Tyrosine sulfation is a post-translational modification mediated by one of two Golgi tyrosylprotein sulfotransferases (TPST-1 and TPST-2), that catalyze the transfer of sulfate from PAPS to lumenally oriented tyrosine residues in secreted and transmembrane proteins. A small number of proteins are known to be tyrosine sulfated in man, including certain adhesion molecules, coagulation factors, serpins, extracellular matrix proteins, and hormones and some of these require sulfation for optimal function. Recently, several G-protein-coupled receptors (GPCRs), including the chemokine receptors CCR2B, CXCR4, CCR5, and CX3CR1, the C5a receptor, and the TSH receptor were shown to be tyrosine sulfated on their N-terminal domains and to require sulfation for optimal binding of their cognate ligands. However, all current data on GPCR sulfation are based on human receptors expressed in cell lines. Thus, the importance of GPCR sulfation in ligand binding in native cells and its relevance in vivo is uncertain. The N-terminal domains of all known chemokine receptors (CCRs) contain tyrosine residues in the context of several flanking acidic amino acids, features shared by all known tyrosine sulfated proteins. This suggests that all CCRs may be tyrosine sulfated and require sulfation for chemokine binding. The goal of this application is to systematically examine whether mouse CCRs are tyrosine sulfated and to explore the role of tyrosine sulfation in CCR function in vitro and in vivo. The following specific aims are proposed.
Aim 1 -Determine if the known CCRs are tyrosine sulfated in the mouse.
Aim 2 - Determine if CCR sulfation is required for optimal function in vitro by assessing chemokine-induced responses of cells derived fromTpst1-/- and Tpst2-/- mice.
Aim 3 - Determine the biological relevance of deficient sulfation of CCRs in vivo by examining biological responses to inflammatory challenges that normally require a particular CCR are abnormal in Tpst1-/- and Tpst2-/- mice. Chemokines and their receptors play crucial roles in a variety of pathophysiological processes, such as bacterial and viral infection (i.e. HIV/AIDS), allergy, atherosclerosis, malignancy, and various autoimmune and chronic inflammatory diseases. These studies will provide new insights into the importance of tyrosine sulfation in innate and adaptive host responses during a wide variety of pathophysiological processes.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
5R01HL074015-03
Application #
6931007
Study Section
Allergy and Immunology Study Section (ALY)
Program Officer
Hasan, Ahmed AK
Project Start
2003-09-29
Project End
2007-08-31
Budget Start
2005-09-01
Budget End
2006-08-31
Support Year
3
Fiscal Year
2005
Total Cost
$309,000
Indirect Cost
Name
Oklahoma Medical Research Foundation
Department
Type
DUNS #
077333797
City
Oklahoma City
State
OK
Country
United States
Zip Code
73104
Westmuckett, Andrew D; Moore, Kevin L (2009) Lack of tyrosylprotein sulfotransferase activity in hematopoietic cells drastically attenuates atherosclerosis in Ldlr-/- mice. Arterioscler Thromb Vasc Biol 29:1730-6
Danan, Lieza M; Yu, Zhihao; Hoffhines, Adam J et al. (2008) Mass spectrometric kinetic analysis of human tyrosylprotein sulfotransferase-1 and -2. J Am Soc Mass Spectrom 19:1459-66
Westmuckett, Andrew D; Hoffhines, Adam J; Borghei, Atefeh et al. (2008) Early postnatal pulmonary failure and primary hypothyroidism in mice with combined TPST-1 and TPST-2 deficiency. Gen Comp Endocrinol 156:145-53
Yu, Yonghao; Hoffhines, Adam J; Moore, Kevin L et al. (2007) Determination of the sites of tyrosine O-sulfation in peptides and proteins. Nat Methods 4:583-8
Borghei, Atefeh; Ouyang, Ying-Bin; Westmuckett, Andrew D et al. (2006) Targeted disruption of tyrosylprotein sulfotransferase-2, an enzyme that catalyzes post-translational protein tyrosine O-sulfation, causes male infertility. J Biol Chem 281:9423-31
Hoffhines, Adam J; Damoc, Eugen; Bridges, Kristie G et al. (2006) Detection and purification of tyrosine-sulfated proteins using a novel anti-sulfotyrosine monoclonal antibody. J Biol Chem 281:37877-87