Effects of anti-inflammatory vagus nerve stimulation in endotoxemic rats on blood and spleen lymphocyte subsets
详细信息    查看全文
  • 作者:S. Mihaylova (1)
    H. Schweigh?fer (1)
    H. Hackstein (2)
    B. Rosengarten (1)
  • 关键词:Vagus nerve stimulation ; Endotoxic shock ; Lymphocytes ; Splenocytes ; Anti ; inflammation ; Rat
  • 刊名:Inflammation Research
  • 出版年:2014
  • 出版时间:August 2014
  • 年:2014
  • 卷:63
  • 期:8
  • 页码:683-690
  • 全文大小:230 KB
  • 参考文献:1. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348:138-0. CrossRef
    2. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med. 2001;29:1303-0. CrossRef
    3. Angus DC, Wax RS. Epidemiology of sepsis: an update. Crit Care Med. 2001;29:S109-6. CrossRef
    4. Nguyen HB, Smith D. Sepsis in the 21st century: recent definitions and therapeutic advances. Am J Emerg Med. 2007;25:564-1. CrossRef
    5. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med. 2003;348:1546-4. CrossRef
    6. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D, et al. 2001 SCCM/ESICM/ACCP/ATS/SIS international sepsis definitions conference. Crit Care Med. 2003;31:1250-. CrossRef
    7. Nathan C. Points of control in inflammation. Nature. 2002;420:846-2. CrossRef
    8. Wang H, Liao H, Ochani M, Justiniani M, Lin X, Yang L, et al. Cholinergic agonists inhibit HMGB1 release and improve survival in experimental sepsis. Nat Med. 2004;10:1216-1. CrossRef
    9. Daubeuf B, Mathison J, Spiller S, Hugues S, Herren S, Ferlin W, et al. TLR4/MD-2 monoclonal antibody therapy affords protection in experimental models of septic shock. J Immunol. 2007;179:6107-4. CrossRef
    10. Calandra T, Echtenacher B, Roy DL, Pugin J, Metz CN, Hultner L, et al. Protection from septic shock by neutralization of macrophage migration inhibitory factor. Nat Med. 2000;6:164-0. CrossRef
    11. Yang H, Ochani M, Li J, Qiang X, Tanovic M, Harris HE, et al. Reversing established sepsis with antagonists of endogenous high-mobility group box 1. Proc Natl Acad Sci USA. 2004;101:296-01. CrossRef
    12. Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458-2. CrossRef
    13. Tracey KJ. Reflex control of immunity. Nat Rev Immunol. 2009;9:418-8. CrossRef
    14. Huston JM, Gallowitsch-Puerta M, Ochani M, Ochani K, Yuan R, Rosas-Ballina M, et al. Transcutaneous vagus nerve stimulation reduces serum high mobility group box 1 levels and improves survival in murine sepsis. Crit Care Med. 2007;35:2762-. CrossRef
    15. Pena G, Cai B, Ramos L, Vida G, Deitch EA, Ulloa L. Cholinergic regulatory lymphocytes re-establish neuromodulation of innate immune responses in sepsis. J Immunol. 2011;187:718-5. CrossRef
    16. Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421:384-. CrossRef
    17. Rosas-Ballina M, Ochani M, Parrish WR, Ochani K, Harris YT, Huston JM, et al. Splenic nerve is required for cholinergic antiinflammatory pathway control of TNF in endotoxemia. Proc Natl Acad Sci USA. 2008;105:11008-3. CrossRef
    18. Huston JM, Ochani M, Rosas-Ballina M, Liao H, Ochani K, Pavlov VA, et al. Splenectomy inactivates the cholinergic antiinflammatory pathway during lethal endotoxemia and polymicrobial sepsis. J Exp Med. 2006;203:1623-. CrossRef
    19. Rosengarten B, Hecht M, Wolff S, Kaps M. Autoregulative function in the brain in an endotoxic rat shock model. Inflamm Res. 2008;57:542-. CrossRef
    20. Holub M, Kluckova Z, Beneda B, Hobstova J, Huzicka I, Prazak J, et al. Changes in lymphocyte subpopulations and CD3+/DR+ expression in sepsis. Clin Microbiol Infect. 2000;6:657-0. CrossRef
    21. Holub M, Kluckova Z, Helcl M, Prihodov J, Rokyta R, Beran O. Lymphocyte subset numbers depend on the bacterial origin of sepsis. Clin Microbiol Infect. 2003;9:202-1. CrossRef
    22. Lin RY, Astiz ME, Saxon JC, Rackow EC. Altered leukocyte immunophenotypes in septic shock. Studies of HLA-DR, CD11b, CD14, and IL-2R expression. Chest. 1993;104:847-3. CrossRef
    23. Nishijima MK, Takezawa J, Hosotsubo KK, Takahashi H, Shimada Y, Yoshiya I. Serial changes in cellular immunity of septic patients with multiple organ-system failure. Crit Care Med. 1986;14:87-1. CrossRef
    24. Hotchkiss RS, Coopersmith CM, McDunn JE, Ferguson TA. The sepsis seesaw: tilting toward immunosuppression. Nat Med. 2009;15:496-. CrossRef
    25. Sharshar T, Hopkinson NS, Orlikowski D, Annane D. Science review: the brain in sepsis–culprit and victim. Crit Care. 2005;9:37-4. CrossRef
    26. Monserrat J, de Pablo R, Reyes E, Diaz D, Barcenilla H, Zapata MR, et al. Clinical relevance of the severe abnormalities of the T cell compartment in septic shock patients. Crit Care. 2009;13:R26. CrossRef
    27. Sherwood ER, Enoh VT, Murphey ED, Lin CY. Mice depleted of CD8+ T and NK cells are resistant to injury caused by cecal ligation and puncture. Lab Investig J Tech Meth Pathol. 2004;84:1655-5. CrossRef
    28. Chang WL, Jones SP, Lefer DJ, Welbourne T, Sun G, Yin L, et al. CD8(+)-T-cell depletion ameliorates circulatory shock in Plasmodium berghei-infected mice. Infect Immun. 2001;69:7341-. CrossRef
    29. Menges T, Engel J, Welters I, Wagner RM, Little S, Ruwoldt R, et al. Changes in blood lymphocyte populations after multiple trauma: association with posttraumatic complications. Crit Care Med. 1999;27:733-0. CrossRef
    30. Sarobe P, Lasarte JJ, Garcia N, Civeira MP, Borras-Cuesta F, Prieto J. Characterization of T-cell responses against immunodominant epitopes from hepatitis C virus E2 and NS4a proteins. J Viral Hepatitis. 2006;13:47-5. CrossRef
    31. Lim B, Sutherland RM, Zhan Y, Deliyannis G, Brown LE, Lew AM. Targeting CD45RB alters T cell migration and delays viral clearance. Int Immunol. 2006;18:291-00. CrossRef
    32. Du X, Zheng G, Jin H, Kang Y, Wang J, Xiao C, et al. The adjuvant effects of co-stimulatory molecules on cellular and memory responses to HBsAg DNA vaccination. J Gene Med. 2007;9:136-6. CrossRef
    33. Karimi K, Bienenstock J, Wang L, Forsythe P. The vagus nerve modulates CD4+ T cell activity. Brain Behav Immun. 2010;24:316-3. CrossRef
    34. McDunn JE, Turnbull IR, Polpitiya AD, Tong A, MacMillan SK, Osborne DF, et al. Splenic CD4+ T cells have a distinct transcriptional response six hours after the onset of sepsis. J Am Coll Surg. 2006;203:365-5. CrossRef
    35. Klehmet J, Harms H, Richter M, Prass K, Volk HD, Dirnagl U, et al. Stroke-induced immunodepression and post-stroke infections: lessons from the preventive antibacterial therapy in stroke trial. Neuroscience. 2009;158:1184-3. CrossRef
    36. Kerr AR, Kirkham LA, Kadioglu A, Andrew PW, Garside P, Thompson H, et al. Identification of a detrimental role for NK cells in pneumococcal pneumonia and sepsis in immunocompromised hosts. Microb Infect Institut Pasteur. 2005;7:845-2. CrossRef
    37. Zhang L, Cardinal JS, Pan P, Rosborough BR, Chang Y, Yan W, et al. Splenocyte apoptosis and autophagy is mediated by interferon regulatory factor-1 during murine endotoxemia. Shock. 2012;37(5):511-. CrossRef
    38. Mazzola S, Forni M, Albertini M, Bacci ML, Zannoni A, Gentilini F, et al. Carbon monoxide pretreatment prevents respiratory derangement and ameliorates hyperacute endotoxic shock in pigs. FASEB J. 2005;19:2045-.
    39. Huang J, Wang Y, Jiang D, Zhou J, Huang X. The sympathetic-vagal balance against endotoxemia. J Neural Transm. 2010;117:729-5. CrossRef
  • 作者单位:S. Mihaylova (1)
    H. Schweigh?fer (1)
    H. Hackstein (2)
    B. Rosengarten (1)

    1. Departments of Neurology, Justus-Liebig University Giessen, Klinikstrasse 33, 35392, Giessen, Germany
    2. Departments of Clinical Immunology and Transfusion Therapy, Justus-Liebig University Giessen, Klinikstrasse 33, 35392, Giessen, Germany
  • ISSN:1420-908X
文摘
Background Anti-inflammatory cytokine effects of vagus nerve stimulation in sepsis syndromes are well established. Effects on immune cells are less clear. Therefore, we studied changes in peripheral and spleen leukocyte subsets in an endotoxic rat sepsis model. Methods Ventilated and sedated adult male SD rats received 5?mg/kg b.w. lipopolysaccharide intravenously to induce endotoxic sepsis. Controls and a group with both-sided vagotomy were compared to animals with both sided vagotomy and left distal vagus nerve stimulation. 4.5?h after sepsis induction immune cell counts and types in the peripheral blood and spleen were determined [T-lymphocytes (CD3+), T-helper cells (CD3+ CD4+), activated T-helper cells (CD3+ CD4+ CD134+), cytotoxic T-cells (CD3+ CD8+), activated cytotoxic T-cells (CD3+ CD8+ CD134+), B-lymphocytes (CD45R+ CD11cneg-dim), dendritic cells (CD11c+ OX-62?+), natural killer cells (CD161+ CD3neg) and granulocytes (His48?+)] together with cytokine and chemokine plasma levels (IL10; IFN-g, TNF-a, Cxcl5, Ccl5). Results Blood cell counts declined in all LPS groups. However, vagus nerve stimulation but not vagotomy activated cytotoxic T-cells. Vagotomy also depleted natural killer cells. In the spleen, vagotomy resulted in a strong decline of all cell types which was not present in the other septic groups where only granulocyte numbers declined. Conclusion Vagotomy strongly declines immune cell counts in the septic spleen. This could not be explained by an evasion or apoptosis of cells. A marginalisation of spleen immune cells into the peripheral microcirculation might be therefore most likely. Further studies are warranted to clear this issue.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700