Неудачи интенсивного лечения полиорганной недостаточности: патофизиология и потребность в персонификации (обзор литературы)
#2019-2
PDF_2019-2_48-57
HTML_2019-2_48-57

Ключевые слова

системный воспалительный ответ
полиорганная недостаточность
алармины
митохондрии
иммунная супрессия
барьерная недостаточность
эндофенотип

Как цитировать

Григорьев Е.В., Шукевич Д.Л., Плотников Г.П., Кудрявцев А.Н., Радивилко А.С. Неудачи интенсивного лечения полиорганной недостаточности: патофизиология и потребность в персонификации (обзор литературы). Вестник интенсивной терапии имени А.И. Салтанова. 2019;(2):48–57. doi:10.21320/1818-474X-2019-2-48-57.

Статистика

Просмотров аннотации: 197
PDF_2019-2_48-57 загрузок: 19
HTML_2019-2_48-57 загрузок: 17
Статистика с 21.01.2023

Аннотация

Полиорганная недостаточность (ПОН) — наиболее тяжелый исход критического состояния любого генеза (сепсис, травма, ишемия и реперфузия), планка летальности при данном синдроме не имеет тенденции к снижению. Обзорная статья предлагает прежде всего знакомство с ключевыми научными направлениями, по которым на данный момент развивается теория ПОН (исследование значимости аларминов, митохондриальная дисфункция, барьерная недостаточность, иммунологическое и неврологическое сопряжение, формы программированной гибели клеток, индуцированная иммуносупрессия, разрешение воспаления). Исследования доказывают целесообразность введения персонифицированного подхода к диагностике ПОН путем обоснования эндофенотипа критического состояния на основании комплекса иммунологических, геномных и клинических показателей.
https://doi.org/10.21320/1818-474X-2019-2-48-57
PDF_2019-2_48-57
HTML_2019-2_48-57

Библиографические ссылки

  1. Ciesla D.J., Moore E.E., Johnson J.L., et al. A 12-year prospective study of postinjury multiple organ failure: has anything changed? Arch. Surg. 2005; 140(5): 432–438. DOI: 10.1001/archsurg.140.5.432
  2. Davidson G.H., Hamlat C.A., Rivara F.P., et al. Long-term survival of adult trauma patients. JAMA. 2011; 305(10): 1001–1007. DOI: 10.1001/jama.2011.259
  3. Eiseman B., Beart R., Norton L. Multiple organ failure. Surg. Gynecol. Obstet. 1977; 144(3): 323–326.
  4. Deitch E.A., Vincent J.L., Windsor A. Sepsis and multiple organ dysfunction: multidisciplinary approach. Philadelphia: WB Sanders company, 2002.
  5. Minei J.P., Cuschieri J., Sperry J., et al. The changing pattern and implications of multiple organ failure after blunt injury with hemorrhagic shock. Crit. Care Med. 2012; 40(4): 1129–1135. DOI: 10.1097/CCM.0b013e3182376e9f
  6. Lelubre C., Vincent J.L. Mechanisms and treatment of organ failure in sepsis. Nature Review. Nephrology. 2018; 14: 417–427. DOI: 10.1038/s41581-018-0005-7
  7. Григорьев Е.В., Плотников Г.П., Шукевич Д.Л., Головкин А.С. Персистирующая полиорганная недостаточность. Патология кровообращения и кардиохирургия. 2014; 18(3): 82–86.DOI: 10.21688/1681-3472-2014-3-82-86. [Grigoryev Ye.V., Plotnikov G.P., Shukevich D.L., Golovkin A.S. Persistent multiorgan failure. Patologiya krovoobrascheniya i kardiohirurgiya. Circulation Pathology and Cardiac Surgery. 2014; 18(3): 82–86. (In Russ)]
  8. Schaefer L. Complexity of danger: The diverse nature of damage-associated molecular patterns. J. Biol. Chem. 2014; 289: 35237–35245. DOI: 10.1074/jbc.R114.619304
  9. Ma K.C., Schenck E.J., Pabon M.A., Choi A.M.K. The role of danger signals in the pathogenesis and perpetuation of critical illness. Am. J. Respir. Crit. Care Med. 2018; 197(3): 300–309. DOI: 10.1164/rccm.201612–2460PP
  10. Zhang Q., Raoof M., Chen Y., et al. Circulating mitochondrial DAMPs cause in inflammatory responses to injury. Nature. 2010; 464: 104–107. DOI: 10.1038/nature08780
  11. Harris H.E., Raucci A. Alarmin(g) news about danger: Workshop on innate danger signals and HMGB1. EMBO Rep. 2006; 7: 774–778. DOI: 10.1038/sj.embor.7400759
  12. Guo H., Callaway J.B., Ting J.P. Infammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015; 21: 677–687. DOI: 10.1038/nm.3893
  13. Cobb J.P., Buchman T.G., Karl I.E., Hotchkiss R.S. Molecular biology of multiple organ dysfunction syndrome: Injury, adaptation, and apoptosis. Surg. Infect (Larchmt). 2000; 1: 207–213; discussion 214. DOI: 10.1089/109629600750018132
  14. Conrad M., Angeli J.P., Vandenabeele P., Stockwell B.R. Regulated necrosis: Disease relevance and therapeutic opportunities. Nat. Rev. Drug. Discov. 2016; 15: 348–366. DOI: 10.1038/nrd.2015.6
  15. Kaczmarek A., Vandenabeele P., Krysko D.V. Necroptosis: The release of damage-associated molecular patterns and its physiological relevance. Immunity. 2013; 38: 209–223. DOI: 10.1016/j.immuni.2013.02.003
  16. Krysko D.V., Agostinis P., Krysko O., et al. Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol. 2011; 32: 157–164. DOI: 10.1016/j.it.2011.01.005
  17. Zhang Q., Raoof M., Chen Y., et al. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature. 2010; 464: 104–107. DOI: 10.1038/nature08780
  18. Deutchman C.S., Tracey K.J. Sepsis: Current dogma and new perspectives. Immunity. 2014; 40: 463–475. DOI: 10.1016/j.immuni.2014.04.001
  19. Bosmann M., Ward P.A. The inflammatory response in sepsis. Trends Immunol. 2013; 34: 129–136. DOI: 10.1016/j.it.2012.09.004
  20. Matsuda N. Alert cell strategy in SIRS-induced vasculitis: sepsis and endothelial cells Journal of Intensive Care. 2016; 4: 21. DOI: 10.1186/s40560-016-0147-2
  21. Johansson P.I., Henriksen H.H., Stensballe J., et al. Traumatic endotheliopathy: a prospective observational study of 424 severely injured patients. Ann. Surg. 2017; 265(3): 597–603. DOI: 10.1097/SLA.0000000000001751
  22. Hirase T, Node K. Endothelial dysfunction as a cellular mechanism for vascular failure. Am. J. Physiol. Heart Circ. Physiol. 2012; 302(3): 499–505. DOI: 10.1152/ajpheart.00325.2011
  23. Aird W.C. The role of the endothelium in severe sepsis and multiple organ dysfunction syndrome. Blood. 2003; 101(10): 3765–3777. DOI: 10.1182/blood-2002-06-1887
  24. Szotowski B., Antoniak S., Rauch U. Alternatively spliced tissue factor: a previously unknown piece in the puzzle of hemostasis. Trends Cardiovasc. Med. 2006; 16(5): 177–182. DOI: 10.1016/j.tcm.2006.03.005
  25. Monroe D.M., Key N.S. The tissue factor-factor VIIa complex: procoagulant activity, regulation, and multitasking. J. Thromb. Haemost. 2007; 5(6): 1097–1105. DOI: 10.1111/j.1538-7836.2007.02435.x
  26. Danese S., Vetrano S., Zhang L., et al. The protein C pathway in tissue inflammation and injury: pathogenic role and therapeutic implications. Blood. 2010; 115(6): 1121–1130. DOI: 10.1182/blood-2009-09-201616
  27. Brinkmann V., Zychlinsky A. Beneficial suicide: why neutrophils die to, make NETs. Nature Rev. 2007; 5: 577–582. DOI: 10.1038/nrmicro1710
  28. Camicia G., Pozner R., de Larrañaga G. Neutrophil extracellular traps in Sepsis. Shock. 2014; 42(4): 286–294. DOI: 10.1097/SHK.0000000000000221
  29. Wang X., Qin W., Sun B. New strategy for sepsis: Targeting a key role of platelet-neutrophil interaction. Burns Trauma. 2014; 2(3): 114–120. DOI: 10.4103/2321–3868.135487
  30. Salmon A.H., Satchell S.C. Endothelial glycocalyx dysfunction in disease: albuminuria and increased microvascular permeability. J. Pathol. 2012; 226: 562–574. DOI: 10.1002/path.3964
  31. Pries A.R., Secomb T.W., Gaehtgens P. The endothelial surface layer. Pflugers Arch. 2000; 440: 653–666. DOI: 10.1007/s004240000307
  32. Reitsma S., Slaaf D.W., Vink H., et al. The endothelial glycocalyx: composition, functions, and visualization. Pflugers Arch. 2007; 454: 345–359. DOI: 10.1007/s00424-007-0212-8
  33. Lekakis J., Abraham P., Balbarini A., et al. Methods for evaluating endothelial function: a position statement from the European Society of Cardiology Working Group on Peripheral Circulation. Eur. J. Cardiovasc. Prev. Rehabil. 2011; 18: 775–789. DOI: 10.1177/1741826711398179
  34. Woodcock T.E., Woodcock T.M. Revised Starling equation and the glycocalyx model of transvascular fluid exchange: an improved paradigm for prescribing intravenous fluid therapy. Br. J. Anaesth. 2012; 108: 384–394. DOI: 10.1093/bja/aer515
  35. Chelazzi C., Villa G., Mancinelli P. Glycocalyx and sepsis-induced alterations in vascular permeability. Crit. Care. 2015; 19(1): 26. DOI: 10.1186/s13054-015-0741-z
  36. Uchimido R., Schmidt E.P., Shapiro N.I. The glycocalyx: a novel diagnostic and therapeutic target in sepsis. Crit. Care. 2019; 23(1): 16. DOI: 10.1186/s13054-018-2292-6
  37. Steppan J., Hofer S., Funke B., et al. Sepsis and major abdominal surgery lead to flaking of the endothelial glycocalyx. J. Surg. Res. 2011; 165: 136–141. DOI: 10.1016/j.jss.2009.04.034
  38. Tracey K.J. The inflammatory reflex. Nature. 2002; 420: 853–859. DOI: 10.1038/nature01321
  39. Tracey K.J. Physiology and immunology of the cholinergic antiinflammatory pathway. J. Clin. Invest. 2007; 117: 289–296. DOI: 10.1172/JCI30555
  40. Григорьев Е.В., Шукевич Д.Л., Плотников Г.П. и др. Нейровоспаление в критических состояниях: механизмы и протективная роль гипотермии. Фундаментальная и клиническая медицина. 2016; 1(3): 88–96. [Grigoryev E.V., Shukevich D.L., Plotnikov G.P., et al. Neuroinflammation in critical care: neuroprotective role role of hypothermia. Fundamental and clinical medicine. 2016; 1(3): 88–96. (In Russ)]
  41. Qin S., Wang H., Yuan R., et al. Role of HMGB1 in apoptosis mediated sepsis lethality. J. Exp. Med. 2006; 203: 1637–1642. DOI: 10.1084/jem.20052203
  42. Lu H., Wen D., Wang X., et al. Host genetic variants in sepsis risk: a field synopsis and meta-analysis. Crit. Care. 2019; 23(1): 26. DOI: 10.1186/s13054-019-2313-0
  43. Thayer J.F., Sternberg E.M. Neural aspects of immunomodulation: focus on the vagus nerve. Brain Behav. Immun. 2010; 24: 1223–1228. DOI: 10.1016/j.bbi.2010.07.247
  44. Karbowski M., Youle R.J. Dynamics of mitochondrial morphology in healthy cells and during apoptosis. Cell Death. Differ. 2003, 10: 870–880. DOI: 10.1038/sj.cdd.4401260
  45. Kuznetsov A.V., Kehrer I., Kozlov A.V., et al. Mitochondrial ROS production under cellular stress: comparison of different detection methods. Anal. Bioanal. Chem. 2011, 400: 2383–2390. DOI: 10.1007/s00216-011-4764-2
  46. Li C., Jackson R.M. Reactive species mechanisms of cellular hypoxia-reoxygenation injury. Am. J. Physiol. Cell Physiol. 2002; 282: 227–241. DOI: 10.1152/ajpcell.00112.2001
  47. Pellegrini M., Baldari C.T. Apoptosis and oxidative stress-related diseases: the p66Shc connection. Curr. Mol. Med. 2009; 9: 392–398. DOI: 10.2174/156652409787847254
  48. Butow R.A., Avadhani N.G. Mitochondrial signalling: the retrograde response. Mol. Cell. 2004, 14: 1–15. DOI: 10.1016/S1097–2765(04)00179–0
  49. Wendel M., Heller A.R. Mitochondrial function and dysfunction in sepsis. Wien. Med. Wochenschr. 2010; 160: 118–123. DOI: 10.1007/s10354-010-0766-5
  50. Basanez G., Zhang J., Chau B.N., et al. Pro-apoptotic cleavage products of Bcl-xL form cytochrome c-conducting pores in pure lipid membranes. J. Biol. Chem. 2001, 276: 31083–31091. DOI: 10.1074/jbc.M103879200
  51. Orrenius S., Gogvadze A., Zhivotovsky B. Mitochondrial oxidative stress: implications for cell death. Annu Rev. Pharmacol. Toxicol. 2007, 47: 143–183. DOI: 10.1146/annurev.pharmtox.47.120505.105122
  52. Glick D., Barth S., Macleod K.F. Autophagy: cellular and molecular mechanisms. J. Pathology. 2010; 221: 3–12. DOI: 10.1002/path.2697
  53. Lee I., Huttemann M. Energy crisis: the role of oxidative phosphorylation in acute inflammation and sepsis. Biochim. Biophys. Acta. 2014; 1842(9): 1579–1586. DOI: 10.1016/j.bbadis.2014.05.031
  54. Merz T.M., Pereira A.J., Schürch R., et al. Mitochondrial function of immune cells in septic shock: A prospective observational cohort study. PLoS One. 2017; 12(6): e0178946. DOI: 10.1371/journal.pone.0178946
  55. Grigoryev E.V., Shukevich D.L., Matveeva V.G., Kornekyuk R.A. Immunosuppression as a component of multiple organ dysfunction syndrome following cardiac surgery. Complex issues of cardiovascular diseases. 2018; 7(4): 84–91. DOI: 10.17802/2306-1278-2018-7-4-84-91
  56. Boomer J.S., Green J.M., Hotchkiss R.S. The changing immune system in sepsis: Is individualized immuno-modulatory therapy the answer? Virulence. 2014; 5(1), 45–56. DOI: 10.4161/viru.26516
  57. Rock K.L., Latz E., Ontiveros F., Kono H. The sterile inflammatory response. Annu Rev. Immunol. 2010; 28: 321–342. DOI: 10.1146/annurev-immunol-030409-101311
  58. Warren O.J., Smith A.J., Alexiou C., et al. The inflammatory response to cardiopulmonary bypass: part 1 — mechanisms of pathogenesis. Journal of cardiothoracic and vascular anaesthesia. 2009; 23(2): 223–231. DOI: 10.1053/j.jvca.2008.08.007
  59. Callahan L.A., Supinski G.S. Sepsis-induced myopathy. Crit. Care Med. 2009; 37(10 Suppl.): 354–367. DOI: 10.1007/s13539-010-0010-6
  60. Hermans G., Van den Berghe G. Clinical review: intensive care unit acquired weakness. Crit. Care. 2015; 19(1): 274. DOI: 10.1186/s13054-015-0993-7
  61. Klaude M., Mori M., Tjader I., et al. Protein metabolism and gene expression in skeletal muscle of critically ill patients with sepsis. Clin. Sci (Lond.). 2012; 122(3): 133–142. DOI: 10.1042/CS20110233
  62. Preiser J.-C. High protein intake during the early phase of critical illness: yes or no? Crit. Care. 2018; 22: 261. DOI: 10.1186/s13054-018-2196-5
  63. Cuenca A.G., Cuenca A.L., Winfield R.D., et al. Novel role for tumor-induced expansion of myeloid-derived cells in cancer cachexia. J. Immunol. 2014; 192(12): 6111–6119. DOI: 10.4049/jimmunol.1302895
  64. Mittal R., Coopersmith C.M. Redefining the gut as the motor of critical illness. Trends Mol. Med. 2014; 20: 214–223. DOI: 10.1016/j.molmed.2013.08.004
  65. Moore F.A., Moore E.E., Poggetti R. Gut bacterial translocation via the portal vein: A clinical perspective with major torso trauma. J. Trauma. 1991; 31: 629–636.
  66. Assimakopoulos S.F., Triantos C., Thomopoulos K., et al. Gut-origin sepsis in the critically ill patient: pathophysiology and treatment. Infection. 2018; 46(6): 751–760. DOI: 10.1007/s15010-018-1178-5
  67. Zahs A., Bird M.D., Ramirez L., et al. Inhibition of long myosin light chain kinase activation alleviates intestinal damage after binge ethanol exposure and burn injury. Am. J. Physiol. Gastrointest Liver Physiol. 2012; 303: G705–G712. DOI: 10.1152/ajpgi.00157.2012
  68. Nathan C., Ding A. Nonresolving inflammation. Cell. 2010; 140 (6): 871–882. DOI: 10.1016/j.cell.2010.02.029
  69. Carcillo J.A., Halstead E.S., Hall M.W., et al. Three Hypothetical Inflammation Pathobiology Phenotypes and Pediatric Sepsis-Induced Multiple Organ Failure Outcome. Pediatric Critical Care Medicine. 2017, 18(6): 513–523. DOI: 10.1097/PCC.0000000000001122
  70. Scicluna B.P., Vught L.A., Zwinderman A.H, et al., on behalf of the MARS consortium. Classification of patients with sepsis according to blood genomic endotype: a prospective cohort study. Lancet Respir. Med. 2017. DOI: 10.1016/S2213–2600(17)30294-1
Лицензия Creative Commons

Это произведение доступно по лицензии Creative Commons «Attribution-NonCommercial-ShareAlike» («Атрибуция — Некоммерческое использование — На тех же условиях») 4.0 Всемирная.