51
|
Romanenko AM, Kinoshita A, Wanibuchi H, Wei M, Zaparin WK, Vinnichenko WI, Vozianov AF, Fukushima S. Involvement of ubiquitination and sumoylation in bladder lesions induced by persistent long-term low dose ionizing radiation in humans. J Urol 2006; 175:739-43. [PMID: 16407042 DOI: 10.1016/s0022-5347(05)00172-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Indexed: 11/18/2022]
Abstract
PURPOSE We determined whether ubiquitination and sumoylation processes are up-regulated in bladder urothelium by chronic, long-term, persistent low doses of ionizing radiation in male patients with benign prostate hyperplasia and females with chronic cystitis living more than 19 years in 137Cs contaminated areas after the Chernobyl accident in Ukraine. MATERIALS AND METHODS Bladder urothelial biopsies from 45 patients were subjected to histopathological and immunohistochemical study of Ub, SUMO1, SUMO E2 conjugating enzyme Ubc9, and the cell cycle inhibitors p53 and p27(Kip1). RESULTS Of 25 group 1 patients from radio contaminated areas chronic proliferative atypical cystitis (Chernobyl cystitis), featuring multiple foci of dysplasia, and carcinoma in situ were observed in 23 (92%) and 19 (76%), respectively, in addition to 1 small pTa grade 1 urothelial carcinoma. Chronic cystitis with areas of dysplasia and urothelial hyperplasia were detected in 2 (10%) and 3 (15%), respectively of the 20 patients in control group 2 from clean (without radio contamination) areas of Ukraine. Greatly increased levels of Ub, SUMO1, Ubc9 and p53 as well as decreased levels of p27(Kip1) were evident in patients in group 1 compared to those in group 2 (all p <0.001). CONCLUSIONS These findings support the hypothesis that up-regulated ubiquitination and sumoylation processes might be an adaptive response to unscheduled proteolysis of aberrant p53 and p27(Kip1) cell cycle regulators occurring with long-term low dose rate ionizing radiation exposure with a possible contribution to urothelial carcinogenesis.
Collapse
Affiliation(s)
- Alina M Romanenko
- Department of Pathology, Institute of Urology, Academy of Medical Sciences of Ukraine, Ukraine
| | | | | | | | | | | | | | | |
Collapse
|
52
|
Edelman MJ. The potential role of bortezomib in combination with chemotherapy and radiation in non-small-cell lung cancer. Clin Lung Cancer 2006; 7 Suppl 2:S64-6. [PMID: 16250930 DOI: 10.3816/clc.2005.s.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The combination of chemotherapy and radiation has been validated for the treatment of locally advanced non-small-cell lung cancer (NSCLC). However, the results are still unsatisfactory, and there is a need to improve current treatment. One approach is to use new agents that have the potential to enhance the efficacy of chemotherapy, radiation therapy (RT), or both. One potential target is the ubiquitin-proteasome pathway. This pathway plays an essential role in the degradation of most short- and long-lived intracellular proteins in eukaryotic cells and therefore regulating the cell cycle, neoplastic growth, and metastasis. Bortezomib is a selective 26S proteasome inhibitor that has been approved for the treatment of multiple myeloma. Bortezomib has demonstrated in vitro chemotherapy- and RT-sensitizing properties as well as single-agent activity in lung cancer. This article will review the rationale for the use of bortezomib as part of the chemotherapy/RT strategy for the treatment of NSCLC.
Collapse
Affiliation(s)
- Martin J Edelman
- University of Maryland Greenebaum Cancer Center, Baltimore, 21201, USA.
| |
Collapse
|
53
|
Abstract
Exposure of cells to ionizing radiation slows the rate of degradation of substrates through the proteasome. Because the 26S proteasome degrades most short-lived cellular proteins, changes in its activity might significantly, and selectively, alter the life span of many signaling proteins and play a role in promoting the biological consequences of radiation exposure, such as cell cycle arrest, DNA repair, and apoptosis. Experiments were therefore undertaken to identify the radiation target that is associated with the proteasome. Regardless of whether they were irradiated before or after extraction and purification from human prostate cancer PC3 cells, 26S proteasomes remained intact but showed a rapid 30% to 50% dose-independent decrease in their three major enzymatic activities following exposure to 1 to 20 Gy. There was no effect on 20S proteasomes, suggesting that the radiation-sensitive target is located in the 19S cap of the 26S proteasome, rather than in the enzymatically active core. Because the base of the 19S cap contains an ATPase ring that mediates substrate unfolding, pore opening, and translocation of substrates into the catalytic chamber, we examined whether the ATPase activity of purified 26S proteasomes was affected. In fact, in vitro irradiation of proteasomes enhanced their ATPase activity. Furthermore, pretreatment with low concentrations of the free radical scavenger tempol was able to prevent both the radiation-induced decrease in proteolytic activity and the increase in ATP utilization, indicating that free radicals are mediators of these radiation-induced phenomena. Finally, we have shown that cell irradiation results in the accumulation of proteasome substrates: polyubiquitinated proteins and ornithine decarboxylase, indicating that the observed decrease in proteasome function is physiologically relevant.
Collapse
Affiliation(s)
- Milena Pervan
- Department of Radiation Oncology, Roy E. Coats Research Laboratories, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA 90095-1714, USA
| | | | | |
Collapse
|
54
|
Fekete MR, McBride WH, Pajonk F. Anthracyclines, proteasome activity and multi-drug-resistance. BMC Cancer 2005; 5:114. [PMID: 16159384 PMCID: PMC1242219 DOI: 10.1186/1471-2407-5-114] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Accepted: 09/13/2005] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND P-glycoprotein is responsible for the ATP-dependent export of certain structurally unrelated compounds including many chemotherapeutic drugs. Amplification of P-glycoprotein activity can result in multi-drug resistance and is a common cause of chemotherapy treatment failure. Therefore, there is an ongoing search for inhibitors of P-glycoprotein. Observations that cyclosporin A, and certain other substances, inhibit both the proteasome and P-glycoprotein led us to investigate whether anthracyclines, well known substrates of P-gp, also inhibit the function of the proteasome. METHODS Proteasome function was measured in cell lysates from ECV304 cells incubated with different doses of verapamil, doxorubicin, daunorubicin, idarubicin, epirubicin, topotecan, mitomycin C, and gemcitabine using a fluorogenic peptide assay. Proteasome function in living cells was monitored using ECV304 cells stably transfected with the gene for an ubiquitin/green fluorescent protein fusion protein. The ability of the proteasome inhibitor MG-132 to affect P-glycoprotein function was monitored by fluorescence due to accumulation of daunorubicin in P-glycoprotein overexpressing KB 8-5 cells. RESULTS Verapamil, daunorubicin, doxorubicin, idarubicin, and epirubicin inhibited 26S chymotrypsin-like function in ECV304 extracts in a dose-dependent fashion. With the exception of daunorubicin, 20S proteasome function was also suppressed. The proteasome inhibitor MG-132 caused a dose-dependent accumulation of daunorubicin in KB 8-5 cells that overexpress P-glycoprotein, suggesting that it blocked P-glycoprotein function. CONCLUSION Our data indicate that anthracyclines inhibit the 26S proteasome as well as P-glycoprotein. Use of inhibitors of either pathway in cancer therapy should take this into consideration and perhaps use it to advantage, for example during chemosensitization by proteasome inhibitors.
Collapse
Affiliation(s)
- Mirela R Fekete
- Department of Neurology, Bürgerhospital, Tunzhofer Str. 14-16, 70191 Stuttgart, Germany
| | - William H McBride
- Department of Radiation Oncology, Roy E. Coats Labs., David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA90095-1714, USA
| | - Frank Pajonk
- Department of Radiation Oncology, Roy E. Coats Labs., David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA90095-1714, USA
| |
Collapse
|
55
|
Pajonk F, van Ophoven A, Weissenberger C, McBride WH. The proteasome inhibitor MG-132 sensitizes PC-3 prostate cancer cells to ionizing radiation by a DNA-PK-independent mechanism. BMC Cancer 2005; 5:76. [PMID: 16001975 PMCID: PMC1177933 DOI: 10.1186/1471-2407-5-76] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Accepted: 07/07/2005] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND By modulating the expression levels of specific signal transduction molecules, the 26S proteasome plays a central role in determining cell cycle progression or arrest and cell survival or death in response to stress stimuli, including ionizing radiation. Inhibition of proteasome function by specific drugs results in cell cycle arrest, apoptosis and radiosensitization of many cancer cell lines. This study investigates whether there is also a concomitant increase in cellular radiosensitivity if proteasome inhibition occurs only transiently before radiation. Further, since proteasome inhibition has been shown to activate caspase-3, which is involved in apoptosis, and caspase-3 can cleave DNA-PKcs, which is involved in DNA-double strand repair, the hypothesis was tested that caspase-3 activation was essential for both apoptosis and radiosensitization following proteasome inhibition. METHODS Prostate carcinoma PC-3 cells were treated with the reversible proteasome inhibitor MG-132. Cell cycle distribution, apoptosis, caspase-3 activity, DNA-PKcs protein levels and DNA-PK activity were monitored. Radiosensitivity was assessed using a clonogenic assay. RESULTS Inhibition of proteasome function caused cell cycle arrest and apoptosis but this did not involve early activation of caspase-3. Short-time inhibition of proteasome function also caused radiosensitization but this did not involve a decrease in DNA-PKcs protein levels or DNA-PK activity. CONCLUSION We conclude that caspase-dependent cleavage of DNA-PKcs during apoptosis does not contribute to the radiosensitizing effects of MG-132.
Collapse
Affiliation(s)
- Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA90095-1714, USA
| | - Arndt van Ophoven
- Department of Urology, University Hospital Münster, Albert-Schweitzer-Straße 33, D-48149 Münster Germany
| | - Christian Weissenberger
- Department of Radiation Oncology, University Hospital Freiburg, Robert-Koch-Straße 3, D-79106 Freiburg, Germany
| | - William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles, CA90095-1714, USA
| |
Collapse
|
56
|
Goel A, Dispenzieri A, Greipp PR, Witzig TE, Mesa RA, Russell SJ. PS-341–mediated selective targeting of multiple myeloma cells by synergistic increase in ionizing radiation-induced apoptosis. Exp Hematol 2005; 33:784-95. [PMID: 15963854 DOI: 10.1016/j.exphem.2005.04.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 03/28/2005] [Accepted: 04/14/2005] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Multiple myeloma remains incurable with current therapy. The proteosome inhibitor, PS-341, has shown objective clinical responses in relapsed refractory myeloma patients. We investigated the potential of enhancing the radiosensitivity of myeloma cells by combining with PS-341; the underlying mechanisms were delineated. MATERIALS AND METHODS Clonogenic assays were used to evaluate cell survival after exposure to PS-341, ionizing radiation (IR), or PS-341 followed by IR. Apoptosis was studied by annexin V-propidium iodide staining and caspase activation. Cell-cycle phase distribution of cells was determined. Nuclear factor-kappaB (NF-kappaB) activity was monitored by enzyme-linked immunosorbent assay and Western blotting. The expression of death receptor Fas/APO-1/CD95 was analyzed by flow cytometry. The consequential caspase-8 activation was detected by Western blotting. RESULTS In clonogenic assays, sequential exposure to nontoxic doses of PS-341 (10 nM) and IR (6 Gy) resulted in synergistic inhibition of proliferation of myeloma cells by modulating the apoptotic sensitivity of these cells. Biochemically, sublethal dose of IR led to potent induction of NF-kappaB activity, and this response was significantly inhibited by pretreatment with PS-341, or by the NF-kappaB inhibitory peptide SN-50. Enhanced Fas expression was seen in myeloma cells exposed sequentially to PS-341 and IR. Finally, PS-341 sensitized primary myeloma (CD138+ve) cells to IR but had little effect on CD138-ve bone marrow cells from myeloma patients. CONCLUSION These data indicate that PS-341 can sensitize myeloma cells to IR by both intrinsic and extrinsic apoptotic pathways. The study indicates improved therapeutic benefits in treatment of multiple myeloma by combining PS-341 with conventional radiotherapy.
Collapse
Affiliation(s)
- Apollina Goel
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
57
|
Szkanderová S, Vávrová J, Hernychová L, Neubauerová V, Lenco J, Stulík J. Proteome alterations in gamma-irradiated human T-lymphocyte leukemia cells. Radiat Res 2005; 163:307-15. [PMID: 15733044 DOI: 10.1667/rr3309] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Analyses of the protein expression profiles of irradiated cells may be beneficial for identification of new biomolecules of radiation-induced cell damage. Therefore, in this study we exploited the proteomic approach to identify proteins whose expression is significantly altered in gamma-irradiated human T-lymphocyte leukemia cells. MOLT-4 cells were irradiated with 7.5 Gy and the cell lysates were collected at different times after irradiation (2, 5 and 12 h). The proteins were separated by two-dimensional electrophoresis and quantified using an image evaluation system. Proteins exhibiting significant radiation-induced alterations in abundance were identified by peptide mass fingerprinting. We identified 14 proteins that were either up- or down-regulated. Cellular levels of four of the proteins (Rho GDP dissociation inhibitor 1 and 2, Ran binding protein 1, serine/threonine protein kinase PAK2) were further analyzed by two-dimensional immunoblotting to confirm the data obtained from proteome analysis. All identified proteins were classified according to their cellular function, including their participation in biochemical and signaling pathways. Taken together, our results suggest the feasibility of the proteome method for monitoring of cellular radiation responses.
Collapse
Affiliation(s)
- Sylva Szkanderová
- Institute of Molecular Pathology, Purkyne Military Medical Academy, Trebesska 1575, Hradec Kralove 500 01, Czech Republic.
| | | | | | | | | | | |
Collapse
|
58
|
Tapio S, Danescu-Mayer J, Asmuss M, Posch A, Gomolka M, Hornhardt S. Combined effects of gamma radiation and arsenite on the proteome of human TK6 lymphoblastoid cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2005; 581:141-52. [PMID: 15725613 DOI: 10.1016/j.mrgentox.2004.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2004] [Revised: 11/18/2004] [Accepted: 11/29/2004] [Indexed: 01/21/2023]
Abstract
Arsenic present in drinking water and mining environments in some areas has been associated with an increased rate of skin and internal cancers. Contrary to the epidemiological evidence in humans, arsenic does not induce cancer in animal models, but is able to enhance the mutagenicity of other agents. In order to achieve a better understanding of the interaction between arsenic and ionising radiation, an investigation was conducted to detect differences at the proteome level of human TK6 lymphoblastoid cells exposed to these agents. Cells were exposed to either a single dose of 1-Gy 137Cs-gamma-rays or to 1 microM arsenite (As(III)) or to both agents in combination. Two-dimensional (2D) electrophoresis and matrix-assisted laser desorption/ionisation-time of flight (MALDI-TOF) were employed for the screening and identification of proteins, respectively. It proved possible to identify seven proteins with significantly affected abundance, three of which showed increased levels and the remaining four showed decreased levels under at least one of the exposure conditions. Following arsenite treatment or irradiation, a significant increase compared with that of the control was observed for glutathione (GSH) transferase omega 1 and proteasome subunit beta type 4 precursor. The combined exposure did not result in an induction of the enzymes. The expression of electron-transfer flavoprotein subunit alpha was found to be enhanced under all three-exposure conditions. Ubiquinol-cytochrome C reductase complex core protein I, adenine phosphoribosyl transferase and endoplasmic reticulum protein hERp29 showed decreased levels after irradiation or arsenite treatment, but not after the combined exposure. The level of serine/threonine protein phosphatase 1 alpha decreased with all treatments. The main conclusions are that both arsenite and gamma-radiation influence the levels of several proteins involved in major metabolic and regulatory pathways, either directly or by triggering the defence mechanisms of the cell. The combined effect of both exposures on the level of some essential proteins such as glutathione transferase, proteasome or serine/threonine phosphatase may contribute to the co-carcinogenic effect of arsenic.
Collapse
Affiliation(s)
- Soile Tapio
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, Ingolstaedter Landstrasse 1, 85764 Neuherberg, Germany.
| | | | | | | | | | | |
Collapse
|
59
|
Liao YP, Wang CC, Butterfield LH, Economou JS, Ribas A, Meng WS, Iwamoto KS, McBride WH. Ionizing radiation affects human MART-1 melanoma antigen processing and presentation by dendritic cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:2462-9. [PMID: 15294960 DOI: 10.4049/jimmunol.173.4.2462] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Radiation is generally considered to be an immunosuppressive agent that acts by killing radiosensitive lymphocytes. In this study, we demonstrate the noncytotoxic effects of ionizing radiation on MHC class I Ag presentation by bone marrow-derived dendritic cells (DCs) that have divergent consequences depending upon whether peptides are endogenously processed and loaded onto MHC class I molecules or are added exogenously. The endogenous pathway was examined using C57BL/6 murine DCs transduced with adenovirus to express the human melanoma/melanocyte Ag recognized by T cells (AdVMART1). Prior irradiation abrogated the ability of AdVMART1-transduced DCs to induce MART-1-specific T cell responses following their injection into mice. The ability of these same DCs to generate protective immunity against B16 melanoma, which expresses murine MART-1, was also abrogated by radiation. Failure of AdVMART1-transduced DCs to generate antitumor immunity following irradiation was not due to cytotoxicity or to radiation-induced block in DC maturation or loss in expression of MHC class I or costimulatory molecules. Expression of some of these molecules was affected, but because irradiation actually enhanced the ability of DCs to generate lymphocyte responses to the peptide MART-1(27-35) that is immunodominant in the context of HLA-A2.1, they were unlikely to be critical. The increase in lymphocyte reactivity generated by irradiated DCs pulsed with MART-1(27-35) also protected mice against growth of B16-A2/K(b) tumors in HLA-A2.1/K(b) transgenic mice. Taken together, these results suggest that radiation modulates MHC class I-mediated antitumor immunity by functionally affecting DC Ag presentation pathways.
Collapse
Affiliation(s)
- Yu-Pei Liao
- Department of Radiation Oncology, Division of Surgical Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
60
|
McBride WH, Chiang CS, Olson JL, Wang CC, Hong JH, Pajonk F, Dougherty GJ, Iwamoto KS, Pervan M, Liao YP. A Sense of Danger from Radiation1. Radiat Res 2004; 162:1-19. [PMID: 15222781 DOI: 10.1667/rr3196] [Citation(s) in RCA: 234] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Tissue damage caused by exposure to pathogens, chemicals and physical agents such as ionizing radiation triggers production of generic "danger" signals that mobilize the innate and acquired immune system to deal with the intrusion and effect tissue repair with the goal of maintaining the integrity of the tissue and the body. Ionizing radiation appears to do the same, but less is known about the role of "danger" signals in tissue responses to this agent. This review deals with the nature of putative "danger" signals that may be generated by exposure to ionizing radiation and their significance. There are a number of potential consequences of "danger" signaling in response to radiation exposure. "Danger" signals could mediate the pathogenesis of, or recovery from, radiation damage. They could alter intrinsic cellular radiosensitivity or initiate radioadaptive responses to subsequent exposure. They may spread outside the locally damaged site and mediate bystander or "out-of-field" radiation effects. Finally, an important aspect of classical "danger" signals is that they link initial nonspecific immune responses in a pathological site to the development of specific adaptive immunity. Interestingly, in the case of radiation, there is little evidence that "danger" signals efficiently translate radiation-induced tumor cell death into the generation of tumor-specific immunity or normal tissue damage into autoimmunity. The suggestion is that radiation-induced "danger" signals may be inadequate in this respect or that radiation interferes with the generation of specific immunity. There are many issues that need to be resolved regarding "danger" signaling after exposure to ionizing radiation. Evidence of their importance is, in some areas, scant, but the issues are worthy of consideration, if for no other reason than that manipulation of these pathways has the potential to improve the therapeutic benefit of radiation therapy. This article focuses on how normal tissues and tumors sense and respond to danger from ionizing radiation, on the nature of the signals that are sent, and on the impact on the eventual consequences of exposure.
Collapse
Affiliation(s)
- William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-1714, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Rödel F, Hantschel M, Hildebrandt G, Schultze-Mosgau S, Rödel C, Herrmann M, Sauer R, Voll RE. Dose-dependent biphasic induction and transcriptional activity of nuclear factor kappa B (NF-kappaB) in EA.hy.926 endothelial cells after low-dose X-irradiation. Int J Radiat Biol 2004; 80:115-23. [PMID: 15164793 DOI: 10.1080/09553000310001654701] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
PURPOSE Low-dose radiotherapy is known to exert an anti-inflammatory effect, but the underlying radiobiological mechanisms are still elusive. It was recently reported that transforming growth factor (TGF) beta1 essentially contributes to the reduced adhesion of peripheral blood mononuclear cells to endothelial cells at low-dose X-irradiation. As the transcription factor nuclear factor kappa B (NF-kappaB) is crucially involved in mediating an inflammatory response by inducing the expression of cytokines and adhesion molecules, NF-kappaB DNA binding and transcriptional activity as well as its impact on the expression of TGF-beta1 in EA.hy.926 endothelial cells were analysed subsequently to low-dose radiotherapy. MATERIALS AND METHODS Human EA.hy.926 endothelial cells were grown to subconfluence. Twenty hours after X-irradiation with single doses ranging from 0.3 to 3 Gy, the cells were activated with tumour necrosis factor-alpha. Four hours later, the cells were harvested. NF-kappaB DNA-binding activity of nuclear extracts was analysed by electrophoretic mobility shift assay. The NF-kappaB subunits p50, p65/RelA, c-Rel and RelB of the NF-kappaB complexes were quantified by enzyme-linked immunoabsorbant assay. The transcriptional activity of NF-kappaB was measured using luciferase reporter gene assays in EA.hy.926 endothelial cells transiently transfected with the plasmid pB2xLuc. To correlate transcriptional activity to TGF-beta1 expression, NF-kappaB decoy oligonucleotides were used to inhibit NF-kappaB activity and TGF-beta1 secretion. RESULTS After low-dose radiotherapy, an increased NF-kappaB DNA-binding activity was observed in stimulated EA.hy.926 endothelial cells with a relative maximum (threefold induction) at 0.5 Gy. The NF-kappaB activation then decreased after X-irradiation at 0.6-0.8 Gy and subsequently increased again at doses of 1 and 3 Gy. This biphasic induction profile of NF-kappaB was confirmed by the analysis of the NF-kappaB-specific transcriptional activity. The latter showed a relative maximum at 0.5 Gy, a relative minimum between 0.5 and 1.0 Gy, and an increase at 3 Gy. Transfection of EA.hy.926 endothelial cells with NF-kappaB decoy oligonucleotides before irradiation resulted in a 50% reduction of TGF-beta1 secretion at 0.5 Gy compared with control oligonucleotides or untreated cells. CONCLUSIONS Low-dose radiotherapy induces a biphasic activation of NF-kappaB with a relative maximum at 0.5 Gy. The induction by NF-kappaB of TGF-beta1 in endothelial cells might contribute to the anti-inflammatory properties of low-dose ionizing irradiation.
Collapse
Affiliation(s)
- F Rödel
- Department of Radiooncology, University of Erlangen-Nuremberg, Universitätsstrasse 27, D-91054 Erlangen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Baust H, Schoke A, Brey A, Gern U, Los M, Schmid RM, Röttinger EM, Seufferlein T. Evidence for radiosensitizing by gliotoxin in HL-60 cells: implications for a role of NF-κB independent mechanisms. Oncogene 2003; 22:8786-96. [PMID: 14647473 DOI: 10.1038/sj.onc.1206969] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Radioresistance markedly impairs the efficacy of tumor radiotherapy and may involve antiapoptotic signal transduction pathways that prevent radiation-induced cell death. A common cellular response to genotoxic stress induced by radiation is the activation of the nuclear factor kappa B (NF-kappaB). NF-kappaB activation in turn can lead to an inhibition of radiation-induced apoptotic cell death. Thus, inhibition of NF-kappaB activation is commonly regarded as an important strategy to abolish radioresistance. Among other compounds, the fungal metabolite gliotoxin (GT) has been reported to be a highly selective inhibitor of NF-kappaB activation. Indeed, low doses of GT were sufficient to significantly enhance radiation-induced apoptosis in HL-60 cells. However, this effect turned out to be largely independent of NF-kappaB activation since radiation of HL-60 cells with clinically relevant doses of radiation induced only a marginal increase in NF-kappaB activity, and selective inhibition of NF-kappaB by SN50 did not result in a marked enhancement of GT-induced apoptosis. GT induced activation of JNKs, cytochrome c release from the mitochondria and potently stimulated the caspase cascade inducing cleavage of caspases -9, -8, -7 and -3. Furthermore, cleavage of the antiapoptotic protein X-linked IAP and downregulation of the G2/M-specific IAP-family member survivin were observed during GT-induced apoptosis. Finally, the radiation-induced G2/M arrest was markedly reduced in GT-treated cells most likely due to the rapid induction of apoptosis. Our data demonstrate that various other pathways apart from the NF-kappaB signaling complex can sensitize tumor cells to radiation and propose a novel mechanism for radiosensitization by GT, the interference with the G2/M checkpoint that is important for repair of radiation-induced DNA damage in p53-deficient tumor cells.
Collapse
Affiliation(s)
- H Baust
- Department of Radiation Oncology, University of Ulm, D-89081 Ulm, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Abstract
Radiation hormesis is reviewed with emphasis on its expression in the immune system. The shape of the dose-response relationship of the immune functions depends on a number of factors, chiefly the target cell under study, experimental design with emphasis on the dose range, dose spacing, dose rate and temporal changes, as well as the animal strain. For mouse and human T lymphocyte functions in the dose range of 0.01 to 10 Gy a J or inverted J-shaped curve is usually observed. For the more radioresistant macrophages, stimulation of many of their functions is often observed in the dose range up to a few grays. The cellular and molecular mechanisms of the enhancement of immunity induced by low-dose radiation were analyzed on the basis of literature published in the last decade of the past century. Intercellular reactions among the APCs and lymphocytes via distinct changes in expression of relevant surface molecules and secretion of regulatory cytokines in response to different doses of radiation were described. The major signal transduction pathways activated in response to these intercellular reactions were illustrated. The suppressive effect of low-dose radiation on cancer induction, growth, and metastasis and its immunologic mechanisms were analyzed. The present status of research in this field gives strong support to radiation hormesis in immunity with low-dose radiation as one of the mechanisms of cancer surveillance. Further research with new techniques using microarray with biochips to fully elucidate the molecular mechanisms is suggested.
Collapse
Affiliation(s)
- Shu-Zheng Liu
- Department of Radiation Biology, Jilin University Norman Bethune Medical Center, 8 Xinmin Street, Changchun 130021, China.
| |
Collapse
|
64
|
McBride WH, Iwamoto KS, Syljuasen R, Pervan M, Pajonk F. The role of the ubiquitin/proteasome system in cellular responses to radiation. Oncogene 2003; 22:5755-73. [PMID: 12947384 DOI: 10.1038/sj.onc.1206676] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the last few years, the ubiquitin(Ub)/proteasome system has become increasingly recognized as a controller of numerous physiological processes, including signal transduction, DNA repair, chromosome maintenance, transcriptional activation, cell cycle progression, cell survival, and certain immune cell functions. This is in addition to its more established roles in the removal of misfolded, damaged, and effete proteins. This review examines the role of the Ub/proteasome system in processes underlying the classical effects of irradiation on cells, such as radiation-induced gene expression, DNA repair and chromosome instability, oxidative damage, cell cycle arrest, and cell death. Furthermore, recent evidence suggests that the proteasome is a redox-sensitive target for ionizing radiation and other oxidative stress signals. In other words, the Ub/proteasome system may not simply be a passive player in radiation-induced responses, but may modulate them. The extent of the modulation will be influenced by the functional and structural diversity that is expressed by the system. Cell types vary in the Ub/proteasome structures they possess and the level at which they function, and this changes as they go from the normal to the cancerous condition. Cancer-related functional changes within the Ub/proteasome system may therefore present unique targets for cancer therapy, especially when targeting agents are used in combination with radio- or chemotherapy. The peptide boronic acid compound PS-341, which was designed to inhibit proteasome chymotryptic activity, is in clinical trials for the treatment of solid and hematogenous tumors. It has shown some efficacy on its own and in combination with chemotherapy. Preclinical studies have shown that PS-341 will also potentiate the cytotoxic effects of radiation therapy. In addition, other drugs in common clinical use have been shown to affect proteasome function, and their activities may be valuably reconsidered from this perspective.
Collapse
Affiliation(s)
- William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | | | | | | | | |
Collapse
|
65
|
Liu SZ. Nonlinear dose-response relationship in the immune system following exposure to ionizing radiation: mechanisms and implications. NONLINEARITY IN BIOLOGY, TOXICOLOGY, MEDICINE 2003; 1:71-92. [PMID: 19330113 PMCID: PMC2651616 DOI: 10.1080/15401420390844483] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The health effects of low-dose radiation (LDR) have been the concern of the academic spheres, regulatory bodies, governments, and the public. Among these effects, the most important is carcinogenesis. In view of the importance of immune surveillance in cancer control, the dose-response relationship of the changes in different cell types of the immune system after whole-body irradiation is analyzed on the basis of systemic data from the author's laboratory in combination with recent reports in the literature. For T lymphocytes J- or inverted J-shaped curves are usually demonstrated after irradiation, while for macrophages dose-response curves of chiefly stimulation with irregular patterns are often observed. The intercellular reactions between the antigen presenting cell (APC) and T lymphocyte (TLC) in the immunologic synapse via expression of surface molecules and secretion of cytokines by the two cell types after different doses of radiation are illustrated. The different pathways of signal transduction thus facilitated in the T lymphocyte by different doses of radiation are analyzed to explain the mechanism of the phenomenon of low-dose stimulation and high-dose suppression of immunity. Experimental and clinical data are cited to show that LDR retards tumor growth, reduces metastasis, increases the efficacy of conventional radiotherapy and chemotherapy as well as alleviates the suppression of immunity due to tumor burden. The incidence of thymic lymphoma after high-dose radiation is lowered by preexposure to low-dose radiation, and its mechanism is supposed to be related to the stimulation of anticancer immunity induced by low-dose radiation. Recent reports on lowering of standardized cancer mortality rate and all cause death rate of cohorts occupationally exposed to low-dose radiation from the US, UK, and Canada are cited.
Collapse
Affiliation(s)
- Shu-Zheng Liu
- Radiobiology Research Unit, Ministry of Health, PRC, Jilin University Norman Bethune Medical Center 8 Xinmin Street, Changchun 130021, China Phone number: +86-431-564-4597. Fax number: +86-431-563-3025.
| |
Collapse
|
66
|
Sonis ST, Scherer J, Phelan S, Lucey CA, Barron JE, O'Donnell KE, Brennan RJ, Pan H, Busse P, Haley JD. The gene expression sequence of radiated mucosa in an animal mucositis model. Cell Prolif 2002; 35 Suppl 1:93-102. [PMID: 12139712 PMCID: PMC6496665 DOI: 10.1046/j.1365-2184.35.s1.10.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Oral mucositis is a common, dose-limiting, acute toxicity of radiation therapy administered for the treatment of cancers of the head and neck. Accumulating data would suggest that the pathogenesis of mucositis is complex and involves the sequential interaction of all cell types of the oral mucosa, as well as a number of cytokines and elements of the oral environment. While a number of studies have reported on gene expression of particular cell types in response to radiation, the overall response of irradiated mucosa has only been evaluated in a limited way. The present study was undertaken to evaluate the expression of a target group of genes using RNA quantification assays and, more broadly, to assess patterns of mucosal gene expression using DNA microarray hybridization. Our results demonstrate the sequential upregulation of a series of genes that, when taken collectively, suggest an intricate functional interaction.
Collapse
Affiliation(s)
- S T Sonis
- Division of Oral Medicine, Oral and Maxillofacial Surgery and Dentistry, Brigham and Women's Hospital and the Department of Otal Medicine and Diagnostic Sciences, Harvard School of Dental Medicine, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Bulteau AL, Moreau M, Nizard C, Friguet B. Impairment of proteasome function upon UVA- and UVB-irradiation of human keratinocytes. Free Radic Biol Med 2002; 32:1157-70. [PMID: 12031900 DOI: 10.1016/s0891-5849(02)00816-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The major environmental influence for epidermal cells is sun exposure and the harmful effect of UV radiation on skin is related to the generation of reactive oxygen species that are altering cellular components including proteins. It is now well established that the proteasome is responsible for the degradation of oxidized proteins. Therefore, the effects of UV-irradiation on proteasome have been investigated in human keratinocyte cultures. Human keratinocytes were irradiated with 10 J/cm(2) of UVA and 0.05 J/cm(2) of UVB and proteasome peptidase activities were measured in cell lysates using fluorogenic peptides. All three peptidase activities were decreased as early as 1 h and up to 24 h after irradiation of the cells. Increased levels of oxidized and ubiquitinated proteins as well as proteins modified by the lipid peroxidation product 4-hydroxy-2-nonenal were also observed in irradiated cells. However, immunopurified 20S proteasome exhibited no difference in both peptidase specific activities and 2D gel pattern of subunits in irradiated cells, ruling out the possibility that the 20S proteasome could be a target for the UV-induced damage. Finally, extracts from irradiated keratinocytes were able to inhibit degradation by the proteasome, demonstrating the presence of endogeneous inhibitors, including 4-hydroxy-2-nonenal modified proteins, generated upon UV-irradiation.
Collapse
Affiliation(s)
- Anne-Laure Bulteau
- Laboratoire de Biologie et Biochimie Cellulaire du Vieillissement, Université Paris, 7-Denis Diderot, Paris, France
| | | | | | | |
Collapse
|
68
|
Pajonk F, Riess K, Sommer A, McBride WH. N-acetyl-L-cysteine inhibits 26S proteasome function: implications for effects on NF-kappaB activation. Free Radic Biol Med 2002; 32:536-43. [PMID: 11958954 DOI: 10.1016/s0891-5849(02)00743-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ionizing radiation shares with cytokines, such as TNF-alpha, an ability to generate free radicals in cells and activate downstream proinflammatory responses through NF-kappaB-dependent signal transduction pathways. Support for the role of free radicals in triggering such responses comes from the use of free radical scavengers like N-acetyl-L-cysteine (NAC). The nature of the link between free radical generation and NF-kappaB activation is, however, unclear. In this study, we explore the possibility that scavenging of free radicals by NAC might not be the mechanism by which it inhibits NF-kappaB activation, but rather that NAC acts through inhibition of proteasome function. The effect of NAC on the chymotryptic function of the 26s and 20s proteasome complex was measured in extracts from EVC 304 bladder carcinoma cells by assessing degradation of fluorogenic substrates. NAC inhibited 26s but not 20s proteasome activity, suggesting that it interferes with 19s regulatory subunit function. NAC blocked radiation-induced NF-kappaB activity in ECV 304 cells and RAW 264.7 macrophages, as measured by a gel shift assay, at doses that inhibited proteasome activity. This provides a possible mechanism whereby NAC could block NF-kappaB activation and affect the expression of other molecules that are dependent on the ubiquitin/proteasome system for their degradation, other than by scavenging free radicals.
Collapse
Affiliation(s)
- Frank Pajonk
- Department of Radiation Therapy, Radiological University Clinic Freiburg, Hugstetter Strasse 55, 79106 Freiburg im Breisgau, Germany.
| | | | | | | |
Collapse
|
69
|
Abstract
During the last 30 years, investigation of the transcriptional and translational mechanisms of gene regulation has been a major focus of molecular cancer biology. More recently, it has become evident that cancer-related mutations and cancer-related therapies also can affect post-translational processing of cellular proteins and that control exerted at this level can be critical in defining both the cancer phenotype and the response to therapeutic intervention. One post-translational mechanism that is receiving considerable attention is degradation of intracellular proteins through the multicatalytic 26S proteasome. This follows growing recognition of the fact that protein degradation is a well-regulated and selective process that can differentially control intracellular protein expression levels. The proteasome is responsible for the degradation of all short-lived proteins and 70-90% of all long-lived proteins, thereby regulating signal transduction through pathways involving factors such as AP1 and NFKB, and processes such as cell cycle progression and arrest, DNA transcription, DNA repair/misrepair, angiogenesis, apoptosis/survival, growth and development, and inflammation and immunity, as well as muscle wasting (e.g. in cachexia and sepsis). In this review, we discuss the potential involvement of the proteasome in both cancer biology and cancer treatment.
Collapse
Affiliation(s)
- F Pajonk
- Department of Radiation Therapy, Radiological University Clinic, Hugstetter Str. 55, 79106 Freiburg i. Brsg., Germany.
| | | |
Collapse
|
70
|
Pervan M, Pajonk F, Sun JR, Withers HR, McBride WH. Molecular pathways that modify tumor radiation response. Am J Clin Oncol 2001; 24:481-5. [PMID: 11586100 DOI: 10.1097/00000421-200110000-00013] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Aberrant expression of signal transduction molecules in pathways controlling cell survival, proliferation, death, or differentiation are a common feature of all tumors. The identification of the molecules that are involved allows the development of novel tumor-specific strategies. Not surprisingly, targeting these pathways often also results in radiosensitization. The efficacy of such directed therapies may, however, be limited by the heterogeneity and the multiple mutations that are associated with the cancerous state. A more robust alternative may be to target global mechanisms of cellular control. The ubiquitin/proteasome degradation pathway is one candidate for such therapeutic intervention. This pathway is the main posttranscriptional mechanism that controls levels of many short-lived proteins involved in regulation of cell cycle progression, DNA transcription, DNA repair, and apoptosis. Many of these proteins are involved in various malignancies and/or radiation responses. In recent years, proteasome inhibitors have gained interest as a promising new group of antitumor drugs. PS-341, a reversible inhibitor of proteasome chymotryptic activity, is currently being tested in phase I clinical trials. In this study, we show that proteasome inhibition by PS-341 can alter cellular radiosensitivity in vitro and in vivo, in addition to having direct antitumor effects.
Collapse
Affiliation(s)
- M Pervan
- Department of Radiation Oncology, Roy E. Coats Research Laboratories and Jonsson Comprehensive Cancer Center, UCLA School of Medicine, Los Angeles, CA 90095-1714, USA
| | | | | | | | | |
Collapse
|