Vis enkel innførsel

dc.contributor.advisorEngh, Richard
dc.contributor.authorAlam, Kazi Asraful
dc.date.accessioned2017-06-27T11:09:38Z
dc.date.available2017-06-27T11:09:38Z
dc.date.issued2017-06-09
dc.description.abstractProtein kinases are involved in many essential cellular processes, and are regulated in a dynamic manner by the movement of domains or motifs via interaction with various proteins and substrates. Protein kinase deregulation can lead to a variety of diseases, including cancer and diabetes. Protein kinase A (PKA) has been a prototype to study the entire family, including for studies in drug discovery research. However, the high sequence similarities in the kinase domain of protein kinases hinders the development of target-specific inhibitors, and the use of PKA alone is insufficient. Mutants of PKA can act as surrogate targets to aid the design of target specificity. In this work, we use a combination of several biophysical methods to investigate the properties of PKA, PKA based surrogate kinases (PKAB3 & PKA-Au6), the cancer target Aurora kinase, including especially their interactions with inhibitors. We characterized a series of 32 enantiomerically pure inhibitors with respect to interactions with protein kinase A (PKA) and its mutant PKAB3 as a PKB surrogate. The ligands bind to the hinge region, ribose pocket, while their substituted aromatic rings (phenyl, thienyl) bind to the glycine-rich loop at the ATP site. Biological assays show their high potency against both PKA and PKAB3, generally with preference to PKA. The crystal structures reveal a multi-facetted network of ligand–glycine-rich loop interactions, with efficient water replacement contributing to the binding strength. Site-directed mutagenesis and biophysical characterization of the glycine–rich loop mutants (T51G and G55A-PKA) show weakened affinities against tested inhibitors: 1i, 1p, 1c and H-89. We also show structural effects of the aromatic residue phenylalanine that is highly conserved among the AGC kinase group, corresponding to Phe327 in PKA, and its role in inhibitor binding. The inhibitor-Phe327 interactions reveal a complex mix of favorable and unfavorable contacts: Phe327 can block inhibitors from occupying the ATP-binding pocket, but may also be displaced to enable tight binding. The PKA-based Aurora kinase B (PKA-AU6) model, which shares this Phe327, provides examples of crystal structures (PDB ID: 5N23) showing effects on the binding affinities of the pan-Aurora inhibitor AT9283, and also the novel tricyclic dianilino-pyrimidine inhibitors (1B & 2A). The pan-kinome inhibitor staurosporine is known to be able to displace Phe327. We also observe how bisubstrate inhibitors can be used for studies of strong protein peptide-interactions. In summary, this work advances the understanding of how subtle aspects of flexibility, water structure, and chemical interactions determine inhibitor binding kinetics and energetics. Such detailed understanding is required for truly effective structure based drug design.en_US
dc.description.doctoraltypeph.d.en_US
dc.description.popularabstractProtein kinases are an essential component of cellular processes in which they control--and are controlled by--the activities of other proteins. These interactions form signaling chains or networks that initiate or stop processes in response to internal or external conditions. The protein kinases themselves are regulated dynamically by movements of domains or motifs, changing interactions with other proteins or small molecules, and by chemical modification. Over 500 protein kinases are encoded in the human genome, and they share a conserved catalytic domain in sequence and structure. Despite the similarities, their activities are regulated by diverse mechanisms. As enzymes, they catalyze chemical reactions, which for protein kinases involve the transfer of a phosphate group from adenosine-5-triphosphate (ATP) to the hydroxyl group of a serine, threonine or tyrosine residue. This changes the activity of the phosphorylated protein, representing the signal transfer toward the final biological function. Kinases have become important drug targets, since defects may cause cancer or other diseases. However, the design of inhibitors that block only the defective kinase is difficult due to the similarities of the 500 kinases. Most therapeutics target the site of ATP-binding. This site is covered by a so-called "glycine-rich loop", one of the most dynamic parts of the kinase. We applied several biophysical methods to study key protein kinases and their interaction with inhibitors, concentrating on dynamic effects that modelling methods cannot predict. Our studies show the changes in dynamics and structure of the glycine-rich loop and other parts of the kinase in response to inhibitor binding and mutations. The structures reveal how inhibitors form networks of subtle interactions, including water molecules, that can explain their diverse binding strengths and kinetics. This, along with analyses of the effects of glycine-rich loop mutations on inhibitor binding, and of interactions with a key selectivity determinant (the "gatekeeper"), help to advance the understanding of how individual protein kinases react to potential new therapeutics, aiding structure based drug design.en_US
dc.description.sponsorshipBiostruct- The National graduate school in structural biology The Norwegian Research Councilen_US
dc.descriptionThe papers II, III, IV and VI of this thesis are not available in Munin. <br> Paper II: Alam, K. A., Gani, O., Christopeit, T., Engh, R. A.: “Flexibility properties of the protein kinase glycine-rich loop are critical for inhibitor binding. (Manuscript). <p> Paper III: Alam, K. A., Rothweiler, U., Gani, O., Engh, R. A.: “Inhibitor induced structural effects involving Phe327 in AGC kinases. (Manuscript). <p> Paper IV: Ivan, T., Enkvist, E., Viira, B., Manoharan, G. B., Raidaru, G., Pflug, A., Alam, K. A., Zaccolo, M., Engh, R. A., Uri, A.: “Bifunctional Ligands for Inhibition of Tight-Binding Protein–Protein Interactions”. Available in <a href=http://dx.doi.org/10.1021/acs.bioconjchem.6b00293> Bioconjugate Chem., 2016, 27(8):1900–1910. </a> <p> Paper VI: Narayanan, D., Alam, K. A., Engh, M. M., Engh, R. A., Gani, O.: “On methionine as a gatekeeper residue for protein kinase inhibition targets”. (Manuscript).en_US
dc.identifier.isbn978-82-8236-264-1 (trykt) og 978-82-8236-265-8 (pdf)
dc.identifier.urihttps://hdl.handle.net/10037/11195
dc.language.isoengen_US
dc.publisherUiT Norges arktiske universiteten_US
dc.publisherUiT The Arctic University of Norwayen_US
dc.rights.accessRightsopenAccessen_US
dc.rights.holderCopyright 2017 The Author(s)
dc.subject.courseIDDOKTOR-004
dc.subjectVDP::Mathematics and natural science: 400::Chemistry: 440en_US
dc.subjectVDP::Matematikk og Naturvitenskap: 400::Kjemi: 440en_US
dc.titleStudies on selectivity determinants of protein kinase inhibitor bindingen_US
dc.typeDoctoral thesisen_US
dc.typeDoktorgradsavhandlingen_US


Tilhørende fil(er)

Thumbnail
Thumbnail
Thumbnail
Thumbnail

Denne innførselen finnes i følgende samling(er)

Vis enkel innførsel