Chemotaxis of leukocytes into various tissues is known to be a critical step in the pathogenesis of several inflammatory disorders. Complement pro-inflammatory peptides C5a and C5a des Arg also have been implicated in disease pathogenesis. C5-derived peptides are very potent chemoattractants for a wide variety of cell types. Much is known about the bioactivities of C5-derived peptides but the regulation of these functions is poorly understood. Previously, we were the first of several groups to demonstrate that the vitamin D binding protein (DBP), also known as Gc-globulin, can enhance the chemotactic activity of C5a and C5a des Arg, i.e., function as a co-chemotaxin. Moreover, the co-chemotactic activity of DBP is specific for the C5-derived peptides. Although DBP appears to be a physiologically important regulator of the chemotactic activity for activated complement, the mechanism of chemotaxis enhancement by DBP is not known. Recently, we have reported several important observations that should help define the mechanism by which DBP acts as a co-chemotactic factor for C5a. (1) DBP needs to be bound to the cell surface in order to function as a co-chemotaxin for C5a. (2) The neutrophil DBP binding site is a chondroitin sulfate proteoglycan. (3) Expression of the DBP binding site is regulated by cell surface-bound neutrophil elastase, which cleaves and sheds the proteoglycan. (4) Preliminary studies have shown that activated platelets modify DBP to an active co-chemotactic form. (5) Clinical samples from patients with inflammatory disorder (ARDS) contain the modified co-chemotactic form of DBP. It is our hypothesis that a modified form of DBP binds to the cell surface and initiates an enhanced chemotactic response to C5a. In this proposal, we endeavor to investigate the mechanism by which DBP augments the leukocyte chemotactic activity of C5a by utilizing human neutrophils and the U937 cell line transfected with the C5a receptor (U937-C5aR). The process of co-chemotaxis will be divided into component parts and examined individually. First, determine how activated platelets modify DBP by focusing on the most likely alteration: extracellular phosphorylation by ubiquitous protein kinases. Second, investigate how modified DBP interacts with cells by examining binding and shedding on the cell surface. Third, determine how cells terminate the co-chemotactic signal by focusing on dephosphorylation by phosphatases. Finally, clinical samples from patients with inflammatory disorders will be analyzed using a proteomic approach to determine if modified DBP is correlated with disease outcome. This study will bridge basic knowledge derived from in vitro biochemical approaches and apply it to examine samples obtained from patients with inflammatory disorders. Results of this study will demonstrate a novel mechanism for a chemotactic cofactor and could serve as a prototype for other cofactors yet to be discovered.

Agency
National Institute of Health (NIH)
Institute
National Institute of General Medical Sciences (NIGMS)
Type
Research Project (R01)
Project #
1R01GM063769-01
Application #
6365025
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Marino, Pamela
Project Start
2001-07-01
Project End
2006-06-30
Budget Start
2001-07-01
Budget End
2002-06-30
Support Year
1
Fiscal Year
2001
Total Cost
$255,850
Indirect Cost
Name
State University New York Stony Brook
Department
Pathology
Type
Schools of Medicine
DUNS #
804878247
City
Stony Brook
State
NY
Country
United States
Zip Code
11794
Kew, Richard R; Tabrizian, Tahmineh; Vosswinkel, James A et al. (2018) Vitamin D-binding protein deficiency in mice decreases systemic and select tissue levels of inflammatory cytokines in a murine model of acute muscle injury. J Trauma Acute Care Surg 84:847-854
Ramadass, Mahalakshmi; Ghebrehiwet, Berhane; Kew, Richard R (2015) Enhanced recognition of plasma proteins in a non-native state by complement C3b. A possible clearance mechanism for damaged proteins in blood. Mol Immunol 64:55-62
Penzo, Marianna; Habiel, David M; Ramadass, Mahalakshmi et al. (2014) Cell migration to CXCL12 requires simultaneous IKK? and IKK?-dependent NF-?B signaling. Biochim Biophys Acta 1843:1796-1804
Ramadass, Mahalakshmi; Ghebrehiwet, Berhane; Smith, Richard J et al. (2014) Generation of multiple fluid-phase C3b:plasma protein complexes during complement activation: possible implications in C3 glomerulopathies. J Immunol 192:1220-30
Ge, Lingyin; Trujillo, Glenda; Miller, Edmund J et al. (2014) Circulating complexes of the vitamin D binding protein with G-actin induce lung inflammation by targeting endothelial cells. Immunobiology 219:198-207
Ghebrehiwet, Berhane; Ji, Yan; Valentino, Alisa et al. (2014) Soluble gC1qR is an autocrine signal that induces B1R expression on endothelial cells. J Immunol 192:377-84
Trujillo, Glenda; Habiel, David M; Ge, Lingyin et al. (2013) Neutrophil recruitment to the lung in both C5a- and CXCL1-induced alveolitis is impaired in vitamin D-binding protein-deficient mice. J Immunol 191:848-56
Kew, Richard R; Penzo, Marianna; Habiel, David M et al. (2012) The IKK?-dependent NF-?B p52/RelB noncanonical pathway is essential to sustain a CXCL12 autocrine loop in cells migrating in response to HMGB1. J Immunol 188:2380-6
Trujillo, Glenda; Zhang, Jianhua; Habiel, David M et al. (2011) Cofactor regulation of C5a chemotactic activity in physiological fluids. Requirement for the vitamin D binding protein, thrombospondin-1 and its receptors. Mol Immunol 49:495-503
Guerriero, Jennifer L; Ditsworth, Dara; Catanzaro, Joseph M et al. (2011) DNA alkylating therapy induces tumor regression through an HMGB1-mediated activation of innate immunity. J Immunol 186:3517-26

Showing the most recent 10 out of 18 publications