Yonsei Med J.  2015 Jan;56(1):112-123. 10.3349/ymj.2015.56.1.112.

The Effect of Bortezomib on Expression of Inflammatory Cytokines and Survival in a Murine Sepsis Model Induced by Cecal Ligation and Puncture

Affiliations
  • 1Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea. jmkim@yuhs.ac
  • 2Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
  • 3Department of Microbiology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
  • 4Brain Korea 21 Project for Medical Science, Brain Research Institute and Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.

Abstract

PURPOSE
Although the proteasome inhibitor known as bortezomib can modulate the inflammatory process through the nuclear factor-kappa B signaling pathway, the immunomodulatory effect of pre-incubated bortezomib has not been fully evaluated for inflammation by infectious agents. Therefore, we evaluated the effect of bortezomib on the expression of inflammatory cytokines and mediators in macrophage cell lines and on survival in a murine peritonitis sepsis model.
MATERIALS AND METHODS
Bortezomib was applied 1 hr before lipopolysaccharide (LPS) stimulation in RAW 264.7 cells. The cecal ligation and puncture (CLP) experiments were performed in C57BL/6J mice.
RESULTS
Pre-incubation with bortezomib (25 nM or 50 nM) prior to LPS (50 ng/mL or 100 ng/mL) stimulation significantly recovered the number of viable RAW 264.7 cells compared to those samples without pre-incubation. Bortezomib decreased various inflammatory cytokines as well as nitric oxide production in LPS-stimulated cells. The 7-day survival rate in mice that had received bortezomib at 0.01 mg/kg concentration 1 hr prior to CLP was significantly higher than in the mice that had only received a normal saline solution of 1 mL 1 hr prior to CLP. In addition, the administration of bortezomib at 0.01 mg/kg concentration 1 hr before CLP resulted in a significant decrease in inflammation of the lung parenchyma. Collectively, pretreatment with bortezomib showed an increase in the survival rate and changes in the levels of inflammatory mediators.
CONCLUSION
These results support the possibility of pretreatment with bortezomib as a new therapeutic target for the treatment of overwhelming inflammation, which is a characteristic of severe sepsis.

Keyword

Bortezomib; proteasome inhibitor; lipopolysaccharide; inflammatory cytokines; cecal ligation and puncture

MeSH Terms

Animals
Boronic Acids/administration & dosage/pharmacology/*therapeutic use
Cecum/*pathology
Cell Adhesion Molecules/metabolism
Cell Line
Cell Proliferation/drug effects
Cell Survival/drug effects
Chymotrypsin/metabolism
Cytokines/*metabolism
Disease Models, Animal
Inflammation Mediators/*metabolism
Ligation
Lipopolysaccharides/pharmacology
Lung/drug effects/metabolism/pathology
Male
Mice, Inbred C57BL
Nitric Oxide/metabolism
Proteasome Inhibitors/pharmacology
Punctures
Pyrazines/administration & dosage/pharmacology/*therapeutic use
Sepsis/*drug therapy
Boronic Acids
Cell Adhesion Molecules
Chymotrypsin
Cytokines
Inflammation Mediators
Lipopolysaccharides
Proteasome Inhibitors
Pyrazines
Nitric Oxide

Figure

  • Fig. 1 The concentrations of viable RAW 264.7 cells measured using the CCK-8 assay to evaluate the effect of pre-incubation with bortezomib 1 hr before LPS treatment on the cell proliferation. On experiment day 3, LPS at 50 ng/mL (A) and 100 ng/mL (B) was administered to the growing RAW 264.7 cell lines of 8×103 cells/mL at 1 hr after the application of bortezomib with various concentrations from 0 to 100 nM. Arrows (↓) indicate the reference values in samples that had received only LPS application with 50 and 100 ng/mL, without pre-incubation with bortezomib. *Statistical significance with p-values less than 0.05 when compared with reference values (LPS alone, arrows). CCK-8, Cell Counting Kit-8; LPS, lipopolysaccharide; OD, optical density.

  • Fig. 2 The chymotrypsin-like activity of proteasomes measured by the bioluminescent proteasome assay. RAW 264.7 cells (arrow) that were not treated with bortezomib and lipopolysaccharide (100 ng/mL) were used as the reference control to determine statistical significance (*p<0.01). †Means statistically significant with a p-value less than 0.01 when compared with bortezomib at a 25 nM concentration (asterisk). The results are reported as mean±standard error of the mean of three independent experiments in triplicate. RLU, relative luminescent units; LPS, lipopolysaccharide; Bor, bortezomib.

  • Fig. 3 The expression levels of inflammatory cytokines and intercellular adhesion molecules based on the administration of bortezomib and LPS at various concentrations. (A) The RT-PCR products expressed by agarose gel electrophoresis. (B-F) The relative expression rate of RT-PCR products by internal control using mouse GAPDH in each cytokine and ICAM-1. These semi-quantitation was performed using the Multi Guage Version 3.0 program. *Statistical significance with p-values less than 0.01. **Statistical significance with p-values less than 0.001. †,‡Reference values for statistical comparison. GAPDH, glyceraldehyde 3-phosphate dehydrogenase; TNF-α, tumor necrosis factor-alpha; IFN-α, interferon-gamma; IL, interleukin; ICAM, intercellular adhesion molecule; LPS, lipopolysaccharide.

  • Fig. 4 The measurement of nitric oxide concentrations using the diazotization assay (Griess method) in order to analyze the effect of bortezomib on nitric oxide production after LPS stimulation. On the third day of RAW 264.7 cell growth, the supernatant of the cell line growth media received bortezomib (0 or 50 nM) and then LPS (0 or 100 ng/mL) with a 1 hr interval. These samples were used for the nitric oxide detection assay 16 and 24 hr after administration of bortezomib and/or LPS to the cell lines. *p-values less than 0.001 compared with the LPS-only treatment group for the 16 and 24 hr time periods. LPS, lipopolysaccharide; Bor, bortezomib.

  • Fig. 5 Comparison of 7-day mortalities after CLP surgery by the Kaplan-Meier survival curve. Bortezomib and normal saline (at 1 mL) were always injected intraperitoneally. CLP, cecal ligation and puncture; NS, normal saline; Bor, bortezomib.

  • Fig. 6 Histologic findings of lung tissue after H&E staining (×200). All lung tissues were harvested from the mice that were alive at 7 days after the surgery. (A) Normal mice (negative control). (B) Normal saline 1 mL injection IP at 1 hr before sham surgery (negative control). (C) Normal saline 1 mL injection IP at 1 hr before CLP surgery (positive control). (D) Bortezomib 0.01 mg/kg injection IP at 1 hr before CLP surgery. (E) Bortezomib 0.1 mg/kg injection, IP at 1 hr before CLP surgery. (F) Bortezomib 0.01 mg/kg injection, IP at 24 hr after CLP surgery. IP, intraperitoneal; CLP, cecal ligation and puncture.


Reference

1. Mao X, Pan X, Cheng T, Zhang X. Therapeutic potential of the proteasome inhibitor Bortezomib on titanium particle-induced inflammation in a murine model. Inflammation. 2012; 35:905–912.
Article
2. Koca SS, Ozgen M, Dagli F, Tuzcu M, Ozercan IH, Sahin K, et al. Proteasome inhibition prevents development of experimental dermal fibrosis. Inflammation. 2012; 35:810–817.
Article
3. Yannaki E, Papadopoulou A, Athanasiou E, Kaloyannidis P, Paraskeva A, Bougiouklis D, et al. The proteasome inhibitor bortezomib drastically affects inflammation and bone disease in adjuvant-induced arthritis in rats. Arthritis Rheum. 2010; 62:3277–3288.
Article
4. Blanco B, Sánchez-Abarca LI, Caballero-Velázquez T, Santamaría C, Inogés S, Pérez-Simón JA. Depletion of alloreactive T-cells in vitro using the proteasome inhibitor bortezomib preserves the immune response against pathogens. Leuk Res. 2011; 35:1412–1415.
Article
5. Brun J. Proteasome inhibition as a novel therapy in treating rheumatoid arthritis. Med Hypotheses. 2008; 71:65–72.
Article
6. Inoue S, Nakase H, Matsuura M, Mikami S, Ueno S, Uza N, et al. The effect of proteasome inhibitor MG132 on experimental inflammatory bowel disease. Clin Exp Immunol. 2009; 156:172–182.
Article
7. Lee SW, Kim JH, Park YB, Lee SK. Bortezomib attenuates murine collagen-induced arthritis. Ann Rheum Dis. 2009; 68:1761–1767.
Article
8. Palombella VJ, Rando OJ, Goldberg AL, Maniatis T. The ubiquitin-proteasome pathway is required for processing the NF-kappa B1 precursor protein and the activation of NF-kappa B. Cell. 1994; 78:773–785.
Article
9. Paramore A, Frantz S. Bortezomib. Nat Rev Drug Discov. 2003; 2:611–612.
Article
10. Meiners S, Ludwig A, Stangl V, Stangl K. Proteasome inhibitors: poisons and remedies. Med Res Rev. 2008; 28:309–327.
Article
11. Moehler T, Goldschmidt H. Therapy of relapsed and refractory multiple myeloma. Recent Results Cancer Res. 2011; 183:239–271.
Article
12. Tsujimoto H, Ono S, Efron PA, Scumpia PO, Moldawer LL, Mochizuki H. Role of Toll-like receptors in the development of sepsis. Shock. 2008; 29:315–321.
Article
13. Christaki E, Anyfanti P, Opal SM. Immunomodulatory therapy for sepsis: an update. Expert Rev Anti Infect Ther. 2011; 9:1013–1033.
Article
14. Qureshi N, Perera PY, Shen J, Zhang G, Lenschat A, Splitter G, et al. The proteasome as a lipopolysaccharide-binding protein in macrophages: differential effects of proteasome inhibition on lipopolysaccharide-induced signaling events. J Immunol. 2003; 171:1515–1525.
Article
15. Safránek R, Ishibashi N, Oka Y, Ozasa H, Shirouzu K, Holecek M. Modulation of inflammatory response in sepsis by proteasome inhibition. Int J Exp Pathol. 2006; 87:369–372.
Article
16. Reis J, Tan X, Yang R, Rockwell CE, Papasian CJ, Vogel SN, et al. A combination of proteasome inhibitors and antibiotics prevents lethality in a septic shock model. Innate Immun. 2008; 14:319–329.
Article
17. Jeong SJ, Lim BJ, Park S, Choi D, Kim HW, Ku NS, et al. The effect of sRAGE-Fc fusion protein attenuates inflammation and decreases mortality in a murine cecal ligation and puncture model. Inflamm Res. 2012; 61:1211–1218.
Article
18. O'Brien MA, Moravec RA, Riss TL, Bulleit RF. Homogeneous, bioluminescent proteasome assays. Methods Mol Biol. 2008; 414:163–181.
19. Ishiyama M, Miyazono Y, Sasamoto K, Ohkura Y, Ueno K. A highly water-soluble disulfonated tetrazolium salt as a chromogenic indicator for NADH as well as cell viability. Talanta. 1997; 44:1299–1305.
Article
20. Isobe I, Michikawa M, Yanagisawa K. Enhancement of MTT, a tetrazolium salt, exocytosis by amyloid beta-protein and chloroquine in cultured rat astrocytes. Neurosci Lett. 1999; 266:129–132.
Article
21. Lee JW, Choi CH, Choi JJ, Park YA, Kim SJ, Hwang SY, et al. Altered MicroRNA expression in cervical carcinomas. Clin Cancer Res. 2008; 14:2535–2542.
Article
22. Kotewicz ML, D'Alessio JM, Driftmier KM, Blodgett KP, Gerard GF. Cloning and overexpression of Moloney murine leukemia virus reverse transcriptase in Escherichia coli. Gene. 1985; 35:249–258.
Article
23. Tsikas D. Analysis of nitrite and nitrate in biological fluids by assays based on the Griess reaction: appraisal of the Griess reaction in the L-arginine/nitric oxide area of research. J Chromatogr B Analyt Technol Biomed Life Sci. 2007; 851:51–70.
Article
24. Rittirsch D, Huber-Lang MS, Flierl MA, Ward PA. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009; 4:31–36.
Article
25. Lutterloh EC, Opal SM, Pittman DD, Keith JC Jr, Tan XY, Clancy BM, et al. Inhibition of the RAGE products increases survival in experimental models of severe sepsis and systemic infection. Crit Care. 2007; 11:R122.
Article
26. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003; 348:138–150.
Article
27. Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Molecular Biology of the Cell. 4th ed. New York: Garland Science;2002.
28. Thijs A, Thijs LG. Pathogenesis of renal failure in sepsis. Kidney Int Suppl. 1998; 66:S34–S37.
29. Goldberg AL. Protein degradation and protection against misfolded or damaged proteins. Nature. 2003; 426:895–899.
Article
30. Naujokat C, Hoffmann S. Role and function of the 26S proteasome in proliferation and apoptosis. Lab Invest. 2002; 82:965–980.
Article
31. Jesenberger V, Jentsch S. Deadly encounter: ubiquitin meets apoptosis. Nat Rev Mol Cell Biol. 2002; 3:112–121.
Article
32. Wang T. The 26S proteasome system in the signaling pathways of TGF-beta superfamily. Front Biosci. 2003; 8:d1109–d1127.
Article
33. Wojcikiewicz RJ. Regulated ubiquitination of proteins in GPCR-initiated signaling pathways. Trends Pharmacol Sci. 2004; 25:35–41.
Article
34. Muratani M, Tansey WP. How the ubiquitin-proteasome system controls transcription. Nat Rev Mol Cell Biol. 2003; 4:192–201.
Article
35. Fuchs SY. The role of ubiquitin-proteasome pathway in oncogenic signaling. Cancer Biol Ther. 2002; 1:337–341.
36. Kloetzel PM, Ossendorp F. Proteasome and peptidase function in MHC-class-I-mediated antigen presentation. Curr Opin Immunol. 2004; 16:76–81.
Article
37. Bowerman B, Kurz T. Degrade to create: developmental requirements for ubiquitin-mediated proteolysis during early C. elegans embryogenesis. Development. 2006; 133:773–784.
Article
38. Elliott PJ, Zollner TM, Boehncke WH. Proteasome inhibition: a new anti-inflammatory strategy. J Mol Med (Berl). 2003; 81:235–245.
Article
39. Mattingly LH, Gault RA, Murphy WJ. Use of systemic proteasome inhibition as an immune-modulating agent in disease. Endocr Metab Immune Disord Drug Targets. 2007; 7:29–34.
Article
40. Amiri KI, Horton LW, LaFleur BJ, Sosman JA, Richmond A. Augmenting chemosensitivity of malignant melanoma tumors via proteasome inhibition: implication for bortezomib (VELCADE, PS-341) as a therapeutic agent for malignant melanoma. Cancer Res. 2004; 64:4912–4918.
Article
41. Fujioka S, Schmidt C, Sclabas GM, Li Z, Pelicano H, Peng B, et al. Stabilization of p53 is a novel mechanism for proapoptotic function of NF-kappaB. J Biol Chem. 2004; 279:27549–27559.
Article
42. Sun K, Wilkins DE, Anver MR, Sayers TJ, Panoskaltsis-Mortari A, Blazar BR, et al. Differential effects of proteasome inhibition by bortezomib on murine acute graft-versus-host disease (GVHD): delayed administration of bortezomib results in increased GVHD-dependent gastrointestinal toxicity. Blood. 2005; 106:3293–3299.
Article
43. Blanco B, Pérez-Simón JA, Sánchez-Abarca LI, Carvajal-Vergara X, Mateos J, Vidriales B, et al. Bortezomib induces selective depletion of alloreactive T lymphocytes and decreases the production of Th1 cytokines. Blood. 2006; 107:3575–3583.
Article
44. Russell JA. Management of sepsis. N Engl J Med. 2006; 355:1699–1713.
Article
45. Shen J, Reis J, Morrison DC, Papasian C, Raghavakaimal S, Kolbert C, et al. Key inflammatory signaling pathways are regulated by the proteasome. Shock. 2006; 25:472–484.
Article
46. Meiners S, Ludwig A, Lorenz M, Dreger H, Baumann G, Stangl V, et al. Nontoxic proteasome inhibition activates a protective antioxidant defense response in endothelial cells. Free Radic Biol Med. 2006; 40:2232–2241.
Article
47. Hassan SW, Doody KM, Hardy S, Uetani N, Cournoyer D, Tremblay ML. Increased susceptibility to dextran sulfate sodium induced colitis in the T cell protein tyrosine phosphatase heterozygous mouse. PLoS One. 2010; 5:e8868.
Article
48. Gray PW, Goeddel DV. Cloning and expression of murine immune interferon cDNA. Proc Natl Acad Sci U S A. 1983; 80:5842–5846.
Article
49. Shaftel SS, Kyrkanides S, Olschowka JA, Miller JN, Johnson RE, O'Banion MK. Sustained hippocampal IL-1 beta overexpression mediates chronic neuroinflammation and ameliorates Alzheimer plaque pathology. J Clin Invest. 2007; 117:1595–1604.
Article
50. Moore KW, Vieira P, Fiorentino DF, Trounstine ML, Khan TA, Mosmann TR. Homology of cytokine synthesis inhibitory factor (IL-10) to the Epstein-Barr virus gene BCRFI. Science. 1990; 248:1230–1234.
Article
51. Colle JH, Falanga PB, Singer M, Hevin B, Milon G. Quantitation of messenger RNA by competitive RT-PCR: a simplified read out assay. J Immunol Methods. 1997; 210:175–184.
Article
52. Fujitani Y, Kanaoka Y, Aritake K, Uodome N, Okazaki-Hatake K, Urade Y. Pronounced eosinophilic lung inflammation and Th2 cytokine release in human lipocalin-type prostaglandin D synthase transgenic mice. J Immunol. 2002; 168:443–449.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr