• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 1
  • Tagged with
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
1

Synthesis of Sulfotyrosine Bearing Peptides and Analogues

Ali, Ahmed Magdy Ahmed Mohamed January 2010 (has links)
Sulfation of tyrosine residues is a post-translational modification that occurs on many secretory as well as transmembrane proteins. This modification is believed to be essential for numerous biological processes. However, one of the factors hindering the study of the significance of sulfotyrosine (sTyr) within a protein is the absence of a general method that enables the synthesis of sTyr peptides in satisfactory yields and purity. A general approach to the synthesis of sTyr-bearing peptides was developed in which the sTyr residue is incorporated into the peptides with the sulfate group protected. For the implementation of this general approach a new protecting group for sulfates, namely, the dichlorovinyl (DCV) group was developed. This was accomplished by conducting a careful analysis of the reaction of a trichloroethyl (TCE)-protected sulfate ester with piperidine and 2-methylpiperidine (2-MP). A unique sulfuryl imidazolium reagent, compound 2.22, was also developed that enabled the ready synthesis of DCV-protected sulfates. This reagent was used to prepare the amino acid building block FmocTyr(SO3DCV)OH (2.23). An alternative and more economical synthesis of FmocTyr(SO3DCV)OH (2.23) was also developed that did not require reagent 2.22. Fmoc-based solid phase peptide synthesis (SPPS) was used to incorporate 2.23 into peptides using 2-MP for Fmoc removal. After cleavage of the peptide from the support, the DCV group was removed by hydrogenolysis to give sTyr peptides in good yield and purity. Using this approach a variety of sTyr peptides were prepared including a tetrasulfated 20-mer corresponding to residues 14-33 in chemokine receptor D6 and a disulfated 35-mer corresponding to residues 8-42 of the N-terminus region of the chemokine receptor DARC and this is the largest multisulfated sTyr–bearing peptide made to date. It was also demonstrated that the incorporation of an important non-hydrolyzable sTyr analog, 4-(sulfonomethyl)phenylalanine (Smp), into peptides can be accomplished in good yield by protecting the sulfonate residue with a TCE group during SPPS. This approach was shown to be superior to the previously reported method where the sulfonate group is unprotected. Finally, a number of sulfotyrosine bearing peptides were synthesized and tested as protein tyrosine phosphatase-1B (PTP1B) inhibitors
2

Synthesis of Sulfotyrosine Bearing Peptides and Analogues

Ali, Ahmed Magdy Ahmed Mohamed January 2010 (has links)
Sulfation of tyrosine residues is a post-translational modification that occurs on many secretory as well as transmembrane proteins. This modification is believed to be essential for numerous biological processes. However, one of the factors hindering the study of the significance of sulfotyrosine (sTyr) within a protein is the absence of a general method that enables the synthesis of sTyr peptides in satisfactory yields and purity. A general approach to the synthesis of sTyr-bearing peptides was developed in which the sTyr residue is incorporated into the peptides with the sulfate group protected. For the implementation of this general approach a new protecting group for sulfates, namely, the dichlorovinyl (DCV) group was developed. This was accomplished by conducting a careful analysis of the reaction of a trichloroethyl (TCE)-protected sulfate ester with piperidine and 2-methylpiperidine (2-MP). A unique sulfuryl imidazolium reagent, compound 2.22, was also developed that enabled the ready synthesis of DCV-protected sulfates. This reagent was used to prepare the amino acid building block FmocTyr(SO3DCV)OH (2.23). An alternative and more economical synthesis of FmocTyr(SO3DCV)OH (2.23) was also developed that did not require reagent 2.22. Fmoc-based solid phase peptide synthesis (SPPS) was used to incorporate 2.23 into peptides using 2-MP for Fmoc removal. After cleavage of the peptide from the support, the DCV group was removed by hydrogenolysis to give sTyr peptides in good yield and purity. Using this approach a variety of sTyr peptides were prepared including a tetrasulfated 20-mer corresponding to residues 14-33 in chemokine receptor D6 and a disulfated 35-mer corresponding to residues 8-42 of the N-terminus region of the chemokine receptor DARC and this is the largest multisulfated sTyr–bearing peptide made to date. It was also demonstrated that the incorporation of an important non-hydrolyzable sTyr analog, 4-(sulfonomethyl)phenylalanine (Smp), into peptides can be accomplished in good yield by protecting the sulfonate residue with a TCE group during SPPS. This approach was shown to be superior to the previously reported method where the sulfonate group is unprotected. Finally, a number of sulfotyrosine bearing peptides were synthesized and tested as protein tyrosine phosphatase-1B (PTP1B) inhibitors
3

Engineering and analysis of protease fine specificity via site-directed mutagenesis

Flowers, Crystal Ann 08 October 2013 (has links)
Altering the substrate specificity of proteases is a powerful process with possible applications in many areas of therapeutics as well as proteomics. Although the field is still developing, several proteases have been successfully engineered to recognize novel substrates. Previously in our laboratory, eight highly active OmpT variants were engineered with novel catalytic sites. The present study examined the roles of several residues surrounding the active site of OmpT while attempting to use rational design to modulate fine specificity enough to create a novel protease that prefers phosphotyrosine containing substrates relative to sulfotyrosine or unmodified tyrosine residues. In particular, a previously engineered sulfotyrosine-specific OmpT variant (Varadarajan et al., 2008) was the starting point for rationally designing fifteen new OmpT variants in an attempt to create a highly active protease that would selectively cleave phosphotyrosine substrates. Our design approach was to mimic the most selective phosphoryl-specific enzymes and binding proteins by increasing positive charge around the active site. Sulfonyl esters have a net overall charge of -1 near neutral pH, while phosphate monoesters have a net overall charge of -2. Selected active site residues were mutated by site-directed mutagenesis to lysine, arginine, and histidine. The catalytic activities and substrate specificities of each variant were characterized. Although several variants displayed altered substrate specificity, none preferred phosphotyrosine over sulfotyrosine containing peptides. Taken together, our results have underscored the subtle nature of protease substrate specificity and how elusive it can be to engineer fine specificity. Apparently, phosphotyrosine specific variants were not possible within the context of our starting sulfotyrosine specific OmpT derivative mutated to have single amino acid changes chosen on the basis of differential charge interactions. / text

Page generated in 0.052 seconds