Sepsis continues to be a leading cause of mortality in the ICU. The root cause of mortality can be traced directly to multisystem organ failure caused by damage to the vasculature that supplies these organs. Vascular damage is due in part to uncontrolled leukocyte interaction with endothelium. The challenge in designing an immuno-centric intervention is to avoid complete suppression. In spite of systemic hyper-inflammation, trials using corticosteroids have failed. A more targeted and deliberate approach is needed. A common feature of the exaggerated inflammatory response during sepsis is the accumulation of activated neutrophils within the microvasculature of organs such as liver, kidney, brain, spleen and lung leading to cell mediated tissue damage and organ failure. The underlying mechanisms that lead to the accumulation of neutrophils in the vasculature with ensuing damage to barrier function are not known. The overarching hypothesis is that protection of the vascular endothelium from the damage induced by the activated neutrophils will facilitate a return to vascular homeostasis and in turn protect the parenchyma from an excessive invasive inflammatory response. We will (1) determine the mechanisms by which the endothelial glycocalyx layer regulates neutrophil-endothelial cell interaction during sepsis, (2) investigate whether neutrophil-derived microparticles protect endothelial barrier function, and (3) investigate whether neutrophil diapedesis contributes to damage of the vascular basement membrane and is associated vascular permeability due to a loss of barrier function. If transmigration and degranulatory activities of neutrophils were controlled without being obviated, a more physiological response to infection/injury would ensue. Thus, understanding how to control vascular damage through manipulation of neutrophil extravasation has potential applications in many acute/chronic inflammatory settings including sepsis.

Public Health Relevance

Severe sepsis, defined as sepsis associated with acute organ dysfunction, is an increasing cause of morbidity and mortality among children and adults, and has been one of the most significant challenges in critical care. The root cause of mortality can be traced directly to multisystem organ failure caused by damage to the vasculature, which is due in part to uncontrolled leukocyte interaction with endothelium. This study will determine the underlying mechanisms that lead to the accumulation of neutrophils in the vasculature with ensuing damage to barrier function. Understanding how to control vascular damage through manipulation of neutrophil extravasation has potential applications in many acute/chronic inflammatory settings including sepsis.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL125265-01
Application #
8799334
Study Section
Special Emphasis Panel (ZHL1-CSR-O (S1))
Program Officer
Sarkar, Rita
Project Start
2014-09-15
Project End
2019-05-31
Budget Start
2014-09-15
Budget End
2015-05-31
Support Year
1
Fiscal Year
2014
Total Cost
$786,272
Indirect Cost
$213,485
Name
University of Rochester
Department
Microbiology/Immun/Virology
Type
Schools of Dentistry
DUNS #
041294109
City
Rochester
State
NY
Country
United States
Zip Code
14627
Dickinson, Catherine M; LeBlanc, Brian W; Edhi, Muhammad M et al. (2018) Leukadherin-1 ameliorates endothelial barrier damage mediated by neutrophils from critically ill patients. J Intensive Care 6:19
Sarangi, Pranita P; Lee, Hyun-Wook; Lerman, Yelena V et al. (2017) Activated Protein C Attenuates Severe Inflammation by Targeting VLA-3high Neutrophil Subpopulation in Mice. J Immunol 199:2930-2936
Johnson, Courtney M; O'Brien, Xian M; Byrd, Angel S et al. (2017) Integrin Cross-Talk Regulates the Human Neutrophil Response to Fungal ?-Glucan in the Context of the Extracellular Matrix: A Prominent Role for VLA3 in the Antifungal Response. J Immunol 198:318-334
O'Brien, Xian M; Biron, Bethany M; Reichner, Jonathan S (2017) Consequences of extracellular trap formation in sepsis. Curr Opin Hematol 24:66-71
Lerman, Yelena V; Kim, Minsoo (2015) Neutrophil migration under normal and sepsis conditions. Cardiovasc Hematol Disord Drug Targets 15:19-28
Lerman, Yelena V; Lim, Kihong; Hyun, Young-Min et al. (2014) Sepsis lethality via exacerbated tissue infiltration and TLR-induced cytokine production by neutrophils is integrin ?3?1-dependent. Blood 124:3515-23