Translational evidence for two distinct patterns of neuroaxonal injury in sepsis: a longitudinal, prospective translational study

Johannes Ehler, Lucinda K Barrett, Valerie Taylor, Michael Groves, Francesco Scaravilli, Matthias Wittstock, Stephan Kolbaske, Annette Grossmann, Jörg Henschel, Martin Gloger, Tarek Sharshar, Fabrice Chretien, Francoise Gray, Gabriele Nöldge-Schomburg, Mervyn Singer, Martin Sauer, Axel Petzold, Johannes Ehler, Lucinda K Barrett, Valerie Taylor, Michael Groves, Francesco Scaravilli, Matthias Wittstock, Stephan Kolbaske, Annette Grossmann, Jörg Henschel, Martin Gloger, Tarek Sharshar, Fabrice Chretien, Francoise Gray, Gabriele Nöldge-Schomburg, Mervyn Singer, Martin Sauer, Axel Petzold

Abstract

Background: Brain homeostasis deteriorates in sepsis, giving rise to a mostly reversible sepsis-associated encephalopathy (SAE). Some survivors experience chronic cognitive dysfunction thought to be caused by permanent brain injury. In this study, we investigated neuroaxonal pathology in sepsis.

Methods: We conducted a longitudinal, prospective translational study involving (1) experimental sepsis in an animal model; (2) postmortem studies of brain from patients with sepsis; and (3) a prospective, longitudinal human sepsis cohort study at university laboratory and intensive care units (ICUs). Thirteen ICU patients with septic shock, five ICU patients who died as a result of sepsis, fourteen fluid-resuscitated Wistar rats with fecal peritonitis, eleven sham-operated rats, and three human and four rat control subjects were included. Immunohistologic and protein biomarker analysis were performed on rat brain tissue at baseline and 24, 48, and 72 h after sepsis induction and in sham-treated rats. Immunohistochemistry was performed on human brain tissue from sepsis nonsurvivors and in control patients without sepsis. The clinical diagnostics of SAE comprised longitudinal clinical data collection and magnetic resonance imaging (MRI) and electroencephalographic assessments. Statistical analyses were performed using SAS software (version 9.4; SAS Institute, Inc., Cary, NC, USA). Because of non-Gaussian distribution, the nonparametric Wilcoxon test general linear models and the Spearman correlation coefficient were used.

Results: In postmortem rat and human brain samples, neurofilament phosphoform, β-amyloid precursor protein, β-tubulin, and H&E stains distinguished scattered ischemic lesions from diffuse neuroaxonal injury in septic animals, which were absent in controls. These two patterns of neuroaxonal damage were consistently found in septic but not control human postmortem brains. In experimental sepsis, the time from sepsis onset correlated with tissue neurofilament levels (R = 0.53, p = 0.045) but not glial fibrillary acidic protein. Of 13 patients with sepsis who had clinical features of SAE, MRI detected diffuse axonal injury in 9 and ischemia in 3 patients.

Conclusions: Ischemic and diffuse neuroaxonal injury to the brain in experimental sepsis, human postmortem brains, and in vivo MRI suggest these two distinct lesion types to be relevant. Future studies should be focused on body fluid biomarkers to detect and monitor brain injury in sepsis. The relationship of neurofilament levels with time from sepsis onset may be of prognostic value.

Trial registration: ClinicalTrials.gov, NCT02442986 . Registered on May 13, 2015.

Keywords: Animal models; Biomarkers; Encephalopathy; Intermediate filaments; Rats; SAE; Sepsis; Sepsis-associated encephalopathy.

Conflict of interest statement

Authors’ information

Not applicable.

Ethics approval and consent to participate

All experiments were performed according to the University College London Ethics Committee and Home Office (UK) guidelines under the 1986 Scientific Procedures Act. The study was approved by the local ethics board at Rostock University (A 2012-0058) and by the Comité Consultatif de Protection des Personnes se Prêtant à la Recherche Biomédicale de Saint Germain en Laye, France. All patients or their legal representatives signed written informed consent forms before study inclusion.

Consent for publication

Not applicable.

Competing interests

AP is supported by the Moorfields Biomedical Research Centre and the Dutch MS Research Foundation and received honoraria from Novartis for quality control reading (PASSOS study). The other authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Brain lesions seen in rat sepsis model. a Central brain white matter immunohistochemistry in sham-treated animals (controls) shows characteristic neuronal soma with restricted staining for β-amyloid precursor protein (βAPP) (arrows). b In septic animals, brain tissue from the same locations showed abnormal and more widespread axonal staining extending from the axonal hillock to the proximal axon (arrows). c Abnormal axonal βAPP staining follows white matter tracts (arrows). d There are also pockets of inflammatory and ischemic brain lesions seen in the rat sepsis model (H&E stain; arrow). e Staining of such lesions shows intense neuronal and axonal staining for βAPP (arrows). f Staining of lesions of sham-treated animals for β-tubulin in the magnification field is crisp and shows integrity of the neuroaxonal compartment (overview, inset; original magnification × 10). g Likewise, the integrity of white matter tracts in sham-treated animals can be seen (×10; inset, original magnification × 40). h There is severe structural disorganization of the β-tubulin network in white matter tracts of the septic animals. i The level of structural β-tubulin disorganization in the septic rat brain is best observed at greater magnification (original magnification × 40) of the neuroaxonal compartment from the same location as that taken from the sham model shown in (f)
Fig. 2
Fig. 2
Brain lesions seen in postmortem human tissue. a Patient 1 (control): Diffuse staining of axons (arrows), extending from the axonal hillock (β-amyloid precursor protein [βAPP]). b Extensive diffuse axonal injury is shown in patient 2 (sepsis). Staining for βAPP is not restricted to the axonal hillock but is seen throughout the white matter tracts. Multiple axonal endbulbs can also be seen (small arrowheads). c Disruption of the deep white matter axons and the presence of axonal end bulbs are widespread based on dephosphorylated neurofilament heavy chain (SMI32). d Patient 3: Small areas of ischemic lesions can be seen throughout the brain (βAPP). e Patient 4: One type of lesion not observed in the animal model is shown. Amyloid plaques (arrow) are present and scattered throughout the brain tissue (βAPP). f In this patient, diffuse deep white matter axonal damage (arrows) is the most severe of this series (βAPP)
Fig. 3
Fig. 3
Brain magnetic resonance imaging of three patients during septic shock. The images are fluid-attenuated inversion recovery (FLAIR; a, c, and e) and echo planar imaging diffusion-weighted imaging (DWI; b, d, and f) scans. a and b An 81-year-old male patient with urosepsis. a Axial FLAIR image obtained on day 5 after the onset of septic shock shows punctiform and confluent white matter hyperintensities (WMH) in both paraventricular and paramedian regions (grade 2 leukoencephalopathy). b DWI study shows subacute ischemic lesion in the left occipital paramedian region. c and d An 80-year-old female patient with urosepsis. (c) Axial FLAIR image obtained 9 days after the onset of septic shock shows confluent WMH in the left periventricular region (grade 2 leukoencephalopathy). d DWI study shows bilateral ischemic lesions in the frontal region. e and f An 80-year-old female patient with urosepsis. e Axial FLAIR performed 8 days after onset of septic shock revealing a single punctiform WMH in the left periventricular region (grade 1 leukoencephalopathy). f DWI study shows punctiform ischemic lesions in the left occipital and parietal (inset) regions

References

    1. Brun-Buisson C, Doyon F, Carlet J, et al. Incidence, risk factors, and outcome of severe sepsis and septic shock in adults: a multicenter prospective study in intensive care units. JAMA. 1995;274:968–74. doi: 10.1001/jama.1995.03530120060042.
    1. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) JAMA. 2016;315:801–10. doi: 10.1001/jama.2016.0287.
    1. Iwashyna TJ, Ely EW, Smith DM, et al. Long-term cognitive impairment and functional disability among survivors of severe sepsis. JAMA. 2010;304:1787–94. doi: 10.1001/jama.2010.1553.
    1. Petzold A, Downie P, Smith M. Critical illness brain syndrome (CIBS): an underestimated entity? Crit Care Med. 2005;33:1464. doi: 10.1097/01.CCM.0000166866.47020.E5.
    1. Tauber SC, Eiffert H, Brück W, et al. Septic encephalopathy and septic encephalitis. Expert Rev Anti Infect Ther. 2017;15:121–32. doi: 10.1080/14787210.2017.1265448.
    1. Flierl MA, Rittirsch D, Huber-Lang MS, et al. Pathophysiology of septic encephalopathy - an unsolved puzzle. Crit Care. 2010;14:165. doi: 10.1186/cc9035.
    1. Sonneville R, Verdonk F, Rauturier C, et al. Understanding brain dysfunction in sepsis. Ann Intensive Care. 2013;3:15. doi: 10.1186/2110-5820-3-15.
    1. Semmler A, Frisch C, Debeir T, et al. Long-term cognitive impairment, neuronal loss and reduced cortical cholinergic innervation after recovery from sepsis in a rodent model. Exp Neurol. 2007;204:733–40. doi: 10.1016/j.expneurol.2007.01.003.
    1. Comim CM, Cassol-Jr OJ, Constantino LS, et al. Alterations in inflammatory mediators, oxidative stress parameters and energetic metabolism in the brain of sepsis survivor rats. Neurochem Res. 2011;36:304–11. doi: 10.1007/s11064-010-0320-2.
    1. Steckert AV, Comim CM, Mina F, et al. Late brain alterations in sepsis-survivor rats. Synapse. 2013;67:786–93. doi: 10.1002/syn.21686.
    1. Weberpals M, Hermes M, Hermann S, et al. NOS2 gene deficiency protects from sepsis-induced long-term cognitive deficits. J Neurosci. 2009;29:14177–84. doi: 10.1523/JNEUROSCI.3238-09.2009.
    1. Semmler A, Okulla T, Sastre M, et al. Systemic inflammation induces apoptosis with variable vulnerability of different brain regions. J Chem Neuroanat. 2005;30:144–57. doi: 10.1016/j.jchemneu.2005.07.003.
    1. d’Avila JC, Santiago AP, Amâncio RT, et al. Sepsis induces brain mitochondrial dysfunction. Crit Care Med. 2008;36:1925–32. doi: 10.1097/CCM.0b013e3181760c4b.
    1. Schwalm MT, Pasquali M, Miguel SP, et al. Acute brain inflammation and oxidative damage are related to long-term cognitive deficits and markers of neurodegeneration in sepsis-survivor rats. Mol Neurobiol. 2014;49:380–5. doi: 10.1007/s12035-013-8526-3.
    1. Haapasalo A, Viswanathan J, Bertram L, et al. Emerging role of Alzheimer’s disease-associated ubiquilin-1 in protein aggregation. Biochem Soc Trans. 2010;38:150–5. doi: 10.1042/BST0380150.
    1. Gentleman SM, Nash MJ, Sweeting CJ, et al. β-Amyloid precursor protein (βAPP) as a marker for axonal injury after head injury. Neurosci Lett. 1993;160:139–44. doi: 10.1016/0304-3940(93)90398-5.
    1. Hoshino S, Kobayashi S, Furukawa T, et al. Multiple immunostaining methods to detect traumatic axonal injury in the rat fluid-percussion brain injury model. Neurol Med Chir (Tokyo) 2003;43:165–73. doi: 10.2176/nmc.43.165.
    1. Oehmichen M, Meissner C, Schmidt V, et al. Axonal injury—a diagnostic tool in forensic neuropathology? A review. Forensic Sci Int. 1998;95:67–83. doi: 10.1016/S0379-0738(98)00075-9.
    1. Hosokawa K, Gaspard N, Su F, et al. Clinical neurophysiological assessment of sepsis-associated brain dysfunction: a systematic review. Crit Care. 2014;18:674. doi: 10.1186/s13054-014-0674-y.
    1. Oddo M, Taccone FS. How to monitor the brain in septic patients? Minerva Anestesiol. 2015;81:776–88.
    1. Sharshar T, Carlier R, Bernard F, et al. Brain lesions in septic shock: a magnetic resonance imaging study. Intensive Care Med. 2007;33:798–806. doi: 10.1007/s00134-007-0598-y.
    1. Finelli PF, Uphoff DF. Magnetic resonance imaging abnormalities with septic encephalopathy. J Neurol Neurosurg Psychiatry. 2004;75:1189–91. doi: 10.1136/jnnp.2003.030833.
    1. Zenaide PV, Gusmao-Flores D. Biomarkers in septic encephalopathy: a systematic review of clinical studies. Rev Bras Ter Intensiva. 2013;25:56–62. doi: 10.1590/S0103-507X2013000100011.
    1. Dal-Pizzol F, Tomasi CD, Ritter C. Septic encephalopathy: does inflammation drive the brain crazy? Rev Bras Psiquiatr. 2014;36:251–8. doi: 10.1590/1516-4446-2013-1233.
    1. Barrett LK, Orie NN, Taylor V, et al. Differential effects of vasopressin and norepinephrine on vascular reactivity in a long-term rodent model of sepsis. Crit Care Med. 2007;35:2337–43. doi: 10.1097/01.CCM.0000281861.72907.17.
    1. Petzold A, Keir G, Green AJ, et al. A specific ELISA for measuring neurofilament heavy chain phosphoforms. J Immunol Methods. 2003;278:179–90. doi: 10.1016/S0022-1759(03)00189-3.
    1. Petzold A, Keir G, Green AJ, et al. An ELISA for glial fibrillary acidic protein. J Immunol Methods. 2004;287:169–77. doi: 10.1016/j.jim.2004.01.015.
    1. Dellinger RP, Levy MM, Rhodes A, et al. Surviving Sepsis Campaign: International Guidelines for Management of Severe Sepsis and Septic Shock: 2012. Crit Care Med. 2013;41:580–637. doi: 10.1097/CCM.0b013e31827e83af.
    1. Vincent JL, Moreno R, Takala J, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. Intensive Care Med. 1996;22:707–10. doi: 10.1007/BF01709751.
    1. Knaus WA, Draper EA, Wagner DP, et al. APACHE II: a severity of disease classification system. Crit Care Med. 1985;13:818–29. doi: 10.1097/00003246-198510000-00009.
    1. Guenther U, Popp J, Koecher L, et al. Validity and reliability of the CAM-ICU Flowsheet to diagnose delirium in surgical ICU patients. J Crit Care. 2010;25:144–51. doi: 10.1016/j.jcrc.2009.08.005.
    1. Khanal K, Bhandari SS, Shrestha N, et al. Comparison of outcome predictions by the Glasgow Coma Scale and the Full Outline of UnResponsiveness score in the neurological and neurosurgical patients in the intensive care unit. Indian J Crit Care Med. 2016;20:473–6. doi: 10.4103/0972-5229.188199.
    1. Truman B, Ely EW. Monitoring delirium in critically ill patients: using the Confusion Assessment Method for the Intensive Care Unit. Crit Care Nurse. 2003;23:25–36.
    1. Sessler CN, Gosnell MS, Grap MJ, et al. The Richmond Agitation-Sedation Scale: validity and reliability in adult intensive care unit patients. Am J Respir Crit Care Med. 2002;166:1338–44. doi: 10.1164/rccm.2107138.
    1. Schefold JC, Bierbrauer J, Weber-Carstens S. Intensive care unit-acquired weakness (ICUAW) and muscle wasting in critically ill patients with severe sepsis and septic shock. J Cachexia Sarcopenia Muscle. 2010;1:147–57. doi: 10.1007/s13539-010-0010-6.
    1. Stevens RD, Marshall SA, Cornblath DR, et al. A framework for diagnosing and classifying intensive care unit-acquired weakness. Crit Care Med. 2009;37(10 Suppl):S299–308. doi: 10.1097/CCM.0b013e3181b6ef67.
    1. Young GB, Bolton CF, Archibald YM, et al. The electroencephalogram in sepsis-associated encephalopathy. J Clin Neurophysiol. 1992;9:145–52. doi: 10.1097/00004691-199201000-00016.
    1. Schmidt R, Fazekas F, Kleinert G, et al. Magnetic resonance imaging signal hyperintensities in the deep and subcortical white matter: a comparative study between stroke patients and normal volunteers. Arch Neurol. 1992;49:825–7. doi: 10.1001/archneur.1992.00530320049011.
    1. Messaris E, Memos N, Chatzigianni E, et al. Time-dependent mitochondrial-mediated programmed neuronal cell death prolongs survival in sepsis. Crit Care Med. 2004;32:1764–70. doi: 10.1097/01.CCM.0000135744.30137.B4.
    1. Sharshar T, Gray F, Poron F, et al. Multifocal necrotizing leukoencephalopathy in septic shock. Crit Care Med. 2002;30:2371–5. doi: 10.1097/00003246-200210000-00031.
    1. Sharshar T, Annane D, de la Grandmaison GL, et al. The neuropathology of septic shock. Brain Pathol. 2004;14:21–33. doi: 10.1111/j.1750-3639.2004.tb00494.x.
    1. Petzold A, Groves M, Leis AA, et al. Neuronal and glial cerebrospinal fluid protein biomarkers are elevated after West Nile virus infection. Muscle Nerve. 2010;41:42–9. doi: 10.1002/mus.21448.
    1. Bacioglu M, Maia LF, Preische O, et al. Neurofilament light chain in blood and CSF as marker of disease progression in mouse models and in neurodegenerative diseases. Neuron. 2016;91:494–6. doi: 10.1016/j.neuron.2016.07.007.
    1. Morandi A, Rogers BP, Gunther ML, et al. The relationship between delirium duration, white matter integrity, and cognitive impairment in intensive care unit survivors as determined by diffusion tensor imaging: the VISIONS prospective cohort magnetic resonance imaging study. Crit Care Med. 2012;40:2182–9. doi: 10.1097/CCM.0b013e318250acdc.
    1. Sen J, Belli A, Petzold A, et al. Extracellular fluid S100B in the injured brain: a future surrogate marker of acute brain injury? Acta Neurochir (Wien) 2005;147:897–900. doi: 10.1007/s00701-005-0526-2.
    1. Calvi MR, Rosa CM, Beretta L, et al. Early prognosis after severe traumatic brain injury with minor or absent computed tomography scan lesions. J Trauma. 2011;70:447–51. doi: 10.1097/TA.0b013e3182095e14.
    1. Ma Y, Ouyang B, Guan X. Use of quantitative electroencephalogram in patients with septic shock [in Chinese] Zhonghua Yi Xue Za Zhi. 2016;96:195–8.
    1. Terry KJ, Anger KE, Szumita PM. Prospective evaluation of unable-to-assess CAM-ICU documentations of critically ill patients. J Intensive Care. 2015;3:52. doi: 10.1186/s40560-015-0119-y.
    1. Petzold A. Glial fibrillary acidic protein is a body fluid biomarker for glial pathology in human disease. Brain Res. 2015;1600:17–31. doi: 10.1016/j.brainres.2014.12.027.
    1. Rundgren M, Friberg H, Cronberg T, et al. Serial soluble neurofilament heavy chain in plasma as a marker of brain injury after cardiac arrest. Crit Care. 2012;16:R45. doi: 10.1186/cc11244.
    1. Huisman TA, Schwamm LH, Schaefer PW, et al. Diffusion tensor imaging as potential biomarker of white matter injury in diffuse axonal injury. AJNR Am J Neuroradiol. 2004;25:370–6.
    1. Gunther ML, Morandi A, Krauskopf E, et al. The association between brain volumes, delirium duration, and cognitive outcomes in intensive care unit survivors: the VISIONS cohort magnetic resonance imaging study. Crit Care Med. 2012;40:2022–32. doi: 10.1097/CCM.0b013e318250acc0.

Source: PubMed

3
Subskrybuj