401
|
Huang K, Duan N, Zhang C, Mo R, Hua Z. Improved antitumor activity of TRAIL fusion protein via formation of self-assembling nanoparticle. Sci Rep 2017; 7:41904. [PMID: 28225020 PMCID: PMC5320504 DOI: 10.1038/srep41904] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/21/2016] [Indexed: 12/29/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been known as a promising agent for cancer therapy due to its specific apoptosis-inducing effect on tumor cells rather than most normal cells. However, systemically delivered TRAIL suffers from a rapid clearance from the body with an extremely short half-life. Thermally responsive elastin-like polypeptides (ELPs) are a promising class of temperature sensitive biopolymers based on the structural motif found in mammalian tropoelastin and retain the advantages of polymeric drug delivery systems. We therefore expressed RGD-TRAIL fused with ELP (RGD-TRAIL-ELP) in E. coli. Purification of RGD-TRAIL-ELP was achieved by the conveniently inverse transition cycling (ITC). The purified RGD-TRAIL-ELP without any chemical conjugation was able to self-assemble into nanoparticle under physiological condition. Non-reducing SDS-PAGE results showed that trimer content of RGD-TRAIL-ELP increased 3.4-fold than RGD-TRAIL. Flow cytometry confirmed that RGD-TRAIL-ELP 3-fold enhanced apoptosis-inducing capacity than RGD-TRAIL. Single intraperitoneal injection of the RGD-TRAIL-ELP nanoparticle induced nearly complete tumor regression in the COLO-205 tumor xenograft model. Histological observation confirmed that RGD-TRAIL-ELP induced significant tumor cell apoptosis without apparent liver toxicity. These findings suggested that a great potential application of the RGD-TRAIL-ELP nanoparticle system as a safe and efficient delivery strategy for cancer therapy.
Collapse
Affiliation(s)
- Kaizong Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and Affiliated Stomatological Hospital, Nanjing University, Nanjing, Jiangsu 210046, P.R. China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Ningjun Duan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and Affiliated Stomatological Hospital, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Chunmei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and Affiliated Stomatological Hospital, Nanjing University, Nanjing, Jiangsu 210046, P.R. China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Zichun Hua
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences and Affiliated Stomatological Hospital, Nanjing University, Nanjing, Jiangsu 210046, P.R. China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China.,Nanjing Industrial Innovation Center for Pharmaceutical Biotechnology, Nanjing, Jiangsu 210019, P.R. China.,Changzhou High-Tech Research Institute of Nanjing University, Changzhou, Jiangsu 213164, P.R. China
| |
Collapse
|
402
|
Artemisinin and its derivatives in cancer therapy: status of progress, mechanism of action, and future perspectives. Cancer Chemother Pharmacol 2017; 79:451-466. [DOI: 10.1007/s00280-017-3251-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/03/2017] [Indexed: 12/21/2022]
|
403
|
Functional Effects of Prebiotic Fructans in Colon Cancer and Calcium Metabolism in Animal Models. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9758982. [PMID: 28293641 PMCID: PMC5331302 DOI: 10.1155/2017/9758982] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/27/2016] [Accepted: 01/11/2017] [Indexed: 12/26/2022]
Abstract
Inulin-type fructans are polymers of fructose molecules and are known for their capacity to enhance absorption of calcium and magnesium, to modulate gut microbiota and energy metabolism, and to improve glycemia. We evaluated and compared the effects of Chicory inulin “Synergy 1®” and inulin from Mexican agave “Metlin®” in two experimental models of colon cancer and bone calcium metabolism in mice and rats. Inulins inhibited the development of dextran sulfate sodium-induced colitis and colon cancer in mice; these fructans reduced the concentration of tumor necrosis factor alpha and prevented the formation of intestinal polyps, villous atrophy, and lymphoid hyperplasia. On the other hand, inulin treatments significantly increased bone densitometry (femur and vertebra) in ovariectomized rats without altering the concentration of many serum biochemical parameters and urinary parameters. Histopathology results were compared between different experimental groups. There were no apparent histological changes in rats treated with inulins and a mixture of inulins-isoflavones. Our results showed that inulin-type fructans have health-promoting properties related to enhanced calcium absorption, potential anticancer properties, and anti-inflammatory effects. The use of inulin as a prebiotic can improve health and prevent development of chronic diseases such as cancer and osteoporosis.
Collapse
|
404
|
Iacopetta D, Carocci A, Sinicropi MS, Catalano A, Lentini G, Ceramella J, Curcio R, Caroleo MC. Old Drug Scaffold, New Activity: Thalidomide-Correlated Compounds Exert Different Effects on Breast Cancer Cell Growth and Progression. ChemMedChem 2017; 12:381-389. [PMID: 28099781 DOI: 10.1002/cmdc.201600629] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/16/2017] [Indexed: 12/17/2022]
Abstract
Thalidomide was first used for relief of morning sickness in pregnant women and then withdrawn from the market because of its dramatic effects on normal fetal development. Over the last decades, it has been used successfully for the treatment of several pathologies, including cancer. Many analogues with improved activity have been synthesized and tested. Herein we report some effects on the growth and progression of MCF-7 and MDA-MB-231 breast cancer cells by a small series of thalidomide-correlated compounds, which are very effective at inducing cancer cell death by triggering TNFα-mediated apoptosis. The most active compounds are able to drastically reduce the migration of breast cancer cells by regulation of the two major proteins involved in epithelial-mesenchymal transition (EMT): vimentin and E-cadherin. Moreover, these compounds diminish the intracellular biosynthesis of vascular endothelial growth factor (VEGF), which is primarily involved in the promotion of angiogenesis, sustaining tumor progression. The multiple features of these compounds that act on various key points of the tumorigenesis process make them good candidates for preclinical studies.
Collapse
Affiliation(s)
- Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Alessia Carocci
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126, Bari, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126, Bari, Italy
| | - Giovanni Lentini
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126, Bari, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Rosita Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| | - Maria Cristina Caroleo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Italy
| |
Collapse
|
405
|
Bianconi D, Unseld M, Prager GW. Integrins in the Spotlight of Cancer. Int J Mol Sci 2016; 17:ijms17122037. [PMID: 27929432 PMCID: PMC5187837 DOI: 10.3390/ijms17122037] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/17/2016] [Accepted: 11/28/2016] [Indexed: 02/07/2023] Open
Abstract
Integrins are heterodimeric cell surface receptors that bind to different extracellular ligands depending on their composition and regulate all processes which enable multicellular life. In cancer, integrins trigger and play key roles in all the features that were once described as the Hallmarks of Cancer. In this review, we will discuss the contribution of integrins to these hallmarks, including uncontrolled and limitless proliferation, invasion of tumor cells, promotion of tumor angiogenesis and evasion of apoptosis and resistance to growth suppressors, by highlighting the latest findings. Further on, given the paramount role of integrins in cancer, we will present novel strategies for integrin inhibition that are starting to emerge, promising a hopeful future regarding cancer treatment.
Collapse
Affiliation(s)
- Daniela Bianconi
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Matthias Unseld
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Gerald W Prager
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
406
|
Singh PR, Priya ES, Balakrishnan S, Arunkumar R, Sharmila G, Rajalakshmi M, Arunakaran J. Nimbolide inhibits androgen independent prostate cancer cells survival and proliferation by modulating multiple pro-survival signaling pathways. Biomed Pharmacother 2016; 84:1623-1634. [DOI: 10.1016/j.biopha.2016.10.076] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/23/2016] [Accepted: 10/24/2016] [Indexed: 10/20/2022] Open
|
407
|
Jemaa AB, Bouraoui Y, Rais NB, Nouira Y, Oueslati R. Cytokine profiling identifies an interaction of IL-6 and IL-1α to drive PSMA-PSA prostate clones. Immunobiology 2016; 221:1424-1431. [DOI: 10.1016/j.imbio.2016.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 07/13/2016] [Indexed: 01/06/2023]
|
408
|
Divella R, De Luca R, Abbate I, Naglieri E, Daniele A. Obesity and cancer: the role of adipose tissue and adipo-cytokines-induced chronic inflammation. J Cancer 2016; 7:2346-2359. [PMID: 27994674 PMCID: PMC5166547 DOI: 10.7150/jca.16884] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue in addition to its ability to keep lipids is now recognized as a real organ with both metabolic and endocrine functions. Recent studies demonstrated that in obese animals is established a status of adipocyte hypoxia and in this hypoxic state interaction between adipocytes and stromal vascular cells contribute to tumor development and progression. In several tumors such as breast, colon, liver and prostate, obesity represents a poor predictor of clinical outcomes. Dysfunctional adipose tissue in obesity releases a disturbed profile of adipokines with elevated levels of pro-inflammatory factors and a consequent alteration of key signaling mediators which may be an active local player in establishing the peritumoral environment promoting tumor growth and progression. Therefore, adipose tissue hypoxia might contribute to cancer risk in the obese population. To date the precise mechanisms behind this obesity-cancer link is not yet fully understood. In the light of information provided in this review that aims to identify the key mechanisms underlying the link between obesity and cancer we support that inflammatory state specific of obesity may be important in obesity-cancer link.
Collapse
Affiliation(s)
- Rosa Divella
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Raffaele De Luca
- Department of Surgery Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Ines Abbate
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Emanuele Naglieri
- Department of Medical Oncology, Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| | - Antonella Daniele
- Clinical Pathology Laboratory, Department of Experimental Oncology. Giovanni Paolo II National Cancer Institute, V.Le Orazio Flacco 65, 70124 -Bari, Italy
| |
Collapse
|
409
|
Pawlikowska M, Jędrzejewski T, Piotrowski J, Kozak W. Fever-range hyperthermia inhibits cells immune response to protein-bound polysaccharides derived from Coriolus versicolor extract. Mol Immunol 2016; 80:50-57. [PMID: 27825050 DOI: 10.1016/j.molimm.2016.10.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/07/2016] [Accepted: 10/27/2016] [Indexed: 01/15/2023]
Abstract
The aim of the study was to explore whether fever-range hyperthermia (FRH) might enhance the anticancer and immunoregulatory activities of protein-bound polysaccharides (PBP), a class of fungus derived immunomodifiers used in the cancer adjuvant therapy. Blood lymphocytes and breast cancer cells (MCF-7) were cultured at 39.5°C in humidified atmosphere containing 5% CO2 for 2h. After rested at 37°C for 6h, the cells were treated with PBP extract at 100- and 300μg/ml concentration. After indicated time, the proliferative response was analyzed and cytokine mRNA expression assessment was performed by qRT-PCR. In animal model, the FRH was induced by placing rats in the Homeothermic Controller with heating blanket. Animals were heated until Tb reached 39.5°C (±0.2°C) and were maintained at this temperature for 30min. The protein-bound polysaccharides solution was injected i.p. at a dose of 100 mg/kg 6h post FRH. Twenty four hours after treatment, the blood was collected and cytokines expression analysis were performed. The results have shown that fever-range hyperthermia has an inhibitory effect on PBP extract-induced proliferative response of blood lymphocytes, as well as IL-1β and IL-6 mRNA expression. Moreover, the temperature of 39.5°C blocks PBP-induced cytotoxicity against MCF-7 cells, which correlates with significant reduction in TNF-α level. Combined treatment of rats (FRH+PBP) results in decrease of IL-1β, IL-6 and TNF-α mRNA expression in peripheral blood mononuclear cells compared to cells derived from rats treated with protein-bound polysaccharides extract alone. This study demonstrates that fever-range temperature inhibits immunostimulatory as well as anticancer effects mediated by protein-bound polysaccharides.
Collapse
Affiliation(s)
- Małgorzata Pawlikowska
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, Lwowska 1 Street, 87-100 Torun, Poland.
| | - Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, Lwowska 1 Street, 87-100 Torun, Poland.
| | - Jakub Piotrowski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, Lwowska 1 Street, 87-100 Torun, Poland.
| | - Wiesław Kozak
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, Lwowska 1 Street, 87-100 Torun, Poland.
| |
Collapse
|
410
|
Khan MI, Czarnecka AM, Lewicki S, Helbrecht I, Brodaczewska K, Koch I, Zdanowski R, Król M, Szczylik C. Comparative Gene Expression Profiling of Primary and Metastatic Renal Cell Carcinoma Stem Cell-Like Cancer Cells. PLoS One 2016; 11:e0165718. [PMID: 27812180 PMCID: PMC5094751 DOI: 10.1371/journal.pone.0165718] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/17/2016] [Indexed: 11/22/2022] Open
Abstract
Background Recent advancement in cancer research has shown that tumors are highly heterogeneous, and multiple phenotypically different cell populations are found in a single tumor. Cancer development and tumor growth are driven by specific types of cells—stem cell-like cancer cells (SCLCCs)—which are also responsible for metastatic spread and drug resistance. This research was designed to verify the presence of SCLCCs in renal cell cancer cell lines. Subsequently, we aimed to characterize phenotype and cell biology of CD105+ cells, defined previously as renal cell carcinoma tumor-initiating cells. The main goal of the project was to describe the gene-expression profile of stem cell-like cancer cells of primary tumor and metastatic origin. Materials and Methods Real-time PCR analysis of stemness genes (Oct-4, Nanog and Ncam) and soft agar colony formation assay were conducted to check the stemness properties of renal cell carcinoma (RCC) cell lines. FACS analysis of CD105+ and CD133+ cells was performed on RCC cells. Isolated CD105+ cells were verified for expression of mesenchymal markers—CD24, CD146, CD90, CD73, CD44, CD11b, CD19, CD34, CD45, HLA-DR and alkaline phosphatase. Hanging drop assay was used to investigate CD105+ cell-cell cohesion. Analysis of free-floating 3D spheres formed by isolated CD105+ was verified, as spheres have been hypothesized to contain undifferentiated multipotent progenitor cells. Finally, CD105+ cells were sorted from primary (Caki-2) and metastatic (ACHN) renal cell cancer cell lines. Gene-expression profiling of sorted CD105+ cells was performed with Agilent’s human GE 4x44K v2 microarrays. Differentially expressed genes were further categorized into canonical pathways. Network analysis and downstream analysis were performed with Ingenuity Pathway Analysis. Results Metastatic RCC cell lines (ACHN and Caki-1) demonstrated higher colony-forming ability in comparison to primary RCC cell lines. Metastatic RCC cell lines harbor numerous CD105+ cell subpopulations and have higher expression of stemness genes (Oct-4 and Nanog). CD105+ cells adopt 3D grape-like floating structures under handing drop conditions. Sorted CD105+ cells are positive for human mesenchymal stem cell (MSC) markers CD90, CD73, CD44, CD146, and alkaline phosphatase activity, but not for CD24 and hematopoietic lineage markers CD34, CD11b, CD19, CD45, and HLA-DR. 1411 genes are commonly differentially expressed in CD105+ cells (both from primary [Caki-2] and metastatic RCC [ACHN] cells) in comparison to a healthy kidney epithelial cell line (ASE-5063). TGF-β, Wnt/β-catenine, epithelial-mesenchymal transition (EMT), Rap1 signaling, PI3K-Akt signaling, and Hippo signaling pathway are deregulated in CD105+ cells. TGFB1, ERBB2, and TNF are the most significant transcriptional regulators activated in these cells. Conclusions All together, RCC-CD105+ cells present stemlike properties. These stem cell-like cancer cells may represent a novel target for therapy. A unique gene-expression profile of CD105+ cells could be used as initial data for subsequent functional studies and drug design.
Collapse
Affiliation(s)
- Mohammed I. Khan
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland
- * E-mail: (MIK); (AMC)
| | - Anna M. Czarnecka
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland
- * E-mail: (MIK); (AMC)
| | - Sławomir Lewicki
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Igor Helbrecht
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland
- Institute of Genetics and Biotechnology, Faculty of Biology, Warsaw University, Warsaw, Poland
| | - Klaudia Brodaczewska
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Irena Koch
- Department of Pathomorphology, Institute of Mother and Child, Warsaw, Poland
| | - Robert Zdanowski
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Magdalena Król
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences—WULS, Warsaw, Poland
| | - Cezary Szczylik
- Molecular Oncology Laboratory, Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
411
|
Munge BS, Stracensky T, Gamez K, DiBiase D, Rusling JF. Multiplex Immunosensor Arrays for Electrochemical Detection of Cancer Biomarker Proteins. ELECTROANAL 2016; 28:2644-2658. [PMID: 28592919 PMCID: PMC5459496 DOI: 10.1002/elan.201600183] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/03/2016] [Indexed: 01/22/2023]
Abstract
Measuring panels of protein biomarkers offer a new personalized approach to early cancer detection, disease monitoring and patients' response to therapy. Multiplex electrochemical methods are uniquely positioned to provide faster, more sensitive, point of care (POC) devices to detect protein biomarkers for clinical diagnosis. Nanomaterials-based electrochemical methods offer sensitivity needed for early cancer detection. This review discusses recent advances in multiplex electrochemical immunosensors for cancer diagnostics and disease monitoring. Different electrochemical strategies including enzyme-based immunoarrays, nanoparticle-based immunoarrays and electrochemiluminescence methods are discussed. Many of these methods have been integrated into microfluidic systems, but measurement of more than 2-4 protein markers in a small single serum sample is still a challenge. For POC applications, a simple, low cost method is required. Major challenges in multiplexed microfluidic immunoassays are reagent additions and washing steps that require creative engineering solutions. 3-D printed microfluidics and paper-based microfluidic devices are also explored.
Collapse
Affiliation(s)
- Bernard S Munge
- Department of Chemistry, Salve Regina University, 100 Ochre Point Avenue, Newport RI 02840, USA
| | - Thomas Stracensky
- Department of Chemistry, Salve Regina University, 100 Ochre Point Avenue, Newport RI 02840, USA
| | - Kathleen Gamez
- Department of Chemistry, Salve Regina University, 100 Ochre Point Avenue, Newport RI 02840, USA
| | - Dimitri DiBiase
- Department of Chemistry, Salve Regina University, 100 Ochre Point Avenue, Newport RI 02840, USA
| | - James F Rusling
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269-3136, USA
- Department of Surgery and Neag Cancer Center, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
- School of Chemistry, National University of Ireland at Galway, Galway, Ireland
| |
Collapse
|
412
|
Li M, Luo Z, Zhu W, Khan RSU, Ummair SU, Shi S. Diagnostic accuracy of tumor necrosis factor-alpha assay for tuberculous pleurisy: A PRISMA-compliant meta-analysis. Medicine (Baltimore) 2016; 95:e5510. [PMID: 27902616 PMCID: PMC5134804 DOI: 10.1097/md.0000000000005510] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The diagnosis of tuberculous pleurisy is difficult and traditional methods are not always helpful. Many studies have focused on the tumor necrosis factor-alpha (TNF-α) assay in pleural effusion for the diagnosis of tuberculous pleurisy, but the results remain controversial. This meta-analysis was conducted to determine the overall diagnostic accuracy of TNF-α. METHODS Relevant studies were searched from PubMed, Embase, Web of Science, Chinese National Knowledge Infrastructure (CNKI), Wangfang, and Weipu. We pooled the published results and computed the accuracy measures, including sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), and diagnostic odds ratio (DOR). Receiver operating characteristic curves (SROC) and the area under the curve (AUC) were used to summarize the overall test performance. RESULTS Twelve studies with 1022 patients met the inclusion criteria. The pooled sensitivity and specificity were 0.85 (95%CI, 0.81-0.89) and 0.80 (95% CI, 0.77-0.83) respectively. The area under the SROC curve was 0.89. CONCLUSIONS The results of meta-analysis suggested that the TNF-α assay plays a vital role in the diagnosis of tuberculous pleurisy, whereas other test results or clinical findings should be interpreted together with the TNF-α assay to improve the overall diagnostic accuracy.
Collapse
Affiliation(s)
- Min Li
- Department of Respiratory Medicine
| | | | - Wenye Zhu
- Department of Pharmacy, First Affiliated Hospital of Kunming Medical University, Kunming
| | | | - Saeed Ummai Ummair
- The Postgraduate School of Kunming Medical University, Kunming, Yunnan, China
| | | |
Collapse
|
413
|
Taghavi Pourianazar N, Gunduz U. Changes in apoptosis-related gene expression and cytokine release in breast cancer cells treated with CpG-loaded magnetic PAMAM nanoparticles. Int J Pharm 2016; 515:11-19. [PMID: 27717915 DOI: 10.1016/j.ijpharm.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/28/2016] [Accepted: 10/01/2016] [Indexed: 02/07/2023]
Abstract
CpG-oligodeoxynucleotide (CpG-ODN) can function as an immune adjuvant. Previously, we showed that stimulation of breast cancer cells with CpG-ODN conjugated with PAMAM dendrimer-coated magnetic nanoparticles (DcMNPs) has induced apoptosis. The aim of the current study was to evaluate the expression levels of some apoptosis-regulating genes in several human breast cancer cells treated with CpG/DcMNPs. Treated MDA-MB231 cells showed an increase in Noxa and Bax gene expression levels, whereas the expression level of Survivin decreased. Similarly, Noxa gene was overexpressed in treated MCF7 cells. In treated SKBR3 cells, a decline in the c-Flip mRNA level was determined. Furthermore, release of cytokines, IL-6, IL-10, and TNF-α, was determined in cell culture supernatants. CpG/DcMNP treatment leads to an increase in the release of IL-6 in MDA-MB231 and SKBR3 cells, whereas release of IL-10 and TNF-α did not change significantly. It is indicated that CpG-ODN may show its cytotoxic effect by regulating the expression of apoptosis-related genes and the release of cytokine in breast cancer cells.
Collapse
Affiliation(s)
| | - Ufuk Gunduz
- Middle East Technical University, Department of Biotechnology, 06800, Ankara, Turkey; Middle East Technical University, Department of Biological Sciences, 06800, Ankara, Turkey
| |
Collapse
|
414
|
Kim HS, Kacew S, Lee BM. Genetic and epigenetic cancer chemoprevention on molecular targets during multistage carcinogenesis. Arch Toxicol 2016; 90:2389-2404. [PMID: 27538406 DOI: 10.1007/s00204-016-1813-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/04/2016] [Indexed: 12/16/2022]
Abstract
The main goal of cancer chemoprevention is to prevent or halt the progression of carcinogenesis with the administration of synthetic or natural compounds. Fundamental chemopreventive strategies include inhibition of genetic damage, anti-proliferation/cell cycle regulation, and induction of apoptosis and anti-inflammatory processes, which may be critical for carcinogenesis intervention. Recently, a new paradigm for identifying chemopreventive agents has been implemented. It focuses on defining new biomarkers that can be used to evaluate chemopreventive efficacy based on multistage carcinogenesis. The functional roles of chemopreventive agents are associated with the modulation of nuclear factor kappa B, nuclear factor erythroid 2-related factor, p53, AMPK/mTOR, phosphatidylinositol 3-kinase, epidermal growth factor receptor, cyclooxygenase-2, chemokine (C-X-C motif) receptor 2, and sphingosine-1-phosphate. This paper summarizes the genetic and epigenetic effects of chemopreventive agents on the expression of cancer-related target genes mediated by epigenetic alterations, such as DNA methylation and histone modifications. This review will provide unique and effective strategies for reducing cancer and aging-related diseases in humans.
Collapse
Affiliation(s)
- Hyung Sik Kim
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi-Do, 440-746, Republic of Korea
| | - Sam Kacew
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON, Canada
| | - Byung Mu Lee
- Division of Toxicology, College of Pharmacy, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi-Do, 440-746, Republic of Korea.
| |
Collapse
|
415
|
Systematic tracking of coordinated differential network motifs identifies novel disease-related genes by integrating multiple data. Neurocomputing 2016. [DOI: 10.1016/j.neucom.2015.12.120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
416
|
Lim YC, Rajabalaya R, Lee SHF, Tennakoon KU, Le QV, Idris A, Zulkipli IN, Keasberry N, David SR. Parasitic Mistletoes of the Genera Scurrula and Viscum: From Bench to Bedside. Molecules 2016; 21:1048. [PMID: 27548121 PMCID: PMC6273404 DOI: 10.3390/molecules21081048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/31/2016] [Accepted: 08/04/2016] [Indexed: 01/16/2023] Open
Abstract
The mistletoes, stem hemiparasites of Asia and Europe, have been used as medicinal herbs for many years and possess sophisticated systems to obtain nutrients from their host plants. Although knowledge about ethnomedicinal uses of mistletoes is prevalent in Asia, systematic scientific study of these plants is still lacking, unlike its European counterparts. This review aims to evaluate the literature on Scurrula and Viscum mistletoes. Both mistletoes were found to have anticancer, antimicrobial, antioxidant and antihypertensive properties. Plants from the genus Scurrula were found to inhibit cancer growth due to presence of phytoconstituents such as quercetin and fatty acid chains. Similar to plants from the genus Viscum, Scurrula also possesses TNFα activity to strengthen the immune system to combat cancer. In line with its anticancer activity, both mistletoes are rich in antioxidants that confer protection against cancer as well as neurodegeneration. Extracts from plants of both genera showed evidence of vasodilation and thus, antihypertensive effects. Other therapeutic effects such as weight loss, postpartum and gastrointestinal healing from different plants of the genus Scurrula are documented. As the therapeutic effects of plants from Scurrula are still in exploration stage, there is currently no known clinical trial on these plants. However, there are few on-going clinical trials for Viscum album that demonstrate the functionalities of these mistletoes. Future work required for exploring the benefits of these plants and ways to develop both parasitic plants as a source of pharmacological drug are explained in this article.
Collapse
Affiliation(s)
- Ya Chee Lim
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Rajan Rajabalaya
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Shirley Huan Fang Lee
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Kushan U Tennakoon
- Institute for Biodiversity and Environmental Research (IBER), Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Quang-Vuong Le
- Institute for Biodiversity and Environmental Research (IBER), Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
- Biology Faculty, Vinh University, 182 Le Duan, 470000 Vinh City, Nghe An Province, Vietnam.
| | - Adi Idris
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Ihsan N Zulkipli
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Natasha Keasberry
- Faculty of Science, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Sheba R David
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| |
Collapse
|
417
|
El-Mesery M, Shaker ME, Elgaml A. The SMAC mimetic BV6 induces cell death and sensitizes different cell lines to TNF-α and TRAIL-induced apoptosis. Exp Biol Med (Maywood) 2016; 241:2015-2022. [PMID: 27465142 DOI: 10.1177/1535370216661779] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The inhibitors of apoptosis proteins are implicated in promoting cancer cells survival and resistance toward immune surveillance and chemotherapy. Second mitochondria-derived activator of caspases (SMAC) mimetics are novel compounds developed to mimic the inhibitory effect of the endogenous SMAC/DIABLO on these IAPs. Here, we examined the potential effects of the novel SMAC mimetic BV6 on different human cancer cell lines. Our results indicated that BV6 was able to induce cell death in different human cancer cell lines. Mechanistically, BV6 dose dependently induced degradation of IAPs, including cIAP1 and cIAP2. This was coincided with activating the non-canonical NF -kappa B (NF-κB) pathway, as indicated by stabilizing NF-κB-inducing kinase (NIK) for p100 processing to p52. More interestingly, BV6 was able to sensitize some of the resistant cancer cell lines to apoptosis induced by the death ligands tumor necrosis factor-α (TNF-α) and TNF-related apoptosis-inducing ligand (TRAIL) that are produced by different cells of the immune system. Such cell death enhancement was mediated by inducing an additional cleavage of caspase-9 to augment that of caspase-8 induced by death ligands. This eventually led to more processing of the executioner caspase-3 and poly (ADP-ribose) polymerase (PARP). In conclusion, therapeutic targeting of IAPs by BV6 might be an effective approach to enhance cancer regression induced by immune system. Our data also open up the future possibility of using BV6 in combination with other antitumor therapies to overcome cancer drug resistance.
Collapse
Affiliation(s)
- Mohamed El-Mesery
- 1 Faculty of Pharmacy, Department of Biochemistry, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed E Shaker
- 2 Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura 35516, Egypt
| | - Abdelaziz Elgaml
- 3 Faculty of Pharmacy, Department of Microbiology and Immunology, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
418
|
Laocharoensuk R. Development of Electrochemical Immunosensors towards Point-of-care Cancer Diagnostics: Clinically Relevant Studies. ELECTROANAL 2016. [DOI: 10.1002/elan.201600248] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Rawiwan Laocharoensuk
- National Nanotechnology Center (NANOTEC); National Science and Technology Development Agency (NSTDA); Pathum Thani 12120 Thailand
| |
Collapse
|
419
|
Panday A, Inda ME, Bagam P, Sahoo MK, Osorio D, Batra S. Transcription Factor NF-κB: An Update on Intervention Strategies. Arch Immunol Ther Exp (Warsz) 2016; 64:463-483. [PMID: 27236331 DOI: 10.1007/s00005-016-0405-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/14/2016] [Indexed: 12/25/2022]
Abstract
The nuclear factor (NF)-κB family of transcription factors are ubiquitous and pleiotropic molecules that regulate the expression of more than 150 genes involved in a broad range of processes including inflammation, immunity, cell proliferation, differentiation, and survival. The chronic activation or dysregulation of NF-κB signaling is the central cause of pathogenesis in many disease conditions and, therefore, NF-κB is a major focus of therapeutic intervention. Because of this, understanding the relationship between NF-κB and the induction of various downstream signaling molecules is imperative. In this review, we provide an updated synopsis of the role of NF-κB in DNA repair and in various ailments including cardiovascular diseases, HIV infection, asthma, herpes simplex virus infection, chronic obstructive pulmonary disease, and cancer. Furthermore, we also discuss the specific targets for selective inhibitors and future therapeutic strategies.
Collapse
Affiliation(s)
- Arvind Panday
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.,Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Maria Eugenia Inda
- Departamento de Microbiología, CONICET, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional Rosario, Suipacha 531, Santa Fe, Argentina
| | - Prathyusha Bagam
- Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology PhD Program, 207 Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA
| | - Malaya K Sahoo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94304, USA
| | - Diana Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Sanjay Batra
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA. .,Laboratory of Pulmonary Immunotoxicology, Environmental Toxicology PhD Program, 207 Health Research Center, Southern University and A&M College, Baton Rouge, LA, 70813, USA.
| |
Collapse
|
420
|
Roy U. Structural biology of tumor necrosis factor demonstrated for undergraduates instruction by computer simulation. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 44:246-255. [PMID: 26638199 DOI: 10.1002/bmb.20937] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 08/10/2015] [Accepted: 10/25/2015] [Indexed: 06/05/2023]
Abstract
This work presents a three-dimensional (3D) modeling exercise for undergraduate students in chemistry and health sciences disciplines, focusing on a protein-group linked to immune system regulation. Specifically, the exercise involves molecular modeling and structural analysis of tumor necrosis factor (TNF) proteins, both wild type and mutant. The structure of the tumor necrosis factor type 1 receptor (TNF-R1) is also briefly explored. TNF and TNF-R1 play major roles in maintaining human immune-system homeostasis. Upon binding with TNFR-1, the TNF can activate the nuclear factor kappa B (NF-κB), eventually resulting in apoptosis or cell death. These essential features of the clinically relevant TNF family is explored within the frame work of a readily adaptable tutorial. © 2015 by The International Union of Biochemistry and Molecular Biology, 44:246-255, 2016.
Collapse
Affiliation(s)
- Urmi Roy
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York, 13699-5820
| |
Collapse
|
421
|
Polysaccharide peptides from Coriolus versicolor exert differential immunomodulatory effects on blood lymphocytes and breast cancer cell line MCF-7 in vitro. Immunol Lett 2016; 174:37-44. [PMID: 27091479 DOI: 10.1016/j.imlet.2016.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/05/2016] [Accepted: 04/13/2016] [Indexed: 01/31/2023]
Abstract
The protein-bound polysaccharides (PBP), isolated from Coriolus versicolor (CV) fungus, are considered as natural compounds with potential therapeutic applications. The immunopotentiating and antitumor activity of polysaccharopeptides has been previously examined, however similar findings could not be achieved. The source of PBP, variations in extraction process as well as environmental factors seems to affect the biological properties of these active CV components. Since further analysis are needed to draw more definite conclusion, the present study aimed to investigate the immunomodulatory properties of the PBP extract, isolated from commercially available capsules of C. versicolor. Our results revealed that the effect mediated by PBP extract depends on the target cells. We reported that the polysaccharopeptides induced a significant decrease in breast cancer MCF-7 cells growth, which was TNF-α-dependent phenomenon. Interestingly, the level of two others cytokines, IL-1β and IL-6 was not affected. On the other hand, in this study we noticed that protein-bound polysaccharides extracted from CV significantly augmented the proliferative response of blood lymphocytes in a time-dependent manner, which was associated with IL-6 and IL-1β mRNA upregulation. Moreover we found that the cells response to PBP stimuli might be inversely related to its concentration.
Collapse
|
422
|
LIU CHANGYING, ZHAO PENGFEI, YANG YUBAO, XU XIAODONG, WANG LIANG, LI BO. Ampelopsin suppresses TNF-α-induced migration and invasion of U2OS osteosarcoma cells. Mol Med Rep 2016; 13:4729-36. [DOI: 10.3892/mmr.2016.5124] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 02/16/2016] [Indexed: 11/05/2022] Open
|
423
|
Fancourt D, Williamon A, Carvalho LA, Steptoe A, Dow R, Lewis I. Singing modulates mood, stress, cortisol, cytokine and neuropeptide activity in cancer patients and carers. Ecancermedicalscience 2016; 10:631. [PMID: 27170831 PMCID: PMC4854222 DOI: 10.3332/ecancer.2016.631] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Indexed: 11/06/2022] Open
Abstract
There is growing evidence that psychosocial interventions can have psychological benefits for people affected by cancer, including improved symptoms of mental health and wellbeing and optimised immune responses. However, despite growing numbers of music interventions, particularly singing, in cancer care, there is less research into their impact. We carried out a multicentre single-arm preliminary study to assess the impact of singing on mood, stress and immune response in three populations affected by cancer: carers (n = 72), bereaved carers (n = 66) and patients (n = 55). Participants were excluded if pregnant or if they were currently being treated with chemotherapy, radiotherapy or oral immunosuppressive drugs. Participants were regular participants in five choirs across South Wales and took part in one hour of group singing. Before and after singing, visual analogue mood scales, stress scales and saliva samples testing for cortisol, beta-endorphin, oxytocin and ten cytokines were taken. Across all five centres and in all four participant groups, singing was associated with significant reductions in negative affect and increases in positive affect (p < .01) alongside significant increases in cytokines including GM-CSF, IL17, IL2, IL4 and sIL-2rα (all p < .01). In addition, singing was associated with reductions in cortisol, beta-endorphin and oxytocin levels. This study provides preliminary evidence that singing improves mood state and modulates components of the immune system. Further work is needed to ascertain how this differs for more specific patient groups and whether repeat exposure could lead to meaningful, longitudinal effects.
Collapse
Affiliation(s)
- Daisy Fancourt
- Centre for Performance Science, Royal College of Music, London SW7 2BS, UK
- Faculty of Medicine, Imperial College, London SW7 2AZ, UK
- Psychobiology Group, Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK
| | - Aaron Williamon
- Centre for Performance Science, Royal College of Music, London SW7 2BS, UK
- Faculty of Medicine, Imperial College, London SW7 2AZ, UK
| | - Livia A Carvalho
- Psychobiology Group, Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK
| | - Andrew Steptoe
- Psychobiology Group, Department of Epidemiology and Public Health, UCL, London WC1E 6BT, UK
| | - Rosie Dow
- Tenovus Cancer Care, Gleider House, Ty-Glas Rd, Cardiff CF14 5BD, Wales, UK
| | - Ian Lewis
- Tenovus Cancer Care, Gleider House, Ty-Glas Rd, Cardiff CF14 5BD, Wales, UK
| |
Collapse
|
424
|
Tavakolpour S, Sali S. Tumor Necrosis Factor-α-308 G/A Polymorphisms and Risk of Hepatocellular Carcinoma: A Meta-Analysis. HEPATITIS MONTHLY 2016; 16:e33537. [PMID: 27257425 PMCID: PMC4888758 DOI: 10.5812/hepatmon.33537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 03/14/2016] [Indexed: 12/11/2022]
Abstract
CONTEXT Hepatocellular carcinoma (HCC) is a common disorder throughout the world that can develop due to various factors, including genetics. Tumor necrosis factor-α (TNF-α) is the most frequently studied cytokine related to the risk of developing HCC, and an association between the 308 position of the TNF-α promoter (TNF-α-308) and HCC risk has been confirmed in various reports. EVIDENCE ACQUISITION The PubMed, Scopus, and Google Scholar databases were searched through July 12, 2015, for studies on associations between TNF-α-308 and the risk of HCC. To determine this association, odds ratios (ORs) and 95% confidence intervals (95% CIs) were calculated. RESULTS A total of 23 case-control studies were investigated, involving 3,389 cases and 4,235 controls. The overall conclusion was that the A allele was more frequent in case groups compared to control groups (13.4% vs. 8.4%). Thus, the A allele was significantly associated with increased HCC risk (OR = 1.77; 95% CI = [1.26-2.50]; P value < 0.002). In addition to the allelic model, the dominant model (AA + AG vs. GG) was significantly associated with HCC risk (OR = 1.80; CI = [1.29-2.51]; P value < 0.001). In the sensitivity analysis for co-dominant (AA vs. GG) and recessive models (AA vs. AG + GG), no trustworthy associations with the risk of HCC development were observed. CONCLUSIONS This meta-analysis indicated that the TNF-α-308 G/A polymorphism is significantly associated with increased susceptibility to HCC. However, to confirm this finding, more studies are needed on TNF-α-308 G/A polymorphisms associated with HCC.
Collapse
Affiliation(s)
- Soheil Tavakolpour
- Iran University of Medical Sciences, Tehran, IR Iran
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Shahnaz Sali
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| |
Collapse
|
425
|
Kabel AM, Elkhoely AA. Ameliorative potential of fluoxetine/raloxifene combination on experimentally induced breast cancer. Tissue Cell 2016; 48:89-95. [PMID: 26881735 DOI: 10.1016/j.tice.2016.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 01/29/2016] [Accepted: 02/03/2016] [Indexed: 02/06/2023]
Abstract
Breast cancer is one of the most common types of malignancies in females worldwide. Targeting the estrogen receptors alone with raloxifene (RAL) reduces the incidence of estrogen receptor positive tumors. Fluoxetine (FLX) is one of selective serotonin reuptake inhibitors that was proven to have anticancer properties. Our aim was to detect the effects of RAL/FLX combination on experimentally induced breast cancer. Eighty female Wistar rats were divided into four equal groups: 7,12-Dimethyl Benzanthracene (DMBA) induced breast cancer group, DMBA+RAL, DMBA+FLX and DMBA+RAL+FLX. Tumor volume, tissue malondialdehyde (MDA), catalase (CAT), superoxide dismutase (SOD), tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6) and transforming growth factor beta1 (TGF-β1) were determined in the tumor tissues. Parts of the tumor were subjected to histopathological examination. RAL or FLX alone or in combination induced significant increase in tumor CAT and SOD with significant decrease in tumor volume, tissue MDA, TNF-α, IL-6 and TGF-β1 and alleviated the histopathological and immunohistochemical changes compared to DMBA group. In conclusion, RAL/FLX combination had a better effect than each of RAL or FLX alone against DMBA-induced breast cancer in rats which may represent a new therapeutic modality for management of breast cancer.
Collapse
Affiliation(s)
- Ahmed M Kabel
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt; Clinical Pharmacy Department, College of Pharmacy, Taif University, Taif, Saudi Arabia.
| | - Abeer A Elkhoely
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Egypt
| |
Collapse
|
426
|
Yamamoto N, Yamasaki T, Takami T, Uchida K, Fujisawa K, Matsumoto T, Saeki I, Terai S, Sakaida I. Deferasirox, an oral iron chelator, prevents hepatocarcinogenesis and adverse effects of sorafenib. J Clin Biochem Nutr 2016; 58:202-9. [PMID: 27257345 PMCID: PMC4865599 DOI: 10.3164/jcbn.15-127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 10/02/2015] [Indexed: 12/18/2022] Open
Abstract
Although sorafenib is expected to have a chemopreventive effect on hepatocellular carcinoma (HCC) recurrence, there are limitations to its use because of adverse effects, including effects on liver function. We have reported that the iron chelator, deferoxamine can prevent liver fibrosis and preneoplastic lesions. We investigated the influence of administering a new oral iron chelator, deferasirox (DFX), on the effects of sorafenib. We used the choline-deficient l-amino acid-defined (CDAA) diet-induced rat liver fibrosis and HCC model. We divided rats into four groups: CDAA diet only (control group), CDAA diet with sorafenib (sorafenib group), CDAA diet with DFX (DFX group), and CDAA diet with DFX and sorafenib (DFX + sorafenib group). Liver fibrosis and development of preneoplastic lesions were assessed. In addition, we assessed adverse effects such as changes in body and liver weight, skin damage (eruption, dryness, and hair loss), which is defined as hand-foot skin syndrome, in the sorafenib and DFX + sorafenib groups. The combination of DFX + sorafenib markedly prevented liver fibrosis and preneoplastic lesions better than the other treatments. Furthermore, the combination therapy significantly decreased adverse effects compared with the sorafenib group. In conclusion, the combination therapy with DFX and sorafenib may be a useful adjuvant therapy to prevent recurrence after curative treatment of HCC.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan; Yamaguchi University Health Administration Center, 1677-1 Yoshida, Yamaguchi, Yamaguchi 753-8511, Japan
| | - Takahiro Yamasaki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan; Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Koichi Uchida
- Department of Human Nutrition Faculty of Nursing and Human Nutrition, Yamaguchi Prefectural University, 3-2-1 Sakurabatake, Yamaguchi, Yamaguchi 753-8502, Japan
| | - Koichi Fujisawa
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan; Center of Research and Education for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Toshihiko Matsumoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan; Department of Oncology and Laboratory Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Issei Saeki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachidori, Chuo-Ku, Niigata 951-8510, Japan
| | - Isao Sakaida
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
427
|
Promotion of a cancer-like phenotype, through chronic exposure to inflammatory cytokines and hypoxia in a bronchial epithelial cell line model. Sci Rep 2016; 6:18907. [PMID: 26759080 PMCID: PMC4725362 DOI: 10.1038/srep18907] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022] Open
Abstract
Globally, lung cancer accounts for approximately 20% of all cancer related deaths. Five-year survival is poor and rates have remained unchanged for the past four decades. There is an urgent need to identify markers of lung carcinogenesis and new targets for therapy. Given the recent successes of immune modulators in cancer therapy and the improved understanding of immune evasion by tumours, we sought to determine the carcinogenic impact of chronic TNF-α and IL-1β exposure in a normal bronchial epithelial cell line model. Following three months of culture in a chronic inflammatory environment under conditions of normoxia and hypoxia (0.5% oxygen), normal cells developed a number of key genotypic and phenotypic alterations. Important cellular features such as the proliferative, adhesive and invasive capacity of the normal cells were significantly amplified. In addition, gene expression profiles were altered in pathways associated with apoptosis, angiogenesis and invasion. The data generated in this study provides support that TNF-α, IL-1β and hypoxia promotes a neoplastic phenotype in normal bronchial epithelial cells. In turn these mediators may be of benefit for biomarker and/or immune-therapy target studies. This project provides an important inflammatory in vitro model for further immuno-oncology studies in the lung cancer setting.
Collapse
|
428
|
Zhang X, Zhou H, Su Y. Targeting truncated RXRα for cancer therapy. Acta Biochim Biophys Sin (Shanghai) 2016; 48:49-59. [PMID: 26494413 DOI: 10.1093/abbs/gmv104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/24/2015] [Indexed: 01/08/2023] Open
Abstract
Retinoid X receptor-alpha (RXRα), a unique member of the nuclear receptor superfamily, is a well-established drug target, representing one of the most important targets for pharmacologic interventions and therapeutic applications for cancer. However, how RXRα regulates cancer cell growth and how RXRα modulators suppress tumorigenesis are poorly understood. Altered expression and aberrant function of RXRα are implicated in the development of cancer. Previously, several studies had demonstrated the presence of N-terminally truncated RXRα (tRXRα) proteins resulted from limited proteolysis of RXRα in tumor cells. Recently, we discovered that overexpression of tRXRα can promote tumor growth by interacting with tumor necrosis factor-alpha-induced phosphoinositide 3-kinase and NF-κB signal transduction pathways. We also identified nonsteroidal anti-inflammatory drug Sulindac and analogs as effective inhibitors of tRXRα activities via a unique binding mechanism. This review discusses the emerging roles of tRXRα and modulators in the regulation of cancer cell survival and death as well as inflammation and our recent understanding of tRXRα regulation by targeting the alternate binding sites on its surface.
Collapse
Affiliation(s)
- Xiaokun Zhang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| | - Hu Zhou
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Ying Su
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China Sanford Burnham Prebys Medical Discovery Institute, Cancer Center, La Jolla, CA 92037, USA
| |
Collapse
|
429
|
Liao J, Hwang SH, Li H, Liu JY, Hammock BD, Yang GY. Inhibition of Chronic Pancreatitis and Murine Pancreatic Intraepithelial Neoplasia by a Dual Inhibitor of c-RAF and Soluble Epoxide Hydrolase in LSL-KrasG¹²D/Pdx-1-Cre Mice. Anticancer Res 2016; 36:27-37. [PMID: 26722025 PMCID: PMC4793710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Mutation of Kirsten rat sarcoma viral oncogene homolog (KRAS) and chronic pancreatitis are the most common pathogenic events involved in human pancreatic carcinogenesis. In the process of long-standing chronic inflammation, aberrant metabolites of arachidonic acid play a crucial role in promoting carcinogenesis, in which the soluble epoxide hydrolase (sEH), as a pro-inflammatory enzyme, generally inactivates anti-inflammatory epoxyeicosatrienoic acids (EETs). Herein, we determined the effect of our newly-synthesized novel compound trans-4-{4-[3-(4-chloro-3-trifluoromethyl-phenyl)-ureido]-cyclohexyloxy}-pyridine-2-carboxylic acid methylamide (t-CUPM), a dual inhibitor of sEH and RAF1 proto-oncogene serine/threonine kinase (c-RAF), on inhibiting the development of pancreatitis and pancreatic intraepithelial neoplasia (mPanIN) in LSL-Kras(G12D)/Pdx1-Cre mice. The results showed that t-CUPM significantly reduced the severity of chronic pancreatitis, as measured by the extent of acini loss, inflammatory cell infiltration and stromal fibrosis. The progression of low-grade mPanIN I to high-grade mPanIN II/III was significantly suppressed. Inhibition of mutant Kras-transmitted phosphorylation of mitogen-activated protein kinase's kinase/extracellular signal-regulated kinases was demonstrated in pancreatic tissues by western blots. Quantitative real-time polymerase chain reaction analysis revealed that t-CUPM treatment significantly reduced the levels of inflammatory cytokines including tumor necrosis facor-α, monocyte chemoattractant protein-1, as well as vascular adhesion molecule-1, and the levels of Sonic hedgehog and Gli transcription factor (Hedgehog pathway). Analysis of the eicosanoid profile revealed a significant increase of the EETs/dihydroxyeicosatrienoic acids ratio, which further confirmed sEH inhibition by t-CUPM. These results indicate that simultaneous inhibition of sEH and c-RAF by t-CUPM is important in preventing chronic pancreatitis and carcinogenesis.
Collapse
Affiliation(s)
- Jie Liao
- Department of Pathology, Northwestern University, Chicago, IL, U.S.A.
| | - Sung Hee Hwang
- Department of Entomology and Cancer Center, University of California, One Shields Avenue, Davis, CA, U.S.A
| | - Haonan Li
- Department of Pathology, Northwestern University, Chicago, IL, U.S.A
| | - Jun-Yan Liu
- Department of Entomology and Cancer Center, University of California, One Shields Avenue, Davis, CA, U.S.A
| | - Bruce D Hammock
- Department of Entomology and Cancer Center, University of California, One Shields Avenue, Davis, CA, U.S.A
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University, Chicago, IL, U.S.A.
| |
Collapse
|
430
|
Gambogic Acid and Its Role in Chronic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 928:375-395. [DOI: 10.1007/978-3-319-41334-1_15] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
431
|
Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. J Periodontal Res 2015; 51:549-66. [DOI: 10.1111/jre.12339] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Affiliation(s)
- K. Algate
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - D. R. Haynes
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - P. M. Bartold
- School of Dentistry; University of Adelaide; Adelaide SA Australia
| | - T. N. Crotti
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - M. D. Cantley
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
- Myeloma Research Laboratory; University of Adelaide; Adelaide SA Australia
| |
Collapse
|
432
|
Kapadia CH, Perry JL, Tian S, Luft JC, DeSimone JM. Nanoparticulate immunotherapy for cancer. J Control Release 2015; 219:167-180. [DOI: 10.1016/j.jconrel.2015.09.062] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 12/11/2022]
|
433
|
Xu J, Xu X, Shi S, Wang Q, Saxton B, He W, Gou X, Jang JH, Nyunoya T, Wang X, Xing C, Zhang L, Lin Y. Autophagy-Mediated Degradation of IAPs and c-FLIP(L) Potentiates Apoptosis Induced by Combination of TRAIL and Chal-24. J Cell Biochem 2015; 117:1136-44. [PMID: 26448608 DOI: 10.1002/jcb.25397] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/06/2015] [Indexed: 12/21/2022]
Abstract
Combination chemotherapy is an effective strategy for increasing anticancer efficacy, reducing side effects and alleviating drug resistance. Here we report that combination of the recently identified novel chalcone derivative, chalcone-24 (Chal-24), and TNF-related apoptosis-inducing ligand (TRAIL) significantly increases cytotoxicity in lung cancer cells. Chal-24 treatment significantly enhanced TRAIL-induced activation of caspase-8 and caspase-3, and the cytotoxicity induced by combination of these agents was effectively suppressed by the pan-caspase inhibitor z-VAD-fmk. Chal-24 and TRAIL combination suppressed expression of cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein large (c-FLIP(L)) and cellular inhibitor of apoptosis proteins (c-IAPs), and ectopic expression of c-FLIP(L) and c-IAPs inhibited the potentiated cytotoxicity. In addition, TRAIL and Chal-24 cooperatively activated autophagy. Suppression of autophagy effectively attenuated cytotoxicity induced by Chal-24 and TRAIL combination, which was associated with attenuation of c-FLIP(L) and c-IAPs degradation. Altogether, these results suggest that Chal-24 potentiates the anticancer activity of TRAIL through autophagy-mediated degradation of c-FLIP(L) and c-IAPs, and that combination of Chal-24 and TRAIL could be an effective approach in improving chemotherapy efficacy.
Collapse
Affiliation(s)
- Jennings Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| | - Xiuling Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| | - Shaoqing Shi
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico.,Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Qiong Wang
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico.,Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Bryanna Saxton
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 510182, P.R. China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 510182, P.R. China
| | - Jun-Ho Jang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China.,Division of Pulmonary and Critical Care Medicine, University of New Mexico and New Mexico VA Health Care System, Albuquerque, New Mexico
| | - Toru Nyunoya
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China.,Division of Pulmonary and Critical Care Medicine, University of New Mexico and New Mexico VA Health Care System, Albuquerque, New Mexico
| | - Xia Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, 55455, Minnesota
| | - Lin Zhang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, 87108, New Mexico
| |
Collapse
|
434
|
Simovic B, Walsh SR, Wan Y. Mechanistic insights into the oncolytic activity of vesicular stomatitis virus in cancer immunotherapy. Oncolytic Virother 2015; 4:157-67. [PMID: 27512679 PMCID: PMC4918393 DOI: 10.2147/ov.s66079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy and oncolytic virotherapy have both shown anticancer efficacy in the clinic as monotherapies but the greatest promise lies in therapies that combine these approaches. Vesicular stomatitis virus is a prominent oncolytic virus with several features that promise synergy between oncolytic virotherapy and immunotherapy. This review will address the cytotoxicity of vesicular stomatitis virus in transformed cells and what this means for antitumor immunity and the virus’ immunogenicity, as well as how it facilitates the breaking of tolerance within the tumor, and finally, we will outline how these features can be incorporated into the rational design of new treatment strategies in combination with immunotherapy.
Collapse
Affiliation(s)
- Boris Simovic
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Scott R Walsh
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
435
|
Liu Y, Yin T, Feng Y, Cona MM, Huang G, Liu J, Song S, Jiang Y, Xia Q, Swinnen JV, Bormans G, Himmelreich U, Oyen R, Ni Y. Mammalian models of chemically induced primary malignancies exploitable for imaging-based preclinical theragnostic research. Quant Imaging Med Surg 2015; 5:708-29. [PMID: 26682141 PMCID: PMC4671963 DOI: 10.3978/j.issn.2223-4292.2015.06.01] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/15/2015] [Indexed: 12/14/2022]
Abstract
Compared with transplanted tumor models or genetically engineered cancer models, chemically induced primary malignancies in experimental animals can mimic the clinical cancer progress from the early stage on. Cancer caused by chemical carcinogens generally develops through three phases namely initiation, promotion and progression. Based on different mechanisms, chemical carcinogens can be divided into genotoxic and non-genotoxic ones, or complete and incomplete ones, usually with an organ-specific property. Chemical carcinogens can be classified upon their origins such as environmental pollutants, cooked meat derived carcinogens, N-nitroso compounds, food additives, antineoplastic agents, naturally occurring substances and synthetic carcinogens, etc. Carcinogen-induced models of primary cancers can be used to evaluate the diagnostic/therapeutic effects of candidate drugs, investigate the biological influential factors, explore preventive measures for carcinogenicity, and better understand molecular mechanisms involved in tumor initiation, promotion and progression. Among commonly adopted cancer models, chemically induced primary malignancies in mammals have several advantages including the easy procedures, fruitful tumor generation and high analogy to clinical human primary cancers. However, in addition to the time-consuming process, the major drawback of chemical carcinogenesis for translational research is the difficulty in noninvasive tumor burden assessment in small animals. Like human cancers, tumors occur unpredictably also among animals in terms of timing, location and the number of lesions. Thanks to the availability of magnetic resonance imaging (MRI) with various advantages such as ionizing-free scanning, superb soft tissue contrast, multi-parametric information, and utility of diverse contrast agents, now a workable solution to this bottleneck problem is to apply MRI for noninvasive detection, diagnosis and therapeutic monitoring on those otherwise uncontrollable animal models with primary cancers. Moreover, it is foreseeable that the combined use of chemically induced primary cancer models and molecular imaging techniques may help to develop new anticancer diagnostics and therapeutics.
Collapse
|
436
|
Loza-Mejía MA, Salazar JR. Sterols and triterpenoids as potential anti-inflammatories: Molecular docking studies for binding to some enzymes involved in inflammatory pathways. J Mol Graph Model 2015; 62:18-25. [PMID: 26342572 DOI: 10.1016/j.jmgm.2015.08.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 08/24/2015] [Accepted: 08/26/2015] [Indexed: 12/12/2022]
Abstract
Triterpenes and sterols are good candidates for the development of anti-inflammatory drugs and use in chemoprevention or chemotherapy of cancer via the interaction with therapeutic targets related to inflammation, such as COX-1 and -2; LOX-5; MPO, PLA2 and i-NOS. In this study, we use molecular docking to evaluate the potential binding of a database of selected sterol and triterpenoid compounds with several skeletons against enzymes related to inflammation to propose structural requirements beneficial for anti-inflammatory activity that can be used for the design of more potent and selective anti-inflammatory and antitumor drugs. Our results suggest that the substitution pattern is important and that there is an important relationship between the class of sterol or triterpenoid skeleton and enzyme binding.
Collapse
Affiliation(s)
- Marco A Loza-Mejía
- Facultad de Ciencias Químicas, Universidad La Salle, Benjamín Franklin 47, 06140 México City, Mexico.
| | - Juan Rodrigo Salazar
- Facultad de Ciencias Químicas, Universidad La Salle, Benjamín Franklin 47, 06140 México City, Mexico.
| |
Collapse
|
437
|
Lebrec H, Ponce R, Preston BD, Iles J, Born TL, Hooper M. Tumor necrosis factor, tumor necrosis factor inhibition, and cancer risk. Curr Med Res Opin 2015; 31:557-74. [PMID: 25651481 DOI: 10.1185/03007995.2015.1011778] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Tumor necrosis factor (TNF) is a highly pleiotropic cytokine with multiple activities other than its originally discovered role of tumor necrosis in rodents. TNF is now understood to play a contextual role in driving either tumor elimination or promotion. Using both animal and human data, this review examines the role of TNF in cancer development and the effect of TNF and TNF inhibitors (TNFis) on malignancy risk. RESEARCH DESIGN A literature review was performed using relevant search terms for TNF and malignancy. RESULTS Although administration of TNF can cause tumor regression in specific rodent tumor models, human expression polymorphisms suggest that TNF can be a tumor-promoting cytokine, whereas blocking the TNF pathway in a variety of tumor models inhibits tumor growth. In addition to direct effects of TNF on tumors, TNF can variously affect immunity and the tumor microenvironment. Whereas TNF can promote immune surveillance designed to eliminate tumors, it can also drive chronic inflammation, autoimmunity, angiogenesis, and other processes that promote tumor initiation, growth, and spread. Key players in TNF signaling that shape this response include NF-κB and JNK, and malignant-inflammatory cell interactions, each of which may have different responses to TNF signaling. Focusing on rheumatoid arthritis (RA) patients, where clinical experience is most extensive, a review of the clinical literature shows no increased risk of overall malignancy or solid tumors such as breast and lung cancers with exposure to TNFis. Lymphoma rates are not increased with use of TNFis. Conflicting data exist regarding the risks of melanoma and nonmelanoma skin cancer. Data regarding the risk of recurrent malignancy are limited. CONCLUSIONS Overall, the available data indicate that elevated TNF is a risk factor for cancer, whereas its inhibition in RA patients is not generally associated with an increased cancer risk. In particular, TNF inhibition is not associated with cancers linked to immune suppression. A better understanding of the tumor microenvironment, molecular events underlying specific tumors, and epidemiologic studies of malignancies within specific disease indications should enable more focused pharmacovigilance studies and a better understanding of the potential risks of TNFis.
Collapse
|
438
|
Synergistic anticancer effect of cisplatin and Chal-24 combination through IAP and c-FLIPL degradation, Ripoptosome formation and autophagy-mediated apoptosis. Oncotarget 2015; 6:1640-51. [PMID: 25682199 PMCID: PMC4359321 DOI: 10.18632/oncotarget.2746] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/08/2014] [Indexed: 01/26/2023] Open
Abstract
Drug resistance is a major hurdle in anticancer chemotherapy. Combined therapy using drugs with distinct mechanisms of function may increase anticancer efficacy. We have recently identified the novel chalcone derivative, chalcone-24 (Chal-24), as a potential therapeutic that kills cancer cells through activation of an autophagy-mediated necroptosis pathway. In this report, we investigated if Chal-24 can be combined with the frontline genotoxic anticancer drug, cisplatin for cancer therapy. The combination of Chal-24 and cisplatin synergistically induced apoptotic cytotoxicity in lung cancer cell lines, which was dependent on Chal-24-induced autophagy. While cisplatin slightly potentiated the JNK/Bcl2/Beclin1 pathway for autophagy activation, its combination with Chal-24 strongly triggered proteasomal degradation of the cellular inhibitor of apoptosis proteins (c-IAPs) and formation of the Ripoptosome complex that contains RIP1, FADD and caspase 8. Furthermore, the cisplatin and Chal-24 combination induced dramatic degradation of cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein large (cFLIPL) which suppresses Ripoptosome-mediated apoptosis activation. These results establish a novel mechanism for potentiation of anticancer activity with the combination of Chal-24 and cisplatin: to enhance apoptosis signaling through Ripoptosome formation and to release the apoptosis brake through c-FLIPL degradation. Altogether, our work suggests that the combination of Chal-24 and cisplatin could be employed to improve chemotherapy efficacy.
Collapse
|
439
|
Nong Y, Wu D, Lin Y, Zhang Y, Bai L, Tang H. Tenascin-C expression is associated with poor prognosis in hepatocellular carcinoma (HCC) patients and the inflammatory cytokine TNF-α-induced TNC expression promotes migration in HCC cells. Am J Cancer Res 2015; 5:782-791. [PMID: 25973315 PMCID: PMC4396033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023] Open
Abstract
Although tenascin-c (TNC) in inflammatory microenvironment contributes to progression in some tumors, its role in hepatocellular carcinoma (HCC) in metastasis and the mechanism by which TNC expression is regulated in HCC cells are elusive. In this study, we examined TNC expression in 100 HCC tissue samples by immunohistochemistry and compared which between the groups with or without metastasis. TNC expression was higher in metastatic HCC tissues than that in the non-metastatic HCC tissues, which was associated with the Knodell inflammation scores. Importantly, high level of TNC expression was associated with lower survival rate and shorter survival time in the HCC patients. We then investigated the mechanism by which TNC expression is regulated in HCC cells with an in vitro cell culture system. The recombinant TNF-α and conditioned medium from macrophages induced TNC expression at both mRNA and protein levels in HepG2 cells. The induction of TNC expression by conditioned medium from macrophages was suppressed by a TNF-α neutralizing antibody. TNF-α-promoted cell migration was inhibited by a TNC siRNA. In addition, TNF-α-induced TNC expression was blocked by a NF-κB pathway inhibitor. These results suggest that TNF-α in the tumor microenvironment induces TNC expression in HCC cells through the NF-κB pathway, which in turn, promotes HCC cell migration. Thus, TNC may play an important role in promoting HCC metastasis and TNC expression could be a predictive factor for poor prognosis in HCC patients.
Collapse
Affiliation(s)
- Yunhong Nong
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute2425 Ridgecrest Dr. SE, Albuquerque, NM 87108, USA
| | - Yongqiang Zhang
- Biorepository, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital, Sichuan UniversityChengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy, Sichuan UniversityChengdu 610041, China
| |
Collapse
|
440
|
Lu L, Shi W, Deshmukh RR, Long J, Cheng X, Ji W, Zeng G, Chen X, Zhang Y, Dou QP. Tumor necrosis factor-α sensitizes breast cancer cells to natural products with proteasome-inhibitory activity leading to apoptosis. PLoS One 2014; 9:e113783. [PMID: 25419573 PMCID: PMC4242672 DOI: 10.1371/journal.pone.0113783] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 10/02/2014] [Indexed: 01/08/2023] Open
Abstract
The inflammatory microenvironment plays an important role in the process of tumor development. Tumor necrosis factor-α (TNF-α), a key pro-inflammatory cytokine, has a significant role in this process. Natural medicinal products such as Withaferin A (WA) and Celastrol (Cel) have shown anti-cancer and anti-inflammatory properties that can be attributed to multiple mechanisms including, but not limited to, apoptosis induction due to the inhibition of proteasomal activities. This study aimed to investigate the effects of TNF-α in combination with WA or Cel in vitro in MDA-MB-231 breast cancer cells. TNF-α, when combined with WA or Cel, activated caspase-3 and -9 and downregulated XIAP in a dose-dependent manner, leading to induction of apoptosis in MDA-MB-231 breast cancer cells. The combination also caused accumulation of the proteasomal target protein IκBα, resulting in inhibition of the nuclear translocation of nuclear factor-κB (NF-κB). Taken together, these results suggest that TNF-α could sensitize breast cancer cells MDA-MB-231 to WA and Cel, at least in part, through inhibiting the activation of NF-κB signaling, leading to XIAP inhibition with subsequent upregulation of caspase-3 and -9 activities. Thus, the anti-cancer activities of TNF-α are enhanced when combined with the natural proteasome inhibitors, WA or Cel.
Collapse
Affiliation(s)
- Li Lu
- Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Pathology and Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Wenli Shi
- Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Rahul R. Deshmukh
- Department of Pathology and Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Jie Long
- Department of Pathology, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Xiaoli Cheng
- Department of Pathophysiology, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Weidong Ji
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China
| | - Guohua Zeng
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Urology, Guangzhou, Guangdong Province, China
| | - Xianliang Chen
- Department of Pathology, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Yajie Zhang
- Department of Pathology, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- * E-mail: (YZ); (QPD)
| | - Q. Ping Dou
- Department of Pathology and Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
- Departments of Oncology and Pharmacology, School of Medicine, Wayne State University, Detroit, Michigan, United States of America
- * E-mail: (YZ); (QPD)
| |
Collapse
|
441
|
Kato H, Wakabayashi H, Naito Y, Kato S, Nakagawa T, Matsumine A, Sudo A. Anti-tumor necrosis factor therapy inhibits lung metastasis in an osteosarcoma cell line. Oncology 2014; 88:139-46. [PMID: 25402182 DOI: 10.1159/000368414] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 09/08/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Osteosarcoma is the most common primary malignancy of bone, and patients often develop pulmonary metastases. In a previous study, tumor necrosis factor (TNF)-α treatment of human osteosarcoma cells increases their metastatic ability in an animal model. TNF-α can act as a tumor necrosis factor and also as a tumor-promoting factor. In the present study, the effect of a TNF-α inhibitor on osteosarcoma aggressiveness and pulmonary metastases was investigated in vitro and in vivo. METHODS The effect of infliximab, a TNF-α inhibitor, on a metastatic osteosarcoma 143B cell growth and motility was investigated in vitro. An orthotopic xenograft model of 143B cell growth and spontaneous metastasis in SCID mice was used to assess the in vivo effect of infliximab. RESULTS Infliximab greatly reduced cell motility and pulmonary metastases in 143B cells. The mechanism of pulmonary metastasis inhibition involved decreased expression of CXC chemokine receptor 4 (CXCR4), Rho (small GTPase protein), and its effector. CONCLUSIONS These results suggest a novel role for TNF-α inhibition in the reduction or prevention of pulmonary metastases of osteosarcoma in this animal model. TNF-α inhibition may become a preventive therapeutic option for the pulmonary metastases of osteosarcoma.
Collapse
Affiliation(s)
- Hiroaki Kato
- Department of Orthopedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | | | | | | | | | | | | |
Collapse
|
442
|
Sarker MMR, Zhong M. Keyhole limpet hemocyanin augmented the killing activity, cytokine production and proliferation of NK cells, and inhibited the proliferation of Meth A sarcoma cells in vitro. Indian J Pharmacol 2014; 46:40-5. [PMID: 24550583 PMCID: PMC3912806 DOI: 10.4103/0253-7613.125164] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Revised: 09/29/2013] [Accepted: 11/12/2013] [Indexed: 12/01/2022] Open
Abstract
Objective: Keyhole limpet hemocyanin (KLH) is a popular tumor vaccine carrier protein and an immunostimulant. The present study aimed to investigate the immunoregulatory activity of KLH on cytotoxicity, cytokines production, and proliferation of natural killer (NK) cells. Moreover, antiproliferative activity of KLH on Meth A sarcoma cells was studied. Materials and Methods: Cytotoxicity was determined with killing ability of NK cells against yeast artificial chromosome (YAC)-1 cells. Interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) productions by NK cells were measured by enzyme-linked immunosorbent assay (ELISA). Proliferations of NK and Meth A cells were determined by [3H]thymidine incorporated proliferation and 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) methods, respectively. Results: KLH at 6.25, 12.5, and 25 μg/well augmented cytotoxicity of NK cells against YAC-1 cells by 2.5, three, and five-times, respectively. KLH at 25 μg/well enhanced IFN-γ and TNF-α productions by 17- and 23-folds, respectively. The proliferation of NK cells was three times stimulated by KLH. The proliferation of Meth A cells was markedly inhibited by all the doses; the highest (4-folds higher) inhibition was observed at a dose of KLH (25 μg/well). Conclusion: The study demonstrated the anticancer activity of KLH acting through the induction of NK cells and inhibition of cancer cells. KLH, therefore, may be a good candidate for an anticancer agent alone or in combination with other chemotherapeutic agents.
Collapse
Affiliation(s)
- Md Moklesur Rahman Sarker
- Department of Immunochemistry, Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Tsushima-naka, Kita-ku, Okayama, Japan ; Clinical Investigation Centre, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ming Zhong
- Department of Immunochemistry, Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Tsushima-naka, Kita-ku, Okayama, Japan
| |
Collapse
|
443
|
Sulindac-derived RXRα modulators inhibit cancer cell growth by binding to a novel site. ACTA ACUST UNITED AC 2014; 21:596-607. [PMID: 24704507 DOI: 10.1016/j.chembiol.2014.02.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/22/2014] [Accepted: 02/19/2014] [Indexed: 12/20/2022]
Abstract
Retinoid X receptor-alpha (RXRα), an intriguing and unique drug target, can serve as an intracellular target mediating the anticancer effects of certain nonsteroidal anti-inflammatory drugs (NSAIDs), including sulindac. We report the synthesis and characterization of two sulindac analogs, K-8008 and K-8012, which exert improved anticancer activities over sulindac in a RXRα-dependent manner. The analogs inhibit the interaction of the N-terminally truncated RXRα (tRXRα) with the p85α subunit of PI3K, leading to suppression of AKT activation and induction of apoptosis. Crystal structures of the RXRα ligand-binding domain (LBD) with K-8008 or K-8012 reveal that both compounds bind to tetrameric RXRα LBD at a site different from the classical ligand-binding pocket. Thus, these results identify K-8008 and K-8012 as tRXRα modulators and define a binding mechanism for regulating the nongenomic action of tRXRα.
Collapse
|
444
|
Stabilization of Snail by HIF-1α and TNF-α is required for hypoxia-induced invasion in prostate cancer PC3 cells. Mol Biol Rep 2014; 41:4573-82. [PMID: 24610352 DOI: 10.1007/s11033-014-3328-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 02/26/2014] [Indexed: 01/01/2023]
Abstract
Hypoxia has been involved in the development of tumor by regulating the expression of invasiveness-associated genes. However, the specific function of hypoxia in cancer cell invasion is still unclear. The aim of the present study was to determine the role of hypoxia in invasion of prostate cancer PC3 cells and to investigate the underlying mechanisms. We found that hypoxia significantly increased the invasive activity of PC3 cells, via up-regulation of the expression of hypoxia inducible factor 1α (HIF-1α) and the autocrine production of tumor necrosis factor α (TNF-α). More important, TNF-α cooperated with HIF-1α in promoting stabilization of Snail, a transcriptional repressor of E-cadherin expression, which lead to the up-regulation of invasiveness-associated genes MMP-9, fibronectin and vimentin. Snail silencing by specific siRNA significantly inhibited hypoxia-induced invasion of PC3 cells, indicating an essential role of Snail in conferring the malignant phenotype to cancer cells under hypoxic conditions. In conclusion, our data demonstrate that hypoxia promoted the invasiveness of prostate cancer PC3 cells via HIF-1α- and TNF-α-induced stabilization of Snail, suggesting a signaling mechanism involving HIF-1α/TNF-α/Snail that mediates invasiveness hypoxic tumor cells in the absence of neoangiogenesis.
Collapse
|
445
|
TNF-α induces CXCL1 chemokine expression and release in human vascular endothelial cells in vitro via two distinct signaling pathways. Acta Pharmacol Sin 2014; 35:339-50. [PMID: 24487964 DOI: 10.1038/aps.2013.182] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/10/2013] [Indexed: 12/13/2022]
Abstract
AIM Chemokines usually direct the movement of circulating leukocytes to sites of inflammation or injury. CXCL1/GRO-α has been shown to be upregulated in atherosclerotic lesions and various cancers. The aim of this study was to investigate the mechanisms underlying the TNF-α-induced release of CXCL1 from human vascular endothelial cells in vitro. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with different proinflam-matory mediators and growth factors. CXCL1 expression and secretion were determined using RT-PCR and ELISA, respectively. TNF-α-induced cell signaling was assayed with Western blotting. Cell viability/growth was determined using MTT assay. Monocyte migration was measured with transwell migration assay. RESULTS Among the 17 mediators and growth factors tested, TNF-α, LPS and thrombin induced marked increase in CXCL1 release from HUVEC cells. TNF-α (2, 5 ng/mL) induced CXCL1 release and mRNA expression in the cells in concentration- and time-dependent manners. TNF-α (5 ng/mL) caused activation of JNK, p38 MAPK, PI3K and Akt, whereas pretreatment with JNK inhibitor (SP600125), p38 MAPK inhibitor (SB202190) or PI-3K inhibitor (LY294002) significantly suppressed TNF-α-induced CXCL1 release from the cells. But only SP600125 significantly reduced TNF-α-induced CXCL1 mRNA expression in the cells. Moreover, dexamethasone (up to 500 nmol/L) failed to affect TNF-α-induced CXCL1 release from the cells. In functional studies, recombinant CXCL1 enhanced HUVEC proliferation, and both recombinant CXCL1 and TNF-α-induced CXCL1 from HUVECs attracted human monocyte migration. CONCLUSION TNF-α stimulates CXCL1 release from human ECs through JNK-mediated CXCL1 mRNA expression and p38 MAPK- and PI-3K-mediated CXCL1 secretory processes.
Collapse
|
446
|
Xu X, Padilla MT, Li B, Wells A, Kato K, Tellez C, Belinsky SA, Kim KC, Lin Y. MUC1 in macrophage: contributions to cigarette smoke-induced lung cancer. Cancer Res 2014; 74:460-70. [PMID: 24282280 PMCID: PMC3947020 DOI: 10.1158/0008-5472.can-13-1713] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Expression of the pro-oncogenic mucin MUC1 is elevated by inflammation in airway epithelial cells, but the contributions of MUC1 to the development of lung cancer are uncertain. In this study, we developed our finding that cigarette smoke increases Muc1 expression in mouse lung macrophages, where we hypothesized MUC1 may contribute to cigarette smoke-induced transformation of bronchial epithelial cells. In human macrophages, cigarette smoke extract (CSE) strongly induced MUC1 expression through a mechanism involving the nuclear receptor PPAR-γ. CSE-induced extracellular signal-regulated kinase (ERK) activation was also required for MUC1 expression, but it had little effect on MUC1 transcription. RNA interference-mediated attenuation of MUC1 suppressed CSE-induced secretion of TNF-α from macrophages, by suppressing the activity of the TNF-α-converting enzyme (TACE), arguing that MUC1 is required for CSE-induced and TACE-mediated TNF-α secretion. Similarly, MUC1 blockade after CSE induction through suppression of PPAR-γ or ERK inhibited TACE activity and TNF-α secretion. Conditioned media from CSE-treated macrophages induced MUC1 expression and potentiated CSE-induced transformation of human bronchial epithelial cells in a TNF-α-dependent manner. Together, our results identify a signaling pathway involving PPAR-γ, ERK, and MUC1 for TNF-α secretion induced by CSE from macrophages. Furthermore, our results show how MUC1 contributes to smoking-induced lung cancers that are driven by inflammatory signals from macrophages.
Collapse
Affiliation(s)
- Xiuling Xu
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| | - Mabel T. Padilla
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| | - Bilan Li
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| | - Alexandria Wells
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| | - Kosuke Kato
- Department of Physiology & Lung Center, Temple University School of Medicine, 3420 N. Broad St., Philadelphia, PA 19140, USA
| | - Carmen Tellez
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| | - Steven A. Belinsky
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| | - Kwang Chul Kim
- Department of Physiology & Lung Center, Temple University School of Medicine, 3420 N. Broad St., Philadelphia, PA 19140, USA
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, 2425 Ridgecrest DR. SE, Albuquerque, NM 87108, USA
| |
Collapse
|
447
|
Xu Y, Shen Y, Ouahab A, Li C, Xiong Y, Tu J. Antitumor activity of TNF-α after intratumoral injection using an in situ thermosensitive hydrogel. Drug Dev Ind Pharm 2013; 41:369-74. [PMID: 24274583 DOI: 10.3109/03639045.2013.861480] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Local drug delivery strategies based on nanoparticles, gels, polymeric films, rods and wafers are increasingly used in cancer chemotherapy in order to enhance therapeutic effect and reduce systemic toxicity. Herein, a biodegradable and biocompatible in situ thermosensitive hydrogel was designed and employed to deliver tumor necrosis factor-α (TNF-α) locally by intratumoral injection. The triblock copolymer was synthesized by ring-opening polymerization (ROP) of β-butyrolactone (β-BL) and lactide (LA) in bulk using polyethylene glycol (PEG) as an initiator and Sn(Oct)2 as the catalyst, the polymer was characterized by NMR, gel permeation chromatography and differential scanning calorimetry. Blood and tumor pharmacokinetics and in vivo antitumor activity of TNF-α after intratumoral administration in hydrogel or solution with the same dose were evaluated on S180 tumor-bearing mice. Compared with TNF-α solution, TNF-α hydrogel exhibited a longer T1/2 (4-fold) and higher AUCtumor (19-fold), but Cmax was lower (0.5-fold), which means that the hydrogel formulation improved the efficacy with a lower systhemic exposure than the solution formation. In addition, TNF-α hydrogel improved the antitumor activity and survival due to lower systemic exposure than the solution. These results demonstrate that the in situ thermosensitive hydrogel-based local delivery system by intratumoral injection is well suited for the administration of TNF-α.
Collapse
Affiliation(s)
- Yourui Xu
- Department of Pharmaceutics, China Pharmaceutical University , Nanjing , China
| | | | | | | | | | | |
Collapse
|
448
|
Hu H, Yin J, Wang M, Liang C, Song H, Wang J, Nie Y, Liang J, Wu K. GX1 targeting delivery of rmhTNFα evaluated using multimodality imaging. Int J Pharm 2013; 461:181-91. [PMID: 24269209 DOI: 10.1016/j.ijpharm.2013.11.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/18/2013] [Accepted: 11/13/2013] [Indexed: 12/12/2022]
Abstract
GX1 is a tumor targeting peptide. In this study, we evaluated the antitumor efficacy of a GX1-derived fusion toxin, GX1-rmhTNFα, and investigated its targeting efficiency and pharmacokinetics in vivo using multimodality imaging. Flow cytometry revealed a greater level of cell apoptosis induced by GX1-rmhTNFα (27.1%) compared with rmhTNFα or a saline control (13.7% and 4.7%, respectively). SPECT (single-photon emission computed tomography) demonstrated high accumulation of GX1-rmhTNFα in tumor site. Biodistribution studies indicated GX1-rmhTNFα was cleared by the liver and kidney, and the drug may not cross the blood-brain barrier. In addition, bioluminescence imaging (BLI) showed that GX1-rmhTNFα caused a satisfactory delay in tumor growth in both subcutaneous and orthotopic cancer models. Contrast-enhanced ultrasound (CEUS) and CD31 staining revealed a loss in blood perfusion and vasculature. TUNEL and Ki67 staining validated the in vivo results. Biochemical analyses revealed limited renal and hepatic toxicity of GX1-rmhTNFα. This study demonstrated that GX1-rmhTNFα is a safe and potent anticancer agent that may have great potential for the targeted therapy of gastric cancer.
Collapse
Affiliation(s)
- Hao Hu
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, China
| | - Jipeng Yin
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, China
| | - Min Wang
- Department of Gastroenterology, Xi'an Children's Hospital, China
| | - Cong Liang
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, China
| | - Hongping Song
- Department of Ultrasound, Xijing Hospital, Fourth Military Medical University, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, China
| | - Jie Liang
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, China.
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology & Xijing Hospital of Digestive Diseases, Fourth Military Medical University, China.
| |
Collapse
|
449
|
Ye YC, Wang HJ, Chen L, Liu WW, Tashiro SI, Onodera S, Xia MY, Ikejima T. Negatively-regulated necroptosis by autophagy required caspase-6 activation in TNFα-treated murine fibrosarcoma L929 cells. Int Immunopharmacol 2013; 17:548-55. [DOI: 10.1016/j.intimp.2013.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 04/23/2013] [Accepted: 05/15/2013] [Indexed: 02/09/2023]
|
450
|
Tian X, Ma P, Sui C, Li Y, Fu L, Jiang T, Wang Y, Jiang Y. Comprehensive assessment of the association between tumor necrosis factor alpha G238A polymorphism and liver cancer risk. Tumour Biol 2013; 35:103-9. [PMID: 23943369 DOI: 10.1007/s13277-013-1012-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 07/08/2013] [Indexed: 12/16/2022] Open
Abstract
Tumor necrosis factor alpha (TNF-α) has been suggested to play an important role in the development and liver cancer. TNF-α 238 G/A polymorphism was hypothesized to increase the risk of liver cancer, but findings from previous studies were controversial. To explore a more precise estimation of the relationship between TNF-α 238 G/A polymorphism and liver cancer, we performed a meta-analysis. PubMed, Embase, and China Biology Medicine databases were searched for all publications on this association through March 12, 2013. Odds ratios (ORs) with its 95% confidence intervals (CIs) were used to assess the strength of this association. Eleven studies with 1,406 liver cancer cases and 2,386 noncancer controls were included into this meta-analysis. Overall, there was a significant association between TNF-α 238 G/A polymorphism and increased risk of liver cancer under all three genetic models (A vs. G, OR 1.51, 95% CI 1.20-1.89, P < 0.001, I(2) = 37.7%; AG vs. GG, OR 1.49, 95% CI 1.01-2.21, P = 0.045, I(2) = 53.2%; AA/AG vs. GG, OR 1.76, 95% CI 1.35-2.30, P < 0.001, I(2) = 36.5%). The sensitivity analysis further strengthened the validity of the positive association. Subgroup analysis of nine studies from Asian countries showed that there was a significant association between TNF-α 238 G/A polymorphism and increased risk of liver cancer in Asians (A vs. G, OR 1.35, 95% CI 1.03-1.76, P = 0.027, I(2) = 40.2%; AA/AG vs. GG, OR 1.56, 95% CI 1.14-2.15, P = 0.006, I(2) = 41.9%). In conclusion, TNF-α 238 G/A polymorphism is significantly associated with increased risk of liver cancer, especially in Asians.
Collapse
Affiliation(s)
- Xin Tian
- Molecular Oncology Department of Cancer Research Institution, The First Hospital, China Medical University, Shenyang, 110001, China
| | | | | | | | | | | | | | | |
Collapse
|