goo blog サービス終了のお知らせ 

救急一直線 特別ブログ Happy保存の法則 福 皆と世界の幸せのために

本ブログは,2002年4月1日に開始され,2004年12月1日よりGOOブログに移転しました。

Mol Pharmacol 2005 67: 1018-1025

2005年03月23日 00時32分50秒 | 論文紹介 全身性炎症反応
誰も敗血症の遺伝子治療など考えていないときに発案した思い出深い研究です。
この第3弾,第4弾は,さらに驚くべき結果となっており,後世に残る研究となると考えております。

Nuclear Factor-kB Decoy Oligodeoxynucleotides Prevent Acute Lung Injury in Mice with Cecal Ligation and Puncture-Induced Sepsis
Naoyuki Matsuda, Yuichi Hattori and Satoshi Gando
Departments of Anesthesiology and Critical Care Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan


Abstract

The transcription factor nuclear factor-kB (NF-kB) plays a key role in expression of many inflammatory genes responsible for the pathophysiology of sepsis-induced acute lung injury. We investigated whether the introduction of synthetic double-stranded oligodeoxynucleotides (ODNs) with consensus NF-kB sequence as transcription factor decoy can prevent acute lung injury with suppression of pulmonary expression of multiple genes involved in its pathological process in a cecal ligation and puncture septic mouse model. NF-kB decoy ODNs were introduced with the aid of the hemagglutinating virus of Japan-envelope vector method. Northern blot analysis indicated that transfection of NF-kB decoy ODN, but not of its scrambled form, resulted in a significant inhibition of sepsis-induced gene overexpression of inducible nitric-oxide synthase (iNOS), cyclooxygenase-2, histamine H1-receptor, platelet-activating factor receptor, and bradykinin B1 and B2 receptors in lung Histological damage in lungs tissues. (wall thickening, inflammatory infiltrate, and hemorrhage), increased pulmonary vascular permeability, and blood gas exchange impairment were clearly documented in mice after cecal ligation and puncture. These changes were strongly eliminated by the introduction of NF-kB decoy but not of scrambled ODN. The effects of the iNOS inhibitor FR260330 on these histological and functional derangements compared unfavorably with those of NF-kB decoy ODN transfection. Our results suggest that ODN decoy, acting as in vivo competitor for the transcription factor's ability to bind to cognate recognition sequence, may represent an effective strategy in the treatment of septic acute lung injury.

  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする

J Clin Invest. 2005 Feb;115(2):221-4. 血管平滑筋傷害

2005年03月19日 18時43分32秒 | 論文紹介 全身性炎症反応
Defining smooth muscle cells and smooth muscle injury.

Mahoney WM, Schwartz SM.

Department of Pathology, University of Washington, Seattle, Washington 98195-7335, USA.

For 3 decades, terms such as synthetic phenotype and contractile phenotype have been used to imply the existence of a specific mechanism for smooth muscle cell (SMC) responses to injury. In this issue of the JCI, Hendrix et al. offer a far more precise approach to examining the mechanisms of SMC responses to injury, focused not on general changes in phenotype but on effects of injury on a single promoter element, the CArG [CC(A/T)6GG] box, in a single gene encoding smooth muscle (SM) alpha-actin. Since CArG box structures are present in some, but not all, SMC genes, these data suggest that we may be progressing toward establishing a systematic, molecular classification of both SMC subsets and the response of SMCs to different injuries.

敗血症病態の血管平滑筋傷害をこの半年,再び,検討し始めております。

  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする

Br J Pharmacol 144: 715-726, 2005 スタチンとエンドセリン

2005年03月19日 11時34分46秒 | 論文紹介 全身性炎症反応
Effects of statins on vascular function of endothelin-1
Fatima Mraiche1, Jonathan Cena1, Debarsi Das1 and Bozena Vollrath1

1Department of Pharmacology, Faculty of Medicine, University of Alberta, Edmonton, Alberta, Canada T6G 2H7
Correspondence: Bozena Vollrath, E-mail: bozena.vollrath@ualberta.ca

Although statins have been reported to inhibit the prepro-endothelin-1 (ET-1) gene transcription in endothelial cells, their effects on the vascular function of ET-1 have not been explored. We, therefore, examined the effects of statins on contraction and DNA synthesis mediated by ET-1 in vascular smooth muscle. The effects of statins on contraction induced by ET-1 were compared to those mediated by noradrenaline (NA) and KCl.
Simvastatin (SV) induced a concentration-dependent relaxation of tonic contraction mediated by ET-1 (10 nM) (IC50 value of 1.3 M). The relaxation was also observed in rings precontracted with NA (0.1 M) and KCl (60 mM). In contrast, pravastatin did not have any effect on the contractions.
Endothelial denudation or pretreatment with L-NAME did not prevent the relaxation, but did reduce the relaxant activity of SV.
SV prevented Rho activation caused by ET-1 and KCl in aortic homogenates, as assessed by a Rho pulldown assay.
The Rho kinase inhibitor HA-1077 mimicked the effects of SV on tonic contractions induced by ET-1, NA and KCl.
Pretreatment with the Kv channels inhibitor, 4-aminopyridine, attenuated the ability of SV to relax contractions mediated by ET-1 and NA.
In quiescent VSM cells, SV significantly inhibited DNA synthesis and Rho translocation stimulated by ET-1, as assessed by [3H]thymidine incorporation and Western blot, respectively.
Inhibition of Rho geranylgeranylation by GGTI-297, or treatment with HA-1077, mimicked the effects of SV on DNA synthesis stimulated by ET-1.
The results show that the statin potently inhibits both ET-1-mediated contraction and DNA synthesis via multiple mechanisms. Clinical benefits of statins may result, in part, from their effects on vascular function of ET-1.

僕の敗血症病態におけるスタチンの研究はほぼ完結に近づいています。

  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする

敗血症の定義に関する重要文献集 Dr.Marshallの論文もあります

2005年03月15日 20時26分48秒 | 論文紹介 全身性炎症反応

 敗血症の定義に関する重要文献集 

1. Members of the American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference Committee (1992) Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med 20:864–874

2. Marshall JC (2000) SIRS and MODS: what is their relevance to the science and practice of intensive care? Shock 14:586–589

3. Vincent J-L (1997) Dear SIRS, I'm sorry to say that I don't like you. Crit Care Med 25:372–374

4. Ramsay G, Gerlach H, Levy MM, et al (2003) An international sepsis survey: a study of doctors' knowledge and perception about sepsis. Crit Care Med 2003

5. Joint International Society and Federation of Cardiology/World Health Organization Task Force on Standardization of Clinical Nomenclature (1979) Nomenclature and criteria for diagnosis of ischemic heart disease. Circulation 59:607–609

6. Falahati A, Sharkey SW, Christensen D, et al (1999) Implementation of serum cardiac troponin I as marker for detection of acute myocardial infarction. Am Heart J 137:332–337

7. Antman EM, Grudzien C, Mitchell RN, et al (2002) Detection of unsuspected myocardial necrosis by rapid bedside assay for cardiac troponin T. Am Heart J 133:596–598

8. Puleo PR, Meyer D, Wathen C, et al (2002) Use of a rapid assay of subforms of creatine kinase MB to diagnose or rule out acute myocardial infarction. N Engl J Med 331:561–566

9. Joint European Society of Cardiology/American College of Cardiology Committee for the redefinition of myocardial infarction (2000) Myocardial infarction redefined–-a consensus document. J Am Coll Cardiol 36:959–969

10. Taniguchi T, Koido Y, Aiboshi J, et al (1999) Change in the ratio of interleukin-6 to interleukin-10 predicts a poor outcome in patients with systemic inflammatory response syndrome. Crit Care Med 27:1262–1264

11. Ueda S, Nishio K, Minamino N, et al (1999) Increased plasma levels of adrenomedullin in patients with systemic inflammatory response syndrome. Am J Respir Crit Care Med 160:132–136

12. Stoiser B, Knapp S, Thalhammer F, et al (1998) Time course of immunological markers in patients with the systemic inflammatory response syndrome: evaluation of sCD14, sVCAM-1, sELAM-1, MIP-1 alpha and TGF-beta 2. Eur J Clin Invest 28:672–678

13. Hietaranta A, Kemppainen E, Puolakkainen P, et al (2002) Extracellular phospholipases A2 in relation to systemic inflammatory response syndrome (SIRS) and systemic complications in severe acute pancreatitis. Pancreas 18:385–391

14. Takala A, Jousela I, Olkkola KT, et al (1999) Systemic inflammatory response syndrome without systemic inflammation in acutely ill patients admitted to hospital in a medical emergency. Clin Sci (Colch) 96:287–295

15. Sablotzki A, Borgermann J, Baulig W, Friedrich I, Spillner J, Silber RE, Czeslick E (2001) Lipopolysaccharide-binding protein (LBP) and markers of acute-phase response in patients with multiple organ dysfunction syndrome (MODS) following open heart surgery. Thorac Cardiovasc Surg 49:273–8]

16. Harbarth S, Holeckova K, Froidevaux C, et al (2001) Diagnostic value of procalcitonin, interleukin-6, and interleukin-8 in critically ill patients admitted with suspected sepsis. Am J Respir Crit Care Med 164:396–340

17. Duflo F, Debon R, Monneret G, et al (2002) Alveolar and serum procalcitonin: diagnostic and prognostic value in ventilator-associated pneumonia. Anesthesiology 96:74–79

18. Angus DC, Linde-Zwirble WT, Lidicer J, et al (2001) Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 29:1303–1310

19. Marshall JC, Cook DJ, Christou NV, et al (1995) Multiple organ dysfunction score: a reliable descriptor of a complex clinical outcome. Crit Care Med 23:1638–1652

20. Ferreira FL, Bota DP, Bross A, et al (2002) Serial evaluation of the SOFA score to predict outcome in critically ill patients. JAMA 286:1754–1758

21. Wilkinson JD, Pollack MM, Ruttiman, et al (1986) Outcome of pediatric patient with multiple organ system failure Crit Care Med 14:271–274

22. Proulx F, Fagan M, Farrell CA, et al (1996) Epidemiology of sepsis and multiple organ dysfunction syndrome in children. Chest 109:1033–1037

23. Doughty LA, Carcillo JA, Kaplan, et al (1996) Plasma nitrite and nitrate concentration and multiple organ failure in pediatric sepsis Crit Care Med 109:1033–1037

24. Leteutre S, Martinot A, Duhamel A, Gauvin F, Grandbastien B, Nam TV, Proulx F LaCroix J, LeClerc Fl (1999) Pediatric logistic dysfunction score. Development of a pediatric multiple organ dysfunction score: use of two strategies. Med Decis Makingaking 19:399-410

25. Carcillo JA, Fields AI (2002) Clinical practice parameters for hemodynamic support of pediatric and neonatal patients in septic shock. Crit Care Med 30:1365–1378

26. Denoix PX (1946) Enquete permanent dans les centres anticancereaux. Bull Inst Natl Hyg 1:70–75

27. Gospodarowicz M, Benedet L, Hutter RV, et al (1998) History and international developments in cancer staging. Cancer Prev Control 2:262–268

28. Renaud B, Brun-Buisson C, ICU-Bacteremia Study Group (2001) Outcomes of primary and catheter-related bacteremia. A cohort and case-control study in critically ill patients. Am J Respir Crit Care Med 163:1584–1590

29. Opal SM, Cohen J (1999) Clinical gram-positive sepsis: does it fundamentally differ from gram-negative bacterial sepsis? Crit Care Med 27:1608–1616

30. Ziegler EJ, Fisher CJ Jr, Sprung CL, et al (1991) Treatment of Gram-negative bacteremia and septic shock with HA-1A human monoclonal antibody against endotoxin: a randomized, double-blind, placebo-controlled trial. N Engl J Med 324:429–436

31. Wortel CH, von der Mohlen MAM, van Deventer SJH, et al (1992) Effectiveness of a human monoclonal anti-endotoxin antibody (HA-1A) in gram-negative sepsis: relationship to endotoxin and cytokine levels. J Infect Dis 166:1367–1374

32. McCloskey RV, Straube RC, Sanders C, et al (1994) Treatment of septic shock with human monoclonal antibody HA-1A: a randomized, double-blind, placebo-controlled trial. Ann Intern Med 121:1–5

33. Hausfater P, Garric S, Ayed SB, et al (2002) Usefulness of procalcitonin as a marker of systemic infection in emergency department patients: a prospective study. Clin Infect Dis 34:895–901

34. Damas P, Ledoux D, Nys M, et al (1992) Cytokine serum level during severe sepsis in human IL-6 as a marker of severity. Ann Surg 215:356–362

35. Panacek EA, Kaul M (1999) IL-6 as a marker of excessive TNF-alpha activity in sepsis. Sepsis 3:65–73

36. Bernard GR, Vincent J-L, Laterre PF, et al (2001) Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med 344:699–709

37. Annane D, Sébille V, Charpentier C, et al (2002) Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA 288:862–871

38. Vincent J-L, Moreno R, Takala J, et al on behalf of the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine (1996) The SOFA (Sepsis-Related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med 22:707–710

39. Marshall JC, Panacek EA, Teoh L, et al (2001) Modeling organ dysfunction as a risk factor, outcome, and measure of biologic effect in sepsis. Crit Care Med 28:A46

40. Eli Lilly and Company (2001) Briefing document for XIGRIS for the treatment of severe sepsis. http:www.fda.gov/ohrms/dockets/ac/01/briefing/3797b1_01_Sponsor.htm, 6 August

41. Cook R, Cook DJ, Tilley J, et al for the Canadian Critical Care Trials Group (2001) Multiple organ dysfunction: baseline and serial component scores. Crit Care Med 29:2046–2050


  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする

J. Biol. Chem. 2005 280: 10040-10046

2005年03月14日 23時04分15秒 | 論文紹介 全身性炎症反応
僕と異なるデータである。培養細胞系で検証しなければない。
しかし,血管内皮細胞にはCD14がない。LPSの反応は極端に弱いか,ないはずであるので,どういうメカニズムでiNOSが上昇するのか自体が疑問である。レフリーはどういう審査をしたのだろうか?

Resistance to Endotoxic Shock in Endothelial Nitric-oxide Synthase (eNOS) Knock-out Mice
A PRO-INFLAMMATORY ROLE FOR eNOS-DERIVED NO IN VIVO*
Linda Connelly, Melanie Madhani, and Adrian J. Hobbs
From the Wolfson Institute for Biomedical Research, University College London, Cruciform Building, Gower Street, London WC1E 6AE, United Kingdom

The expression of inducible nitric-oxide synthase (iNOS) and subsequent "high-output" nitric oxide (NO) production underlies the systemic hypotension, inadequate tissue perfusion, and organ failure associated with septic shock. Therefore, modulators of iNOS expression and activity, both endogenous and exogenous, are important in determining the magnitude and time course of this condition. We have shown previously that NO from the constitutive endothelial NOS (eNOS) is necessary to obtain maximal iNOS expression and activity following exposure of murine macrophages to lipopolysaccharide (LPS). Thus, eNOS represents an important regulator of iNOS expression in vitro. Herein, we validate this hypothesis in vivo using a murine model of sepsis. A temporal reduction in iNOS expression and activity was observed in LPS-treated eNOS knock-out (KO) mice as compared with wild-type animals; this was reflected in a more stable hemodynamic profile in eNOS KO mice during endotoxaemia. Furthermore, in human umbilical vein endothelial cells, LPS leads to the activation of eNOS through phosphoinositide 3-kinase- and Akt/protein kinase B-dependent enzyme phosphorylation. These data indicate that the pathogenesis of sepsis is characterized by an initial eNOS activation, with the resultant NO acting as a co-stimulus for the expression of iNOS, and therefore highlight a novel pro-inflammatory role for eNOS.

  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする

J Clin Invest. 2005 Feb;115(2):233-6

2005年03月12日 17時36分45秒 | 論文紹介 全身性炎症反応
Knock your SOCS off!

Leroith D, Nissley P.

Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Cancer Institute, NIH, Bethesda, Maryland 20892-1758, USA. derek@helix.nih.gov

The growth hormone/IGF-1-signaling (GH/IGF-1-signaling) system is involved in numerous physiological processes during normal growth and development and also in the aging process. Understanding the regulation of this system is therefore of importance to the biologist. Studies conducted over the past decade have shown that the JAK/STAT pathways are involved in GH signaling to the nucleus. More recently, evidence has been presented that a member of the SOCS family, SOCS2, is a negative regulator of GH signaling. This story began several years ago with the dramatic demonstration of gigantism in the SOCS2-knockout mouse. A more specific definition of the role of SOCS2 in GH signaling is provided in this issue of the JCI by the demonstration that the overgrowth phenotype of the SOCS2-/- mouse is dependent upon the presence of endogenous GH and that administration of GH to mice lacking both endogenous GH and SOCS2 produced excessive growth.

Figure 1
Model of JAK/STAT signaling and negative feedback by SOCS proteins. Cytokine signaling involves ligand binding and activation of the cell surface cytokine receptor. Recruitment and activation of JAK in turn facilitates phosphorylation of a STAT tyrosine residue, and subsequent STAT activation induces dimerization. This activation is tightly controlled by multiple negative regulators of phosphorylation such as phosphatases, SOCS, and PIAS. SOCS proteins are also induced by cytokine signaling and form a closed-loop, negative-feedback mechanism.

Figure 2
GH activates the GHR, which leads to multiple tissue effects, including IGF-1 gene expression. In turn, IGF-1, via IGF-1R, induces cellular effects. SOCS2 may negatively regulate these events at multiple levels. A series of papers from the Walter and Eliza Hall Institute of Medical Research, including the paper by Greenhalgh et al. in this issue of the JCI (3), provide convincing evidence that SOCS2 is a negative regulator of GH receptor signaling. Negative regulation of IGF-1R signaling by SOCS2 is more speculative.

  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする

Cancer Res. 2003 Oct 1;63(19):6424-31

2005年03月12日 16時44分03秒 | 論文紹介 全身性炎症反応
Interferon regulatory factor 5, a novel mediator of cell cycle arrest and cell death.

Barnes BJ, Kellum MJ, Pinder KE, Frisancho JA, Pitha PM.

Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD 21231, USA. barnebe@jhmi.edu

We have previously shown a critical role for IFN regulatory factor 5 (IRF-5) in the innate immune response to virus infection. For the first time, we now show that although IRF-5 is a direct target of p53, its cell cycle regulatory and proapoptotic effects are p53 independent. IRF-5 inhibits both in vitro and in vivo B-cell lymphoma tumor growth in the absence of wild-type p53. The molecular mechanism(s) of IRF-5-mediated growth inhibition is associated with a G(2)-M cell cycle arrest and modulation of growth regulatory and proapoptotic genes, including p21, Bak, DAP kinase 2, and Bax. Taken together, these data indicate that although IRF-5 is a downstream target of p53, its growth inhibitory and proapoptotic effects are independent of p53.

  • X
  • Facebookでシェアする
  • はてなブックマークに追加する
  • LINEでシェアする