1
|
Hübbers A, Hennings J, Lambertz D, Haas U, Trautwein C, Nevzorova YA, Sonntag R, Liedtke C. Pharmacological Inhibition of Cyclin-Dependent Kinases Triggers Anti-Fibrotic Effects in Hepatic Stellate Cells In Vitro. Int J Mol Sci 2020; 21:ijms21093267. [PMID: 32380742 PMCID: PMC7246535 DOI: 10.3390/ijms21093267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a wound healing process in response to chronic liver injury, which is characterized by the accumulation of extracellular collagen produced by Hepatic Stellate Cells (HSCs). This process involves cell cycle re-entry and proliferation of normally quiescent HSCs controlled by cyclins and associated cyclin-dependent kinases (Cdks). Cdk2 mediates the entry and progression through S-phase in complex with E-and A-type cyclins. We have demonstrated that cyclin E1 is essential for liver fibrogenesis in mice, but it is not known if this is dependent on Cdk2 or related Cdks. Here, we aimed to evaluate the benefit of the pan-Cdk inhibitor CR8 for treatment of liver fibrosis in vitro. CR8-treatment reduced proliferation and survival in immortalized HSC lines and in addition attenuated pro-fibrotic properties in primary murine HSCs. Importantly, primary murine hepatocytes were much more tolerant against the cytotoxic and anti-proliferative effects of CR8. We identified CR8 dosages mediating anti-fibrotic effects in primary HSCs without affecting cell cycle activity and survival in primary hepatocytes. In conclusion, the pharmacological pan-Cdk inhibitor CR8 restricts the pro-fibrotic properties of HSCs, while preserving proliferation and viability of hepatocytes at least in vitro. Therefore, CR8 and related drugs might be beneficial for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Anna Hübbers
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Julia Hennings
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Daniela Lambertz
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Ute Haas
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
| | - Yulia A. Nevzorova
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
- Department of Genetics, Physiology, and Microbiology, Faculty of Biology, Complutense University Madrid, 28040 Madrid, Spain
| | - Roland Sonntag
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
- Correspondence: (R.S.); (C.L.)
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital, RWTH Aachen University, D-52074 Aachen, Germany; (A.H.); (J.H.); (D.L.); (U.H.); (C.T.); (Y.A.N.)
- Correspondence: (R.S.); (C.L.)
| |
Collapse
|
2
|
Ma B, Chen K, Liu P, Li M, Liu J, Sideras K, Sprengers D, Biermann K, Wang W, IJzermans JNM, Cao W, Kwekkeboom J, Peppelenbosch MP, Pan Q. Dichotomal functions of phosphorylated and unphosphorylated STAT1 in hepatocellular carcinoma. J Mol Med (Berl) 2018; 97:77-88. [PMID: 30456450 PMCID: PMC6326978 DOI: 10.1007/s00109-018-1717-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 09/23/2018] [Accepted: 11/05/2018] [Indexed: 01/12/2023]
Abstract
Abstract Interferons (IFNs) with antiviral and immune-stimulatory functions have been widely used in prevention and treatment of hepatocellular carcinoma (HCC). Signal transducer and activator of transcription 1 (STAT1) is a key element of the IFN signaling, and the function of STAT1 is critically determined by its phosphorylation state. This study aims to understand the functions of phosphorylated (p-) and unphosphorylated (u-) STAT1 in HCC. We found that u-STAT1 is significantly elevated in patient HCC tumor tissues and predominantly expressed in cytoplasm; while p-STAT1 is absent. Loss of u-STAT1 potently arrested cell cycle and inhibited cell growth in HCC cells. Induction of p-STAT1 by IFN-α treatment effectively triggers the expression of interferon-stimulated genes (ISGs), but has moderate effect on HCC cell growth. Interestingly, both u-STAT1 and p-STAT1 are induced by IFN-α, through with distinct time-dependent process. Furthermore, the ISG induction patterns mediated by p-STAT1 and u-STAT1 are also distinct. Importantly, artificial blocking of the induction of u-STAT1, but not p-STAT1, sensitizes HCC cells to treatment of IFNs. Therefore, p-STAT1 and u-STAT1 exert dichotomal functions and coordinately regulate the responsiveness to IFN treatment in HCC. Key Messages STAT1 is upregulated and predominantly presented as u-STAT1 in HCC, while p-STAT1 is absent. U-STAT1 sustains but p-STAT1 inhibits HCC growth. The dynamic change of phosphorylation state of STAT1 control the responsiveness to IFN treatment.
Electronic supplementary material The online version of this article (10.1007/s00109-018-1717-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Buyun Ma
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Kan Chen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands.,College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Pengyu Liu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Meng Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Jiaye Liu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Kostandinos Sideras
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Katharina Biermann
- Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, 3015CE, The Netherlands
| | - Wenshi Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Jan N M IJzermans
- Department of Surgery, Erasmus MC-University Medical Centre, Rotterdam, 3015CE, The Netherlands
| | - Wanlu Cao
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Jaap Kwekkeboom
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-617, 's-Gravendijkwal 230, 3015CE, Rotterdam, The Netherlands.
| |
Collapse
|
3
|
Essential role of HCMV deubiquitinase in promoting oncogenesis by targeting anti-viral innate immune signaling pathways. Cell Death Dis 2017; 8:e3078. [PMID: 28981114 PMCID: PMC5680583 DOI: 10.1038/cddis.2017.461] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/06/2017] [Accepted: 07/24/2017] [Indexed: 01/08/2023]
Abstract
Cancer is a multifactorial disease and virus-mediated carcinogenesis is one of the crucial factors, which is poorly understood. Human cytomegalovirus (HCMV) is a herpesvirus and its components have been evidenced to be associated with cancer of different tissue origin. However, its role in cancer remains unknown. Here, we identified a conserved herpesviral tegument protein known as pUL48 of HCMV, encoding deubiquitinase enzyme, as having a key role in carcinogenesis. We show using deubiquitinase sufficient- and deficient-HCMV that HCMV deubiquitinase is a key in inducing enhanced cellular metabolic activity through upregulation of several anti-apoptotic genes and downregulation of several pro-apoptotic genes expression. Furthermore, HCMV deubiquitinase acquires pro-tumor functions by inhibiting PRR-mediated type I interferon via deubiquitination of TRAF6, TRAF3, IRAK1, IRF7 and STING. Taken together, our results suggest that HCMV infection may promote oncogenesis by inhibiting innate immunity of the host.
Collapse
|
4
|
Meissl K, Macho-Maschler S, Müller M, Strobl B. The good and the bad faces of STAT1 in solid tumours. Cytokine 2015; 89:12-20. [PMID: 26631912 DOI: 10.1016/j.cyto.2015.11.011] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 12/13/2022]
Abstract
Signal transducer and activator of transcription (STAT) 1 is part of the Janus kinase (JAK)/STAT signalling cascade and is best known for its essential role in mediating responses to all types of interferons (IFN). STAT1 regulates a variety of cellular processes, such as antimicrobial activities, cell proliferation and cell death. It exerts important immune modulatory activities both in the innate and the adaptive arm of the immune system. Based on studies in mice and data from human patients, STAT1 is generally considered a tumour suppressor but there is growing evidence that it can also act as a tumour promoter. This review aims at contrasting the two faces of STAT1 in tumourigenesis and providing an overview on the current knowledge of the underlying mechanisms or pathways.
Collapse
Affiliation(s)
- Katrin Meissl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Sabine Macho-Maschler
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria.
| |
Collapse
|
5
|
Wandrer F, Falk CS, John K, Skawran B, Manns MP, Schulze-Osthoff K, Bantel H. Interferon-Mediated Cytokine Induction Determines Sustained Virus Control in Chronic Hepatitis C Virus Infection. J Infect Dis 2015; 213:746-54. [PMID: 26503984 DOI: 10.1093/infdis/jiv505] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/08/2015] [Indexed: 12/14/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a major cause of chronic liver disease and associated complications such as liver cirrhosis and hepatocellular carcinoma. Interferons (IFNs) are crucial for HCV clearance and a sustained virological response (SVR), but a significant proportion of patients do not respond to IFNα. The underlying mechanisms of an insufficient IFN response remain largely unknown. In this study, we found that patients responding to IFNα with viral clearance had significantly higher serum levels of TNF-related apoptosis inducing ligand (TRAIL), compared with patients who failed to control HCV. In addition, upon direct IFNα exposure, peripheral blood mononuclear cells (PBMCs) from patients with SVR upregulated TRAIL, as well as IFN-γ and the chemokines CXCL9 and CXCL10, much more strongly than cells from patients with antiviral treatment failure. As a possible mechanism of the stronger IFNα-induced cytokine response, we identified higher levels of expression and phosphorylation of the transcription factor STAT1 in PBMCs from patients with SVR. Increased TRAIL expression additionally involved the NF-κB and JNK signaling pathways. Thus, SVR in chronic HCV infection is associated with a strong IFNα-induced cytokine response, which might allow for the early prediction of treatment efficacy in HCV infection.
Collapse
Affiliation(s)
| | | | - Katharina John
- Department of Gastroenterology, Hepatology, and Endocrinology
| | - Britta Skawran
- Institute for Cell and Molecular Pathology, Hannover Medical School
| | - Michael P Manns
- Department of Gastroenterology, Hepatology, and Endocrinology
| | - Klaus Schulze-Osthoff
- Interfaculty Institute for Biochemistry, University of Tübingen German Cancer Consortium (DKTK)German Research Cancer Center (DKFZ), Heidelberg, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, and Endocrinology
| |
Collapse
|
6
|
Petersen SL, Chen TT, Lawrence DA, Marsters SA, Gonzalvez F, Ashkenazi A. TRAF2 is a biologically important necroptosis suppressor. Cell Death Differ 2015; 22:1846-57. [PMID: 25882049 DOI: 10.1038/cdd.2015.35] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/11/2015] [Accepted: 02/24/2015] [Indexed: 12/26/2022] Open
Abstract
Tumor necrosis factor α (TNFα) triggers necroptotic cell death through an intracellular signaling complex containing receptor-interacting protein kinase (RIPK) 1 and RIPK3, called the necrosome. RIPK1 phosphorylates RIPK3, which phosphorylates the pseudokinase mixed lineage kinase-domain-like (MLKL)-driving its oligomerization and membrane-disrupting necroptotic activity. Here, we show that TNF receptor-associated factor 2 (TRAF2)-previously implicated in apoptosis suppression-also inhibits necroptotic signaling by TNFα. TRAF2 disruption in mouse fibroblasts augmented TNFα-driven necrosome formation and RIPK3-MLKL association, promoting necroptosis. TRAF2 constitutively associated with MLKL, whereas TNFα reversed this via cylindromatosis-dependent TRAF2 deubiquitination. Ectopic interaction of TRAF2 and MLKL required the C-terminal portion but not the N-terminal, RING, or CIM region of TRAF2. Induced TRAF2 knockout (KO) in adult mice caused rapid lethality, in conjunction with increased hepatic necrosome assembly. By contrast, TRAF2 KO on a RIPK3 KO background caused delayed mortality, in concert with elevated intestinal caspase-8 protein and activity. Combined injection of TNFR1-Fc, Fas-Fc and DR5-Fc decoys prevented death upon TRAF2 KO. However, Fas-Fc and DR5-Fc were ineffective, whereas TNFR1-Fc and interferon α receptor (IFNAR1)-Fc were partially protective against lethality upon combined TRAF2 and RIPK3 KO. These results identify TRAF2 as an important biological suppressor of necroptosis in vitro and in vivo.
Collapse
Affiliation(s)
- S L Petersen
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - T T Chen
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - D A Lawrence
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - S A Marsters
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - F Gonzalvez
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - A Ashkenazi
- Cancer Immunology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
7
|
McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol 2015; 15:87-103. [PMID: 25614319 DOI: 10.1038/nri3787] [Citation(s) in RCA: 1911] [Impact Index Per Article: 191.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type I interferons (IFNs) have diverse effects on innate and adaptive immune cells during infection with viruses, bacteria, parasites and fungi, directly and/or indirectly through the induction of other mediators. Type I IFNs are important for host defence against viruses. However, recently, they have been shown to cause immunopathology in some acute viral infections, such as influenza virus infection. Conversely, they can lead to immunosuppression during chronic viral infections, such as lymphocytic choriomeningitis virus infection. During bacterial infections, low levels of type I IFNs may be required at an early stage, to initiate cell-mediated immune responses. High concentrations of type I IFNs may block B cell responses or lead to the production of immunosuppressive molecules, and such concentrations also reduce the responsiveness of macrophages to activation by IFNγ, as has been shown for infections with Listeria monocytogenes and Mycobacterium tuberculosis. Recent studies in experimental models of tuberculosis have demonstrated that prostaglandin E2 and interleukin-1 inhibit type I IFN expression and its downstream effects, demonstrating that a cross-regulatory network of cytokines operates during infectious diseases to provide protection with minimum damage to the host.
Collapse
Affiliation(s)
- Finlay McNab
- 1] Allergic Inflammation Discovery Performance Unit, Respiratory Disease Respiratory Research and Development, GlaxoSmithKline, Stevenage, Hertfordshire SG1 2NY, UK. [2] Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Katrin Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Andreas Wack
- Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Anne O'Garra
- 1] Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK. [2] National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
8
|
Fayyaz S, Yaylim I, Turan S, Kanwal S, Farooqi AA. Hepatocellular carcinoma: targeting of oncogenic signaling networks in TRAIL resistant cancer cells. Mol Biol Rep 2014; 41:6909-17. [PMID: 25037270 DOI: 10.1007/s11033-014-3577-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/01/2014] [Indexed: 01/18/2023]
Abstract
Apoptotic response in hepatocellular carcinoma (HCC) cells is impaired because of interconnectivity of proteins into complexes and signaling networks that are highly divergent in time and space. TNF-related apoptosis-inducing ligand (TRAIL) has emerged as an attractive anticancer agent reported to selectively induce apoptosis in cancer cells. Although diametrically opposed roles of TRAIL are reported both as an inducer of apoptosis and regulator of metastasis, overwhelmingly accumulating experimental evidence highlighting apoptosis inducing activity of TRAIL is directing TRAIL into clinical trials. Insights from TRAIL mediated signaling in HCC research are catalyzing new lines of study that should not only explain molecular mechanisms of disease but also highlight emerging paradigms in restoration of TRAIL mediated apoptosis in resistant cancer cells. It is becoming progressively more understandable that phytochemicals derived from edible plants have shown potential in modelling their interactions with their target proteins. Rapidly accumulating in vitro and in-vivo evidence indicates that phytonutrients have anticancer activity in rodent models of hepatocellular carcinoma. In this review we bring to limelight how phytonutrients restore apoptosis in hepatocellular carcinoma cells by rebalancing pro-apoptotic and anti-apoptotic proteins. Evidence has started to emerge, that reveals how phytonutrients target pharmacologically intractable proteins to suppress cancer. Target-based small-molecule discovery has entered into the mainstream research in the pharmaceutical industry and a better comprehension of the genetics of patients will be essential for identification of responders and non-responders.
Collapse
Affiliation(s)
- Sundas Fayyaz
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, Pakistan
| | | | | | | | | |
Collapse
|
9
|
Elia A, Powley IR, MacFarlane M, Clemens MJ. Modulation of the sensitivity of Jurkat T-cells to inhibition of protein synthesis by tumor necrosis factor α-related apoptosis-inducing ligand. J Interferon Cytokine Res 2014; 34:769-77. [PMID: 24731196 DOI: 10.1089/jir.2013.0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor α-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis in Jurkat T lymphoma cells. One of the characteristics of the phase preceding overt apoptosis is the marked downregulation of protein synthesis. We have investigated factors that can influence this response and have explored some of the signaling pathways involved. We show that interferon-α (IFNα) pretreatment desensitizes Jurkat cells to TRAIL-induced inhibition of protein synthesis, such that the concentration of TRAIL required for 50% inhibition is increased by 10-fold. The inhibition of translation is characterized by dephosphorylation of the eIF4E-binding protein 4E-BP1 and IFNα desensitizes Jurkat cells to this effect. IFNα also inhibits TRAIL-mediated dephosphorylation of the growth-promoting protein kinase B (Akt). Since Jurkat cells are defective for phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and therefore have constitutive phosphoinositide 3-kinase (PI3K) activity, we investigated the consequences for protein synthesis of inhibiting PI3K using LY294002. Inhibition of PI3K partially inhibits translation, but also enhances the effect of a suboptimal concentration of TRAIL. However, LY294002 does not block the ability of IFNα to protect protein synthesis from TRAIL-induced inhibition. Data are presented suggesting that IFNα impairs the process of activation of caspase-8 within the TRAIL death-inducing signaling complex.
Collapse
Affiliation(s)
- Androulla Elia
- 1 Translational Control Group, Molecular Cell Sciences Research Centre, St George's, University of London , London, United Kingdom
| | | | | | | |
Collapse
|
10
|
Hensley P, Mishra M, Kyprianou N. Targeting caspases in cancer therapeutics. Biol Chem 2013; 394:831-43. [PMID: 23509217 DOI: 10.1515/hsz-2013-0128] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 03/15/2013] [Indexed: 02/01/2023]
Abstract
The identification of the fundamental role of apoptosis in the growth balance and normal homeostasis against cell proliferation led to the recognition of its loss contributing to tumorigenesis. The mechanistic significance of reinstating apoptosis signaling towards selective targeting of malignant cells heavily exploits the caspase family of death-inducing molecules as a powerful therapeutic platform for the development of potent anticancer strategies. Some apoptosis inhibitors induce caspase expression and activity in preclinical models and clinical trials by targeting both the intrinsic and extrinsic apoptotic pathways and restoring the apoptotic capacity in human tumors. Furthermore, up-regulation of caspases emerges as a sensitizing mechanism for tumors exhibiting therapeutic resistance to radiation and adjuvant chemotherapy. This review provides a comprehensive discussion of the functional involvement of caspases in apoptosis control and the current understanding of reactivating caspase-mediated apoptosis signaling towards effective therapeutic modalities in cancer treatment.
Collapse
Affiliation(s)
- Patrick Hensley
- Department of Urology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536, USA
| | | | | |
Collapse
|
11
|
Shadrick WR, Mukherjee S, Hanson AM, Sweeney NL, Frick DN. Aurintricarboxylic acid modulates the affinity of hepatitis C virus NS3 helicase for both nucleic acid and ATP. Biochemistry 2013; 52:6151-9. [PMID: 23947785 DOI: 10.1021/bi4006495] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aurintricarboxylic acid (ATA) is a potent inhibitor of many enzymes needed for cell and virus replication, such as polymerases, helicases, nucleases, and topoisomerases. This study examines how ATA interacts with the helicase encoded by the hepatitis C virus (HCV) and reveals that ATA interferes with both nucleic acid and ATP binding to the enzyme. We show that ATA directly binds HCV helicase to prevent the enzyme from interacting with nucleic acids and to modulate the affinity of HCV helicase for ATP, the fuel for helicase action. Amino acid substitutions in the helicase DNA binding cleft or its ATP binding site alter the ability of ATA to disrupt helicase-DNA interactions. These data, along with molecular modeling results, support the notion that an ATA polymer binds between Arg467 and Glu493 to prevent the helicase from binding either ATP or nucleic acids. We also characterize how ATA affects the kinetics of helicase-catalyzed ATP hydrolysis, and thermodynamic parameters describing the direct interaction between HCV helicase and ATA using microcalorimetry. The thermodynamics of ATA binding to HCV helicase reveal that ATA binding does not mimic nucleic acid binding in that ATA binding is driven by a smaller enthalpy change and an increase in entropy.
Collapse
Affiliation(s)
- William R Shadrick
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee , 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| | | | | | | | | |
Collapse
|
12
|
Type I interferons induce apoptosis by balancing cFLIP and caspase-8 independent of death ligands. Mol Cell Biol 2012; 33:800-14. [PMID: 23230268 DOI: 10.1128/mcb.01430-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Interferons induce a pleiotropy of responses through binding the same cell surface receptor. Here we investigated the molecular mechanism driving interferon-induced apoptosis. Using a nonbiased small interfering RNA (siRNA) screen, we show that silencing genes whose products are directly engaged in the initiation of interferon signaling completely abrogate the interferon antiproliferative response. Apoptosis-related genes such as the caspase-8, cFLIP, and DR5 genes specifically interfere with interferon-induced apoptosis, which we found to be independent of the activity of death ligands. The one gene for which silencing resulted in the strongest proapoptotic effect upon interferon signaling is the cFLIP gene, where silencing shortened the time of initiation of apoptosis from days to hours and increased dramatically the population of apoptotic cells. Thus, cFLIP serves as a regulator for interferon-induced apoptosis. A shift over time in the balance between cFLIP and caspase-8 results in downstream caspase activation and apoptosis. While gamma interferon (IFN-γ) also causes caspase-8 upregulation, we suggest that it follows a different path to apoptosis.
Collapse
|
13
|
Liedtke C, Trautwein C. The role of TNF and Fas dependent signaling in animal models of inflammatory liver injury and liver cancer. Eur J Cell Biol 2011; 91:582-9. [PMID: 22153863 DOI: 10.1016/j.ejcb.2011.10.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/23/2011] [Accepted: 10/06/2011] [Indexed: 02/04/2023] Open
Abstract
Tumor Necrosis Factor (TNF) alpha is a pleiotropic cytokine triggering either pro-inflammatory effects via NF-κB related pathways or apoptosis through activation of caspase-8. The related death ligands Fas and TRAIL use homologous receptors and similar signaling cascades but predominantly induce apoptosis. Here, we summarize our experimental approaches to analyze the mechanisms and consequences of TNF and Fas signaling with the ultimate aim to define molecular targets for the treatment of inflammatory liver disease and liver cancer. By using conditional knockout technology in mice we genetically dissected the I-kappa B kinase (IKK) complex consisting of IKK1/IKKα, IKK2/IKKβ and IKKγ/NEMO. We demonstrated that IKK2/IKKβ, but not IKKγ/NEMO might be a promising target for the prevention of liver injury after ischemia and reperfusion or treating steatohepatitis. Genetic inactivation of IKKγ/NEMO defined a new animal model of spontaneous hepatitis and hepatocarcinogenesis involving constitutive activation of caspase-8 and basal apoptosis. We further show that caspase-8 is not only regulated by post-translational modifications as suggested earlier, but also by complex transcriptional regulation. Targeted stimulation of the caspase-8 promoter by interferons alpha and gamma, cytotoxic drugs or p53 can substantially sensitize hepatoma cells for apoptosis, whereas hepatocellular carcinoma frequently present an inactive caspase-8 gene promoter. In conclusion, our work demonstrates that therapeutic intervention in the TNF-NF-κB-caspase-8 network is technically feasible and could be of potential benefit in inflammatory liver disease.
Collapse
Affiliation(s)
- Christian Liedtke
- Department of Medicine III, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, D-52074 Aachen, Germany.
| | | |
Collapse
|
14
|
Zhao K, Wang X, Wang Y, Ma Y. Functional identification of a non-fusion TRAIL extracellular protein and preparation of its polyclonal antibody. Hybridoma (Larchmt) 2011; 30:217-21. [PMID: 21707355 DOI: 10.1089/hyb.2011.0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Human tumor necrosis factor related apoptosis inducing ligand (TRAIL) can selectively induce apoptosis in a variety of transformed cells and is currently being developed as a cancer therapeutic drug. Here we expressed the TRAIL protein including extracellular (114-281aa) without any tag protein named TRAIL-NT, and prepared anti-TRAIL polyclonal antibodies (Poly-Ab). The human TRAIL extracellular gene was amplified from PBMC and cloned into pGEM-T-Easy vector for sequence analysis. The expression vector pET-28a/TRAIL was constructed using the DNA recombinant method, and the recombinant protein without any tag protein was expressed in Escherichia coli BL21(DE3). The TRAIL-NT protein was purified by cation ion-exchange column and identified by SDS-PAGE and Western blot analysis. The proliferation inhibition activity of TRAIL-NT was detected by the MTT method, Wright-Giemsa staining assay, and FACS. The polyclonal antibody of TRAIL-NT was obtained after the BALB/C mice were immunized with purificated TRAIL-NT protein. Results showed that the target protein expressed in E. coli BL21(DE3) has the same molecular weight as that expected and could be recognized by anti-TRIAL Poly-Ab. The TRAIL-NT protein could also inhibit proliferation and induced apoptosis of Jurkat cells but no cytotoxicity to human liver cells and PBMC was observed. This preliminary research laid a solid foundation for further research on its biological activity and application in anti-tumor therapy.
Collapse
Affiliation(s)
- Kunpeng Zhao
- Institute of Immunology, Medical College, Henan University, PR China
| | | | | | | |
Collapse
|
15
|
Tanimoto Y, Tashiro H, Aikata H, Amano H, Oshita A, Kobayashi T, Kuroda S, Tazawa H, Takahashi S, Itamoto T, Chayama K, Ohdan H. Impact of pegylated interferon therapy on outcomes of patients with hepatitis C virus-related hepatocellular carcinoma after curative hepatic resection. Ann Surg Oncol 2011; 19:418-25. [PMID: 21710324 DOI: 10.1245/s10434-011-1866-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Indexed: 12/13/2022]
Abstract
BACKGROUND Several published reports investigating the effects of interferon (IFN) therapy on survival and tumor recurrence after curative resection of hepatocellular carcinoma (HCC) have been inconclusive. The aim of this study is to investigate the efficacy of pegylated-IFN (peg-IFN) therapy after curative hepatic resection for HCC in patients infected with hepatitis C virus (HCV). METHODS Data from 175 patients who underwent curative hepatic resection for HCC associated with HCV were retrospectively collected and analyzed; 75 patients received peg-IFN therapy after surgery, whereas 100 patients did not receive IFN therapy. To overcome biases resulting from the different distribution of covariates in the two groups, a one-to-one match was created using propensity score analysis. After matching, patient outcomes were analyzed. RESULTS After one-to-one matching, patients (n = 38) who received peg-IFN therapy after surgery and patients (n = 38) who did not receive IFN therapy had the same preoperative and operative characteristics. The 3- and 5-year overall survival rates of patients who received peg-IFN therapy after hepatic resection were significantly higher than those of patients who did not receive IFN therapy (P = 0.00135). The 3- and 5-year overall survival rates were 100 and 91.7% and 76.6 and 50.6% in the peg-IFN group and non-IFN group, respectively. There was no significant difference in disease-free survival between the two matched groups (P = 0.886). CONCLUSION Peg-IFN therapy may be effective as an adjuvant chemopreventive agent after hepatic resection in patients with HCV-related HCC.
Collapse
Affiliation(s)
- Yoshisato Tanimoto
- Department of Gastroenterological Surgery, Hiroshima University Hospital, Hiroshima, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Roos WP, Jöst E, Belohlavek C, Nagel G, Fritz G, Kaina B. Intrinsic Anticancer Drug Resistance of Malignant Melanoma Cells Is Abrogated by IFN-β and Valproic Acid. Cancer Res 2011; 71:4150-60. [DOI: 10.1158/0008-5472.can-10-3498] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
17
|
Lu G, Liu Y, Ji B, Wei F, Hao C, Wang G. Synergistic effect of celecoxib on TRAIL-induced apoptosis in hepatocellular carcinoma cells. Cancer Invest 2010; 28:629-634. [PMID: 20394499 DOI: 10.3109/07357900903095631] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is currently under clinical development as a cancer therapeutic because it can induce apoptosis selectively in cancer cells. The vast majority of hepatocellular carcinomas (HCC), however, are resistant to TRAIL. In search of cancer therapeutics that can overcome TRAIL resistance, we show here that celecoxib and camptothecin can sensitize TRAIL-resistant HCC cell lines, HepG2 and Hep3B, to TRAIL-induced apoptosis through downregulation of cellular Fas-associated death domain-like interleukin-1beta-converting enzyme-inhibitory protein (c-FLIP) and cleavage of caspase-8 and caspase-3 in the HCC cells. The study suggests a framework for TRAIL-based combination treatment of HCC.
Collapse
Affiliation(s)
- Guoyue Lu
- Department of General Surgery, The First Clinical Hospital, Jilin University, 71 Xinmin Street, Changchun, Jilin 130021, People's Republic of China
| | | | | | | | | | | |
Collapse
|
18
|
Tomimaru Y, Nagano H, Eguchi H, Kobayashi S, Marubashi S, Wada H, Tanemura M, Umeshita K, Hiramatsu N, Takehara T, Doki Y, Mori M. Effects of preceding interferon therapy on outcome after surgery for hepatitis C virus-related hepatocellular carcinoma. J Surg Oncol 2010; 102:308-14. [DOI: 10.1002/jso.21633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
19
|
Abstract
A number of tumors are still resistant to the antiproliferative activity of human interferon (IFN)-alpha. The Janus kinases/Signal Transducers and Activators of Transcription (JAK-STAT) pathway plays an important role in initial IFN signaling. To enhance the antiproliferative activity of IFN-alpha, it is important to elucidate which factors in the JAK-STAT pathway play a key role in eliciting this activity. In human ovarian adenocarcinoma OVCAR3 cells sensitive to both IFN-alpha and IFN-gamma, only IFN regulatory factor 9 (IRF9)-RNA interference (RNAi) completely inhibited the antiproliferative activity of IFN-alpha among the intracellular JAK-STAT pathway factors. Conversely, Stat1-RNAi did not inhibit the antiproliferative activity of IFN-alpha, whereas it partially inhibited that of IFN-gamma. As a cell death pathway, it is reported that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis through TRAIL-receptor (R) 1 and TRAIL-R2. In IFN-alpha-treated OVCAR3 cells, IRF9-RNAi inhibited transcription of TRAIL whereas Stat1-RNAi did not, suggesting that the transcription of TRAIL induced by IFN-alpha predominantly required IRF9. Furthermore, IFN-stimulated response element-like motifs of TRAIL bound to IFN-stimulated gene factor 3 (ISGF3) complex after IFN-alpha treatment. Subsequently, TRAIL-R2-RNAi inhibited both antiproliferative activities of IFN-alpha and TRAIL, suggesting that TRAIL-R2 mediated both IFN-alpha and TRAIL signals to elicit their antiproliferative activities. Finally, IRF9 overexpression facilitated IFN-alpha-induced apoptosis in T98G (human glioblastoma multiforme) cells, which were resistant to IFN-alpha. Thus, this study suggests that IRF9 is the key factor for eliciting the antiproliferative activity of IFN-alpha and TRAIL may be one of the potential mediators.
Collapse
|
20
|
Yang S, Wu X. Identification and functional characterization of a human sTRAIL homolog, CasTRAIL, in an invertebrate oyster Crassostrea ariakensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2010; 34:538-545. [PMID: 20045024 DOI: 10.1016/j.dci.2009.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Revised: 12/23/2009] [Accepted: 12/23/2009] [Indexed: 05/28/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2L) is one of the tumor necrosis factor (TNF) superfamily members, participating in many biological processes including apoptosis and immune responses. In present study, a novel human soluble TRAIL (sTRAIL) homolog, CasTRAIL was identified from the oyster, Crassostrea ariakensis. CasTRAIL has a 99% and 98% similarity to human sTRAIL over the cDNA sequence and the amino acid sequence, respectively. It mostly distributes in tissues of the oyster defense system and was mainly localized at cell membrane, and has no cytotoxicity to normal hemocytes of oyster. The phosphorylation state of MAP kinases revealed that CasTRAIL induced a rapid increase in the phospho-ERK and phospho-p38 levels, which indicated that the MAPK pathway was involved in CasTRAIL-mediated signaling. In addition, CasTRAIL also showed an ability of anti-RLO infection which might be through the p38-MAPK activation pathway. Present studies provide an understanding and insight of the biological functions of CasTRAIL.
Collapse
Affiliation(s)
- Shoubao Yang
- Laboratory of Marine Life Science and Technology, College of Animal Sciences, Zhejiang University, No. 268 Kaixuan Road, Hangzhou, Zhejiang, China
| | | |
Collapse
|
21
|
Tong H, Lu C, Yang Y, Zhang J, Zhang J. Role of caspase 8 as a determinant in trail sensitivity of neuroblastoma cell lines. Pediatr Hematol Oncol 2009; 26:549-59. [PMID: 19954364 DOI: 10.3109/08880010903271663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Resistance of neuroblastoma (NB) cells to tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-mediated apoptosis is thought to be caused by loss of caspase 8 expression. The aim of this study is to investigate if induction of caspase 8 by gamma-interferon (IFN gamma) renders NB cells sensitive to TRAIL. METHODS Caspase 8 expression was monitored by RT-PCR and Western blot analysis. The effects of IFN gamma, TRAIL, IFN gamma + TRAIL, and caspase 8 inhibitor + TRAIL on the growth and apoptosis of NB cells were detected with the methods of reduction rate of Alamar blue assay and flow cytometry. The relative caspase 8 activity was measured with colorimetric assay. RESULTS An increased expression of caspase 8 mRNA and protein was found after treatment with IFN gamma. IFN gamma in combination with TRAIL decreased proliferation of NB cell line SH-SY5Y cells. The killing effect of TRAIL on NB cells expressing caspase 8 was depressed by caspase 8 inhibitor. The relative caspase 8 activity of NB cells expressing caspase 8 increased with the prolongation of TRAIL action time. CONCLUSIONS TRAIL induced apoptosis in NB cells expressing caspase 8.
Collapse
Affiliation(s)
- Haixia Tong
- Laboratory of Hematology, Shengjing Hospital of China Medical University, Shenyang, China.
| | | | | | | | | |
Collapse
|
22
|
Fulda S. Caspase-8 in cancer biology and therapy. Cancer Lett 2009; 281:128-33. [DOI: 10.1016/j.canlet.2008.11.023] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 11/12/2008] [Accepted: 11/13/2008] [Indexed: 02/08/2023]
|
23
|
HCV induces oxidative and ER stress, and sensitizes infected cells to apoptosis in SCID/Alb-uPA mice. PLoS Pathog 2009; 5:e1000291. [PMID: 19242562 PMCID: PMC2647842 DOI: 10.1371/journal.ppat.1000291] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Accepted: 01/08/2009] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) is a blood-borne pathogen and a major cause of liver disease worldwide. Gene expression profiling was used to characterize the transcriptional response to HCV H77c infection. Evidence is presented for activation of innate antiviral signaling pathways as well as induction of lipid metabolism genes, which may contribute to oxidative stress. We also found that infection of chimeric SCID/Alb-uPA mice by HCV led to signs of hepatocyte damage and apoptosis, which in patients plays a role in activation of stellate cells, recruitment of macrophages, and the subsequent development of fibrosis. Infection of chimeric mice with HCV H77c also led an inflammatory response characterized by infiltration of monocytes and macrophages. There was increased apoptosis in HCV-infected human hepatocytes in H77c-infected mice but not in mice inoculated with a replication incompetent H77c mutant. Moreover, TUNEL reactivity was restricted to HCV-infected hepatocytes, but an increase in FAS expression was not. To gain insight into the factors contributing specific apoptosis of HCV infected cells, immunohistological and confocal microscopy using antibodies for key apoptotic mediators was done. We found that the ER chaperone BiP/GRP78 was increased in HCV-infected cells as was activated BAX, but the activator of ER stress-mediated apoptosis CHOP was not. We found that overall levels of NF-kappaB and BCL-xL were increased by infection; however, within an infected liver, comparison of infected cells to uninfected cells indicated both NF-kappaB and BCL-xL were decreased in HCV-infected cells. We conclude that HCV contributes to hepatocyte damage and apoptosis by inducing stress and pro-apoptotic BAX while preventing the induction of anti-apoptotic NF-kappaB and BCL-xL, thus sensitizing hepatocytes to apoptosis.
Collapse
|
24
|
Tong HX, Lu CW, Zhang JH, Zhang JH. Enhanced effect of IFNgamma on the induced apoptosis of neuroblastoma cells by cytotoxic drugs. Pediatr Hematol Oncol 2008; 25:549-58. [PMID: 18728974 DOI: 10.1080/08880010802244092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The objective of this study was to explore the antitumor effects of cytotoxic drugs combined with IFNgamma on neuroblastoma cell line SH-SY5Y cells. The expression of caspase 8 mRNA and protein was detected with RT-PCR and Western blot analysis. The effects of cytotoxic drugs and IFNgamma combined with cytotoxic drugs on the growth and apoptosis of SH-SY5Y cells were detected with the methods of MTT and flow cytometry. Caspase 8 activity was measured by colorimetric assay. Caspase 8 was undetectable in SH-SY5Y cells with an increased expression of caspase 8 after the treatment of IFNgamma. SH-SY5Y cells were sensitive to Adriamycin relatively but resistant to TNFalpha and TRAIL, while IFNgamma-pretreated SH-SY5Y cells were more sensitive to the cytotoxic drugs with an increase of caspase 8 activity. The authors conclude that IFNgamma can sensitize SH-SY5Y cells to Adriamycin-, TNFalpha-, and TRAIL-induced apoptosis and this may be realized by the upregulation of caspase 8.
Collapse
Affiliation(s)
- Hai-xia Tong
- Laboratory of Hematology, Shengjing Hospital of China Medical University, Shenyang, China.
| | | | | | | |
Collapse
|
25
|
Dzieran J, Beck JF, Sonnemann J. Differential responsiveness of human hepatoma cells versus normal hepatocytes to TRAIL in combination with either histone deacetylase inhibitors or conventional cytostatics. Cancer Sci 2008; 99:1685-92. [PMID: 18754884 PMCID: PMC11158644 DOI: 10.1111/j.1349-7006.2008.00868.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising candidate for the treatment of cancer because it elicits cell death in many tumor cells while sparing most normal cells. Liver cancer, however, is largely resistant to TRAIL and, thus, requires sensitization for TRAIL-mediated cytotoxicity. Sensitization may be achieved by cotreatment with chemotherapeutic agents. In this study, we comparatively investigated the treatment efficacy of TRAIL in combination with histone deacetylase inhibitors (HDI) versus TRAIL in combination with conventional cytostatics in the hepatocellular carcinoma cell line HepG2 and in the childhood hepatoblastoma cell line Huh6. We found that TRAIL resistance could be overcome by cotreatment with the HDI vorinostat, sodium butyrate and MS-275, but not by cotreatment with the cytostatics carboplatin and etoposide. However, TRAIL combination treatment bears the risk of sensitizing otherwise TRAIL-resistant normal cells. We thus explored a potential cytotoxic effect of combined HDI/TRAIL treatment in normal hepatocytes: TRAIL in conjunction with HDI did not impose any cytotoxicity on the non-malignant cells. In searching for the determinants of HDI-mediated TRAIL sensitization in hepatoma cells, we observed that HDI treatment did not increase cell-surface expression of proapoptotic TRAIL receptors. Instead, HDI treatment enhanced TRAIL-induced cleavage of Bid. In conclusion, our data suggest that HDI are potent sensitizers to TRAIL in hepatoma cells and that the combination of HDI and TRAIL is selectively active in hepatoma cells without affecting normal hepatocytes, indicating that the combination of HDI and TRAIL may be an effective approach for the treatment of advanced liver cancer.
Collapse
Affiliation(s)
- Johanna Dzieran
- Research Center of Pharmacology and Experimental Therapeutics, Ernst Moritz Arndt University, Greifswald, Germany
| | | | | |
Collapse
|
26
|
Piekarska A, Kubiak R, Omulecka A, Szymczak W, Piekarski J. Expression of tumour necrosis factor-related apoptosis-inducing ligand and caspase-3 in relation to grade of inflammation and stage of fibrosis in chronic hepatitis C. Histopathology 2007; 51:597-604. [PMID: 17927580 DOI: 10.1111/j.1365-2559.2007.02863.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIM To assess whether the distribution of the recently described proapoptotic ligand, tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), and the apoptosis effector, caspase-3 alters with the degree of inflammation and fibrosis present in liver biopsy specimens from patients with chronic hepatitis C virus infection. METHODS AND RESULTS Expression of TRAIL and caspase-3 was assessed immunohistochemically in liver biopsy specimens obtained from 89 adults with chronic hepatitis C. Expression of TRAIL in hepatocytes correlated inversely with stage of fibrosis (P = 0.001), classified according to the Scheuer score; expression of caspase-3 in hepatocytes correlated with grade of inflammation (P = 0.012). Expression of TRAIL in hepatocytes was not correlated with grade of inflammation (P > 0.05); expression of caspase-3 was not correlated with stage of fibrosis (P > 0.05). Maximum expression of proapoptotic TRAIL protein was observed in cases with low grade inflammation (G0) and low stage fibrosis (S1). Maximum expression of caspase-3 in hepatocytes was observed in cases with high grade inflammation (G3-4) and high stage fibrosis (S3), but not with liver cirrhosis (S4). CONCLUSIONS There is a significant decrease in TRAIL expression with increasing grade of inflammation, whereas caspase-3 expression is significantly increased with advanced fibrosis, short of cirrhosis.
Collapse
Affiliation(s)
- A Piekarska
- Department of Infectious Diseases and Hepatology, Chair of Infectious Diseases, Medical University of Lodz, Lodz, Poland.
| | | | | | | | | |
Collapse
|
27
|
Shashkova EV, Kuppuswamy MN, Wold WSM, Doronin K. Anticancer activity of oncolytic adenovirus vector armed with IFN-alpha and ADP is enhanced by pharmacologically controlled expression of TRAIL. Cancer Gene Ther 2007; 15:61-72. [PMID: 17992200 DOI: 10.1038/sj.cgt.7701107] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have previously described oncolytic adenovirus (Ad) vectors KD3 and KD3-interferon (IFN) that were rendered cancer-specific by mutations in the E1A region of Ad; these mutations abolish binding of E1A proteins to p300/CBP and pRB. The antitumor activity of the vectors was enhanced by overexpression of the Adenovirus Death Protein (ADP, E3-11.6K) and by replication-linked expression of IFN-alpha. We hypothesized that the anticancer efficacy of the KD3-IFN vector could be further improved by expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). E1-deleted Ad vectors were constructed carrying reporter genes for enhanced green fluorescent protein or secreted placental alkaline phosphatase (SEAP) and a therapeutic gene for TRAIL under control of the TetON system. Expression of the genes was increased in the presence of a helper virus and the inducer doxycycline such that up to 231-fold activation of expression for the TetON-SEAP vector was obtained. Coinfection with TetON-TRAIL augmented oncolytic activity of KD3 and KD3-IFN in vitro. Induction of TRAIL expression did not reduce the yield of progeny virus. Combination of TetON-TRAIL and KD3-IFN produced superior antitumor activity in vivo as compared with either vector alone demonstrating the efficacy of a four-pronged cancer gene therapy approach, which includes Ad oncolysis, ADP overexpression, IFN-alpha-mediated immunotherapy, and pharmacologically controlled TRAIL activity.
Collapse
|
28
|
Phenotypic characterization of resistant Val36 variants of hepatitis C virus NS3-4A serine protease. Antimicrob Agents Chemother 2007; 52:110-20. [PMID: 17938182 DOI: 10.1128/aac.00863-07] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In patients chronically infected with hepatitis C virus (HCV) strains of genotype 1, rapid and dramatic antiviral activity has been observed with telaprevir (VX-950), a highly selective and potent inhibitor of the HCV NS3-4A serine protease. HCV variants with substitutions in the NS3 protease domain were observed in some patients during telaprevir dosing. In this study, purified protease domain proteins and reconstituted HCV subgenomic replicons were used for phenotypic characterization of many of these substitutions. V36A/M or T54A substitutions conferred less than eightfold resistance to telaprevir. Variants with double substitutions at Val36 plus Thr54 had approximately 20-fold resistance to telaprevir, and variants with double substitutions at Val36 plus Arg155 or Ala156 had >40-fold resistance to telaprevir. An X-ray structure of the HCV strain H protease domain containing the V36M substitution in a cocomplex with an NS4A cofactor peptide was solved at a 2.4-A resolution. Except for the side chain of Met36, the V36M variant structure is identical to that of the wild-type apoenzyme. The in vitro replication capacity of most variants was significantly lower than that of the wild-type replicon in cells, which is consistent with the impaired in vivo fitness estimated from telaprevir-dosed patients. Finally, the sensitivity of these replicon variants to alpha interferon or ribavirin remained unchanged compared to that of the wild-type.
Collapse
|
29
|
Molecular mechanism of Mitomycin C-dependent caspase-8 regulation: implications for apoptosis and synergism with interferon-α signalling. Apoptosis 2007; 12:2259-70. [DOI: 10.1007/s10495-007-0145-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
30
|
Antitumor effect of interferon-on U937 human acute leukemia cells in vitro and its molecular mechanism. ACTA ACUST UNITED AC 2007; 27:513-5. [DOI: 10.1007/s11596-007-0509-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Indexed: 10/22/2022]
|
31
|
Kronenberger B, Zeuzem S, Sarrazin C, Mihm U, Von Wagner M, Hofmann WP, Piiper A, Herrmann E. Dynamics of Apoptotic Activity during Antiviral Treatment of Patients with Chronic Hepatitis C. Antivir Ther 2007. [DOI: 10.1177/135965350701200503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Introduction Cell death during antiviral therapy of patients with chronic hepatitis C is not well understood. Methods In the present study, apoptotic activity was monitored by quantification of apoptotic cytokeratin-18 neoepitopes in serum from patients with chronic hepatitis C before and 4, 12, 24 and 48 weeks after initiation of antiviral therapy with pegylated interferon-α2a and ribavirin and was compared with viral kinetic parameters. Results After 4 weeks of treatment apoptotic activity decreased significantly compared with baseline. Later during treatment, however, apoptotic activity increased again to levels similar to baseline. Alanine aminotransferase (ALT) activity also showed a significant decrease at week 4 compared with baseline but, in contrast to apoptotic activity, ALT remained at a reduced level during the treatment period. Baseline apoptotic activity was inversely correlated with the infected cell loss while an increase of apoptotic activity within the first 4 treatment weeks compared with baseline was positively correlated with the infected cell loss. Conclusions Apoptosis appears to be an important form of cell death during interferon-α-based treatment and is associated with infected cell loss and underestimated by ALT activity.
Collapse
Affiliation(s)
- Bernd Kronenberger
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt/Main, Germany
| | - Stefan Zeuzem
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt/Main, Germany
| | - Christoph Sarrazin
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt/Main, Germany
| | - Ulrike Mihm
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt/Main, Germany
| | - Michael Von Wagner
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt/Main, Germany
| | - W Peter Hofmann
- Medizinische Klinik I, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt/Main, Germany
| | - Albrecht Piiper
- Innere Medizin, Medizinische Fakultät, Universität des Saarlandes, Homburg/Saar, Germany
| | - Eva Herrmann
- Innere Medizin, Medizinische Fakultät, Universität des Saarlandes, Homburg/Saar, Germany
| |
Collapse
|
32
|
Zhang X, Chen ZG, Khuri FR, Shin DM. Induction of cell cycle arrest and apoptosis by a combined treatment with 13-cis-retinoic acid, interferon-α2a, and α-tocopherol in squamous cell carcinoma of the head and neck. Head Neck 2007; 29:351-61. [PMID: 17163463 DOI: 10.1002/hed.20525] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND We have previously conducted phase II trials with a combination of 13-cis-retinoic acid (13-cRA), interferon-alpha2a (IFN-alpha2a), and alpha-tocopherol (alpha-TF) in patients with advanced oral premalignant lesions and locally advanced head and neck cancer in the adjuvant settings and achieved promising outcomes. The present study was conducted in vitro to elucidate the mechanisms of anti-tumor activity of this 3-drug combination in squamous cell carcinoma of the head and neck (SCCHN). METHODS Five SCCHN cell lines were treated with 13-cRA, IFN-alpha2a, and alpha-TF as single agents or 2- to 3-drug combinations for 72 hours. Inhibition of cell growth and cell cycle progression and induction of apoptosis by the treatments were evaluated. RESULTS Our results demonstrated that although each single-agent and 2-drug combination showed a certain level of cell growth inhibition, the 3-drug combination apparently further inhibited cell growth in comparison to any single agents and 2-drug combinations in the 5 SCCHN cell lines. Cell cycle analysis on Tu212 and 886LN cells by flow cytometry exhibited significant accumulation of the cells at S phase in the 3-drug combination. On the other hand, Annexin-V binding assay demonstrated that the 3-drug combination induced more profound apoptosis than any of the single agents or 2-drug combinations. In parallel, proteolytic cleavages of pro-caspase-8, -9, -3 and poly (ADP ribose) polymerase as well as caspase-3 activity induced by the 3-drug treatment were observed. CONCLUSIONS Our data suggests that 3-drug combination biochemopreventive regimen has cooperative inhibitory effect on the growth of SCCHN cells. Both cell cycle arrest and apoptosis contribute to cell growth inhibition of this 3-drug combination therapy.
Collapse
Affiliation(s)
- Xin Zhang
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | |
Collapse
|
33
|
Jeffrey IW, Elia A, Bornes S, Tilleray VJ, Gengatharan K, Clemens MJ. Interferon-alpha induces sensitization of cells to inhibition of protein synthesis by tumour necrosis factor-related apoptosis-inducing ligand. FEBS J 2006; 273:3698-708. [PMID: 16911520 DOI: 10.1111/j.1742-4658.2006.05374.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tumour cells are often sensitized by interferons to the effects of tumour necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL). We have demonstrated previously that TRAIL has an inhibitory effect on protein synthesis [Jeffrey IW, Bushell M, Tilleray VJ, Morley S & Clemens MJ (2002) Cancer Res62, 2272-2280] and we have therefore examined the consequences of prior interferon-alpha treatment for the sensitivity of translation to inhibition by TRAIL. Interferon treatment alone has only a minor effect on protein synthesis but it sensitizes both MCF-7 cells and HeLa cells to the downregulation of translation by TRAIL. The inhibition of translation is characterized by increased phosphorylation of the alpha subunit of eukaryotic initiation factor eIF2 and dephosphorylation of the eIF4E-binding protein 4E-BP1. Both of these effects, as well as the decrease in overall protein synthesis, require caspase-8 activity, although they precede overt apoptosis by several hours. Interferon-alpha enhances the level and/or the extent of activation of caspase-8 by TRAIL, thus providing a likely explanation for the sensitization of cells to the inhibition of translation.
Collapse
Affiliation(s)
- Ian W Jeffrey
- Translational Control Group, Centre for Molecular and Metabolic Signalling, Division of Basic Medical Sciences, St George's, University of London, UK
| | | | | | | | | | | |
Collapse
|