCLUSION Our results show that GEM sensitizes pancreatic cancer cells to the CTLs antitumor response, and the sensitization is associated with upregulation of Fas on pancreatic cancer cells.
Collapse
Affiliation(s)
- Qingshan Pei
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China; Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China
| | - Jianmei Pan
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xiwei Ding
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China
| | - Jing Wang
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China
| | - Xiaoping Zou
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China.
| | - Ying Lv
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China.
| |
Collapse
|
83
|
Zhang W, Gao R, Yu Y, Guo K, Hou P, Yu M, Liu Y, Yang A. Iodine-131 induces apoptosis in HTori-3 human thyrocyte cell line and G2/M phase arrest in a p53-independent pathway. Mol Med Rep 2015; 11:3148-54. [PMID: 25515142 DOI: 10.3892/mmr.2014.3096] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 11/07/2014] [Indexed: 11/05/2022] Open
Abstract
Iodine‑131 is known to destroy residual thyroid tissue following surgical resection of differentiated thyroid carcinoma and is widely used to treat hyperthyroidism. However, the mechanism by which iodine‑131 induces apoptosis and cell cycle arrest in the human thyrocyte cell line, Htori‑3, remains to be elucidated. In the present study, the cytotoxic effect of iodine‑131 on the HTori‑3 cell line and the underlying mechanism of iodine‑131‑induced cell apoptosis were investigated. Cell viability was analyzed using an MTT assay, while cell apoptosis and cell cycle arrest were determined using flow cytometry. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analyses were performed to determine the changes in the expression levels of p53, B‑cell lymphoma 2 (Bcl‑2), Fas and growth arrest and DNA damage‑inducible 45 (GADD45), following iodine‑131 treatment. The results demonstrated that iodine‑131 may inhibit HTori‑3 cell growth via cell apoptosis and G2/M phase arrest in a time‑ and dose‑dependent manner. The iodine‑131 dose required for 50% growth inhibition of HTori‑3 cell viability 48 h after treatment was 27.75±2.22 MBq/ml. Upregulation of Fas and downregulation of Bcl‑2 expression levels were observed following iodine‑131 treatment. The results of RT‑qPCR revealed an increase in the GADD45 mRNA expression following HTori‑3 cell exposure to iodine‑131. Notably, the mRNA and protein expression levels of p53 were not altered following iodine‑131 treatment. In conclusion, iodine‑131 may induce apoptosis in HTori‑3 cells by downregulating the expression of Bcl‑2 and upregulating the expression of Fas. In addition, iodine‑131 may upregulate GADD45 mRNA expression in HTori‑3 cells, resulting in G2/M phase arrest in a p53‑independent pathway.
Collapse
Affiliation(s)
- Weixiao Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Gao
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Yu
- Department of Public Health, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Kun Guo
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Hou
- Endocrinology Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Mingqi Yu
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Aimin Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
84
|
Modulation of SIV and HIV DNA vaccine immunity by Fas-FasL signaling. Viruses 2015; 7:1429-53. [PMID: 25807052 PMCID: PMC4379579 DOI: 10.3390/v7031429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 03/10/2015] [Accepted: 03/15/2015] [Indexed: 01/14/2023] Open
Abstract
Signaling through the Fas/Apo-1/CD95 death receptor is known to affect virus-specific cell-mediated immune (CMI) responses. We tested whether modulating the Fas-apoptotic pathway can enhance immune responses to DNA vaccination or lymphocytic choriomeningitis virus (LCMV) infection. Mice were electroporated with plasmids expressing a variety of pro- or anti-apoptotic molecules related to Fas signaling and then either LCMV-infected or injected with plasmid DNA expressing SIV or HIV antigens. Whereas Fas or FasL knockout mice had improved CMI, down-regulation of Fas or FasL by shRNA or antibody failed to improve CMI and was accompanied by increases in regulatory T cells (Treg). Two “adjuvant” plasmids were discovered that significantly enhanced plasmid immunizations. The adjuvant effects of Fas-associated death domain (FADD) and of cellular FLICE-inhibitory protein (cFLIP) were consistently accompanied by increased effector memory T lymphocytes and increased T cell proliferation. This adjuvant effect was also observed when comparing murine infections with LCMV-Armstrong and its persisting variant LCMV-Clone 13. LCMV-Armstrong was cleared in 100% of mice nine days after infection, while LCMV-Clone 13 persisted in all mice. However, half of the mice pre-electroporated with FADD or cFLIP plasmids were able to clear LCMV-Clone 13 by day nine, and, in the case of cFLIP, increased viral clearance was accompanied by higher CMI. Our studies imply that molecules in the Fas pathway are likely to affect a number of events in addition to the apoptosis of cells involved in immunity.
Collapse
|
85
|
Chen J. Signaling pathways in HPV-associated cancers and therapeutic implications. Rev Med Virol 2015; 25 Suppl 1:24-53. [PMID: 25752815 DOI: 10.1002/rmv.1823] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 10/15/2014] [Accepted: 12/27/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Jiezhong Chen
- School of Biomedical Sciences and Australian Institute for Bioengineering and Nanotechnology; The University of Queensland; Brisbane Queensland Australia
| |
Collapse
|
86
|
Benito-Martin A, Di Giannatale A, Ceder S, Peinado H. The new deal: a potential role for secreted vesicles in innate immunity and tumor progression. Front Immunol 2015; 6:66. [PMID: 25759690 PMCID: PMC4338782 DOI: 10.3389/fimmu.2015.00066] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Accepted: 02/02/2015] [Indexed: 12/30/2022] Open
Abstract
Tumors must evade the immune system to survive and metastasize, although the mechanisms that lead to tumor immunoediting and their evasion of immune surveillance are far from clear. The first line of defense against metastatic invasion is the innate immune system that provides immediate defense through humoral immunity and cell-mediated components, mast cells, neutrophils, macrophages, and other myeloid-derived cells that protect the organism against foreign invaders. Therefore, tumors must employ different strategies to evade such immune responses or to modulate their environment, and they must do so prior metastasizing. Exosomes and other secreted vesicles can be used for cell–cell communication during tumor progression by promoting the horizontal transfer of information. In this review, we will analyze the role of such extracellular vesicles during tumor progression, summarizing the role of secreted vesicles in the crosstalk between the tumor and the innate immune system.
Collapse
Affiliation(s)
- Alberto Benito-Martin
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Weill Cornell Medical College , New York, NY , USA
| | - Angela Di Giannatale
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Weill Cornell Medical College , New York, NY , USA
| | - Sophia Ceder
- Department of Oncology and Pathology, Karolinska Institutet , Stockholm , Sweden
| | - Héctor Peinado
- Children's Cancer and Blood Foundation Laboratories, Department of Pediatrics, Weill Cornell Medical College , New York, NY , USA ; Microenvironment and Metastasis Laboratory, Department of Molecular Oncology, Spanish National Cancer Research Centre (CNIO) , Madrid , Spain
| |
Collapse
|
87
|
NF-κB regulates caspase-4 expression and sensitizes neuroblastoma cells to Fas-induced apoptosis. PLoS One 2015; 10:e0117953. [PMID: 25695505 PMCID: PMC4335045 DOI: 10.1371/journal.pone.0117953] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/01/2022] Open
Abstract
Found in neurons and neuroblastoma cells, Fas-induced apoptosis and accompanied activation of NF-κB signaling were thought to be associated with neurodegenerative diseases. However, the detailed functions of NF-κB activation in Fas killing and the effect of NF-κB activation on its downstream events remain unclear. Here, we demonstrated that agonistic Fas antibody induces cell death in a dose-dependent way and NF-κB signaling is activated as well, in neuroblastoma cells SH-EP1. Unexpectedly, NF-κB activation was shown to be pro-apoptotic, as suggested by the reduction of Fas-induced cell death with either a dominant negative form of IκBα (DN-IκBα) or an IκB kinase-specific inhibitor. To our interest, when analyzing downstream events of NF-κB signaling, we found that DN-IκBα only suppressed the expression of caspase-4, but not other caspases. Vice versa, enhancement of NF-κB activity by p65 (RelA) overexpression increased the expression of caspase-4 at both mRNA and protein levels. More directly, results from dual luciferase reporter assay demonstrated the regulation of caspase-4 promoter activity by NF-κB. When caspase-4 activity was blocked by its dominant negative (DN) form, Fas-induced cell death was substantially reduced. Consistently, the cleavage of PARP and caspase-3 induced by Fas was also reduced. In contrast, the cleavage of caspase-8 remained unaffected in caspase-4 DN cells, although caspase-8 inhibitor could rescue Fas-induced cell death. Collectively, these data suggest that caspase-4 activity is required for Fas-induced cell apoptosis and caspase-4 may act upstream of PARP and caspase-3 and downstream of caspase-8. Overall, we demonstrate that NF-κB can mediate Fas-induced apoptosis through caspase-4 protease, indicating that caspase-4 is a new mediator of NF-κB pro-apoptotic pathway in neuroblastoma cells.
Collapse
|
88
|
Saigusa S, Tanaka K, Ohi M, Toiyama Y, Yasuda H, Kitajima T, Okugawa Y, Inoue Y, Mohri Y, Kusunoki M. Clinical implications of Fas/Fas ligand expression in patients with esophageal squamous cell carcinoma following neoadjuvant chemoradiotherapy. Mol Clin Oncol 2014; 3:151-156. [PMID: 25469286 DOI: 10.3892/mco.2014.431] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/10/2014] [Indexed: 12/23/2022] Open
Abstract
Recent epidemiological studies demonstrated that the incidence of esophageal squamous cell carcinoma (ESCC) is on the increase. Although neoadjuvant chemoradiotherapy (CRT) followed by surgery may improve long-term survival and reduce local recurrence in patients with esophageal cancer, the overall cure rate of esophageal cancer is low. Fas/Fas ligand (FasL) signaling initiates the cell death pathway. The roles of FasL in tumor growth, progression and resistance to treatment have been demonstrated in several malignancies. The aim of this preliminary study was to evaluate Fas/FasL expression in ESCC with neoadjuvant CRT. A total of 20 patients who received neoadjuvant CRT (30-40 Gy; 5-fluorouracil plus cisplatin followed by surgery) were enrolled. We evaluated the expression of Fas, FasL and Ki67 (a proliferative marker) using immunohistochemistry and analyzed the correlations between their expression and clinical outcomes. Additionally, we investigated the association of Fas/FasL expression with peritumoral immune CD8-positive and Foxp3-positive cells. High FasL expression was significantly correlated with disease recurrence (P=0.0134). Patients with high FasL expression exhibited poorer recurrence-free and overall survival (P=0.0102 and 0.0385, respectively). Patients with low Fas and high FasL exhibited significantly poorer recurrence-free survival (P=0.0035). Although statistical significance was not reached, Fas expression appeared to be inversely correlated with Foxp3-positive cells and FasL expression appeared to be inversely correlated with CD8-positive cells. In conclusion, FasL expression was associated with tumor relapse and poor prognosis in patients with ESCC following CRT. Pharmacological control of Fas/FasL signaling may improve therapeutic efficacy and outcome in ESCC patients receiving preoperative CRT.
Collapse
Affiliation(s)
- Susumu Saigusa
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Koji Tanaka
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masaki Ohi
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Hiromi Yasuda
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Takahito Kitajima
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshinaga Okugawa
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yasuhiro Inoue
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yasuhiko Mohri
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Masato Kusunoki
- Department of Gastrointestinal and Pediatric Surgery, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
89
|
Tegillarca granosa Extract Haishengsu Induces Apoptosis in Human Hepatocellular Carcinoma Cell Line BEL-7402 Via Fas-Signaling Pathways. Cell Biochem Biophys 2014; 71:837-44. [DOI: 10.1007/s12013-014-0271-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
90
|
Li L, Yao YC, Fang SH, Ma CQ, Cen Y, Xu ZM, Dai ZY, Li C, Li S, Zhang T, Hong HH, Qi WW, Zhou T, Li CY, Yang X, Gao GQ. Pigment epithelial-derived factor (PEDF)-triggered lung cancer cell apoptosis relies on p53 protein-driven Fas ligand (Fas-L) up-regulation and Fas protein cell surface translocation. J Biol Chem 2014; 289:30785-30799. [PMID: 25225287 DOI: 10.1074/jbc.m114.590000] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF), a potent antiangiogenesis agent, has recently attracted attention for targeting tumor cells in several types of tumors. However, less is known about the apoptosis-inducing effect of PEDF on human lung cancer cells and the underlying molecular events. Here we report that PEDF has a growth-suppressive and proapoptotic effect on lung cancer xenografts. Accordingly, in vitro, PEDF apparently induced apoptosis in A549 and Calu-3 cells, predominantly via the Fas-L/Fas death signaling pathway. Interestingly, A549 and Calu-3 cells are insensitive to the Fas-L/Fas apoptosis pathway because of the low level of cell surface Fas. Our results revealed that, in addition to the enhancement of Fas-L expression, PEDF increased the sensitivity of A549 and Calu-3 cells to Fas-L-mediated apoptosis by triggering the translocation of Fas protein to the plasma membrane in a p53- and FAP-1-dependent manner. Similarly, the up-regulation of Fas-L by PEDF was also mediated by p53. Furthermore, peroxisome proliferator-activated receptor γ was determined to be the upstream regulator of p53. Together, these findings uncover a novel mechanism of tumor cell apoptosis induced by PEDF and provide a potential therapeutic strategy for tumors that are insensitive to Fas-L/Fas-dependent apoptosis because of a low level of cell surface Fas.
Collapse
Affiliation(s)
- Lei Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; Department of Reproductive Medicine Center, Key Laboratory for Reproductive Medicine of Guangdong Province, Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Ya-Chao Yao
- Laboratory Center of Guangdong No. 2 Provincial People's Hospital, Guangzhou 510317, Guangdong Province, China
| | - Shu-Huan Fang
- DME Center, Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Cai-Qi Ma
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Cen
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zu-Min Xu
- Cancer Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524000, China
| | - Zhi-Yu Dai
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Cen Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Shuai Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hong-Hai Hong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei-Wei Qi
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Chao-Yang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China,.
| | - Xia Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; China Key Laboratory of Tropical Disease Control, Sun Yat-sen University, Ministry of Education, Guangzhou 510080, China, and.
| | - Guo-Quan Gao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China,; Key Laboratory of Functional Molecules from Marine Microorganisms, Sun Yat-sen University, Department of Education of Guangdong Province, Guangdong 510080, China.
| |
Collapse
|
91
|
Kidney tubular β-catenin signaling controls interstitial fibroblast fate via epithelial-mesenchymal communication. Sci Rep 2014; 3:1878. [PMID: 23698793 PMCID: PMC3662012 DOI: 10.1038/srep01878] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/01/2013] [Indexed: 12/11/2022] Open
Abstract
Activation of β-catenin, the principal mediator of canonical Wnt signaling, is a common pathologic finding in a wide variety of chronic kidney diseases (CKD). While β-catenin is induced predominantly in renal tubular epithelium in CKD, surprisingly, depletion of tubular β-catenin had little effect on the severity of renal fibrosis. Interestingly, less apoptosis was detected in interstitial fibroblasts in knockout mice, which was accompanied by a decreased expression of Bax and Fas ligand (FasL). Tubule-specific knockout of β-catenin diminished renal induction of matrix metalloproteinase (MMP-7), which induced FasL expression in interstitial fibroblasts and potentiated fibroblast apoptosis in vitro. These results demonstrate that loss of tubular β-catenin resulted in enhanced interstitial fibroblast survival due to decreased MMP-7 expression. Our studies uncover a novel role of the tubular β-catenin/MMP-7 axis in controlling the fate of interstitial fibroblasts via epithelial-mesenchymal communication.
Collapse
|
92
|
Knockdown of microRNA-155 in Kupffer cells results in immunosuppressive effects and prolongs survival of mouse liver allografts. Transplantation 2014; 97:626-35. [PMID: 24531847 DOI: 10.1097/tp.0000000000000061] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Our previous studies have shown that Kupffer cells (KCs) play a crucial role in postoperative pathologic changes. Recent reports have demonstrated that microRNA-155 (miR-155) is associated with inflammation and upregulation of proinflammatory mediators in the peripheral blood and allografts of transplant patients. However, the precise mechanism for this remains unknown. METHODS KCs isolated from BALB/c mice were transfected with miR-155 mimic or inhibitor. Levels of suppressor of cytokine signaling 1/Janus kinase/signal transducer and activator of transcription (SOCS1/JAK/STAT) proteins and surface molecules (MHC-II, CD40, and CD86) were then measured. T-cell proliferation and apoptosis were evaluated in mixed lymphocyte reactions. Orthotopic liver transplantation was performed in mice after miR-155 short hairpin RNA lentivirus treatment, and postoperative survival, liver function and histology, and mRNA and protein expression were analyzed. RESULTS miR-155 knockdown in KCs decreased MHC-II, CD40, and CD86 expression, suppressed antigen-presenting function, and affected SOCS1/JAK/STAT inflammatory pathways. In addition, KCs transfected with miR-155 inhibitor and cocultured with T lymphocytes showed reduced T-cell responses but a greater number of apoptotic T cells. Finally, miR-155 suppression in graft liver prolonged liver allograft survival and improved liver function. The changes were closely associated with the levels of T helper 1 and 2 (Th1/Th2) cytokines and T-cell apoptosis, but a direct mechanistic link in vivo was not established. CONCLUSION These data suggest miR-155 regulates the balance of Th1/Th2 cytokines and the maturation and function of KCs in mice. miR-155 repression in KCs positively regulates KC function toward immunosuppression and prolongs liver allograft survival.
Collapse
|
93
|
Thorburn A, Thamm DH, Gustafson DL. Autophagy and cancer therapy. Mol Pharmacol 2014; 85:830-8. [PMID: 24574520 PMCID: PMC4014668 DOI: 10.1124/mol.114.091850] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 02/26/2014] [Indexed: 12/13/2022] Open
Abstract
Autophagy is the process by which cellular material is delivered to lysosomes for degradation and recycling. There are three different types of autophagy, but macroautophagy, which involves the formation of double membrane vesicles that engulf proteins and organelles that fuse with lysosomes, is by far the most studied and is thought to have important context-dependent roles in cancer development, progression, and treatment. The roles of autophagy in cancer treatment are complicated by two important discoveries over the past few years. First, most (perhaps all) anticancer drugs, as well as ionizing radiation, affect autophagy. In most, but not all cases, these treatments increase autophagy in tumor cells. Second, autophagy affects the ability of tumor cells to die after drug treatment, but the effect of autophagy may be to promote or inhibit cell death, depending on context. Here we discuss recent research related to autophagy and cancer therapy with a focus on how these processes may be manipulated to improve cancer therapy.
Collapse
Affiliation(s)
- Andrew Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado (A.T.); and Flint Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado (D.H.T., D.L.G.)
| | | | | |
Collapse
|
94
|
Cohen M, Pierredon S, Wuillemin C, Delie F, Petignat P. Acellular fraction of ovarian cancer ascites induce apoptosis by activating JNK and inducing BRCA1, Fas and FasL expression in ovarian cancer cells. Oncoscience 2014; 1:262-71. [PMID: 25594018 PMCID: PMC4278302 DOI: 10.18632/oncoscience.31] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 04/28/2014] [Indexed: 12/26/2022] Open
Abstract
Acellular fraction of ascites might play an active role in tumor development. Nevertheless the mechanisms involved in the tumor-modulating properties are still controversial. Here, we demonstrate that malignant ascites from 8 patients with epithelial ovarian cancer did not influence proliferative or invasive properties of ovarian cancer cells, but promoted H2O2-induced apoptosis and increased sensitivity to paclitaxel. Malignant ascites induced BRCA1, Fas and FasL expression and phosphorylation of JNK, but not the activation of caspase pathway. Ascites-induced apoptosis of ovarian cancer cells was strongly inhibited by a JNK inhibitor suggesting a critical role of JNK pathway in ascite-induced apoptosis. The use of siRNA JNK confirmed the importance of JNK in ascites-induced Fas and FasL expression. These results demonstrate that malignant ascites induce apoptosis of ovarian cancer cells and encourage us to think about the clinical management of ovarian cancer patients with malignant ascites.
Collapse
Affiliation(s)
- Marie Cohen
- Department of Gynecology-Obstetrics, faculty of medicine, Geneva, Switzerland
| | - Sandra Pierredon
- Department of Gynecology-Obstetrics, faculty of medicine, Geneva, Switzerland
| | - Christine Wuillemin
- Department of Gynecology-Obstetrics, faculty of medicine, Geneva, Switzerland
| | - Florence Delie
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva,Switzerland
| | - Patrick Petignat
- Department of Gynecology-Obstetrics, faculty of medicine, Geneva, Switzerland
| |
Collapse
|
95
|
Nikitovic D, Papoutsidakis A, Karamanos NK, Tzanakakis GN. Lumican affects tumor cell functions, tumor–ECM interactions, angiogenesis and inflammatory response. Matrix Biol 2014; 35:206-14. [DOI: 10.1016/j.matbio.2013.09.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/16/2013] [Accepted: 09/16/2013] [Indexed: 11/17/2022]
|
96
|
Liu Q, Tan Q, Zheng Y, Chen K, Qian C, Li N, Wang Q, Cao X. Blockade of Fas signaling in breast cancer cells suppresses tumor growth and metastasis via disruption of Fas signaling-initiated cancer-related inflammation. J Biol Chem 2014; 289:11522-11535. [PMID: 24627480 DOI: 10.1074/jbc.m113.525014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mechanisms for cancer-related inflammation remain to be fully elucidated. Non-apoptotic functions of Fas signaling have been proposed to play an important role in promoting tumor progression. It has yet to be determined if targeting Fas signaling can control tumor progression through suppression of cancer-related inflammation. In the current study we found that breast cancer cells with constitutive Fas expression were resistant to apoptosis induction by agonistic anti-Fas antibody (Jo2) ligation or Fas ligand cross-linking. Higher expression of Fas in human breast cancer tissue has been significantly correlated with poorer prognosis in breast cancer patients. To determine whether blockade of Fas signaling in breast cancer could suppress tumor progression, we prepared an orthotopic xenograft mouse model with mammary cancer cells 4T1 and found that blockade of Fas signaling in 4T1 cancer cells markedly reduced tumor growth, inhibited tumor metastasis in vivo, and prolonged survival of tumor-bearing mice. Mechanistically, blockade of Fas signaling in cancer cells significantly decreased systemic or local recruitment of myeloid derived suppressor cells (MDSCs) in vivo. Furthermore, blockade of Fas signaling markedly reduced IL-6, prostaglandin E2 production from breast cancer cells by impairing p-p38, and activity of the NFκB pathway. In addition, administration of a COX-2 inhibitor and anti-IL-6 antibody significantly reduced MDSC accumulation in vivo. Therefore, blockade of Fas signaling can suppress breast cancer progression by inhibiting proinflammatory cytokine production and MDSC accumulation, indicating that Fas signaling-initiated cancer-related inflammation in breast cancer cells may be a potential target for treatment of breast cancer.
Collapse
Affiliation(s)
- Qiuyan Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China,.
| | - Qinchun Tan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and
| | - Yuanyuan Zheng
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Kun Chen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and
| | - Cheng Qian
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Nan Li
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China,; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and; National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China.
| |
Collapse
|
97
|
Abstract
AIM: To investigate the effect of casticin on apoptosis of hepatic stellate cells (HSCs) in vitro and the possible mechanisms involved.
METHODS: Rat HSC-T6 cells were cultured in high-glucose DMEM and then treated with different concentrations of casticin (0, 0.5, 1.0, and 2.0 μmol/L) for 12, 24 and 48 h. HSC-T6 apoptosis was identified by flow cytometry (FCM) and agarose gel electrophoresis. The mRNA expression of apoptosis-related genes Fas/FasL and Bcl-2 was examined by RT-PCR. The expression of Caspase3 was studied by immunocytochemical staining assay (SABC).
RESULTS: Casticin treatment significantly increased the apoptosis of HSC-T6 in a dose- and time-dependent manner compared with the control group (P < 0.01). The highest apoptosis rate was observed in HSC-T6 cells treated with 2.0 μmol/L of casticin for 48 h (55.70% ± 5.56%). An oligonucleosomal DNA ladder was demonstrated by SABC, indicating DNA break in HSC-T6 cells. The expression of Fas/FasL mRNA was increased, while expression of Bcl-2 mRNA was reduced. After HSC-T6 cells were treated with casticin at concentrations of 0.5, 1.0, and 2.0 μmol/L for 48 h, the positive rates of Caspase3 protein expression were 12.78% ± 0.74%, 41.00% ± 1.51% and 71.33% ± 2.68%, respectively. Casticin treatment significantly increased Caspase3 protein expression in a dose- and time-dependent manner compared with the control group (P < 0.001).
CONCLUSION: Casticin induced apoptosis of HSC-T6 cells may involve mitochondrial pathways and Bcl-2 family proteins. Casticin might be a potential Chinese medical component for inhibiting liver fibrosis.
Collapse
|
98
|
Villa-Morales M, Cobos MA, González-Gugel E, Álvarez-Iglesias V, Martínez B, Piris MA, Carracedo A, Benítez J, Fernández-Piqueras J. FAS system deregulation in T-cell lymphoblastic lymphoma. Cell Death Dis 2014; 5:e1110. [PMID: 24603338 PMCID: PMC3973220 DOI: 10.1038/cddis.2014.83] [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: 10/16/2013] [Revised: 01/17/2014] [Accepted: 02/04/2014] [Indexed: 11/09/2022]
Abstract
The acquisition of resistance towards FAS-mediated apoptosis may be required for tumor formation. Tumors from various histological origins exhibit FAS mutations, the most frequent being hematological malignancies. However, data regarding FAS mutations or FAS signaling alterations are still lacking in precursor T-cell lymphoblastic lymphomas (T-LBLs). The available data on acute lymphoblastic leukemia, of precursor origin as well, indicate a low frequency of FAS mutations but often report a serious reduction in FAS-mediated apoptosis as well as chemoresistance, thus suggesting the occurrence of mechanisms able to deregulate the FAS signaling pathway, different from FAS mutation. Our aim at this study was to determine whether FAS-mediated apoptotic signaling is compromised in human T-LBL samples and the mechanisms involved. This study on 26 T-LBL samples confirms that the FAS system is impaired to a wide extent in these tumors, with 57.7% of the cases presenting any alteration of the pathway. A variety of mechanisms seems to be involved in such alteration, in order of frequency the downregulation of FAS, the deregulation of other members of the pathway and the occurrence of mutations at FAS. Considering these results together, it seems plausible to think of a cumulative effect of several alterations in each T-LBL, which in turn may result in FAS/FASLG system deregulation. Since defective FAS signaling may render the T-LBL tumor cells resistant to apoptotic cell death, the correct prognosis, diagnosis and thus the success of anticancer therapy may require such an in-depth knowledge of the complete scenario of FAS-signaling alterations.
Collapse
Affiliation(s)
- M Villa-Morales
- 1] Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain [2] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain [3] Instituto de Investigación Sanitario Fundación Jiménez Díaz, ISCIII, Madrid, Spain
| | - M A Cobos
- 1] Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain [2] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain [3] Instituto de Investigación Sanitario Fundación Jiménez Díaz, ISCIII, Madrid, Spain
| | - E González-Gugel
- Musculoskeletal Research Center, NYU Hospital for Joint Diseases, New York, NY, USA
| | - V Álvarez-Iglesias
- Grupo de Medicina Xenómica, CIBERER, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - B Martínez
- 1] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain [2] Instituto de Investigación de Enfermedades Raras, ISCIII, Madrid, Spain
| | - M A Piris
- Hospital Universitario Marqués de Valdecilla, Fundación IFIMAV, Santander, Spain
| | - A Carracedo
- 1] Grupo de Medicina Xenómica, CIBERER, Universidade de Santiago de Compostela, Santiago de Compostela, Spain [2] Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, KSA
| | - J Benítez
- 1] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain [2] Human Genetics Group, CNIO, Madrid, Spain
| | - J Fernández-Piqueras
- 1] Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain [2] Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain [3] Instituto de Investigación Sanitario Fundación Jiménez Díaz, ISCIII, Madrid, Spain
| |
Collapse
|
99
|
Koehler BC, Jäger D, Schulze-Bergkamen H. Targeting cell death signaling in colorectal cancer: Current strategies and future perspectives. World J Gastroenterol 2014; 20:1923-1934. [PMID: 24587670 PMCID: PMC3934462 DOI: 10.3748/wjg.v20.i8.1923] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/06/2013] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
The evasion from controlled cell death induction has been considered as one of the hallmarks of cancer cells. Defects in cell death signaling are a fundamental phenomenon in colorectal cancer. Nearly any non-invasive cancer treatment finally aims to induce cell death. However, apoptosis resistance is the major cause for insufficient therapeutic success and disease relapse in gastrointestinal oncology. Various compounds have been developed and evaluated with the aim to meet with this obstacle by triggering cell death in cancer cells. The aim of this review is to illustrate current approaches and future directions in targeting cell death signaling in colorectal cancer. The complex signaling network of apoptosis will be demonstrated and the “druggability” of targets will be identified. In detail, proteins regulating mitochondrial cell death in colorectal cancer, such as Bcl-2 and survivin, will be discussed with respect to potential therapeutic exploitation. Death receptor signaling and targeting in colorectal cancer will be outlined. Encouraging clinical trials including cell death based targeted therapies for colorectal cancer are under way and will be demonstrated. Our conceptual understanding of cell death in cancer is rapidly emerging and new types of controlled cellular death have been identified. To meet this progress in cell death research, the implication of autophagy and necroptosis for colorectal carcinogenesis and therapeutic approaches will also be depicted. The main focus of this topic highlight will be on the revelation of the complex cell death concepts in colorectal cancer and the bridging from basic research to clinical use.
Collapse
|
100
|
Zhu LB, Zhao ST, Xu TZ, Wang H. Tumor necrosis factor-α-induced a disintegrin and metalloprotease 10 increases apoptosis resistance in prostate cancer cells. Oncol Lett 2014; 7:897-901. [PMID: 24520307 PMCID: PMC3919938 DOI: 10.3892/ol.2014.1810] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/20/2013] [Indexed: 11/28/2022] Open
Abstract
In developed countries, prostate cancer (PCa) is the second most frequently diagnosed type of cancer and the third most common cause of cancer-related mortality in males. Compared with western countries, the morbidity rate of PCa in China is markedly lower, however, it is rising annually. The etiology of PCa is unclear, therefore, to investigate how a disintegrin and metalloprotease 10 (ADAM10) functions in PCa, ADAM10 mRNA and protein levels induced by tumor necrosis factor (TNF)-α were identified using polymerase chain reaction and flow cytometry, respectively. To investigate the mechanism of ADAM10 activity in PCa, specific inhibitors were used, and DNA transfection and RNA interference technology were employed to identify the interaction between ADAM10 and the Fas ligand (L). The results indicated that TNF-α induced ADAM10 expression in a time- and dose-dependent manner through the p38 mitogen-activated protein kinase/necrosis factor-κB signaling pathway. ADAM10 hydrolyzed FasL and contributed to apoptosis resistance of the tumor cells. These observations indicate a promising therapeutic modality for the treatment of apoptosis-resistant PCa, by targeting ADAM10 sheddase activity.
Collapse
Affiliation(s)
- Li Bing Zhu
- Department of Urology, The People's Liberation Army Mount Lu Sanatorium, Jiujiang, Jiangxi 332000, P.R. China ; Department of Urology, Fuzhou General Hospital of Nanjing Military Area Command, Fuzhou, Fujian 350000, P.R. China
| | - Sheng Tao Zhao
- Department of Respiratory Medicine, Kunming General Hospital of Chengdu Military Area Command, Kunming, Yunnan 650000, P.R. China
| | - Ting Zhao Xu
- Department of Urology, Fuzhou General Hospital of Nanjing Military Area Command, Fuzhou, Fujian 350000, P.R. China
| | - He Wang
- Department of Urology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|