Intensive care unit-acquired weakness (ICUAW) and muscle wasting in critically ill patients with severe sepsis and septic shock

Joerg C Schefold, Jeffrey Bierbrauer, Steffen Weber-Carstens, Joerg C Schefold, Jeffrey Bierbrauer, Steffen Weber-Carstens

Abstract

Sepsis presents a major health care problem and remains one of the leading causes of death within the intensive care unit (ICU). Therapeutic approaches against severe sepsis and septic shock focus on early identification. Adequate source control, administration of antibiotics, preload optimization by fluid resuscitation and further hemodynamic stabilisation using vasopressors whenever appropriate are considered pivotal within the early-golden-hours of sepsis. However, organ dysfunction develops frequently in and represents a significant comorbidity of sepsis. A considerable amount of patients with sepsis will show signs of severe muscle wasting and/or ICU-acquired weakness (ICUAW), which describes a frequently observed complication in critically ill patients and refers to clinically weak ICU patients in whom there is no plausible aetiology other than critical illness. Some authors consider ICUAW as neuromuscular organ failure, caused by dysfunction of the motor unit, which consists of peripheral nerve, neuromuscular junction and skeletal muscle fibre. Electrophysiologic and/or biopsy studies facilitate further subclassification of ICUAW as critical illness myopathy, critical illness polyneuropathy or critical illness myoneuropathy, their combination. ICUAW may protract weaning from mechanical ventilation and impede rehabilitation measures, resulting in increased morbidity and mortality. This review provides an insight on the available literature on sepsis-mediated muscle wasting, ICUAW and their potential pathomechanisms.

Figures

Fig. 1
Fig. 1
Risk factors involved in muscle wasting and ICUAW. Both complications may overlap in septic patients, yet they present two distinct entities that should not be used synonymously. Whereas ICUAW is most likely accompanied by muscle wasting, muscle wasting is not necessarily associated with ICUAW
Fig. 2
Fig. 2
Suggested beneficial effects of electrical muscle stimulation (EMS) with regard to muscle hypertrophy, atrophy, aerobic capacity, membrane excitability and membrane translocation of GLUT4. EMS may preserve membrane excitability. Membrane translocation of GLUT4 is regulated by IGF-1, AMPK, PGC-1α and its downstream targets, which may all be affected by EMS. Atrophy gene expression (MuRF-1, atrogin-1) increases upon desphosphorylation of FoxO3 trancription factors, which is inhibited by downstream insulin signalling. EMS electrical muscle stimulation, IGF-1 insulin growth factor-1, GLUT4 glucose transporter 4, IRS-1 insulin receptor substrate 1, AMPK AMP-activated protein kinase, PI3K phopshoinositide 3-kinase, PPAR peroxisome proliferator-activated receptor, PGC-1α PPAR-γ coactivator 1α, pAKT phosphorylated Akt protein kinase B, mTOR mammalian target of rapamycin, FoxO3 forkhead box O3, MuRF muscle-specific ring finger protein
Fig. 3
Fig. 3
Muscle histologies (vastus lateralis muscle) from an ICU patient with critical illness myopathy (subclassification of ICUAW) and an ICU patient without this complication, referred to as ICU control. ATPase/Toludine blue staining differentiates type I, IIa and IIb muscle fibres as indicated

References

    1. Annane D, Bellissant E, Cavaillon J. Septic shock. Lancet. 2005;365:63–78. doi: 10.1016/S0140-6736(04)17667-8.
    1. Annane D, Aegerter P, Jars-Guincestre MC, Guidet B. Current epidemiology of septic shock: the CUB-Réa Network. Am J Respir Crit Care Med. 2003;168:165–172. doi: 10.1164/rccm.2201087.
    1. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003;348:138–150. doi: 10.1056/NEJMra021333.
    1. Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med. 2008;36:296–327. doi: 10.1097/01.CCM.0000298158.12101.41.
    1. American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med 1992;20:864–74.
    1. Bone RC. Sepsis, the sepsis syndrome, multi-organ failure: a plea for comparable definitions. Ann Intern Med. 1991;114:332–333.
    1. Bone RC. Sir Isaac Newton, sepsis, SIRS, and CARS. Crit Care Med. 1996;24:1125–1128. doi: 10.1097/00003246-199607000-00010.
    1. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med. 2001;29:1303–1310. doi: 10.1097/00003246-200107000-00002.
    1. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med. 2003;348:1546–1554. doi: 10.1056/NEJMoa022139.
    1. Engel C, Brunkhorst FM, Bone H, Brunkhorst R, Gerlach H, Grond S, et al. Epidemiology of sepsis in Germany: results from a national prospective multicenter study. Intensive Care Med. 2007;33:606–618. doi: 10.1007/s00134-006-0517-7.
    1. Moerer O, Schmid A, Hofmann M, Herklotz A, Reinhart K, Werdan K, et al. Direct costs of severe sepsis in three German intensive care units based on retrospective electronic patient record analysis of resource use. Intensive Care Med. 2002;28:1440–1446. doi: 10.1007/s00134-002-1429-9.
    1. Schmid A, Burchardi H, Clouth J, Schneider H. Burden of illness imposed by severe sepsis in Germany. Eur J Health Econ. 2002;3:77–82. doi: 10.1007/s10198-002-0095-8.
    1. Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, et al. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001;345:1368–1377. doi: 10.1056/NEJMoa010307.
    1. Annane D, Sébille V, Charpentier C, Bollaert P, François B, Korach J, et al. Effect of treatment with low doses of hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA. 2002;288:862–871. doi: 10.1001/jama.288.7.862.
    1. Van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, et al. Intensive insulin therapy in the medical ICU. N Engl J Med. 2006;354:449–461. doi: 10.1056/NEJMoa052521.
    1. van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, et al. Intensive insulin therapy in the critically ill patients. N Engl J Med. 2001;345:1359–1367. doi: 10.1056/NEJMoa011300.
    1. Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez-Rodriguez A, et al. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001;344:699–709. doi: 10.1056/NEJM200103083441001.
    1. Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K, et al. Hydrocortisone therapy for patients with septic shock. N Engl J Med. 2008;358:111–124. doi: 10.1056/NEJMoa071366.
    1. Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, et al. Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009;360:1283–1297. doi: 10.1056/NEJMoa0810625.
    1. Finfer S, Heritier S. The NICE-SUGAR (normoglycaemia in intensive care evaluation and survival using glucose algorithm regulation) study: statistical analysis plan. Crit Care Resusc. 2009;11:46–57.
    1. Cohen J. The immunopathogenesis of sepsis. Nature. 2002;420:885–891. doi: 10.1038/nature01326.
    1. Opal SM, Girard TD, Ely EW. The immunopathogenesis of sepsis in elderly patients. Clin Infect Dis. 2005;41:S504–S512. doi: 10.1086/432007.
    1. Schefold JC, Hasper D, Volk HD, Reinke P. Sepsis: time has come to focus on the later stages. Med Hypotheses. 2008;71:203–208. doi: 10.1016/j.mehy.2008.03.022.
    1. Höflich C, Volk HD. Immunomodulation in sepsis. Chirurg. 2002;73:1100–1104. doi: 10.1007/s00104-002-0559-5.
    1. Kox WJ, Volk T, Kox SN, Volk HD. Immunomodulatory therapies in sepsis. Intensive Care Med. 2000;26:S124–S128. doi: 10.1007/s001340051129.
    1. Schefold JC, Hasper D, Reinke P, Monneret G, Volk H. Consider delayed immunosuppression into the concept of sepsis. Crit Care Med. 2008;36:3118. doi: 10.1097/CCM.0b013e31818bdd8f.
    1. Volk HD, Reinke P, Döcke WD. Clinical aspects: from systemic inflammation to 'immunoparalysis’. Chem Immunol. 2000;74:162–177. doi: 10.1159/000058753.
    1. Nierhaus A, Montag B, Timmler N, Frings DP, Gutensohn K, Jung R, et al. Reversal of immunoparalysis by recombinant human granulocyte-macrophage colony-stimulating factor in patients with severe sepsis. Intensive Care Med. 2003;29:646–651.
    1. Döcke WD, Randow F, Syrbe U, Krausch D, Asadullah K, Reinke P, et al. Monocyte deactivation in septic patients: restoration by IFN-gamma treatment. Nat Med. 1997;3:678–681. doi: 10.1038/nm0697-678.
    1. Meisel C, Schefold JC, Pschowski R, Baumann T, Hetzger K, Gregor J, et al. Granulocyte-macrophage colony-stimulating factor to reverse sepsis-associated immunosuppression: a double-blind, randomized, placebo-controlled multicenter trial. Am J Respir Crit Care Med. 2009;180:640–648. doi: 10.1164/rccm.200903-0363OC.
    1. Schefold JC, von Haehling S, Corsepius M, Pohle C, Kruschke P, Zuckermann H, et al. A novel selective extracorporeal intervention in sepsis: immunoadsorption of endotoxin, interleukin 6, and complement-activating product 5a. Shock. 2007;28:418–425. doi: 10.1097/shk.0b013e31804f5921.
    1. Bercker S, Weber-Carstens S, Deja M, Grimm C, Wolf S, Behse F, et al. Critical illness polyneuropathy and myopathy in patients with acute respiratory distress syndrome. Crit Care Med. 2005;33:711–715. doi: 10.1097/01.CCM.0000157969.46388.A2.
    1. Osler SW. The principles and practice of medicine: designed for the use of practitioners and students of medicine. New York: D. Appleton and Company; 1910.
    1. de Jonghe B, Lacherade J, Sharshar T, Outin H. Intensive care unit-acquired weakness: risk factors and prevention. Crit Care Med. 2009;37:S309–S315. doi: 10.1097/CCM.0b013e3181b6e64c.
    1. Stevens RD, Marshall SA, Cornblath DR, Hoke A, Needham DM, de Jonghe B, et al. A framework for diagnosing and classifying intensive care unit-acquired weakness. Crit Care Med. 2009;37:S299–S308. doi: 10.1097/CCM.0b013e3181b6ef67.
    1. Callahan LA, Supinski GS. Sepsis-induced myopathy. Crit Care Med. 2009;37:S354–S367. doi: 10.1097/CCM.0b013e3181b6e439.
    1. Lang CH, Frost RA, Vary TC. Regulation of muscle protein synthesis during sepsis and inflammation. Am J Physiol Endocrinol Metab. 2007;293:E453–E459. doi: 10.1152/ajpendo.00204.2007.
    1. Vary TC, Kimball SR. Sepsis-induced changes in protein synthesis: differential effects on fast- and slow-twitch muscles. Am J Physiol. 1992;262:C1513–C1519.
    1. Thorell A, Rooyackers O, Myrenfors P, Soop M, Nygren J, Ljungqvist OH. Intensive insulin treatment in critically ill trauma patients normalizes glucose by reducing endogenous glucose production. J Clin Endocrinol Metab. 2004;89:5382–5386. doi: 10.1210/jc.2004-1118.
    1. Agwunobi AO, Reid C, Maycock P, Little RA, Carlson GL. Insulin resistance and substrate utilization in human endotoxemia. J Clin Endocrinol Metab. 2000;85:3770–3778. doi: 10.1210/jc.85.10.3770.
    1. Carlson GL. Insulin resistance in sepsis. Br J Surg. 2003;90:259–260. doi: 10.1002/bjs.4081.
    1. Carlson GL. Hunterian Lecture: insulin resistance in human sepsis: implications for the nutritional and metabolic care of the critically ill surgical patient. Ann R Coll Surg Engl. 2004;86:75–81. doi: 10.1308/003588404322827428.
    1. Mesotten D, Delhanty PJD, Vanderhoydonc F, Hardman KV, Weekers F, Baxter RC, et al. Regulation of insulin-like growth factor binding protein-1 during protracted critical illness. J Clin Endocrinol Metab. 2002;87:5516–5523. doi: 10.1210/jc.2002-020664.
    1. Zauner A, Nimmerrichter P, Anderwald C, Bischof M, Schiefermeier M, Ratheiser K, et al. Severity of insulin resistance in critically ill medical patients. Metab Clin Exp. 2007;56:1–5.
    1. Spranger J, Kroke A, Möhlig M, Hoffmann K, Bergmann MM, Ristow M, et al. Inflammatory cytokines and the risk to develop type 2 diabetes: results of the prospective population-based European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam Study. Diabetes. 2003;52:812–817. doi: 10.2337/diabetes.52.3.812.
    1. Xu J, Kim HT, Ma Y, Zhao L, Zhai L, Kokorina N, et al. Trauma and hemorrhage-induced acute hepatic insulin resistance: dominant role of tumor necrosis factor-alpha. Endocrinology. 2008;149:2369–2382. doi: 10.1210/en.2007-0922.
    1. Lang CH, Frost RA. Sepsis-induced suppression of skeletal muscle translation initiation mediated by tumor necrosis factor alpha. Metab Clin Exp. 2007;56:49–57.
    1. Phillips T, Leeuwenburgh C. Muscle fiber specific apoptosis and TNF-alpha signaling in sarcopenia are attenuated by life-long calorie restriction. FASEB J. 2005;19:668–670.
    1. Steffen BT, Lees SJ, Booth FW. Anti-TNF treatment reduces rat skeletal muscle wasting in monocrotaline-induced cardiac cachexia. J Appl Physiol. 2008;105:1950–1958. doi: 10.1152/japplphysiol.90884.2008.
    1. Ebisui C, Tsujinaka T, Morimoto T, Kan K, Iijima S, Yano M, et al. Interleukin-6 induces proteolysis by activating intracellular proteases (cathepsins B and L, proteasome) in C2C12 myotubes. Clin Sci. 1995;89:431–439.
    1. Frost RA, Nystrom GJ, Jefferson LS, Lang CH. Hormone, cytokine, and nutritional regulation of sepsis-induced increases in atrogin-1 and MuRF1 in skeletal muscle. Am J Physiol Endocrinol Metab. 2007;292:E501–E512. doi: 10.1152/ajpendo.00359.2006.
    1. Vary TC, Deiter G, Lang CH. Cytokine-triggered decreases in levels of phosphorylated eukaryotic initiation factor 4G in skeletal muscle during sepsis. Shock. 2006;26:631–636. doi: 10.1097/01.shk.0000230299.78515.2c.
    1. Kobayashi H, Kato H, Hirabayashi Y, Murakami H, Suzuki H. Modulations of muscle protein metabolism by branched-chain amino acids in normal and muscle-atrophying rats. J Nutr. 2006;136:234S–236S.
    1. Freyssenet D, Berthon P, Denis C, Barthelemy JC, Guezennec CY, Chatard JC. Effect of a 6-week endurance training programme and branched-chain amino acid supplementation on histomorphometric characteristics of aged human muscle. Arch Physiol Biochem. 1996;104:157–162. doi: 10.1076/apab.104.2.157.12879.
    1. Ventrucci G, Ramos Silva LG, Roston Mello MA, Gomes Marcondes MCC. Effects of a leucine-rich diet on body composition during nutritional recovery in rats. Nutrition. 2004;20:213–217. doi: 10.1016/j.nut.2003.10.014.
    1. Sugawara T, Ito Y, Nishizawa N, Nagasawa T. Supplementation with dietary leucine to a protein-deficient diet suppresses myofibrillar protein degradation in rats. J Nutr Sci Vitaminol. 2007;53:552–555. doi: 10.3177/jnsv.53.552.
    1. Lang CH, Frost RA. Glucocorticoids and TNFalpha interact cooperatively to mediate sepsis-induced leucine resistance in skeletal muscle. Mol Med. 2006;12:291–299. doi: 10.2119/2006-00071.Lang.
    1. Mitch WE, Goldberg AL. Mechanisms of muscle wasting. The role of the ubiquitin-proteasome pathway. N Engl J Med. 1996;335:1897–1905. doi: 10.1056/NEJM199612193352507.
    1. Hasselgren P, Menconi MJ, Fareed MU, Yang H, Wei W, Evenson A. Novel aspects on the regulation of muscle wasting in sepsis. Int J Biochem Cell Biol. 2005;37:2156–2168. doi: 10.1016/j.biocel.2005.01.017.
    1. Rabuel C, Renaud E, Brealey D, Ratajczak P, Damy T, Alves A, et al. Human septic myopathy: induction of cyclooxygenase, heme oxygenase and activation of the ubiquitin proteolytic pathway. Anesthesiology. 2004;101:583–590. doi: 10.1097/00000542-200409000-00006.
    1. Klaude M, Fredriksson K, Tjäder I, Hammarqvist F, Ahlman B, Rooyackers O, et al. Proteasome proteolytic activity in skeletal muscle is increased in patients with sepsis. Clin Sci. 2007;112:499–506. doi: 10.1042/CS20060265.
    1. Baracos V, Rodemann HP, Dinarello CA, Goldberg AL. Stimulation of muscle protein degradation and prostaglandin E2 release by leukocytic pyrogen (interleukin-1). A mechanism for the increased degradation of muscle proteins during fever. N Engl J Med. 1983;308:553–558. doi: 10.1056/NEJM198303103081002.
    1. Hummel RP, James JH, Warner BW, Hasselgren PO, Fischer JE. Evidence that cathepsin B contributes to skeletal muscle protein breakdown during sepsis. Arch Surg. 1988;123:221–224.
    1. Voisin L, Breuillé D, Combaret L, Pouyet C, Taillandier D, Aurousseau E, et al. Muscle wasting in a rat model of long-lasting sepsis results from the activation of lysosomal, Ca2+ -activated, and ubiquitin-proteasome proteolytic pathways. J Clin Invest. 1996;97:1610–1617. doi: 10.1172/JCI118586.
    1. Huang J, Forsberg NE. Role of calpain in skeletal-muscle protein degradation. Proc Natl Acad Sci USA. 1998;95:12100–12105. doi: 10.1073/pnas.95.21.12100.
    1. Du J, Wang X, Miereles C, Bailey JL, Debigare R, Zheng B, et al. Activation of caspase-3 is an initial step triggering accelerated muscle proteolysis in catabolic conditions. J Clin Invest. 2004;113:115–123.
    1. Tan FC, Goll DE, Otsuka Y. Some properties of the millimolar Ca2 + -dependent proteinase from bovine cardiac muscle. J Mol Cell Cardiol. 1988;20:983–997. doi: 10.1016/0022-2828(88)90576-7.
    1. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab. 2007;6:472–483. doi: 10.1016/j.cmet.2007.11.004.
    1. Sandri M, Sandri C, Gilbert A, Skurk C, Calabria E, Picard A, et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell. 2004;117:399–412. doi: 10.1016/S0092-8674(04)00400-3.
    1. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science. 2001;294:1704–1708. doi: 10.1126/science.1065874.
    1. Gomes MD, Lecker SH, Jagoe RT, Navon A, Goldberg AL. Atrogin-1, a muscle-specific F-box protein highly expressed during muscle atrophy. Proc Natl Acad Sci USA. 2001;98:14440–14445. doi: 10.1073/pnas.251541198.
    1. Clarke BA, Drujan D, Willis MS, Murphy LO, Corpina RA, Burova E, et al. The E3 Ligase MuRF1 degrades myosin heavy chain protein in dexamethasone-treated skeletal muscle. Cell Metab. 2007;6:376–385. doi: 10.1016/j.cmet.2007.09.009.
    1. Cohen S, Brault JJ, Gygi SP, Glass DJ, Valenzuela DM, Gartner C, et al. During muscle atrophy, thick, but not thin, filament components are degraded by MuRF1-dependent ubiquitylation. J Cell Biol. 2009;185:1083–1095. doi: 10.1083/jcb.200901052.
    1. Tintignac LA, Lagirand J, Batonnet S, Sirri V, Leibovitch MP, Leibovitch SA. Degradation of MyoD mediated by the SCF (MAFbx) ubiquitin ligase. J Biol Chem. 2005;280:2847–2856. doi: 10.1074/jbc.M411346200.
    1. Lagirand-Cantaloube J, Offner N, Csibi A, Leibovitch MP, Batonnet-Pichon S, Tintignac LA, et al. The initiation factor eIF3-f is a major target for atrogin1/MAFbx function in skeletal muscle atrophy. EMBO J. 2008;27:1266–1276. doi: 10.1038/emboj.2008.52.
    1. Attaix D, Baracos VE. MAFbx/Atrogin-1 expression is a poor index of muscle proteolysis. Curr Opin Clin Nutr Metab. 2010;13:223–224. doi: 10.1097/MCO.0b013e328338b9a6.
    1. Vincent J, Norrenberg M. Intensive care unit-acquired weakness: framing the topic. Crit Care Med. 2009;37:S296–S298. doi: 10.1097/CCM.0b013e3181b6f1e1.
    1. Spitzer AR, Giancarlo T, Maher L, Awerbuch G, Bowles A. Neuromuscular causes of prolonged ventilator dependency. Muscle Nerve. 1992;15:682–686. doi: 10.1002/mus.880150609.
    1. Latronico N, Fenzi F, Recupero D, Guarneri B, Tomelleri G, Tonin P, et al. Critical illness myopathy and neuropathy. Lancet. 1996;347:1579–1582. doi: 10.1016/S0140-6736(96)91074-0.
    1. Garnacho-Montero J, Madrazo-Osuna J, García-Garmendia JL, Ortiz-Leyba C, Jiménez-Jiménez FJ, Barrero-Almodóvar A, et al. Critical illness polyneuropathy: risk factors and clinical consequences. A cohort study in septic patients. Intensive Care Med. 2001;27:1288–1296. doi: 10.1007/s001340101009.
    1. Stevens RD, Dowdy DW, Michaels RK, Mendez-Tellez PA, Pronovost PJ, Needham DM. Neuromuscular dysfunction acquired in critical illness: a systematic review. Intensive Care Med. 2007;33:1876–1891. doi: 10.1007/s00134-007-0772-2.
    1. De Jonghe B, Bastuji-Garin S, Sharshar T, Outin H, Brochard L. Does ICU-acquired paresis lengthen weaning from mechanical ventilation? Intensive Care Med. 2004;30:1117–1121. doi: 10.1007/s00134-004-2174-z.
    1. De Jonghe B, Lacherade J, Durand M, Sharshar T. Critical illness neuromuscular syndromes. Crit Care Clin. 2007;23:55–69. doi: 10.1016/j.ccc.2006.11.001.
    1. De Jonghe B, Bastuji-Garin S, Durand M, Malissin I, Rodrigues P, Cerf C, et al. Respiratory weakness is associated with limb weakness and delayed weaning in critical illness. Crit Care Med. 2007;35:2007–2015. doi: 10.1097/01.ccm.0000281450.01881.d8.
    1. Herridge MS. Long-term outcomes after critical illness. Curr Opin Crit Care. 2002;8:331–336. doi: 10.1097/00075198-200208000-00010.
    1. Herridge MS. Legacy of intensive care unit-acquired weakness. Crit Care Med. 2009;37:S457–S461. doi: 10.1097/CCM.0b013e3181b6f35c.
    1. Kleyweg RP, van der Meché FG, Schmitz PI. Interobserver agreement in the assessment of muscle strength and functional abilities in Guillain-Barré syndrome. Muscle Nerve. 1991;14:1103–1109. doi: 10.1002/mus.880141111.
    1. Latronico N, Rasulo FA. Presentation and management of ICU myopathy and neuropathy. Curr Opin Crit Care [Internet]. 2010 Jan 13. Available from: . Accessed 4 Feb 2010
    1. Weber-Carstens S, Koch S, Spuler S, Spies CD, Bubser F, Wernecke KD, et al. Nonexcitable muscle membrane predicts intensive care unit-acquired paresis in mechanically ventilated, sedated patients. Crit Care Med. 2009;37:2632–2637. doi: 10.1097/CCM.0b013e3181a92f28.
    1. Cruz DN, Antonelli M, Fumagalli R, Foltran F, Brienza N, Donati A, et al. Early use of polymyxin B hemoperfusion in abdominal septic shock: the EUPHAS randomized controlled trial. JAMA. 2009;301:2445–2452. doi: 10.1001/jama.2009.856.
    1. Cruz DN, Perazella MA, Bellomo R, de Cal M, Polanco N, Corradi V, et al. Effectiveness of polymyxin B-immobilized fiber column in sepsis: a systematic review. Crit Care. 2007;11:R47. doi: 10.1186/cc5780.
    1. Schefold JC, Hasper D, Jörres A. Organ crosstalk in critically ill patients: hemofiltration and immunomodulation in sepsis. Blood Purif. 2009;28:116–123. doi: 10.1159/000223361.
    1. Stegmayr B. Apheresis in patients with severe sepsis and multi organ dysfunction syndrome. Transfus Apher Sci. 2008;38:203–208. doi: 10.1016/j.transci.2008.03.009.
    1. Haase M, Bellomo R, Morgera S, Morger S, Baldwin I, Boyce N. High cut-off point membranes in septic acute renal failure: a systematic review. Int J Artif Organs. 2007;30:1031–1041.
    1. Schweickert WD, Pohlman MC, Pohlman AS, Nigos C, Pawlik AJ, Esbrook CL, et al. Early physical and occupational therapy in mechanically ventilated, critically ill patients: a randomised controlled trial. Lancet. 2009;373:1874–1882. doi: 10.1016/S0140-6736(09)60658-9.
    1. Gerovasili V, Stefanidis K, Vitzilaios K, Karatzanos E, Politis P, Koroneos A, et al. Electrical muscle stimulation preserves the muscle mass of critically ill patients: a randomized study. Crit Care. 2009;13:R161. doi: 10.1186/cc8123.
    1. Strasser EM, Stättner S, Karner J, Klimpfinger M, Freynhofer M, Zaller V, et al. Neuromuscular electrical stimulation reduces skeletal muscle protein degradation and stimulates insulin-like growth factors in an age- and current-dependent manner: a randomized, controlled clinical trial in major abdominal surgical patients. Ann Surg. 2009;249:738–743. doi: 10.1097/SLA.0b013e3181a38e71.
    1. Van den Berghe G, Schoonheydt K, Becx P, Bruyninckx F, Wouters PJ. Insulin therapy protects the central and peripheral nervous system of intensive care patients. Neurology. 2005;64:1348–1353.
    1. Hermans G, Wilmer A, Meersseman W, Milants I, Wouters PJ, Bobbaers H, et al. Impact of intensive insulin therapy on neuromuscular complications and ventilator dependency in the medical intensive care unit. Am J Respir Crit Care Med. 2007;175:480–489. doi: 10.1164/rccm.200605-665OC.
    1. De Jonghe B, Sharshar T, Lefaucheur J, Authier F, Durand-Zaleski I, Boussarsar M, et al. Paresis acquired in the intensive care unit: a prospective multicenter study. JAMA. 2002;288:2859–2867. doi: 10.1001/jama.288.22.2859.
    1. Sander HW, Golden M, Danon MJ. Quadriplegic areflexic ICU illness: selective thick filament loss and normal nerve histology. Muscle Nerve. 2002;26:499–505. doi: 10.1002/mus.10233.
    1. Amaya-Villar R, Garnacho-Montero J, García-Garmendía JL, Madrazo-Osuna J, Garnacho-Montero MC, Luque R, et al. Steroid-induced myopathy in patients intubated due to exacerbation of chronic obstructive pulmonary disease. Intensive Care Med. 2005;31:157–161. doi: 10.1007/s00134-004-2509-9.
    1. Weber-Carstens S, Deja M, Koch S, Spranger J, Bubser F, Wernecke K, et al. Risk factors in Critical illness myopathy (CIM) during the early course of critical illness: a prospective observational study. Crit Care. 2010;14:R119. doi: 10.1186/cc9074.
    1. Segredo V, Caldwell JE, Matthay MA, Sharma ML, Gruenke LD, Miller RD. Persistent paralysis in critically ill patients after long-term administration of vecuronium. N Engl J Med. 1992;327:524–528. doi: 10.1056/NEJM199208203270804.
    1. Koch S, Spuler S, Deja M, Bierbrauer J, Dimroth A, Behse F, et al. Critical illness myopathy is frequent—accompanying neuropathy protracts ICU discharge. JNNP. 2010 (in press).
    1. Larsson L, Li X, Edström L, Eriksson LI, Zackrisson H, Argentini C, et al. Acute quadriplegia and loss of muscle myosin in patients treated with nondepolarizing neuromuscular blocking agents and corticosteroids: mechanisms at the cellular and molecular levels. Crit Care Med. 2000;28:34–45. doi: 10.1097/00003246-200001000-00006.
    1. Lacomis D, Giuliani MJ, Van Cott A, Kramer DJ. Acute myopathy of intensive care: clinical, electromyographic, and pathological aspects. Ann Neurol. 1996;40:645–654. doi: 10.1002/ana.410400415.
    1. Rich MM, Teener JW, Raps EC, Schotland DL, Bird SJ. Muscle is electrically inexcitable in acute quadriplegic myopathy. Neurology. 1996;46:731–736.
    1. Rich MM, Bird SJ, Raps EC, McCluskey LF, Teener JW. Direct muscle stimulation in acute quadriplegic myopathy. Muscle Nerve. 1997;20:665–673. doi: 10.1002/(SICI)1097-4598(199706)20:6<665::AID-MUS2>;2-6.
    1. Hirano M, Ott BR, Raps EC, Minetti C, Lennihan L, Libbey NP, et al. Acute quadriplegic myopathy: a complication of treatment with steroids, nondepolarizing blocking agents, or both. Neurology. 1992;42:2082–2087.
    1. Ramsay DA, Zochodne DW, Robertson DM, Nag S, Ludwin SK. A syndrome of acute severe muscle necrosis in intensive care unit patients. J Neuropathol Exp Neurol. 1993;52:387–398. doi: 10.1097/00005072-199307000-00006.
    1. Showalter CJ, Engel AG. Acute quadriplegic myopathy: analysis of myosin isoforms and evidence for calpain-mediated proteolysis. Muscle Nerve. 1997;20:316–322. doi: 10.1002/(SICI)1097-4598(199703)20:3<316::AID-MUS8>;2-E.
    1. Sher JH, Shafiq SA, Schutta HS. Acute myopathy with selective lysis of myosin filaments. Neurology. 1979;29:100–106.
    1. Lin MC, Ebihara S, El Dwairi Q, Hussain SN, Yang L, Gottfried SB, et al. Diaphragm sarcolemmal injury is induced by sepsis and alleviated by nitric oxide synthase inhibition. Am J Respir Crit Care Med. 1998;158:1656–1663.
    1. Comtois AS, Barreiro E, Huang PL, Marette A, Perrault M, Hussain SN. Lipopolysaccharide-induced diaphragmatic contractile dysfunction and sarcolemmal injury in mice lacking the neuronal nitric oxide synthase. Am J Respir Crit Care Med. 2001;163:977–982.
    1. Ebihara S, Hussain SNA, Danialou G, Cho W, Gottfried SB, Petrof BJ. Mechanical ventilation protects against diaphragm injury in sepsis: interaction of oxidative and mechanical stresses. Am J Respir Crit Care Med. 2002;165:221–228.
    1. Supinski G, Stofan D, Callahan LA, Nethery D, Nosek TM, DiMarco A. Peroxynitrite induces contractile dysfunction and lipid peroxidation in the diaphragm. J Appl Physiol. 1999;87:783–791.
    1. Supinski G, Nethery D, Nosek TM, Callahan LA, Stofan D, DiMarco A. Endotoxin administration alters the force vs. pCa relationship of skeletal muscle fibers. Am J Physiol Regul Integr Comp Physiol. 2000;278:R891–R896.
    1. Callahan LA, She ZW, Nosek TM. Superoxide, hydroxyl radical, and hydrogen peroxide effects on single-diaphragm fiber contractile apparatus. J Appl Physiol. 2001;90:45–54.
    1. Callahan LA, Nethery D, Stofan D, DiMarco A, Supinski G. Free radical-induced contractile protein dysfunction in endotoxin-induced sepsis. Am J Respir Cell Mol Biol. 2001;24:210–217.
    1. Reid MB, Lännergren J, Westerblad H. Respiratory and limb muscle weakness induced by tumor necrosis factor-alpha: involvement of muscle myofilaments. Am J Respir Crit Care Med. 2002;166:479–484. doi: 10.1164/rccm.2202005.
    1. Hardin BJ, Campbell KS, Smith JD, Arbogast S, Smith J, Moylan JS, et al. TNF-alpha acts via TNFR1 and muscle-derived oxidants to depress myofibrillar force in murine skeletal muscle. J Appl Physiol. 2008;104:694–699. doi: 10.1152/japplphysiol.00898.2007.
    1. Fredriksson K, Hammarqvist F, Strigård K, Hultenby K, Ljungqvist O, Wernerman J, et al. Derangements in mitochondrial metabolism in intercostal and leg muscle of critically ill patients with sepsis-induced multiple organ failure. Am J Physiol Endocrinol Metab. 2006;291:E1044–E1050. doi: 10.1152/ajpendo.00218.2006.
    1. von Haehling S, Morley JE, Coats AJ, Anker SD. Ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. J Cachexia Sarcopenia Muscle 2010;1:7–8.

Source: PubMed

3
Iratkozz fel