Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

CRITICAL CARE AND TRAUMA

SECTION EDITOR
PETER M. SUTER

Ketamine Suppresses Proinflammatory Cytokine


Production in Human Whole Blood In Vitro
Takashi Kawasaki, MD, Masanori Ogata, MD, Chika Kawasaki, MD, Jun-ichi Ogata, MD,
Yoshitaka Inoue, MD, and Akio Shigematsu, MD
Department of Anesthesiology, University of Occupational and Environmental Health, Kitakyushu, Japan
Downloaded from https://journals.lww.com/anesthesia-analgesia by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC1y0abggQZXdtwnfKZBYtws= on 12/15/2021

The production of proinflammatory cytokines, such as assay or an enzyme-linked immunoassay. Ketamine


tumor necrosis factor (TNF) a, interleukin (IL)-6, and significantly suppressed LPS-induced TNF-a produc-
IL-8, increases in patients with sepsis; marked produc- tion at concentrations .20 mg/mL. At concentrations
tion causes organ failure and septic shock. We previ- .100 mg/mL, ketamine also significantly suppressed
ously reported that ketamine suppressed lipopolysac- both LPS-induced and rhTNF-induced IL-6 and IL-8
charide (LPS)-induced TNF-a production in mice. production. In this study, we demonstrated that ket-
However, there are no reports on the effect of ketamine amine directly inhibits the production of proinflamma-
on cytokine production in human whole blood. There- tory cytokines such as TNF-a, IL-6, and IL-8 in human
fore, in this study, we investigated the efficacy of ket- whole blood. Implications: We found that ketamine
amine on LPS-induced TNF-a, IL-6, and IL-8 produc- suppressed lipopolysaccharide-induced tumor necro-
tion and recombinant human (rh) TNF-a–induced IL-6 sis factor a, interleukin (IL)-6, and IL-8 production and
and IL-8 production in human whole blood. After add- recombinant human tumor necrosis factor-induced
ing different doses of ketamine to whole blood, the IL-6 and IL-8 production in human whole blood. Ket-
blood was stimulated with LPS or rhTNF. After incuba- amine directly suppresses proinflammatory cytokine
tion, the plasma TNF-a activity and IL-6 and IL-8 con- production.
centrations were measured using the L929 cell cytotoxic (Anesth Analg 1999;89:665–9)

C
ytokines are essential for hematopoiesis and im- and mortality in carrageenan-sensitized endotoxin shock
mune responses, and they play a key role in the mice (9,10). However, there are no reports on the effects
defense against infection. Lipopolysaccharide of ketamine on the production of other proinflammatory
(LPS) is a potent inducer of the inflammation involved in cytokines. Not only does LPS trigger the production of
the pathogenesis of septic shock. It has been demon- TNF-a, IL-6, and IL-8, it also stimulates IL-6 and IL-8
strated that proinflammatory cytokines, such as tumor production (11).
necrosis factor (TNF) a, interleukin (IL)-6, and IL-8, in- Therefore, in this study, we investigated the effects
crease in patients with sepsis, trauma, and burns (1–6). of ketamine in human whole blood on LPS-induced
IL-6 mediates the acute-phase response, and IL-8 is a production of TNF-a, IL-6, and IL-8 and on TNF-a–
potent chemotactic agent for neutrophils. These cyto- induced IL-6 and IL-8 production.
kines are associated with the development of septic
shock and organ dysfunction. Ketamine, an IV anes-
thetic, has been advocated for anesthesia in septic or
severely ill patients because of its cardiovascular- Methods
stimulating effects (7,8). We previously reported that Phenol-extracted Escherichia coli (serotype 0127: B8)
ketamine suppressed LPS-induced TNF-a production LPS was purchased from Difco Laboratories (Detroit,
MI). Ketamine (10 mg/mL) was purchased from
This work was supported in part by Grant-in-Aid for Scientific Sankyo Pharmaceutical Co. (Tokyo, Japan). Recombi-
Research 10671453 from the Ministry of Education, Science, Sport nant human (rh) TNF-a was purchased from Gen-
and Culture of Japan.
Presented in part at the annual meeting of the American Society zyme Co. (Cambridge, MA).
of Anesthesiologists, October 17–21, 1998, Orlando, FL. After approval of our human investigations commit-
Accepted for publication May 10, 1999. tee, informed consent was obtained from 15 healthy
Address correspondence and reprint requests to Masanori Ogata, male volunteers not taking any medication. Blood sam-
MD, Department of Anesthesiology, University of Occupational and
Environmental Health, 1–1-1 Iseigaoka Yahata-nishi-ku Kitakyushu, ples were drawn into tubes containing heparin and di-
807-8555, Japan. Address e-mail to mogata@med.uoeh-u.ac.jp. luted with 5 vol of RPMI 1640 (Nissui Pharmaceutical,

©1999 by the International Anesthesia Research Society


0003-2999/99 Anesth Analg 1999;89:665–9 665
666 CRITICAL CARE AND TRAUMA KAWASAKI ET AL. ANESTH ANALG
KETAMINE SUPPRESSES TNF-a, IL-6, AND IL-8 PRODUCTION IN HUMAN BLOOD 1999;89:665–9

Tokyo, Japan) (12). One milliliter of diluted blood per for 5 h at 37°C in a 95% air/5% CO2 incubator. After
well was placed into 24-well tissue culture plates. incubation, blood was centrifuged at 700g for 10 min.
After different doses (0 –500 mg/mL) of ketamine Buffy coats were isolated and NH4Cl lysis of red blood
were added to each well, whole blood was stimulated cells was performed. The white blood cells were re-
with LPS (10 ng/mL). The blood was then incubated suspended in RPMI 1640 medium containing 5% fetal
for 6 or 12 h at 37°C in a 95% air/5% CO2 incubator. calf serum, and the cells were stained with 0.2%
After incubation, the blood was centrifuged at 700g for trypan blue. The cell survival rate was assessed by
10 min to remove blood cells. Supernatant samples microscope.
were collected and stored at 280°C until assayed. All data are presented as the mean 6 sem. The
To assess the direct effect of ketamine on IL-6 and repeated paired t-test was used for statistical analysis
IL-8 production, we studied the effect of ketamine on to compare values with the control value. A significant
rhTNF-induced IL-6 and IL-8 production. After differ- difference was presumed at a probability value of
ent doses (0 –500 mg/mL) of ketamine were added to ,0.05.
each well, whole blood was stimulated with rhTNF-a
(104 U/mL). The dose of rhTNF-a chosen induced the
same level of TNF-a activity as when 10 ng/mL LPS
was used on L929 cells in the preliminary study. The Results
blood was then incubated for 6 h at 37°C in a 95% Whole blood was stimulated using different doses of
air/5% CO2 incubator. After incubation, the blood was LPS (0 –100 ng/mL). LPS induced TNF-a production
centrifuged at 700g for 10 min to remove blood cells. in human whole blood in a dose-dependent manner at
The supernatant was collected and stored at 280°C concentrations of 0.1–10 ng/mL. TNF-a production
until assayed. reached a plateau with LPS doses $10 ng/mL (Fig-
The L929 cell cytotoxic assay (described previously) ure 1). Therefore, we used an LPS concentration of 10
was used to determine the plasma TNF-a activity (13). ng/mL in our experiments.
Briefly, L929 cells in RPMI 1640 medium containing After adding different doses (0–500 mg/mL) of ket-
5% fetal calf serum were seeded at 3 3 105 cells/well amine, whole blood was stimulated with LPS (10 ng/
in 96-well flat-bottomed microtiter plates (Becton mL) (n 5 15). Figure 2A shows the effect of ketamine on
Dickinson, Lincoln Park, NJ) and incubated overnight LPS-induced TNF-a production. When the blood was
at 37°C in an atmosphere of 5% CO2 in air. Serial incubated for 6 h, ketamine significantly suppressed
1:2 dilutions of samples were made in this me- LPS-induced TNF-a production in a dose-dependent
dium containing 1 mg/mL actinomycin D (Banyu manner at concentrations of 20–500 mg/mL, compared
Pharmaceutical Co., Tokyo, Japan), and 0.1 mL of each with the control (P , 0.05). Concentrations ,4 mg/mL
dilution was added to different wells. On the follow- ketamine had no effect on LPS-induced TNF-a produc-
ing day, cell survival was assessed by fixing and stain- tion. When the blood was incubated for 12 h, LPS-
ing the cells with crystal violet (0.2% in 20% metha- induced TNF-a production increased from 1152 6 176
nol), and 1% sodium dodecyl sulfate was added to U/mL (6-h incubation) to 2176 6 174 U/mL. At concen-
each well to solubilize the stained cells. The absor- trations of 20–500 mg/mL, ketamine significantly sup-
bance of each well was determined at 490 nm using a pressed LPS-induced TNF-a production (P , 0.05).
microplate reader (Bio-Rad Laboratories, Richmond, Figure 2B shows the effect of ketamine on LPS-
CA). TNF activity was expressed in units per milliliter, induced IL-6 production. Ketamine (100 –500 mg/mL)
which is the reciprocal of the dilution necessary for suppressed LPS-induced IL-6 production in the 6-h
50% lysis of the cells. incubation (P , 0.0001). Ketamine (100 –500 mg/mL)
The plasma IL-6 concentration was measured in also suppressed LPS-induced IL-8 production com-
duplicate using a commercially available enzyme- pared with the control (P , 0.05) (Figure 2C). Concen-
linked immunoassay (IL-6 Enzyme Immunoassay Kit; trations ,20 mg/mL ketamine had no effect on IL-6 or
Advanced Magnetics, Inc., Cambridge, MA). The IL-8 production in the 6-h whole blood incubation
intra- and interassay precision was 9% and 6%, respec- (Figure 2, B and C). When the blood was incubated for
tively, at an IL-6 concentration of 88 pg/mL. The 12 h, LPS-induced IL-6 production increased from
plasma IL-8 concentration was measured in duplicate 555 6 33 pg/mL (6-h incubation) to 1195 6 59 pg/mL,
using the same enzyme-linked immunoassay. The and IL-8 production increased from 907 6 56 pg/mL
intra- and interassay precision was 7% and 4%, respec- (6-h incubation) to 1502 6 165 pg/mL. Ketamine (100 –
tively, at an IL-8 concentration of 76 pg/mL. Accord- 500 mg/mL) also significantly suppressed LPS-
ing to the manufacturer, cross-reactivity with other induced IL-6 and IL-8 production when the blood was
cytokines is negligible in both assays. incubated for 12 h (P , 0.0001) (Figure 2, B and C).
To assess the effect of ketamine on leukocyte viabil- After adding different doses (0 –500 mg/mL) of ket-
ity, different doses (0 –500 mg/mL) of ketamine were amine, whole blood was stimulated with rhTNF
added to diluted human whole blood and incubated (104 U/mL) (n 5 8). Figure 3 shows the cytokine
ANESTH ANALG CRITICAL CARE AND TRAUMA KAWASAKI ET AL. 667
1999;89:665–9 KETAMINE SUPPRESSES TNF-a, IL-6, AND IL-8 PRODUCTION IN HUMAN BLOOD

Figure 1. Lipopolysaccharide (LPS)-induced tumor necrosis factor


(TNF) a production in human whole blood. Values are expressed as
mean 6 sem (n 5 5).

production when the whole blood was incubated with


rhTNF for 6 h. Ketamine 250 and 500 mg/mL signifi-
cantly suppressed rhTNF-induced IL-6 production
(P , 0.05) (Figure 3A), whereas ,100 mg/mL ket-
amine had no effect on rhTNF-induced IL-6 produc-
tion (Figure 3A). Ketamine (100 –500 mg/mL) also sig-
nificantly suppressed rhTNF-induced IL-8 production
(P , 0.0001) (Figure 3B), whereas ,20 mg/mL ket-
amine had no effect on rhTNF-induced IL-8 produc-
tion (Figure 3B).
Ketamine had no effect on L929 cell viability. Ket-
amine also had no effect on white blood cell viability, as
assessed by the exclusion of the vital stain trypan blue.

Discussion
In this study, we demonstrated that ketamine sup-
pressed both LPS-induced TNF-a, IL-6, and IL-8 produc-
tion and rhTNF-induced IL-6 and IL-8 production in
human whole blood. TNF-a is the first cytokine ex-
pressed after LPS stimulation, after which it stimulates
IL-6 and IL-8 secretion from macrophages, monocytes,
neutrophils, and endothelial cells. We suspected that the
suppressive effect of ketamine on LPS-induced IL-6 and Figure 2. Effect of ketamine on lipopolysaccharide (LPS)-induced tu-
IL-8 production might be caused by the inhibiting effect mor necrosis factor (TNF) a (A), interleukin-6 (B), and interleukin-8 (C)
production. After ketamine (0–500 mg/mL) was added, human whole
of ketamine on LPS-induced TNF-a production. How- blood was stimulated by LPS (10 ng/mL) and incubated for 6 or 12 h.
ever, our results demonstrated that ketamine suppressed Values are expressed as mean 6 sem (n 5 15). *P , 0.05, #P , 0.0001
rhTNF-induced IL-6 and IL-8 production, which sug- compared with control.
gests that the suppression of IL-6 and IL-8 production is
dependent not only on the suppression of TNF-a, but after the LPS stimulation effectively suppressed TNF
also on the direct effect of ketamine on the production of production (9). We assume that ketamine may regu-
these cytokines in whole blood. late LPS-induced TNF-a production at a posttran-
The mechanism for the suppressive effect of ket- scriptional level. Further study is required to elucidate
amine on cytokine production is not clear. A detect- the mechanism for the suppressive effect of ketamine
able increase in the TNF-a mRNA level is seen min- on TNF-a, IL-6, and IL-8.
utes after the LPS injection and reaches a peak two In this study, human whole blood was used as an ex
hours after the LPS injection (11). In a previous study, vivo model of cytokine production in human tissue. Us-
we reported that the addition of ketamine two hours ing the whole blood model reduces the confounding
668 CRITICAL CARE AND TRAUMA KAWASAKI ET AL. ANESTH ANALG
KETAMINE SUPPRESSES TNF-a, IL-6, AND IL-8 PRODUCTION IN HUMAN BLOOD 1999;89:665–9

protein complex to CD14 on the monocyte surface,


whereas the response to LPS concentrations .10 ng/mL
can occur in the absence of either LPS binding protein or
CD14 (15). We assume that different cytokine responses
may be caused by the differences in the LPS concen-
tration.
It has been demonstrated that ketamine, an IV an-
esthetic, has protective effects in septic patients and
in an animal septic shock model (7–10,17,18). The
cardiovascular-stimulating effects of ketamine have
been advocated for anesthesia in septic patients (7,8).
The concentration of ketamine in human plasma
reached 110 mM by the IV administration of ketamine
2–2.2 mg/kg (19). Shimaoka et al. (20) showed that
ketamine (30 – 600 mM) inhibits nitric oxide production
in mouse-activated macrophage-like cells via inhibi-
tion of TNF-a production, and Li et al. (21) showed
that .10 mM ketamine also inhibits nitric oxide pro-
duction in LPS-treated rat alveolar macrophages.
Schmidt et al. (22) showed that ketamine (10 mg/kg
body weight) also inhibits endotoxin-induced leuko-
cyte adherence due to the decreased production of
TNF-a in rat mesenteric venules. We previously dem-
onstrated that ketamine has a potent suppressive ef-
fect on LPS-induced TNF-a production in vitro and in
vivo (9,10).
In addition, in the present study, we showed that
.20 mg/mL ketamine (73 mM) suppressed LPS-
induced TNF-a production and that .100 mg/mL ket-
amine (365 mM) had a potent suppressive effect on
Figure 3. Effect of ketamine on recombinant human tumor necrosis
factor (rhTNF) a-induced interleukin-6 (A) and interleukin-8 (B)
IL-6 and IL-8 production in human whole blood.
production. After ketamine (0 –500 mg/mL) was added, human These studies suggest that the protective effects of
whole blood was stimulated by rhTNF-a and incubated for 6 h. ketamine in septic patients and in animal septic shock
Values are expressed as mean 6 sem (n 5 8). *P , 0.05, #P , 0.0001 models are due to suppression of the excessive pro-
compared with control.
duction of proinflammatory cytokines.
Roytblat et al. (23) reported that a single dose of
factors that may be associated with the isolation of ketamine 0.25 mg/kg administered before cardiopul-
monocytes, such as the adherence-induced expression of monary bypass suppressed the increase in serum IL-
cell-surface TNF or TNF mRNA (14). Moreover, whole 6 during and after coronary artery bypass surgery,
blood is a more physiologic environment in which to and that a subanesthetic dose of ketamine suppressed
examine cytokine production in response to LPS because IL-6 production in women undergoing hysterectomy
the cellular interactions are preserved and the presence (24). However, in our study, such a small dose of
of LPS-binding protein is maintained (15). ketamine did not suppress LPS- and rhTNF-induced
In our study, after a 12-hour incubation, LPS-induced IL-6 production. The reason for this is not clear but
TNF-a, IL-6, and IL-8 production in human whole blood may lie in the differences between in vivo and in vitro
increased significantly compared with a 6-hour incuba- experimental conditions.
tion. In contrast, Deforge et al. (11) demonstrated that In conclusion, we demonstrated that ketamine di-
IL-6 reached a plateau six hours after the LPS challenge. rectly inhibits the production of proinflammatory cy-
They also showed that IL-8 increased after LPS stimula- tokines such as TNF-a, IL-6, and IL-8 in human whole
tion, reached a plateau between 6 and 12 hours, then blood. Further study is required to elucidate the mech-
continued to increase for .24 hours (16). The time course anism of the suppressive effect of ketamine.
for LPS-induced cytokine production that we observed
differed from theirs. In this study, we stimulated human
whole blood at an LPS concentration of 10 ng/mL,
whereas Deforge et al. used an LPS concentration of References
10 mg/mL. The response to a low concentration of LPS 1. Marshall WG, Dimick AR. Natural history of major burns with
has been attributed to the binding of an LPS-LPS binding multiple subsystem organ failure. J Trauma 1983;23:102–5.
ANESTH ANALG CRITICAL CARE AND TRAUMA KAWASAKI ET AL. 669
1999;89:665–9 KETAMINE SUPPRESSES TNF-a, IL-6, AND IL-8 PRODUCTION IN HUMAN BLOOD

2. Goris RJA, Boekholtz WKF, van Bebber IPT, et al. Multiple 14. Desch CE, Kovach NL, Present W, et al. Production of human
organ failure and sepsis without bacteria. Arch Surg 1986;121: tumor necrosis factor from whole blood ex vivo. Lymphokine
897–901. Res 1989;8:141– 6.
3. Marano MA, Fong Y, Moldawer LL, et al. Serum cachectin/ 15. Wright SD, Ramos RA, Tobias PS, et al. CD14, a receptor for
tumor necrosis factor in critically ill patients with burns corre- complexes of LPS and LPS binding protein. Science 1990;249:
lates with infection and mortality. Surg Gynecol Obstet 1990; 1431–3.
170:32– 8. 16. DeForge LE, Kenney JS, Jones ML, et al. Biphasic production of
4. Meade P, Shoemaker WC, Donnelly TJ, et al. Temporal patterns IL-8 in LPS-stimulated human whole blood. J Immunol 1992;
of hemodynamics, oxygen transport, cytokine activity and com- 148:2133– 41.
plement activity in the development of adult respiratory dis-
17. Bodai BI, Harms BA, Nottingham PB, et al. The effect of ket-
tress syndrome after severe injury. J Trauma 1994;36:651–7.
amine on endotoxin-induced lung injury. Anesth Analg 1983;
5. Waage A, Brandtzaeg P, Halstensen A, et al. The complex
pattern of cytokines in serum from patients with meningococcal 62:398 – 403.
septic shock. J Exp Med 1989;169:333– 8. 18. Van der Linder P, Gilbart E, Engelman E, et al. Comparison of
6. Tracey KJ, Beutler B, Lowry SF, et al. Shock and tissue injury by halothane, isoflurane, alfentanil, and ketamine in experimental
recombinant human cachectin. Science 1986;234:470 – 4. septic shock. Anesth Analg 1990;70:608 –17.
7. Lippmann M, Appel PL, Mok MS, Shoemaker WC. Sequential 19. Domino EF, Zsigmond EK, Domino LE, et al. Plasma levels of
cardiorespiratory patterns of anesthetic induction with ket- ketamine and two of its metabolites in surgical patients using a
amine in critically ill patients. Crit Care Med 1983;11:730 – 4. gas chromatographic mass fragmentographic assay. Anesth
8. Yli-Hankala, Kirvela M, Randell T, et al. Ketamine anaesthsia in Analg 1982;61:87–92.
a patient with septic shock. Acta Anaesthsiol Scand 1992;36: 20. Shimaoka M, Iida T, Ohara A, et al. Ketamine inhibits nitric
483–5. oxide production in mouse-activated macrophage-like cells. Br J
9. Takenaka I, Ogata M, Koga K, et al. Ketamine suppresses Anaesth 1996;77:238 – 42.
endotoxin-induced tumor necrosis factor alpha production in 21. Li C-Y, Chou T-C, Wong C-S, et al. Ketamine inhibits nitric
mice. Anesthesiology 1994;80:402– 8. oxide synthase in lipopolysaccharide-treated rat alveolar mac-
10. Koga K, Ogata M, Takenaka I, et al. Ketamine suppresses tumor rophages. Can J Anaesth 1997;44:989 –95.
necrosis factor-a activity and mortality in carrageenan- 22. Schmidt H, Ebeling D, Bauer H, et al. Ketamine attenuates
sensitized endotoxin shock model. Circ Shock 1995;44:160 – 8. endotoxin-induced leukocyte adherence in rat mesenteric
11. DeForge LE, Remick DG. Kinetics of TNF, IL-6, and IL-8 gene
venules. Crit Care Med 1995;23:2008 –14.
expression in LPS-stimulated human whole blood. Biochem
23. Roytblat L, Talmor D, Rachinsky M, et al. Ketamine attenuates
Biophys Res Commun 1991;174:18 –24.
12. Ogata M, Fletcher MF, Kloczewiak M, et al. Effect of anticoagulants the interleukin-6 response after cardiopulmonary bypass.
on binding and neutralization of lipopolysaccharide by the peptide Anesth Analg 1998;87:266 –71.
immunoglobulin conjugate CAP18106 –138-immunoglobulin G in 24. Roytblat L, Roy-Shapira A, Greemberg L, et al. Preoperative low
whole blood. Infect Immunol 1997;65:2160 –7. dose ketamine reduces serum interleukin-6 response after ab-
13. Ruff MR, Gifford GE. Purification and physiochemical charac- dominal hysterectomy. Pain Clin 1996;9:327–34.
terization of rabbit tumor necrosis factor. J Immunol 1980;125:
1671–7.

You might also like