Abstract

BackgroundSystemic inflammation associated with sepsis can induce neuronal hyperexcitability, leading to enhanced seizure predisposition and occurrence. Brain microglia are rapidly activated in response to systemic inflammation and, in this activated state, release multiple cytokines and signaling factors that amplify the inflammatory response and increase neuronal excitability. NADPH oxidase (NOX) enzymes promote microglial activation through the generation of reactive oxygen species (ROS), such as superoxide anion. We hypothesized that NOX isoforms, particularly NOX2, are potential targets for prevention of sepsis-associated seizures.MethodsTo reduce NADPH oxidase 2-derived ROS production, mice with deficits of NOX regulatory subunit/NOX2 organizer p47phox (p47phox−/−) or NOX2 major subunit gp91phox (gp91phox−/−) were used or the NOX2-selective inhibitor diphenyleneiodonium (DPI) was used to treat wild-type (WT) mice. Systemic inflammation was induced by intraperitoneal injection of lipopolysaccharide (LPS). Seizure susceptibility was compared among mouse groups in response to intraperitoneal injection of pentylenetetrazole (PTZ). Brain tissues were assayed for proinflammatory gene and protein expression, and immunofluorescence staining was used to estimate the proportion of activated microglia.ResultsIncreased susceptibility to PTZ-induced seizures following sepsis was significantly attenuated in gp91phox−/− and p47phox−/− mice compared with WT mice. Both gp91phox−/− and p47phox−/− mice exhibited reduced microglia activation and lower brain induction of multiple proconvulsive cytokines, including TNFα, IL-1β, IL-6, and CCL2, compared with WT mice. Administration of DPI following LPS injection significantly attenuated the increased susceptibility to PTZ-induced seizures and reduced both microglia activation and brain proconvulsive cytokine concentrations compared with vehicle-treated controls. DPI also inhibited the upregulation of gp91phox transcripts following LPS injection.ConclusionsOur results indicate that NADPH oxidases contribute to the development of increased seizure susceptibility in mice after sepsis. Pharmacologic inhibition of NOX may be a promising therapeutic approach to reducing sepsis-associated neuroinflammation, neuronal hyperexcitability, and seizures.

Highlights

  • Systemic inflammation associated with sepsis can induce neuronal hyperexcitability, leading to enhanced seizure predisposition and occurrence

  • NADPH oxidase 2 (NOX2) deletion reduces the increased seizure susceptibility to pentylenetetrazole following LPS-induced systemic inflammation To elucidate the role of NOX2 in seizure incidence following systemic inflammation, we induced systemic inflammation in adult (8–9-week-old) wild-type, gp91phox−/−, and p47phox−/− mice with a single dose of LPS via i.p. injection and subsequently determined PTZ

  • The results showed that the seizure duration was 2.14 ± 0.73, 18.00 ± 2.67, 8.14 ± 1.44, and 8.43 ± 1.36 mins, for the vehicle-treated WT, LPStreated WT, LPS-treated gp91phox−/−, and LPS-treated p47phox−/− groups, respectively (Fig. 1d)

Read more

Summary

Introduction

Systemic inflammation associated with sepsis can induce neuronal hyperexcitability, leading to enhanced seizure predisposition and occurrence. It is believed that hyperexcitation of neurons, resulting from the release of signaling factors such as cytokines and reactive oxygen species (ROS), leads to enhanced seizure predisposition [3,4,5] and subsequent neuroplastic changes and neurodegeneration, which may eventually induce a chronic seizure syndrome [6, 7]. Brief peripheral inflammation usually does not damage the mature brain, it can induce transient functional deficits in behavior and an acute inflammatory response in the brain similar in some respects to that in the periphery, including elevation of the same proinflammatory cytokines [9, 10] This induced neuroinflammation can be transient or long-lasting depending on the age at insult [6, 7, 11,12,13]. Such studies indicate that both immature and mature brains can be permanently modified after a single brief episode of systemic inflammation, resulting in increased neuronal excitability and behavior changes

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call