Sterile post-traumatic immunosuppression

Md Nahidul Islam, Benjamin A Bradley, Rhodri Ceredig, Md Nahidul Islam, Benjamin A Bradley, Rhodri Ceredig

Abstract

After major trauma, the human immune system initiates a series of inflammatory events at the injury site that is later followed by suppression of local inflammation favoring the repair and remodeling of the damaged tissues. This local immune response involves complex interactions between resident cells such as macrophages and dendritic cells, soluble mediators such as cytokines and chemokines, and recruited cells such as neutrophils, monocytes and mesenchymal stromal cells. If of sufficient magnitude, these initial immune responses nevertheless have systemic consequences resulting in a state called post-traumatic immunosuppression (PTI). However, controversy exists regarding the exact immunological changes occurring in systemic compartments triggered by these local immune responses. PTI is one of the leading causes of post-surgical mortality and makes patients vulnerable to hospital-acquired infections, multiple organ failure and many other complications. In addition, hemorrhage, blood transfusion, immunesenescence and immunosuppressant drugs aggravate PTI. PTI has been intensively studied, but published results are frequently cloudy. The purpose of this review is to focus on the contributions made by different responsive modalities to immunosuppression following sterile trauma and to try to integrate these into an overall scheme of PTI.

Figures

Figure 1
Figure 1
Schematic representation of PTI after sterile trauma. This figure schematically illustrates the sequence of events following major trauma. In brief, at the local injury site, release of different DAMPs by the damaged tissues induces viable cells to secrete chemokines such as IL-8, MCP-1 and MIP-1α, and the immediate secretion of inflammatory cytokines such as TNF-α. DAMPs release results in IL-6 and TNF-α production that activate the HPE axis to release ACTH, cortisol and also PGE2. These events then trigger the secretion of anti-inflammatory biomolecules such as IL-1RA, IL-10 and sTNF-R. Later, IL-6, by the virtue of its ability to trigger the release of acute phase proteins (APPs) by the liver, indirectly involved in reducing the inflammatory events at the injured site. In parallel with the immediate release of DAMPs, inflammatory cytokines and the activities of resident immune cells, there is recruitment of neutrophils, monocytes and MSCs from the blood to the injured site. Bone marrow and spleen act as reservoirs for the egression of these cells to the site of injury via the blood. By releasing antimicrobial peptides and helping in hemostasis, damaged skin also activates inflammasomes to release DAMPs at the local site. On the other hand, ischemia–reperfusion injury associated with the surgical procedure increases HIF and ROS expression that also trigger the production of DAMPs, and thus have an additional role in recruiting immune cells to the injured site. The ultimate goal of the above events following a major trauma is tissue remodeling and promotion of wound healing.
Figure 2
Figure 2
Measuring immune status helps in choosing appropriate immunomodulatory drug. Measuring a patient's immune status based on particular profile of specific pro- and anti-inflammatory proteins will help to indicate if the patient: (i) has a stable immune status and medication is not necessary, (ii) is at the stage of cytokine storm following trauma or infection when both pro- and anti-inflammatory cytokines rise, (iii) requires immediate attention with treatment (either immunostimulatory or immunosuppressive drug will be administered) or (iv) is not in need of medication but requires monitoring over the coming days. Knowing immune status will provide information following trauma or sepsis that can be monitored throughout the patient's recovery, thereby preventing the risk of unnecessary danger from incorrect therapy.

References

    1. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 2010; 10: 826–837.
    1. Hunter J. A Treatise on the Blood, Inflammation, and Gun-Shot Wounds, by the Late John Hunter. To which is Prefixed, a Short Account of the Author's Life, by his Brother-in-law, Everard Home. John Richardson, For George Nicol: London. 1794.
    1. Nightingale F. Florence Nightingale: Measuring Hospital Care Outcomes: Excerpts From the Books Notes on Matters Affecting the Health, Efficiency, and Hospital Administration of the British Army Founded Chiefly on the Experience of the Late War, and Notes on Hospitals. Oakbrook Terrace, Ill: Joint Commission on Accreditation of Healthcare Organizations: Oakbrook Terrace, Il, USA. 1999.
    1. Warkentien T, Rodriguez C, Lloyd B, Wells J, Weintrob A, Dunne JR et al. Invasive mold infections following combat-related injuries. Clin Infect Dis 2012; 55: 1441–1449.
    1. Murray CK. Infectious disease complications of combat-related injuries. Crit Care Med 2008; 36: 358–364.
    1. Oztuna V, Ersoz G, Ayan I, Eskandari MM, Colak M, Polat A. Early internal fracture fixation prevents bacterial translocation. Clin Orthop Relat Res 2006; 446: 253–258.
    1. Oztuna V, Ersoz G, Ayan I, Eskandari MM, Uguz K, Kuyurtar F. Head injury-associated bone fractures induce bacterial translocation: an experimental study. J Orthop Trauma 2004; 18: 92–95.
    1. Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest 1992; 101: 1644–1655.
    1. Bone RC. Immunologic dissonance: a continuing evolution in our understanding of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS). Ann Intern Med 1996; 125: 680–687.
    1. Levy MM, Fink MP, Marshall JC, Abraham E, Angus D, Cook D et al. SCCM/ESICM/ACCP/ATS/SIS International Sepsis Definitions Conference. Crit Care Med 2003; 31: 1250–1256.
    1. Costa A, Benedetto V, Ricci C, Merlin P, Borelli P, Fadda E et al. Endocrine, hematological and immunological changes in surgical patients undergoing general anesthesia. Ital J Surg Sci 1989; 19: 41–49.
    1. Philippou A, Maridaki M, Theos A, Koutsilieris M. Cytokines in muscle damage. Adv Clin Chem 2012; 58: 49–87.
    1. Burton D, Nicholson G, Hall G. Endocrine and metabolic response to surgery. Contin Educ Anaesth Crit Care Pain 2004; 4: 144–147.
    1. Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): role as guardians of inflammation. Mol Ther 2012; 20: 14–20.
    1. Buvanendran A, Kroin JS, Berger RA, Hallab NJ, Saha C, Negrescu C et al. Upregulation of prostaglandin E2 and interleukins in the central nervous system and peripheral tissue during and after surgery in humans. Anesthesiology 2006; 104: 403–410.
    1. Lu J, Goh SJ, Tng PY, Deng YY, Ling EA, Moochhala S. Systemic inflammatory response following acute traumatic brain injury. Front Biosci (Landmark Ed) 2009; 14: 3795–3813.
    1. Cruickshank AM, Fraser WD, Burns HJ, Van Damme J, Shenkin A. Response of serum interleukin-6 in patients undergoing elective surgery of varying severity. Clin Sci (London) 1990; 79: 161–165.
    1. Neidhardt R, Keel M, Steckholzer U, Safret A, Ungethuem U, Trentz O et al. Relationship of interleukin-10 plasma levels to severity of injury and clinical outcome in injured patients. J Trauma 1997; 42: 863–871.
    1. Rock KL, Latz E, Ontiveros F, Kono H. The sterile inflammatory response. Annu Rev Immunol 2010; 28: 321–342.
    1. Sorensen OE, Thapa DR, Roupe KM, Valore EV, Sjobring U, Roberts AA et al. Injury-induced innate immune response in human skin mediated by transactivation of the epidermal growth factor receptor. J Clin Invest 2006; 116: 1878–1885.
    1. Sjogren F, Anderson C. Sterile trauma to normal human dermis invariably induces IL1beta, IL6 and IL8 in an innate response to ‘danger'. Acta Derm Venereol 2009; 89: 459–465.
    1. Lin ZQ, Kondo T, Ishida Y, Takayasu T, Mukaida N. Essential involvement of IL-6 in the skin wound-healing process as evidenced by delayed wound healing in IL-6-deficient mice. J Leukoc Biol 2003; 73: 713–721.
    1. Kaelin WGJr, Ratcliffe PJ. Oxygen sensing by metazoans: the central role of the HIF hydroxylase pathway. Mol Cell 2008; 30: 393–402.
    1. Kuhlicke J, Frick JS, Morote-Garcia JC, Rosenberger P, Eltzschig HK. Hypoxia inducible factor (HIF)-1 coordinates induction of Toll-like receptors TLR2 and TLR6 during hypoxia. PLoS ONE 2007; 2: e1364.
    1. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med 2011; 364: 656–665.
    1. Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 2001; 414: 916–920.
    1. van Golen RF, Reiniers MJ, Olthof PB, van Gulik TM, Heger M. Sterile inflammation in hepatic ischemia/reperfusion injury: present concepts and potential therapeutics. J Gastroenterol Hepatol 2013; 28: 394–400.
    1. Besedovsky HO, del Rey A, Klusman I, Furukawa H, Monge Arditi G, Kabiersch A. Cytokines as modulators of the hypothalamus-pituitary-adrenal axis. J Steroid Biochem Mol Biol 1991; 40: 613–618.
    1. Griffin GD, Charron D, Al-Daccak R. Post-traumatic stress disorder: revisiting adrenergics, glucocorticoids, immune system effects and homeostasis. Clin Transl Immunol 2014; 3: e27.
    1. Griffin GD. Stroke, mTBI, infection, antibiotics and beta blockade: Connecting the dots. Med Hypotheses 2015; 85: 224–229.
    1. McDonald B, Kubes P. Cellular and molecular choreography of neutrophil recruitment to sites of sterile inflammation. J Mol Med 2011; 89: 1079–1088.
    1. Tsou CL, Peters W, Si Y, Slaymaker S, Aslanian AM, Weisberg SP et al. Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow and recruitment to inflammatory sites. J Clin Invest 2007; 117: 902–909.
    1. Seebach C, Henrich D, Tewksbury R, Wilhelm K, Marzi I. Number and proliferative capacity of human mesenchymal stem cells are modulated positively in multiple trauma patients and negatively in atrophic nonunions. Calcif Tissue Int 2007; 80: 294–300.
    1. Kimura F, Shimizu H, Yoshidome H, Ohtsuka M, Miyazaki M. Immunosuppression following surgical and traumatic injury. Surg Today 2010; 40: 793–808.
    1. Suratt BT, Petty JM, Young SK, Malcolm KC, Lieber JG, Nick JA et al. Role of the CXCR4/SDF-1 chemokine axis in circulating neutrophil homeostasis. Blood 2004; 104: 565–571.
    1. Majetschak M, Zedler S, Hostmann A, Sorell LT, Patel MB, Novar LT et al. Systemic ubiquitin release after blunt trauma and burns: association with injury severity, posttraumatic complications, and survival. J Trauma 2008; 64: 586–596.
    1. Majetschak M. Extracellular ubiquitin: immune modulator and endogenous opponent of damage-associated molecular pattern molecules. J Leukoc Biol 2011; 89: 205–219.
    1. Arancibia SA, Beltran CJ, Aguirre IM, Silva P, Peralta AL, Malinarich F et al. Toll-like receptors are key participants in innate immune responses. Biol Res 2007; 40: 97–112.
    1. Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 2013; 13: 159–175.
    1. Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW. Emerging mechanisms of neutrophil recruitment across endothelium. Trends Immunol 2011; 32: 461–469.
    1. Drifte G, Dunn-Siegrist I, Tissieres P, Pugin J. Innate immune functions of immature neutrophils in patients with sepsis and severe systemic inflammatory response syndrome. Crit Care Med 2013; 41: 820–832.
    1. El-Maallem H, Fletcher J. Effects of surgery on neutrophil granulocyte function. Infect Immun 1981; 32: 38–41.
    1. Köller M, Wick M, Muhr G. Decreased leukotriene release from neutrophils after severe trauma: role of immature cells. Inflammation 2001; 25: 53–59.
    1. Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 2009; 325: 612–616.
    1. Henderson RB, Hobbs JA, Mathies M, Hogg N. Rapid recruitment of inflammatory monocytes is independent of neutrophil migration. Blood 2003; 102: 328–335.
    1. Hensler T, Hecker H, Heeg K, Heidecke CD, Bartels H, Barthlen W et al. Distinct mechanisms of immunosuppression as a consequence of major surgery. Infect Immun 1997; 65: 2283–2291.
    1. Kawasaki T, Fujimi S, Lederer JA, Hubbard WJ, Choudhry MA, Schwacha MG et al. Trauma-hemorrhage induces depressed splenic dendritic cell functions in mice. J Immunol 2006; 177: 4514–4520.
    1. Ho CS, Lopez JA, Vuckovic S, Pyke CM, Hockey RL, Hart DN. Surgical and physical stress increases circulating blood dendritic cell counts independently of monocyte counts. Blood 2001; 98: 140–145.
    1. Henrich D, Maier M, Relja B, Trendafilov P, Schiessling S, Wirth M et al. Significant Decline of Peripheral Myeloid Dendritic Cells Following Multiple Trauma. J Surg Res 2009; 154: 239–245.
    1. Maier M, Geiger EV, Henrich D, Ebrahimi R, Wutzler S, Lehnert M et al. Apoptosis differs in dendritic cell subsets early after severe trauma. Hum Immunol 2009; 70: 803–808.
    1. Geiger EV, Maier M, Schiessling S, Wutzler S, Lehnert M, Marzi I et al. Subsequent gene expression pattern in dendritic cells following multiple trauma. Langenbecks Arch Surg 2013; 398: 327–333.
    1. Maier M, Geiger EV, Henrich D, Bendt C, Wutzler S, Lehnert M et al. Platelet factor 4 is highly upregulated in dendritic cells after severe trauma. Mol Med 2009; 15: 384–391.
    1. Maier M, Wutzler S, Bauer M, Trendafilov P, Henrich D, Marzi I. Altered gene expression patterns in dendritic cells after severe trauma: implications for systemic inflammation and organ injury. Shock 2008; 30: 344–351.
    1. Albertsmeier M, Quaiser D, von Dossow-Hanfstingl V, Winter H, Faist E, Angele MK. Major surgical trauma differentially affects T-cells and APC. Innate Immun 2015; 21: 55–64.
    1. Filipazzi P, Valenti R, Huber V, Pilla L, Canese P, Iero M et al. Identification of a new subset of myeloid suppressor cells in peripheral blood of melanoma patients with modulation by a granulocyte-macrophage colony-stimulation factor-based antitumor vaccine. J Clin Oncol 2007; 25: 2546–2553.
    1. Cuenca AG, Delano MJ, Kelly-Scumpia KM, Moreno C, Scumpia PO, Laface DM et al. A paradoxical role for myeloid-derived suppressor cells in sepsis and trauma. Mol Med 2011; 17: 281–292.
    1. Mansilla E, Marin GH, Drago H, Sturla F, Salas E, Gardiner C et al. Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc 2006; 38: 967–969.
    1. Marik PE, Flemmer M. The immune response to surgery and trauma: Implications for treatment. J Trauma Acute Care Surg 2012; 73: 801–808.
    1. Munoz M, Cobos A, Campos A, Ariza D, Munoz E, Gomez A. Post-operative unwashed shed blood transfusion does not modify the cellular immune response to surgery for total knee replacement. Acta Anaesthesiol Scand 2006; 50: 443–450.
    1. 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: 1184–1193.
    1. Gharehbaghian A, Haque KM, Truman C, Evans R, Morse R, Newman J et al. Effect of autologous salvaged blood on postoperative natural killer cell precursor frequency. Lancet 2004; 363: 1025–1030.
    1. Blazar BA, Rodrick ML, O'Mahony JB, Wood JJ, Bessey PQ, Wilmore DW et al. Suppression of natural killer-cell function in humans following thermal and traumatic injury. J Clin Immunol 1986; 6: 26–36.
    1. Roche M, Thorn GW, Hills AG. The levels of circulating eosinophils and their response to ACTH in surgery. N Engl J Med 1950; 242: 307–314.
    1. Laroche D, Chrysanthou S, Lefrançois C, Gérard JL, Dubois F, Malet M et al. Evidence of a progressive degranulation of basophil in subjects operated on without adverse reaction. Agents Actions 1992; 36: C199–C200.
    1. Buvanendran A, Mitchell K, Kroin JS, Iadarola MJ. Cytokine gene expression after total hip arthroplasty: surgical site versus circulating neutrophil response. Anesth Analg 2009; 109: 959–964.
    1. Abe T, Sakamoto K, Mita S, Kamohara H, Hirano Y-i, Kuwahara N et al. Surgical trauma induces group ii phospholipase a2 production by neutrophils at a local site after surgery. Clin Biochem 1998; 31: 101–106.
    1. Laudanski K, Miller-Graziano C, Xiao W, Mindrinos MN, Richards DR, De A et al. Cell-specific expression and pathway analyses reveal alterations in trauma-related human T cell and monocyte pathways. Proc Natl Acad Sci USA 2006; 103: 15564–15569.
    1. Sugimoto M, Shimaoka M, Hosotsubo K, Tanigami H, Taenaka N, Kiyono H et al. Up-regulation of Fas ligand (FasL) mRNA expression in peripheral blood mononuclear cells (PBMC) after major surgery. Clin Exp Immunol 1998; 112: 120–125.
    1. Miller AC, Rashid RM, Elamin EM. The ‘T' in trauma: the helper T-cell response and the role of immunomodulation in trauma and burn patients. J Trauma 2007; 63: 1407–1417.
    1. Gale LM, McColl SR. Chemokines: extracellular messengers for all occasions? Bioessays 1999; 21: 17–28.
    1. Manson J, Thiemermann C, Brohi K. Trauma alarmins as activators of damage-induced inflammation. Br J Surg 2012; 99 (Suppl 1): 12–20.
    1. Khurana A, Zafar S, Abdul W, Mukhopadhyay S, Mohanty K. Postoperative reduction in haemoglobin levels is related to rise in troponin T following hip fracture surgery. Orthopaedic Proceedings of Bone and Joint 2012; 94B (Supp XVII): 024.
    1. Burk AM, Martin M, Flierl MA, Rittirsch D, Helm M, Lampl L et al. Early complementopathy after multiple injuries in humans. Shock 2012; 37: 348–354.
    1. Kanse SM, Gallenmueller A, Zeerleder S, Stephan F, Rannou O, Denk S et al. Factor VII-activating protease is activated in multiple trauma patients and generates anaphylatoxin C5a. J Immunol 2012; 188: 2858–2865.
    1. Bastian D, Tamburstuen MV, Lyngstadaas SP, Reikeras O. LBP and sCD14 patterns in total hip replacement surgery performed during combined spinal/epidural anaesthesia. Scand J Clin Lab Invest 2011; 71: 486–491.
    1. Kristiansson M, Soop M, Sundqvist KG, Soop A, Suontaka AM, Blomback M. Local vs. systemic immune and haemostatic response to hip arthroplasty. Eur J Anaesthesiol 1998; 15: 260–270.
    1. Corbi P, Rahmati M, Delwail A, Potreau D, Menu P, Wijdenes J et al. Circulating soluble gp130, soluble IL-6R, and IL-6 in patients undergoing cardiac surgery, with or without extracorporeal circulation. Eur J Cardiothorac Surg 2000; 18: 98–103.
    1. Johnson SB, Lissauer M, Bochicchio GV, Moore R, Cross AS, Scalea TM. Gene expression profiles differentiate between sterile SIRS and early sepsis. Ann Surg 2007; 245: 611–621.
    1. Stuber F, Klaschik S, Lehmann LE, Schewe JC, Weber S, Book M. Cytokine promoter polymorphisms in severe sepsis. Clin Infect Dis 2005; 41 (Suppl 7): S416–S420.
    1. Silber JH, Rosenbaum PR, Kelz RR, Reinke CE, Neuman MD, Ross RN et al. Medical and financial risks associated with surgery in the elderly obese. Ann Surg 2012; 256: 79–86.
    1. Haider AH, Chang DC, Efron DT, Haut ER, Crandall M, Cornwell EE3rd. Race and insurance status as risk factors for trauma mortality. Arch Surg 2008; 143: 945–949.
    1. Jellusova J, Wellmann U, Amann K, Winkler TH, Nitschke L. CD22 x Siglec-G double-deficient mice have massively increased B1 cell numbers and develop systemic autoimmunity. J Immunol 2010; 184: 3618–3627.
    1. Islam N, Whitehouse M, Mehendale S, Hall M, Tierney J, O'Connell E et al. Post-traumatic Immunosuppression is Reversed by Anti-coagulated Salvaged Blood Transfusion; Deductions from studying Immune Status after Knee Arthroplasty. Clin Exp Immunol 2014; 177: 509–520.
    1. Islam N. Characterization of post-traumatic immunosuppression and its partial reversal by autologous salvaged blood transfusion. PhD Thesis, Aran Library, NUI Galway, Galway, Ireland, 2015.
    1. Darrah E, Andrade F. NETs: the missing link between cell death and systemic autoimmune diseases? Front Immunol 2012; 3: 428.
    1. Jawa RS, Anillo S, Huntoon K, Baumann H, Kulaylat M. Analytic Review: Interleukin-6 in Surgery, Trauma, and Critical Care: Part I: Basic Science. J Intensive Care Med 2011; 26: 3–12.
    1. Heinrich PC, Castell JV, Andus T. Interleukin-6 and the acute phase response. Biochem J 1990; 265: 621–636.
    1. Gabay C, Kushner I. eLS. John Wiley & Sons, Ltd: Chichester, UK. 2001.
    1. Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med 1999; 340: 448–454.
    1. Tilg H, Dinarello CA, Mier JW. IL-6 and APPs: anti-inflammatory and immunosuppressive mediators. Immunol Today 1997; 18: 428–432.
    1. Arredouani MS, Kasran A, Vanoirbeek JA, Berger FG, Baumann H, Ceuppens JL. Haptoglobin dampens endotoxin-induced inflammatory effects both in vitro and in vivo. Immunology 2005; 114: 263–271.
    1. Pape HC, Dwenger A, Regel G, Auf'm'Kolck M, Gollub F, Wisner D et al. Increased gut permeability after multiple trauma. Br J Surg 1994; 81: 850–852.
    1. Shorr AF, Jackson WL. Transfusion practice and nosocomial infection: assessing the evidence. Curr Opin Crit Care 2005; 11: 468–472.
    1. Faruquzzaman. Positive associations of nosocomial infections in surgical ward with etiological clinical factors. Bratisl Lek Listy 2011; 112: 273–277.
    1. Meisel C, Schwab JM, Prass K, Meisel A, Dirnagl U. Central nervous system injury-induced immune deficiency syndrome. Nat Rev Neurosci 2005; 6: 775–786.
    1. Chamorro A, Urra X, Planas AM. Infection after acute ischemic stroke: a manifestation of brain-induced immunodepression. Stroke 2007; 38: 1097–1103.
    1. Rabinowitz RP, Caplan ES. Management of infections in the trauma patient. Surg Clin North Am 1999; 79: 1373–1383.
    1. Richards MJ, Edwards JR, Culver DH, Gaynes RP. Nosocomial infections in combined medical-surgical intensive care units in the United States. Infect Control Hosp Epidemiol 2000; 21: 510–515.
    1. Alanis AJ. Resistance to antibiotics: are we in the post-antibiotic era? Arch Med Res 2005; 36: 697–705.
    1. British Broadcasting Corporation Antibiotic resistance: World on cusp of 'post-antibiotic era'. BBC News Health, 2015. Available at . Accessed on 19 November 2015 .
    1. Ponce de Leon-Rosales SP, Molinar-Ramos F, Dominguez-Cherit G, Rangel-Frausto MS, Vazquez-Ramos VG. Prevalence of infections in intensive care units in Mexico: a multicenter study. Crit Care Med 2000; 28: 1316–1321.
    1. GOINS WA, RODRIGUEZ A, JOSHI M, JACOBS D. Intra-abdominal Abscess after blunt abdominal trauma. Ann Surg 1990; 212: 60–65.
    1. Ivatury RR, Zubowski R, Psarras P, Nallathambi M, Rohman M, Stahl WM. Intra-abdominal abscess after penetrating abdominal trauma. J Trauma 1988; 28: 1238–1243.
    1. Civetta JM, Hudson-Civetta J, Ball S. Decreasing catheter-related infection and hospital costs by continuous quality improvement. Crit Care Med 1996; 24: 1660–1665.
    1. Gong MN, Thompson BT, Williams P, Pothier L, Boyce PD, Christiani DC. Clinical predictors of and mortality in acute respiratory distress syndrome: potential role of red cell transfusion. Crit Care Med 2005; 33: 1191–1198.
    1. Silverboard H, Aisiku I, Martin GS, Adams M, Rozycki G, Moss M. The role of acute blood transfusion in the development of acute respiratory distress syndrome in patients with severe trauma. J Trauma 2005; 59: 717–723.
    1. Karam O, Tucci M, Toledano BJ, Robitaille N, Cousineau J, Thibault L et al. Length of storage and in vitro immunomodulation induced by prestorage leukoreduced red blood cells. Transfusion (Paris) 2009; 49: 2326–2334.
    1. Stephan RN, Kupper TS, Geha AS, Baue AE, Chaudry IH. HEmorrhage without tissue trauma produces immunosuppression and enhances susceptibility to sepsis. Arch Surg 1987; 122: 62–68.
    1. Abraham E, Chang YH. Haemorrhage-induced alterations in function and cytokine production of T cells and T cell subpopulations. Clin Exp Immunol 1992; 90: 497–502.
    1. Hogan BV, Peter MB, Shenoy HG, Horgan K, Hughes TA. Surgery induced immunosuppression. Surgeon 2011; 9: 38–43.
    1. Kawasaki T, Ogata M, Kawasaki C, Okamoto K, Sata T. Effects of epidural anaesthesia on surgical stress-induced immunosuppression during upper abdominal surgery. Br J Anaesth 2007; 98: 196–203.
    1. Ali T, Kaitha S, Mahmood S, Ftesi A, Stone J, Bronze MS. Clinical use of anti-TNF therapy and increased risk of infections. Drug Healthcare Patient Saf 2013; 5: 79–99.
    1. Gea-Banacloche JC, Opal SM, Jorgensen J, Carcillo JA, Sepkowitz KA, Cordonnier C. Sepsis associated with immunosuppressive medications: an evidence-based review. Crit Care Med 2004; 32: S578–S590.
    1. Hansen BL, Rohr N, Svendsen V, Olsen H, Birkeland SA. Bacterial urinary tract infection in cyclosporine-a immunosuppressed renal transplant recipients. Scand J Infect Dis 1988; 20: 425–427.
    1. Aw D, Silva AB, Palmer DB. Immunosenescence: emerging challenges for an ageing population. Immunology 2007; 120: 435–446.
    1. Ginaldi L, Loreto MF, Corsi MP, Modesti M, De Martinis M. Immunosenescence and infectious diseases. Microbes Infect 2001; 3: 851–857.
    1. Zacks SI, Sheff MF. Age-related impeded regeneration of mouse minced anterior tibial muscle. Muscle Nerve 1982; 5: 152–161.
    1. Kudsk KA. Immunonutrition in surgery and critical care. Annu Rev Nutr 2006; 26: 463–479.
    1. Helminen H, Raitanen M, Kellosalo J. Immunonutrition in elective gastrointestinal surgery patients. Scand J Surg 2007; 96: 46–50.
    1. Stechmiller JK, Childress B, Porter T. Arginine immunonutrition in critically ill patients: a clinical dilemma. Am J Crit Care 2004; 13: 17–23.
    1. Zheng Y, Li F, Qi B, Luo B, Sun H, Liu S et al. Application of perioperative immunonutrition for gastrointestinal surgery: a meta-analysis of randomized controlled trials. Asia Pac J Clin Nutr 2007; 16 (Suppl 1): 253–257.
    1. Calder PC. Immunonutrition in surgical and critically ill patients. Br J Nutr 2007; 98 (Suppl 1): S133–S139.
    1. Klaenhammer TR, Kleerebezem M, Kopp MV, Rescigno M. The impact of probiotics and prebiotics on the immune system. Nat Rev Immunol 2012; 12: 728–734.
    1. Boomer JS, Green JM, Hotchkiss RS. The changing immune system in sepsis: Is individualized immuno-modulatory therapy the answer? Virulence 2014; 5: 45–56.
    1. Hotchkiss RS, Monneret G, Payen D. Immunosuppression in sepsis: a novel understanding of the disorder and a new therapeutic approach. Lancet Infect Dis 2013; 13: 260–268.
    1. Hancock RE, Nijnik A, Philpott DJ. Modulating immunity as a therapy for bacterial infections. Nat Rev Microbiol 2012; 10: 243–254.
    1. Scanzello C, Figgie M, Nestor B, Goodman S. Perioperative management of medications used in the treatment of rheumatoid arthritis. HSS J 2006; 2: 141–147.
    1. Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A. The new normal: immunomodulatory agents against sepsis immune suppression. Trends Mol Med 2014; 20: 224–233.
    1. Savage CD, Lopez-Castejon G, Denes A, Brough D. NLRP3-inflammasome activating DAMPs stimulate an inflammatory response in glia in the absence of priming which contributes to brain inflammation after injury. Front Immunol 2012; 3: 288.
    1. Liu Y, Chen GY, Zheng P. CD24-Siglec G/10 discriminates danger- from pathogen-associated molecular patterns. Trends Immunol 2009; 30: 557–561.
    1. Chen GY, Chen X, King S, Cavassani KA, Cheng J, Zheng X et al. Amelioration of sepsis by inhibiting sialidase-mediated disruption of the CD24-SiglecG interaction. Nat Biotechnol 2011; 29: 428–435.
    1. Chen GY, Tang J, Zheng P, Liu Y. CD24 and Siglec-10 selectively repress tissue damage-induced immune responses. Science 2009; 323: 1722–1725.
    1. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol 2011; 11: 762–774.
    1. Han Z, Jing Y, Zhang S, Liu Y, Shi Y, Wei L. The role of immunosuppression of mesenchymal stem cells in tissue repair and tumor growth. Cell Biosci 2012; 2: 8.
    1. McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CC et al. Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 2010; 330: 362–366.
    1. Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, Plonquet A et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med 2007; 204: 1057–1069.
    1. Kaczmarek A, Vandenabeele P, Krysko DV. Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 2013; 38: 209–223.
    1. Medzhitov R. Origin and physiological roles of inflammation. Nature 2008; 454: 428–435.

Source: PubMed

3
Subscribe