HOMEPRODUCTSCOMPANYCONTACTFAQResearchDictionaryPharmaSign Up FREE or Login

Protein kinase C-delta inhibition protects blood-brain barrier from sepsis-induced vascular damage.

AbstractBACKGROUND:
Neuroinflammation often develops in sepsis leading to activation of cerebral endothelium, increased permeability of the blood-brain barrier (BBB), and neutrophil infiltration. We have identified protein kinase C-delta (PKCδ) as a critical regulator of the inflammatory response and demonstrated that pharmacologic inhibition of PKCδ by a peptide inhibitor (PKCδ-i) protected endothelial cells, decreased sepsis-mediated neutrophil influx into the lung, and prevented tissue damage. The objective of this study was to elucidate the regulation and relative contribution of PKCδ in the control of individual steps in neuroinflammation during sepsis.
METHODS:
The role of PKCδ in mediating human brain microvascular endothelial (HBMVEC) permeability, junctional protein expression, and leukocyte adhesion and migration was investigated in vitro using our novel BBB on-a-chip (B3C) microfluidic assay and in vivo in a rat model of sepsis induced by cecal ligation and puncture (CLP). HBMVEC were cultured under flow in the vascular channels of B3C. Confocal imaging and staining were used to confirm tight junction and lumen formation. Confluent HBMVEC were pretreated with TNF-α (10 U/ml) for 4 h in the absence or presence of PKCδ-i (5 μM) to quantify neutrophil adhesion and migration in the B3C. Permeability was measured using a 40-kDa fluorescent dextran in vitro and Evans blue dye in vivo.
RESULTS:
During sepsis, PKCδ is activated in the rat brain resulting in membrane translocation, a step that is attenuated by treatment with PKCδ-i. Similarly, TNF-α-mediated activation of PKCδ and its translocation in HBMVEC are attenuated by PKCδ-i in vitro. PKCδ inhibition significantly reduced TNF-α-mediated hyperpermeability and TEER decrease in vitro in activated HBMVEC and rat brain in vivo 24 h after CLP induced sepsis. TNF-α-treated HBMVEC showed interrupted tight junction expression, whereas continuous expression of tight junction protein was observed in non-treated or PKCδ-i-treated cells. PKCδ inhibition also reduced TNF-α-mediated neutrophil adhesion and migration across HBMVEC in B3C. Interestingly, while PKCδ inhibition decreased the number of adherent neutrophils to baseline (no-treatment group), it significantly reduced the number of migrated neutrophils below the baseline, suggesting a critical role of PKCδ in regulating neutrophil transmigration.
CONCLUSIONS:
The BBB on-a-chip (B3C) in vitro assay is suitable for the study of BBB function as well as screening of novel therapeutics in real-time. PKCδ activation is a key signaling event that alters the structural and functional integrity of BBB leading to vascular damage and inflammation-induced tissue damage. PKCδ-TAT peptide inhibitor has therapeutic potential for the prevention or reduction of cerebrovascular injury in sepsis-induced vascular damage.
AuthorsYuan Tang, Fariborz Soroush, Shuang Sun, Elisabetta Liverani, Jordan C Langston, Qingliang Yang, Laurie E Kilpatrick, Mohammad F Kiani
JournalJournal of neuroinflammation (J Neuroinflammation) Vol. 15 Issue 1 Pg. 309 (Nov 06 2018) ISSN: 1742-2094 [Electronic] England
PMID30400800 (Publication Type: Journal Article)
Chemical References
  • Peptides
  • Protein Kinase Inhibitors
  • Zonula Occludens-1 Protein
  • Protein Kinase C-delta
Topics
  • Animals
  • Blood-Brain Barrier (drug effects, metabolism, physiopathology)
  • Capillary Permeability (drug effects, physiology)
  • Cell Adhesion (drug effects)
  • Cell Movement (drug effects)
  • Cells, Cultured
  • Disease Models, Animal
  • Endothelial Cells (drug effects, physiology)
  • Humans
  • Male
  • Neutrophil Infiltration (drug effects, physiology)
  • Peptides (pharmacology)
  • Phosphorylation (drug effects)
  • Protein Kinase C-delta (antagonists & inhibitors, metabolism)
  • Protein Kinase Inhibitors (pharmacology)
  • Protein Transport (drug effects)
  • Rats
  • Rats, Sprague-Dawley
  • Sepsis (pathology)
  • Zonula Occludens-1 Protein (metabolism)

Join CureHunter, for free Research Interface BASIC access!

Take advantage of free CureHunter research engine access to explore the best drug and treatment options for any disease. Find out why thousands of doctors, pharma researchers and patient activists around the world use CureHunter every day.
Realize the full power of the drug-disease research graph!


Choose Username:
Email:
Password:
Verify Password:
Enter Code Shown: