Elsevier

Experimental Neurology

Volume 297, November 2017, Pages 82-91
Experimental Neurology

Research Paper
Endotoxemia induces lung-brain coupling and multi-organ injury following cerebral ischemia-reperfusion

https://doi.org/10.1016/j.expneurol.2017.07.016Get rights and content

Highlights

  • Transient endotoxemia exacerbates CNS injury when combined with global ischemia.

  • Global ischemia and LPS exhibit synergistic effects on PMN activation.

  • Combined 3VO/LPS induces PMN homing to the brain, lung, and non-ischemic organs.

  • Lung-brain coupling modulates post-ischemic injury and innate immune responses.

Abstract

Post-ischemic neurodegeneration remains the principal cause of mortality following cardiac resuscitation. Recent studies have implicated gastrointestinal ischemia in the sepsis-like response associated with the post-cardiac arrest syndrome (PCAS). However, the extent to which the resulting low-grade endotoxemia present in up to 86% of resuscitated patients affects cerebral ischemia-reperfusion injury has not been investigated. Here we report that a single injection of low-dose lipopolysaccharide (50 μg/kg, IP) delivered after global cerebral ischemia (GCI) induces blood-brain barrier permeability, microglial activation, cortical injury, and functional decline in vivo, compared to ischemia alone. And while GCI was sufficient to induce neutrophil (PMN) activation and recruitment to the post-ischemic CNS, minimal endotoxemia exhibited synergistic effects on markers of systemic inflammation including PMN priming, lung damage, and PMN burden within the lung and other non-ischemic organs including the kidney and liver. Our findings predict that acute interventions geared towards blocking the effects of serologically occult endotoxemia in survivors of cardiac arrest will limit delayed neurodegeneration, multi-organ dysfunction and potentially other features of PCAS. This work also introduces lung-brain coupling as a novel therapeutic target with broad effects on innate immune priming and post-ischemic neurodegeneration following cardiac arrest and related cerebrovascular conditions.

Introduction

An estimated 500,000 patients suffer cardiac arrest in the United States annually, and despite the introduction of induced hypothermia, the prognosis for meaningful neurological recovery remains poor in over 80% of cases (Bernard et al., 2002, Hoesch et al., 2008). These dire clinical outcomes reflect the exquisite vulnerability of the central nervous system (CNS) to acute ischemic challenge as well as the delayed effects of reperfusion in the context of systemic injury. Collectively referred to as the post-cardiac arrest syndrome (PCAS), this disorder is characterized by changes in hemostasis and microvascular rheology, adrenal insufficiency with resulting shock physiology, heightened levels of inflammation, and delayed damage to both the CNS and peripheral organs (Adrie et al., 2004, Neumar et al., 2008).

The link between systemic inflammation and poor outcomes in patients presenting with this and other forms of acute neurological injury is well recognized (Becker et al., 2011, Kliper et al., 2013, Rodriguez et al., 2013). In the case of cardiac arrest, tissue ischemia stimulates the activity of transcription factors including hypoxia-inducible factor-1α and nuclear factor-κB that stimulate the production of pro-inflammatory cytokines and enhance other aspects of host immune function (Paradis, 2007, Ridder and Schwaninger, 2009). Ischemic damage also induces a sterile inflammatory response through the release of damage-associated molecular patterns (DAMPs) from the brain and other tissues (Pittman and Kubes, 2013, Tadie et al., 2013, Wang et al., 2015). More recently, studies have revealed detectable levels of endotoxin in the serum of up to 86% of patients within 24–48 h following return of spontaneous circulation (Adrie et al., 2002, Grimaldi et al., 2013, Grimaldi et al., 2015, L'Her et al., 2005). The presence of endotoxin and other pathogen-associated molecular patterns (PAMPs) has been linked with ischemia-induced growth of gram-negative intestinal flora and loss of mucosal barrier function with secondary bacteremia (Adrie et al., 2002, Karhausen et al., 2013, Wang et al., 2012). While endotoxemia has been associated with durable neurocognitive deficits in patients surviving Escherichia coli sepsis (Annane and Sharshar, 2015), the levels of lipopolysaccharide (LPS) detected in cases of sepsis are typically higher than those reported following cardiac arrest (2.6 vs. < 0.6 EU/ml) (Casey et al., 1993, Grimaldi et al., 2015). And while linked to increased risk of shock and organ failure (Grimaldi et al., 2013), the effects and consequences of serologically undetectable endotoxemia on cerebral ischemia-reperfusion injury remain unexplored.

The pathological changes in the brain caused by global cerebral ischemia (GCI) have been widely studied and involve both the cell-autonomous and non-cell-autonomous effects of cardiac arrest (Blomqvist and Wieloch, 1985, Hossmann and Hossmann, 1973). Models of GCI are often used to model delayed neurodegeneration within the CA1 field of the hippocampus; however, additional regions of the brain including the thalamus, basal ganglia, and cortex may also be affected (Deierborg et al., 2008, Thal et al., 2010). And while selective neuron vulnerability is the sine qua non of GCI, associated post-ischemic changes include microgliosis, and damage to the cerebrovasculature characterized by endothelial dysfunction, and loss of blood-brain barrier (BBB) integrity (Yao et al., 2015). Preclinical models of GCI typically involve the isolation of the cerebral and systemic circulation via occlusion of 2 or more extracranial vessels (Ito et al., 2013, Pulsinelli and Brierley, 1979, Thal et al., 2010, Yonekura et al., 2004). While intrinsic CNS mechanisms leading to neuronal loss remain an important therapeutic target (Coultrap et al., 2011), GCI models do not recapitulate multi-organ injury and other features of PCAS. Thus, making headway in identifying therapies to disrupt post-arrest neurological decline will require a better understanding of the complex interplay between the brain's intrinsic response to ischemia and the extrinsic changes in the periphery involving cellular immunity as well as the production of other factors with long-range signaling properties.

To better understand the mechanism(s) underlying catastrophic brain injury in PCAS, we studied the effect of low-dose endotoxin challenge on brain injury in a mouse model of GCI. Our results indicate that serologically undetectable endotoxemia increased levels of cortical injury, microglial activation, microvascular injury, and behavioral compromise beyond what is observed after ischemic injury alone. We also observed marked neutrophil (PMN) activation, lung injury, and the accumulation of PMNs in the brain, lung, and other non-ischemic tissues with combined challenge. Our data suggest that therapies geared towards blocking the effects of systemic endotoxin in patients surviving cardiac arrest may be an effective strategy to limit multi-organ dysfunction commonly observed in the post-resuscitation period. Furthermore, these results implicate lung-brain coupling as an important determinant of post-ischemic neurodegeneration mediated through effects on innate immunity.

Section snippets

Endotoxin titration

All mouse work was performed according to federal regulations with approval by the University Committee on Animal Resources. C57BL/6 mice (5–8-week-old males, 25–30 g, n = 3) received intraperitoneal (IP) injections of either saline (SAL) or LPS (50 μg/kg, 4 mg/kg, or 20 mg/kg; derived from E. coli O111:B4; Sigma-Aldrich, St. Louis, MO) to mimic endotoxin leak during gut injury. LPS doses were tested to determine which yielded serum endotoxin levels comparable to those observed in patients admitted

Physiological and behavioral effects of 3VO/LPS treatment

Our initial goal was to determine which LPS dose(s) would recapitulate levels of endotoxemia observed clinically. Intraperitoneal administration of 4 mg/kg LPS produced peak serum endotoxin levels (0.95 ± 0.14 EU) within 6 h (Fig. 1A). After correcting for sample dilution, this was comparable to levels seen in patients within the first 24–72 h following cardiac arrest (Adrie et al., 2002, Grimaldi et al., 2013). However, treatment with doses as low as 1 mg/kg LPS resulted in significant mortality when

Discussion

In the present study, we tested the hypothesis that occult endotoxemia has important effects on the CNS when associated with global ischemia. Our results revealed three main findings. First, we found that low-dose LPS challenge enhances post-ischemic damage in the CNS with secondary effects on microvascular inflammation, injury to the BBB, and a shift in both microglial and PMN activation. Second, we found that combined 3VO/LPS challenge exerted distinct effects on the activation and homing of

Sources of funding

These studies were supported by the Schmitt Program on Integrative Brain Research and awards from the National Institutes of Health to MWH (R01-NS092455) and the Institutional MSTP training grant (NIH/T32-GM007356) and fellowship funding to NM (F30-NS092168) from the NIH.

Conflicts of interest/disclosures

None.

Acknowledgements

The authors would like to thank Minsoo Kim, Yelena Lerman, and Alison Gaylo for their assistance in conducting flow cytometric measurements of neutrophil activation, Rupal Mehta and Orestes Solis for hematoxylin and eosin staining of peripheral organs, Craig Morrell for assistance with granulocyte counts, and Kevin Mazurek for assistance with MATLAB.

References (71)

  • I.K. Hwang et al.

    Ischemia-induced changes of platelet endothelial cell adhesion molecule-1 in the hippocampal ca1 region in gerbils

    Brain Res.

    (2005)
  • S.T. Kocjancic et al.

    Impact of intensified postresuscitation treatment on outcome of comatose survivors of out-of-hospital cardiac arrest according to initial rhythm

    Resuscitation

    (2014)
  • J. Kofler et al.

    Histopathological and behavioral characterization of a novel model of cardiac arrest and cardiopulmonary resuscitation in mice

    J. Neurosci. Methods

    (2004)
  • E. L'Her et al.

    Gut dysfunction and endoscopic lesions after out-of-hospital cardiac arrest

    Resuscitation

    (2005)
  • D.A. Partrick et al.

    Cardiopulmonary bypass renders patients at risk for multiple organ failure via early neutrophil priming and late neutrophil disability

    J. Surg. Res.

    (1999)
  • L.S. Rallidis et al.

    Inflammatory markers and in-hospital mortality in acute ischaemic stroke

    Atherosclerosis

    (2006)
  • S.F. Ribaric et al.

    Prophylactic versus clinically-driven antibiotics in comatose survivors of out-of-hospital cardiac arrest-a randomized pilot study

    Resuscitation

    (2017)
  • D.A. Ridder et al.

    Nf-kappab signaling in cerebral ischemia

    Neuroscience

    (2009)
  • J.A. Rodriguez et al.

    Prometalloproteinase-10 is associated with brain damage and clinical outcome in acute ischemic stroke

    J. Thromb. Haemost.

    (2013)
  • L.C. Schmued et al.

    Fluoro-jade c results in ultra high resolution and contrast labeling of degenerating neurons

    Brain Res.

    (2005)
  • S.C. Thal et al.

    Characterization of a 3-vessel occlusion model for the induction of complete global cerebral ischemia in mice

    J. Neurosci. Methods

    (2010)
  • N. Ullrich et al.

    The temporo-spatial localization of polymorphonuclear cells related to the neurovascular unit after transient focal cerebral ischemia

    Brain Res.

    (2014)
  • J. Vaahersalo et al.

    Admission interleukin-6 is associated with post resuscitation organ dysfunction and predicts long-term neurological outcome after out-of-hospital ventricular fibrillation

    Resuscitation

    (2014)
  • C. Wang et al.

    High mobility group box 1 contributes to anti-neutrophil cytoplasmic antibody-induced neutrophils activation through receptor for advanced glycation end products (rage) and toll-like receptor 4

    Arthritis Res. Ther.

    (2015)
  • H. Xaio et al.

    Effect of lps on the permeability of the blood-brain barrier to insulin

    Brain Res.

    (2001)
  • X. Yao et al.

    Reduced brain edema and infarct volume in aquaporin-4 deficient mice after transient focal cerebral ischemia

    Neurosci. Lett.

    (2015)
  • B.S. Abella et al.

    Intra-arrest cooling improves outcomes in a murine cardiac arrest model

    Circulation

    (2004)
  • E. Abraham

    Neutrophils and acute lung injury

    Crit. Care Med.

    (2003)
  • C. Adrie et al.

    Successful cardiopulmonary resuscitation after cardiac arrest as a “sepsis-like” syndrome

    Circulation

    (2002)
  • C. Adrie et al.

    Postresuscitation disease after cardiac arrest: a sepsis-like syndrome?

    Curr. Opin. Crit. Care

    (2004)
  • M. Aoyama-Ishikawa et al.

    Intravenous immunoglobulin-induced neutrophil apoptosis in the lung during murine endotoxemia

    Surg. Infect.

    (2014)
  • K.J. Becker et al.

    Autoimmune responses to the brain after stroke are associated with worse outcome

    Stroke

    (2011)
  • S.A. Bernard et al.

    Treatment of comatose survivors of out-of-hospital cardiac arrest with induced hypothermia

    N. Engl. J. Med.

    (2002)
  • P. Blomqvist et al.

    Ischemic brain damage in rats following cardiac arrest using a long-term recovery model

    J. Cereb. Blood Flow Metab.

    (1985)
  • R.D. Cardiff et al.

    Manual hematoxylin and eosin staining of mouse tissue sections

    Cold Spring Harb Protoc

    (2014)
  • Cited by (21)

    • Effects of 9-t-butyl doxycycline on the innate immune response to CNS ischemia-reperfusion injury

      2021, Experimental and Molecular Pathology
      Citation Excerpt :

      Our decision to focus on cortical immune transmigration three days following reperfusion was based on several considerations. Comparable to the initial report on the 3VO model by Thal et al. (Thal et al., 2010), the 3VO/LPS paradigm produced consisetent injury across CA1, the cortex, and putamen (Mai et al., 2017). PMN transmigration to the cortex was more robust relative to that seen in the hippocampus, which likely reflects the effect of basilar occlusion on perfusion through the transverse and longitudinal hippocampal arteries (Xiong et al., 2017).

    • Neuroinflammation, Resolution, and Neuroprotection in the Brain

      2021, Neuroinflammation, Resolution, and Neuroprotection in the Brain
    • Lung SOD3 limits neurovascular reperfusion injury and systemic immune activation following transient global cerebral ischemia

      2020, Journal of Stroke and Cerebrovascular Diseases
      Citation Excerpt :

      In all cases, the investigator conducting quantitative image analyses was blinded to the background genotype and treatment arm by coding both video sequences, slides and representative images. To investigate the role of lung inflammation on cerebral reperfusion injury, we used a model of ischemia-reperfusion that combines the immune priming effects of damage-associated molecules released following transient global ischemia with LPS-induced systemic inflammation.21 Mice were randomized into SHAM/SAL and 3VO/LPS groups with LPS delivered by IP injection following cerebral reperfusion (Fig. 1A).

    • Characterization of neutrophil-neuronal co-cultures to investigate mechanisms of post-ischemic immune-mediated neurotoxicity

      2020, Journal of Neuroscience Methods
      Citation Excerpt :

      However, we selected endotoxin as a PMN priming agent for these studies because it has been previously detected in the serum of up to 86 % of cardiac arrest patients within 24–48 h of resuscitation (L’Her et al., 2005; Grimaldi et al., 2015; Adrie et al., 2002). We had also shown that low-dose LPS priming produced sustained systemic neutrophil activation, CNS transmigration, and neurovascular injury in a mouse model of the post cardiac arrest syndrome (PCAS) (Mai et al., 2017). It is important to note that the dose of LPS used in the current and past work was undetectable in serum (<0.01 ng endotoxin/mL) after IP injection (25 μg/kg) at 1/1000th of the dose typically reported in mouse models of sepsis.

    • Post-cardiac arrest syndrome

      2020, AACN Advanced Critical Care
    View all citing articles on Scopus
    View full text