Skip to main content
Erschienen in: Intensive Care Medicine 9/2012

01.09.2012 | Experimental

Effects of anesthetic regimes on inflammatory responses in a rat model of acute lung injury

verfasst von: Spyridon Fortis, Peter M. Spieth, Wei-Yang Lu, Matteo Parotto, Jack J. Haitsma, Arthur S. Slutsky, Nanshan Zhong, C. David Mazer, Haibo Zhang

Erschienen in: Intensive Care Medicine | Ausgabe 9/2012

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter through activation of GABA receptors. Volatile anesthetics activate type-A (GABAA) receptors resulting in inhibition of synaptic transmission. Lung epithelial cells have been recently found to express GABAA receptors that exert anti-inflammatory properties. We hypothesized that the volatile anesthetic sevoflurane (SEVO) attenuates lung inflammation through activation of lung epithelial GABAA receptors.

Methods

Sprague–Dawley rats were anesthetized with SEVO or ketamine/xylazine (KX). Acute lung inflammation was induced by intratracheal instillation of endotoxin, followed by mechanical ventilation for 4 h at a tidal volume of 15 mL/kg without positive end-expiratory pressure (two-hit lung injury model). To examine the specific effects of GABA, healthy human lung epithelial cells (BEAS-2B) were challenged with endotoxin in the presence and absence of GABA with and without addition of the GABAA receptor antagonist picrotoxin.

Results

Anesthesia with SEVO improved oxygenation and reduced pulmonary cytokine responses compared to KX. This phenomenon was associated with increased expression of the π subunit of GABAA receptors and glutamic acid decarboxylase (GAD). The endotoxin-induced cytokine release from BEAS-2B cells was attenuated by the treatment with GABA, which was reversed by the administration of picrotoxin.

Conclusion

Anesthesia with SEVO suppresses pulmonary inflammation and thus protects the lung from the two-hit injury. The anti-inflammatory effect of SEVO is likely due to activation of pulmonary GABAA signaling pathways.
Literatur
1.
Zurück zum Zitat Phua J, Badia JR, Adhikari NK, Friedrich JO, Fowler RA, Singh JM, Scales DC, Stather DR, Li A, Jones A, Gattas DJ, Hallett D, Tomlinson G, Stewart TE, Ferguson ND (2009) Has mortality from acute respiratory distress syndrome decreased over time? A systematic review. Am J Respir Crit Care Med 179:220–227PubMedCrossRef Phua J, Badia JR, Adhikari NK, Friedrich JO, Fowler RA, Singh JM, Scales DC, Stather DR, Li A, Jones A, Gattas DJ, Hallett D, Tomlinson G, Stewart TE, Ferguson ND (2009) Has mortality from acute respiratory distress syndrome decreased over time? A systematic review. Am J Respir Crit Care Med 179:220–227PubMedCrossRef
2.
Zurück zum Zitat The Acute Respiratory Distress Syndrome Network (2000) Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 342:1301–1308CrossRef The Acute Respiratory Distress Syndrome Network (2000) Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med 342:1301–1308CrossRef
3.
Zurück zum Zitat Brun-Buisson C, Minelli C, Bertolini G, Brazzi L, Pimentel J, Lewandowski K, Bion J, Romand J, Villar J, Thorsteinsson A, Damas P, Armaganidis A, Lemaire F, Alive Study Group (2004) Epidemiology and outcome of acute lung injury in European intensive care units. Results from the ALIVE study. Intensive Care Med 30:51–61PubMedCrossRef Brun-Buisson C, Minelli C, Bertolini G, Brazzi L, Pimentel J, Lewandowski K, Bion J, Romand J, Villar J, Thorsteinsson A, Damas P, Armaganidis A, Lemaire F, Alive Study Group (2004) Epidemiology and outcome of acute lung injury in European intensive care units. Results from the ALIVE study. Intensive Care Med 30:51–61PubMedCrossRef
4.
Zurück zum Zitat Tremblay L, Valenza F, Ribeiro SP, Li J, Slutsky AS (1997) Injurious ventilatory strategies increase cytokines and c-fos m-RNA expression in an isolated rat lung model 4. J Clin Invest 99:944–952PubMedCrossRef Tremblay L, Valenza F, Ribeiro SP, Li J, Slutsky AS (1997) Injurious ventilatory strategies increase cytokines and c-fos m-RNA expression in an isolated rat lung model 4. J Clin Invest 99:944–952PubMedCrossRef
5.
Zurück zum Zitat Malhotra A (2007) Low-tidal-volume ventilation in the acute respiratory distress syndrome. N Engl J Med 357:1113–1120PubMedCrossRef Malhotra A (2007) Low-tidal-volume ventilation in the acute respiratory distress syndrome. N Engl J Med 357:1113–1120PubMedCrossRef
6.
Zurück zum Zitat Voigtsberger S, Lachmann RA, Leutert AC, Schlapfer M, Booy C, Reyes L, Urner M, Schild J, Schimmer RC, Beck-Schimmer B (2009) Sevoflurane ameliorates gas exchange and attenuates lung damage in experimental lipopolysaccharide-induced lung injury. Anesthesiology 111:1238–1248PubMedCrossRef Voigtsberger S, Lachmann RA, Leutert AC, Schlapfer M, Booy C, Reyes L, Urner M, Schild J, Schimmer RC, Beck-Schimmer B (2009) Sevoflurane ameliorates gas exchange and attenuates lung damage in experimental lipopolysaccharide-induced lung injury. Anesthesiology 111:1238–1248PubMedCrossRef
7.
Zurück zum Zitat De Conno E, Steurer MP, Wittlinger M, Zalunardo MP, Weder W, Schneiter D, Schimmer RC, Klaghofer R, Neff TA, Schmid ER, Spahn DR, Z’Graggen BR, Urner M, Beck-Schimmer B (2009) Anesthetic-induced improvement of the inflammatory response to one-lung ventilation. Anesthesiology 110:1316–1326PubMedCrossRef De Conno E, Steurer MP, Wittlinger M, Zalunardo MP, Weder W, Schneiter D, Schimmer RC, Klaghofer R, Neff TA, Schmid ER, Spahn DR, Z’Graggen BR, Urner M, Beck-Schimmer B (2009) Anesthetic-induced improvement of the inflammatory response to one-lung ventilation. Anesthesiology 110:1316–1326PubMedCrossRef
8.
Zurück zum Zitat Boost KA, Leipold T, Scheiermann P, Hoegl S, Sadik CD, Hofstetter C, Zwissler B (2009) Sevoflurane and isoflurane decrease TNF-alpha-induced gene expression in human monocytic THP-1 cells: potential role of intracellular I kappa B alpha regulation. Int J Mol Med 23:665–671PubMedCrossRef Boost KA, Leipold T, Scheiermann P, Hoegl S, Sadik CD, Hofstetter C, Zwissler B (2009) Sevoflurane and isoflurane decrease TNF-alpha-induced gene expression in human monocytic THP-1 cells: potential role of intracellular I kappa B alpha regulation. Int J Mol Med 23:665–671PubMedCrossRef
9.
Zurück zum Zitat Hirota K, Roth SH (1997) Sevoflurane modulates both GABAA and GABAB receptors in area CA1 of rat hippocampus. Br J Anaesth 78:60–65PubMedCrossRef Hirota K, Roth SH (1997) Sevoflurane modulates both GABAA and GABAB receptors in area CA1 of rat hippocampus. Br J Anaesth 78:60–65PubMedCrossRef
10.
Zurück zum Zitat Sebel LE, Richardson JE, Singh SP, Bell SV, Jenkins A (2006) Additive effects of sevoflurane and propofol on gamma-aminobutyric acid receptor function. Anesthesiology 104:1176–1183PubMedCrossRef Sebel LE, Richardson JE, Singh SP, Bell SV, Jenkins A (2006) Additive effects of sevoflurane and propofol on gamma-aminobutyric acid receptor function. Anesthesiology 104:1176–1183PubMedCrossRef
11.
Zurück zum Zitat Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, Steinman L (2010) Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci USA 107:2580–2585PubMedCrossRef Bhat R, Axtell R, Mitra A, Miranda M, Lock C, Tsien RW, Steinman L (2010) Inhibitory role for GABA in autoimmune inflammation. Proc Natl Acad Sci USA 107:2580–2585PubMedCrossRef
12.
Zurück zum Zitat Duthey B, Hubner A, Diehl S, Boehncke S, Pfeffer J, Boehncke WH (2010) Anti-inflammatory effects of the GABAB receptor agonist baclofen in allergic contact dermatitis. Exp Dermatol 19:661–666PubMedCrossRef Duthey B, Hubner A, Diehl S, Boehncke S, Pfeffer J, Boehncke WH (2010) Anti-inflammatory effects of the GABAB receptor agonist baclofen in allergic contact dermatitis. Exp Dermatol 19:661–666PubMedCrossRef
13.
Zurück zum Zitat Jin N, Kolliputi N, Gou D, Weng T, Liu L (2006) A novel function of ionotropic gamma-aminobutyric acid receptors involving alveolar fluid homeostasis. J Biol Chem 281:36012–36020PubMedCrossRef Jin N, Kolliputi N, Gou D, Weng T, Liu L (2006) A novel function of ionotropic gamma-aminobutyric acid receptors involving alveolar fluid homeostasis. J Biol Chem 281:36012–36020PubMedCrossRef
14.
Zurück zum Zitat Jin N, Narasaraju T, Kolliputi N, Chen J, Liu L (2005) Differential expression of GABAA receptor pi subunit in cultured rat alveolar epithelial cells. Cell Tissue Res 321:173–183PubMedCrossRef Jin N, Narasaraju T, Kolliputi N, Chen J, Liu L (2005) Differential expression of GABAA receptor pi subunit in cultured rat alveolar epithelial cells. Cell Tissue Res 321:173–183PubMedCrossRef
15.
Zurück zum Zitat Xiang YY, Wang S, Liu M, Hirota JA, Li J, Ju W, Fan Y, Kelly MM, Ye B, Orser B, O’Byrne PM, Inman MD, Yang X, Lu WY (2007) A GABAergic system in airway epithelium is essential for mucus overproduction in asthma. Nat Med 13:862–867PubMedCrossRef Xiang YY, Wang S, Liu M, Hirota JA, Li J, Ju W, Fan Y, Kelly MM, Ye B, Orser B, O’Byrne PM, Inman MD, Yang X, Lu WY (2007) A GABAergic system in airway epithelium is essential for mucus overproduction in asthma. Nat Med 13:862–867PubMedCrossRef
16.
Zurück zum Zitat Mizuta K, Osawa Y, Mizuta F, Xu D, Emala CW (2008) Functional expression of GABAB receptors in airway epithelium. Am J Respir Cell Mol Biol 39:296–304PubMedCrossRef Mizuta K, Osawa Y, Mizuta F, Xu D, Emala CW (2008) Functional expression of GABAB receptors in airway epithelium. Am J Respir Cell Mol Biol 39:296–304PubMedCrossRef
17.
Zurück zum Zitat Voglis S, Quinn K, Tullis E, Liu M, Henriques M, Zubrinich C, Penuelas O, Chan H, Silverman F, Cherepanov V, Orzech N, Khine AA, Cantin A, Slutsky AS, Downey GP, Zhang H (2009) Human neutrophil peptides and phagocytic deficiency in bronchiectatic lungs. Am J Respir Crit Care Med 180:159–166PubMedCrossRef Voglis S, Quinn K, Tullis E, Liu M, Henriques M, Zubrinich C, Penuelas O, Chan H, Silverman F, Cherepanov V, Orzech N, Khine AA, Cantin A, Slutsky AS, Downey GP, Zhang H (2009) Human neutrophil peptides and phagocytic deficiency in bronchiectatic lungs. Am J Respir Crit Care Med 180:159–166PubMedCrossRef
18.
Zurück zum Zitat Zhang H, Kim YK, Govindarajan A, Baba A, Binnie M, Marco Ranieri V, Liu M, Slutsky AS (1999) Effect of adrenoreceptors on endotoxin-induced cytokines and lipid peroxidation in lung explants. Am J Respir Crit Care Med 160:1703–1710PubMed Zhang H, Kim YK, Govindarajan A, Baba A, Binnie M, Marco Ranieri V, Liu M, Slutsky AS (1999) Effect of adrenoreceptors on endotoxin-induced cytokines and lipid peroxidation in lung explants. Am J Respir Crit Care Med 160:1703–1710PubMed
19.
Zurück zum Zitat Racher AJ, Looby D, Griffiths JB (1990) Use of lactate dehydrogenase release to assess changes in culture viability. Cytotechnology 3:301–307PubMedCrossRef Racher AJ, Looby D, Griffiths JB (1990) Use of lactate dehydrogenase release to assess changes in culture viability. Cytotechnology 3:301–307PubMedCrossRef
20.
Zurück zum Zitat Green CJ, Knight J, Precious S, Simpkin S (1981) Ketamine alone and combined with diazepam or xylazine in laboratory animals: a 10 year experience. Lab Anim 15:163–170PubMedCrossRef Green CJ, Knight J, Precious S, Simpkin S (1981) Ketamine alone and combined with diazepam or xylazine in laboratory animals: a 10 year experience. Lab Anim 15:163–170PubMedCrossRef
21.
Zurück zum Zitat Narita M, Yoshizawa K, Aoki K, Takagi M, Miyatake M, Suzuki T (2001) A putative sigma1 receptor antagonist NE-100 attenuates the discriminative stimulus effects of ketamine in rats. Addict Biol 6:373–376PubMedCrossRef Narita M, Yoshizawa K, Aoki K, Takagi M, Miyatake M, Suzuki T (2001) A putative sigma1 receptor antagonist NE-100 attenuates the discriminative stimulus effects of ketamine in rats. Addict Biol 6:373–376PubMedCrossRef
22.
Zurück zum Zitat Hevers W, Hadley SH, Luddens H, Amin J (2008) Ketamine, but not phencyclidine, selectively modulates cerebellar GABAA receptors containing alpha6 and delta subunits. J Neurosci 28:5383–5393PubMedCrossRef Hevers W, Hadley SH, Luddens H, Amin J (2008) Ketamine, but not phencyclidine, selectively modulates cerebellar GABAA receptors containing alpha6 and delta subunits. J Neurosci 28:5383–5393PubMedCrossRef
23.
Zurück zum Zitat Hirota K, Sikand KS, Lambert DG (1999) Interaction of ketamine with mu2 opioid receptors in SH-SY5Y human neuroblastoma cells. J Anesth 13:107–109PubMedCrossRef Hirota K, Sikand KS, Lambert DG (1999) Interaction of ketamine with mu2 opioid receptors in SH-SY5Y human neuroblastoma cells. J Anesth 13:107–109PubMedCrossRef
24.
Zurück zum Zitat Kounenis G, Koutsoviti-Papadopoulou M, Elezoglou V (1995) Ketamine may modify intestinal motility by acting at GABAA-receptor complex; an in vitro study on the guinea pig intestine. Pharmacol Res 31:337–340PubMedCrossRef Kounenis G, Koutsoviti-Papadopoulou M, Elezoglou V (1995) Ketamine may modify intestinal motility by acting at GABAA-receptor complex; an in vitro study on the guinea pig intestine. Pharmacol Res 31:337–340PubMedCrossRef
25.
Zurück zum Zitat Vutskits L, Gascon E, Potter G, Tassonyi E, Kiss JZ (2007) Low concentrations of ketamine initiate dendritic atrophy of differentiated GABAergic neurons in culture. Toxicology 234:216–226PubMedCrossRef Vutskits L, Gascon E, Potter G, Tassonyi E, Kiss JZ (2007) Low concentrations of ketamine initiate dendritic atrophy of differentiated GABAergic neurons in culture. Toxicology 234:216–226PubMedCrossRef
26.
Zurück zum Zitat Sonner JM, Zhang Y, Stabernack C, Abaigar W, Xing Y, Laster MJ (2003) GABAA receptor blockade antagonizes the immobilizing action of propofol but not ketamine or isoflurane in a dose-related manner. Anesth Analg 96:706–712PubMed Sonner JM, Zhang Y, Stabernack C, Abaigar W, Xing Y, Laster MJ (2003) GABAA receptor blockade antagonizes the immobilizing action of propofol but not ketamine or isoflurane in a dose-related manner. Anesth Analg 96:706–712PubMed
27.
Zurück zum Zitat Zhang Y, Behrens MM, Lisman JE (2008) Prolonged exposure to NMDAR antagonist suppresses inhibitory synaptic transmission in prefrontal cortex. J Neurophysiol 100:959–965PubMedCrossRef Zhang Y, Behrens MM, Lisman JE (2008) Prolonged exposure to NMDAR antagonist suppresses inhibitory synaptic transmission in prefrontal cortex. J Neurophysiol 100:959–965PubMedCrossRef
28.
Zurück zum Zitat Irifune M, Sato T, Kamata Y, Nishikawa T, Dohi T, Kawahara M (2000) Evidence for GABAA receptor agonistic properties of ketamine: convulsive and anaesthetic behavioural models in mice. Anesth Analg 91:230–236PubMed Irifune M, Sato T, Kamata Y, Nishikawa T, Dohi T, Kawahara M (2000) Evidence for GABAA receptor agonistic properties of ketamine: convulsive and anaesthetic behavioural models in mice. Anesth Analg 91:230–236PubMed
29.
Zurück zum Zitat Garcia-Villar R, Toutain PL, Alvinerie M, Ruckebusch Y (1981) The pharmacokinetics of xylazine hydrochloride: an interspecific study. J Vet Pharmacol Ther 4:87–92PubMedCrossRef Garcia-Villar R, Toutain PL, Alvinerie M, Ruckebusch Y (1981) The pharmacokinetics of xylazine hydrochloride: an interspecific study. J Vet Pharmacol Ther 4:87–92PubMedCrossRef
30.
Zurück zum Zitat Riker RR, Shehabi Y, Bokesch PM, Ceraso D, Wisemandle W, Koura F, Whitten P, Margolis BD, Byrne DW, Ely EW, Rocha MG (2009) Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. JAMA 301:489–499PubMedCrossRef Riker RR, Shehabi Y, Bokesch PM, Ceraso D, Wisemandle W, Koura F, Whitten P, Margolis BD, Byrne DW, Ely EW, Rocha MG (2009) Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. JAMA 301:489–499PubMedCrossRef
31.
Zurück zum Zitat Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, Nusrat A (2003) Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol 171:6164–6172PubMed Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, Nusrat A (2003) Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol 171:6164–6172PubMed
32.
Zurück zum Zitat Choi H, Fleming NW, Serikov VB (2007) Contact activation via ICAM-1 induces changes in airway epithelial permeability in vitro. Immunol Invest 36:59–72PubMedCrossRef Choi H, Fleming NW, Serikov VB (2007) Contact activation via ICAM-1 induces changes in airway epithelial permeability in vitro. Immunol Invest 36:59–72PubMedCrossRef
33.
Zurück zum Zitat Lee HT, Kim M, Jan M, Emala CW (2006) Anti-inflammatory and antinecrotic effects of the volatile anaesthetic sevoflurane in kidney proximal tubule cells. Am J Physiol Renal Physiol 291:F67–F78PubMedCrossRef Lee HT, Kim M, Jan M, Emala CW (2006) Anti-inflammatory and antinecrotic effects of the volatile anaesthetic sevoflurane in kidney proximal tubule cells. Am J Physiol Renal Physiol 291:F67–F78PubMedCrossRef
34.
Zurück zum Zitat Jia F, Yue M, Chandra D, Homanics GE, Goldstein PA, Harrison NL (2008) Isoflurane is a potent modulator of extrasynaptic GABAA receptors in the thalamus. J Pharmacol Exp Ther 324:1127–1135PubMedCrossRef Jia F, Yue M, Chandra D, Homanics GE, Goldstein PA, Harrison NL (2008) Isoflurane is a potent modulator of extrasynaptic GABAA receptors in the thalamus. J Pharmacol Exp Ther 324:1127–1135PubMedCrossRef
35.
Zurück zum Zitat Erickson MA, Banks WA (2011) Cytokine and chemokine responses in serum and brain after single and repeated injections of lipopolysaccharide: multiplex quantification with path analysis. Brain Behav Immun 25:1637–1648PubMedCrossRef Erickson MA, Banks WA (2011) Cytokine and chemokine responses in serum and brain after single and repeated injections of lipopolysaccharide: multiplex quantification with path analysis. Brain Behav Immun 25:1637–1648PubMedCrossRef
36.
Zurück zum Zitat Ward JL, Harting MT, Cox CS Jr, Mercer DW (2011) Effects of ketamine on endotoxin and traumatic brain injury induced cytokine production in the rat. J Trauma 70:1471–1479PubMedCrossRef Ward JL, Harting MT, Cox CS Jr, Mercer DW (2011) Effects of ketamine on endotoxin and traumatic brain injury induced cytokine production in the rat. J Trauma 70:1471–1479PubMedCrossRef
37.
Zurück zum Zitat Faller S, Strosing KM, Ryter SW, Buerkle H, Loop T, Schmidt R, Hoetzel A (2012) The volatile anaesthetic isoflurane prevents ventilator-induced lung injury via phosphoinositide 3-kinase/Akt signaling in mice. Anesth Analg 114:747–756PubMedCrossRef Faller S, Strosing KM, Ryter SW, Buerkle H, Loop T, Schmidt R, Hoetzel A (2012) The volatile anaesthetic isoflurane prevents ventilator-induced lung injury via phosphoinositide 3-kinase/Akt signaling in mice. Anesth Analg 114:747–756PubMedCrossRef
38.
Zurück zum Zitat Porcher C, Hatchett C, Longbottom RE, McAinch K, Sihra TS, Moss SJ, Thomson AM, Jovanovic JN (2011) Positive feedback regulation between gamma-aminobutyric acid type A (GABAA) receptor signaling and brain-derived neurotrophic factor (BDNF) release in developing neurons. J Biol Chem 286:21667–21677PubMedCrossRef Porcher C, Hatchett C, Longbottom RE, McAinch K, Sihra TS, Moss SJ, Thomson AM, Jovanovic JN (2011) Positive feedback regulation between gamma-aminobutyric acid type A (GABAA) receptor signaling and brain-derived neurotrophic factor (BDNF) release in developing neurons. J Biol Chem 286:21667–21677PubMedCrossRef
39.
Zurück zum Zitat Kaufman DA (2009) Time-dependent behavioral recovery after sepsis in rats. Intensive Care Med 35:576; author reply 577PubMedCrossRef Kaufman DA (2009) Time-dependent behavioral recovery after sepsis in rats. Intensive Care Med 35:576; author reply 577PubMedCrossRef
40.
Zurück zum Zitat Kitamura T, Ogawa M, Kawamura G, Sato K, Yamada Y (2009) The effects of sevoflurane and propofol on glucose metabolism under aerobic conditions in fed rats. Anesth Analg 109:1479–1485PubMedCrossRef Kitamura T, Ogawa M, Kawamura G, Sato K, Yamada Y (2009) The effects of sevoflurane and propofol on glucose metabolism under aerobic conditions in fed rats. Anesth Analg 109:1479–1485PubMedCrossRef
41.
Zurück zum Zitat Honiden S, Gong MN (2009) Diabetes, insulin, and development of acute lung injury. Crit Care Med 37:2455–2464PubMedCrossRef Honiden S, Gong MN (2009) Diabetes, insulin, and development of acute lung injury. Crit Care Med 37:2455–2464PubMedCrossRef
Metadaten
Titel
Effects of anesthetic regimes on inflammatory responses in a rat model of acute lung injury
verfasst von
Spyridon Fortis
Peter M. Spieth
Wei-Yang Lu
Matteo Parotto
Jack J. Haitsma
Arthur S. Slutsky
Nanshan Zhong
C. David Mazer
Haibo Zhang
Publikationsdatum
01.09.2012
Verlag
Springer-Verlag
Erschienen in
Intensive Care Medicine / Ausgabe 9/2012
Print ISSN: 0342-4642
Elektronische ISSN: 1432-1238
DOI
https://doi.org/10.1007/s00134-012-2610-4

Weitere Artikel der Ausgabe 9/2012

Intensive Care Medicine 9/2012 Zur Ausgabe

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.