Lack of effect of glutamine administration to boost the innate immune system response in trauma patients in the intensive care unit

Jon Pérez-Bárcena, Catalina Crespí, Verónica Regueiro, Pedro Marsé, Joan M Raurich, Jordi Ibáñez, Abelardo García de Lorenzo-Mateos, José A Bengoechea, Jon Pérez-Bárcena, Catalina Crespí, Verónica Regueiro, Pedro Marsé, Joan M Raurich, Jordi Ibáñez, Abelardo García de Lorenzo-Mateos, José A Bengoechea

Abstract

Introduction: The use of glutamine as a dietary supplement is associated with a reduced risk of infection. We hypothesized that the underlying mechanism could be an increase in the expression and/or functionality of Toll-like receptors (TLR), key receptors sensing infections. The objective of this study was to evaluate whether glutamine supplementation alters the expression and functionality of TLR2 and TLR4 in circulating monocytes of trauma patients admitted to the intensive care unit (ICU).

Methods: We designed a prospective, randomized and single-blind study. Twenty-three patients received parenteral nutrition (TPN) with a daily glutamine supplement of 0.35 g/kg. The control group (20 patients) received an isocaloric-isonitrogenated TPN. Blood samples were extracted before treatment, at 6 and 14 days. Expression of TLR2 and TLR4 was determined by flow cytometry. Monocytes were stimulated with TLR specific agonists and cytokines were measured in cell culture supernatants. Phagocytic ability of monocytes was also determined.

Results: Basal characteristics were similar in both groups. Monocytes from patients treated with glutamine expressed the same TLR2 levels as controls before treatment (4.9 ± 3.5 rmfi vs. 4.3 ± 1.9 rmfi, respectively; P = 0.9), at Day 6 (3.8 ± 2.3 rmfi vs. 4.0 ± 1.7 rmfi, respectively; P = 0.7) and at Day 14 (4.1 ± 2.1 rfim vs. 4.6 ± 1.9 rmfi, respectively; P = 0.08). TLR4 levels were not significantly different between the groups before treatment: (1.1 ± 1 rmfi vs 0.9 ± 0.1 rmfi respectively; P = 0.9), at Day 6 (1.1 ± 1 rmfi vs. 0.7 ± 0.4 rmfi respectively; P = 0.1) and at Day 14 (1.4 ± 1.9 rmfi vs. 1.0 ± 0.6 rmfi respectively; P = 0.8). No differences in cell responses to TLR agonists were found between groups. TLR functionality studied by phagocytosis did not vary between groups.

Conclusions: In trauma patients in the intensive care unit, TPN supplemented with glutamine does not improve the expression or the functionality of TLRs in peripheral blood monocytes.

Trial registration: ClinicalTrials.gov Identifier: NCT01250080.

Figures

Figure 1
Figure 1
Expression of TLR2 in trauma patients treated with and without glutamine. The expression of TLR2 was analyzed in CD14 positive peripheral blood mononuclear cells. rmfi are shown for 23 trauma patients treated with glutamine (black bars) and 20 trauma patients without glutamine and used as controls (white bars). Samples were obtained at the beginning of the treatment (Day 0); at the end of the treatment (Day 6) and at Day 14. Data are given as mean ± SEM.
Figure 2
Figure 2
Expression of TLR4 in trauma patients treated with and without glutamine. The expression of TLR4 was analyzed in CD14 positive peripheral blood mononuclear cells. rmfi are shown for 23 trauma patients treated with glutamine (black bars) and 20 trauma patients without glutamine and used as controls (white bars). Samples were obtained at the beginning of the treatment (Day 0); at the end of the treatment (Day 6) and at Day 14. Data are given as mean ± SEM.
Figure 3
Figure 3
Concentration of TNFα in cell culture supernatants in trauma patients treated with and without glutamine. TLR functionality. Levels of TNFα analyzed by a bead array ELISA (CBA Kit, BD Biosciences), in response to lipopolysaccharide (LPS-100 ng/mL), Pam3CSK4 (PAM-10 pg/mL) and zymosan (ZYM-10 pg/mL) at the beginning of the treatment (Figure 3A); at Day 6 (Figure 3B) and at Day 14 (Figure 3C). Monocytes from trauma patients treated with glutamine subjects (black bars, n = 23) and trauma patients without glutamine (white bars, n = 20). Control bars are samples production of cytokines by unstimulated monocytes. Data are given as mean ± SEM.
Figure 4
Figure 4
Concentration of IL1β in cell culture supernatants in trauma patients treated with and without glutamine. TLR functionality. Levels of IL1β analyzed by a bead array ELISA (CBA Kit, BD Biosciences), in response to lipopolysaccharide (LPS-100 ng/ml), Pam3CSK4 (PAM-10 pg/mL) and zymosan (ZYM-10 pg/mL) at the beginning of the treatment (Figure 4A); at Day 6 (Figure 4B) and at Day 14 (Figure 4C). Monocytes from trauma patients treated with glutamine subjects (black bars, n = 23) and trauma patients without glutamine (white bars, n = 20). Control bars are samples production of cytokines by unstimulated monocytes. Data are given as mean ± SEM.
Figure 5
Figure 5
Concentration of IL6 in cell culture supernatants in trauma patients treated with and without glutamine. TLR functionality. Levels of Cytokines IL 6 analyzed by a bead array ELISA (CBA Kit, BD Biosciences), in response to lipopolysaccharide (LPS-100 ng/ml), Pam3CSK4 (PAM-10 pg/mL) and zymosan (ZYM-10 pg/mL)) at the beginning of the treatment (Figure 5A); at Day 6 (Figure 5B) and at Day 14 (Figure 5C). Monocytes from trauma patients treated with glutamine subjects (black bars, n = 23) and trauma patients without glutamine (white bars, n = 20). Control bars are samples production of cytokines by unstimulated monocytes. Data are given as mean ± SEM.
Figure 6
Figure 6
Concentration of IL10 in cell culture supernatants in trauma patients treated with and without glutamine. TLR functionality. Levels of Cytokines IL 10 analyzed by a bead array ELISA (CBA Kit, BD Biosciences), in response to lipopolysaccharide (LPS-100 ng/ml), Pam3CSK4 (PAM-10 pg/mL) and zymosan (ZYM-10 pg/mL)) at the beginning of the treatment (Figure 6A); at Day 6 (Figure 6B) and at Day 14 (Figure 6C). Monocytes from trauma patients treated with glutamine subjects (black bars, n = 23) and trauma patients without glutamine (white bars, n = 20). Control bars are samples production of cytokines by unstimulated monocytes. Data are given as mean ± SEM.

References

    1. Roth E. Nonnutritive effects of glutamine. J Nutr. 2008;138:2025S–2031S.
    1. Eliasen MM, Brabec M, Gerner C, Pollheimer J, Auer H, Zellner M, Weingartmann G, Garo F, Roth E, Oehler R. Reduced stress tolerance of glutamine-deprived human monocytic cells is associated with selective down-regulation of Hsp70 by decreased mRNA stability. J Mol Med. 2006;84:147–158. doi: 10.1007/s00109-005-0004-6.
    1. Singleton KD, Wischmeyer PE. Glutamine's protection against sepsis and lung injury is dependent on heat shock protein 70 expression. Am J Physiol Regul Integr Comp Physiol. 2007;292:R1839–1845.
    1. Roth E, Oehler R, Manhart N, Exner R, Wessner B, Strasser E, Spittler A. Regulative potential of glutamine-relation to gluthatione metabolism. Nutrition. 2002;18:217–221. doi: 10.1016/S0899-9007(01)00797-3.
    1. Hong RW, Rounds JD, Helton WS, Robinson MK, Wilmore DK. Glutamine preserves liver glutathione after lethal hepatic injury. Ann Surg. 1992;215:114–119. doi: 10.1097/00000658-199202000-00004.
    1. Haussinger D, Roth E, Lang F, Gerok W. Cellular hydratation state: an important determinant of protein catabolism in health and disease. Lancet. 1993;341:1330–1332. doi: 10.1016/0140-6736(93)90828-5.
    1. Oehler R, Zellner M, Hefel B, Weingartmann G, Spittler A, Struse HM, Roth E. Influence of heat shock protein on cell volume regulation: protection from hypertonic challenge in a human monocyte cell line. FASEB J. 1998;12:553–560.
    1. Eliasen MM, Winkler W, Jordan V, Pokar M, Marchetti M, Roth E, Allmaier G, Oehler R. Adaptative cellular mechanisms in response to glutamine starvation. Front Biosci. 2006;11:3199–3211. doi: 10.2741/2043.
    1. Janeway CA Jr, Medzhitoz R. Innate immune recognition. Annu Rev Immunol. 2002;20:197–216. doi: 10.1146/annurev.immunol.20.083001.084359.
    1. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol. 2001;2:675–680. doi: 10.1038/90609.
    1. Van Amersfoort ES, Van Berkel TJC, Kuiper J. Receptors, mediators, and mechanisms involved in bacterial sepsis and septic shock. Clin Microbiol Rev. 2003;16:379–414. doi: 10.1128/CMR.16.3.379-414.2003.
    1. Pérez-Bárcena J, Regueiro V, Crespí C, Pierola J, Oliver A, Llompart-Pou JA, Ayestarán JI, Raurich JM, Marsé P, Ibáñez J, Bengoechea JA. Expression of Toll-Like Receptors 2 and 3 is upregulated during hospital admission in traumatic patients. Lack of correlation with blunted innate immune responses. Ann Surg. 2010;251:521–527.
    1. Adib-Conquy M, Moine P, Asehnoune K, Edouard A, Espevik T, Miyake K, Werts C, Cavaillon JM. Toll-like receptor mediated tumor necrsosis factor and interleukin-10 production differ during systemic inflammation. Am J Respir Crit Care Med. 2003;168:158–164. doi: 10.1164/rccm.200209-1077OC.
    1. Lendemans S, Kreuzfelder E, Rani M, Bayeeh E, Schade FU, Flohé SB, Waydhas C, Flohé S. Toll-like receptor 2 and 4 expression after severe injury is not involved in the dysregulation of the innate immune system. J Trauma. 2007;63:740–746. doi: 10.1097/01.ta.0000240451.42238.d1.
    1. Laudanski K, De A, Brouxhon S, Kyrkanides S, Miller-Graziano C. Abnormal PGE (2) regulation of monocyte TNF-alpha levels in trauma patients parallels development of a more macrophage-like phenotype. Shock. 2004;22:204–212. doi: 10.1097/.
    1. Déchelotte P, Hasselmann M, Cynober L, Allaouchiche B, Coëffier M, Hecketsweiler B, Merle V, Mazerolles M, Samba D, Guillou YM, Petit J, Mansoor O, Colas G, Cohendy R, Barnoud D, Czernichow P, Bleichner G. L-alanyl-L-Glutamine dipeptide supplemented total parenteral nutrition reduces infectious complications and glucose intolerance in critically ill patients: The French controlled, randomized, double-blind, multicenter study. Crit Care Med. 2006;34:598–604.
    1. Griffiths RD, Allen KD, Andrews FJ, Jones C. Infection, multiple organ failure, and survival in the intensive care unit: influence of glutamine-supplemented parenteral nutrition on acquired infection. Nutrition. 2002;18:546–552. doi: 10.1016/S0899-9007(02)00817-1.
    1. Pérez-Bárcena J, Regueiro V, Marsé P, Raurich JM, Rodríguez A, Ibáñez J, de Lorenzo Mateos AG, Bengoechea JA. Glutamine as a modulator of the immune system of critical care patients: effect on Toll-Like receptor expression. A preliminary study. Nutrition. 2008;24:522–527.
    1. ASPEN Board of Directors and the Clinical Guidelines TASK Force. Guidelines for the use of enteral and parenteral nutrition in adult and pediatric patients. JPEN J Parenter Enteral Nutr. 2002;26:1sa–138sa. doi: 10.1177/0148607102026001011.
    1. Goeters C, Wenn A, Mertes N, Wempe C, Van Aken H, Stehle P, Bone HG. Parenteral L-alanyl-L-glutamine improves 6-month outcome in critically ill patients. Crit Care Med. 2002;30:2032–2037. doi: 10.1097/00003246-200209000-00013.
    1. Mertes N, Schulzki C, Goeters C, Winde G, Benzing S, Kuhn KS, Van Aken H, Stehle P, Fürst P. Cost containment though L-alanyl-L-Glutamine supplemented total parenteral nutrition after major abdominal surgery: a prospective randomized double-blind controlled study. Clin Nutr. 2000;19:395–401. doi: 10.1054/clnu.2000.0142.
    1. Alvarez-Lerma F, Palomar M, Olaechea P, Otal JJ, Insausti J, Cerdá E. Grupo de Estudio de Vigilacia de Infección Nosocomial en UCI. National Study of Control of Nosocomial Infection in Intensive Care Units. Evolutive report of the years 2003-2005. Med Intensiva. 2007;31:6–17. doi: 10.1016/S0210-5691(07)74764-2.
    1. Guidelines for the management of severe sepsis and septic shock. The International Sepsis Forum. Intensive Care Med. 2001;27:S1–S134. doi: 10.1007/s001340000767.
    1. Garner JS, Jarvis WR, Emori TG, Horan TC, Hughes JM. CDC definitions for nosocomial infections. Am J Infect Control. 1988;16:128–140. doi: 10.1016/0196-6553(88)90053-3.
    1. Murray PR, Baron EJ, Jorgensen JH, Pfaller MA, Yolken RH. Manual of Clinical Microbiology. 8. ASM Press, Washington DC; 2003.
    1. Deering RP, Orange JS. Development of a clinical assay to evaluate toll-like receptor function. Clin Vaccine Immunol. 2006;13:68–76. doi: 10.1128/CVI.13.1.68-76.2006.
    1. Hirschfeld M, Ma Y, Weis JH, Vogel SN, Weis JJ. Cutting edge: repurification of lipopolysaccharide eliminates signaling through both human and murine toll-like receptor 2. J Immunol. 2000;165:618–622.
    1. Blander JM, Medzhitov R. Regulation of phagosome maturation by signals from Toll-like receptors. Science. 2004;304:1014–1018. doi: 10.1126/science.1096158.
    1. Novak F, Hekland DK, Avenell A, Drover JW, Su X. Glutamine supplementation in serious illness: a systematic review of the evidence. Crit Care Med. 2002;30:2022–2029. doi: 10.1097/00003246-200209000-00011.
    1. Singer P, Berger MM, Vanden Berghe G, Biolo G, Calder P, Forbes A, Griffiths R, Kreyman G, Leverve X, Pichard C. ESPEN guidelines on parenteral nutrition: Intensive Care. Clin Nutr. 2009;28:387–400. doi: 10.1016/j.clnu.2009.04.024.
    1. Kessel A, Toubi E, Pavlotzky E, Mogilner J, Coran AG, Lurie M, Karry R, Sukhotnik I. Treatment with glutamine is associated with down-regulation of Toll-like receptor-4 and myeloid differentiation factor 88 expression and decrease in intestinal mucosal injury caused by lipopolysaccharide endotoxaemia in a rat. Clin Exp Immunol. 2008;151:341–347. doi: 10.1111/j.1365-2249.2007.03571.x.
    1. Medzhitov R, Janeway C. Innate immune recognition: mechanisms and pathways. Immunol Rev. 2000;173:89–97. doi: 10.1034/j.1600-065X.2000.917309.x.
    1. Qureshi ST, Medzhitov R. Toll-like receptors and their role in experimental models of microbial infection. Genes Immun. 2003;4:87–94. doi: 10.1038/sj.gene.6363937.
    1. Akira S. Toll-like receptor signalling. J Biol Chem. 2003;278:38105–108. doi: 10.1074/jbc.R300028200.
    1. Brandl K, Gluck T, Huber C, Salzberger B, Falk W, Hartmann P. TLR4 surface display is increased in septic patients. Eur J Med Res. 2005;10:319–324.
    1. Adib-Conquy M, Adrie C, Fittings C, Gasttolliat O, Beyaert R, Cavaillon JM. Up-regulation of MyD88s and SIGIRR, molecules inhibiting Toll-like receptor signaling, in monocytes from septic patients. Crit Care Med. 2006;34:2377–2385. doi: 10.1097/01.CCM.0000233875.93866.88.
    1. Bihl F, Salez L, Beaubier M, Torres D, Larivière L, Laroche L, Benedetto A, Martel D, Lapointe JM, Ryffel B, Malo D. Overexpression of Toll-like receptor 4 amplifies the host response to lipopolysaccharide and provides a survival advantage in transgenic mice. J Immunol. 2003;170:6141–6150.
    1. Kalis C, Kanzler B, Lembo A, Poltorak A, Galanos C, Freudenberg MA. Toll-like receptor 4 expression levels determine the degree of LPS-susceptibility in mice. Eur J Immunol. 2003;33:798–805. doi: 10.1002/eji.200323431.
    1. Fabian TC, Croce MA, Fabian MJ, Trenthem LL, Yockey JM, Boscarino R, Proctor KG. Reduced tumor necrosis factor production in endotoxin-spiked whole blood after trauma: experimental results and clinical correlation. Surgery. 1995;118:63–72. doi: 10.1016/S0039-6060(05)80011-X.
    1. Majetschak M, Flach R, Heukamp T, Jennissen V, Obertacke U, Neudeck F, Schmit-Neuerburg KP, Schade FU. Regulation of whole blood tumor necrosis factor production upon endotoxin stimulation after severe blunt trauma. J Trauma. 1997;43:880–887. doi: 10.1097/00005373-199712000-00002.
    1. Ogle CK, Ogle JD, Mao JX, Simon J, Noel JG, Li BG, Alexander JW. Effect of glutamine on phagocytosis and bacterial killing by normal and pediatric burn patient neutrophils. JPEN J Parenter Enteral Nutr. 1994;18:128–133. doi: 10.1177/0148607194018002128.

Source: PubMed

3
Sottoscrivi