Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria A. Argiriadi is active.

Publication


Featured researches published by Maria A. Argiriadi.


Journal of Biological Chemistry | 2000

Biochemical evidence for the involvement of tyrosine in epoxide activation during the catalytic cycle of epoxide hydrolase.

Takashi Yamada; Christophe Morisseau; Joseph E. Maxwell; Maria A. Argiriadi; David W. Christianson; Bruce D. Hammock

Epoxide hydrolases (EH) catalyze the hydrolysis of epoxides and arene oxides to their corresponding diols. The crystal structure of murine soluble EH suggests that Tyr465and Tyr381 act as acid catalysts, activating the epoxide ring and facilitating the formation of a covalent intermediate between the epoxide and the enzyme. To explore the role of these two residues, mutant enzymes were produced and the mechanism of action was analyzed. Enzyme assays on a series of substrates confirm that both Tyr465 and Tyr381 are required for full catalytic activity. The kinetics of chalcone oxide hydrolysis show that mutation of Tyr465 and Tyr381 decreases the rate of binding and the formation of an intermediate, suggesting that both tyrosines polarize the epoxide moiety to facilitate ring opening. These two tyrosines are, however, not implicated in the hydrolysis of the covalent intermediate. Sequence comparisons showed that Tyr465 is conserved in microsomal EHs. The substitution of analogous Tyr374 with phenylalanine in the human microsomal EH dramatically decreases the rate of hydrolysis ofcis-stilbene oxide. These results suggest that these tyrosines perform a significant mechanistic role in the substrate activation by EHs.


Journal of Biological Chemistry | 2015

Tricyclic Covalent Inhibitors Selectively Target Jak3 through an Active Site Thiol

Eric R. Goedken; Maria A. Argiriadi; David Banach; Bryan A. Fiamengo; Sage E. Foley; Kristine E. Frank; Jonathan S. George; Christopher M. Harris; Adrian D. Hobson; David C. Ihle; Douglas Marcotte; Philip Merta; Mark Michalak; Sara Murdock; Medha J. Tomlinson; Jeffrey W. Voss

Background: Janus kinase 3 (Jak3) inhibitors hold promise for treatment of autoimmunity, but developing selective inhibitors is challenging. Results: We designed Jak3 inhibitors that avoid inhibition of the other JAKs. Conclusion: Our inhibitors possess high selectivity against other kinases and can potently inhibit Jak3 activity in cell-based assays. Significance: This class of irreversible inhibitors may be useful as selective agents of Jak3 inhibition. The action of Janus kinases (JAKs) is required for multiple cytokine signaling pathways, and as such, JAK inhibitors hold promise for treatment of autoimmune disorders, including rheumatoid arthritis, inflammatory bowel disease, and psoriasis. However, due to high similarity in the active sites of the four members (Jak1, Jak2, Jak3, and Tyk2), developing selective inhibitors within this family is challenging. We have designed and characterized substituted, tricyclic Jak3 inhibitors that selectively avoid inhibition of the other JAKs. This is accomplished through a covalent interaction between an inhibitor containing a terminal electrophile and an active site cysteine (Cys-909). We found that these ATP competitive compounds are irreversible inhibitors of Jak3 enzyme activity in vitro. They possess high selectivity against other kinases and can potently (IC50 < 100 nm) inhibit Jak3 activity in cell-based assays. These results suggest irreversible inhibitors of this class may be useful selective agents, both as tools to probe Jak3 biology and potentially as therapies for autoimmune diseases.


Journal of Biological Chemistry | 2009

Unusual Water-mediated Antigenic Recognition of the Proinflammatory Cytokine Interleukin-18

Maria A. Argiriadi; Tao Xiang; Chengbin Wu; Tariq Ghayur; David W. Borhani

The unique cytokine interleukin-18 (IL-18) acts synergistically with IL-12 to regulate T-helper 1 and 2 lymphocytes and, as such, seems to underlie the pathogenesis of various autoimmune and allergic diseases. Several anti-IL-18 agents are in clinical development, including the recombinant human antibody ABT-325, which is entering trials for autoimmune diseases. Given competing cytokine/receptor and cytokine/receptor decoy interactions, understanding the structural basis for recognition is critical for effective development of anti-cytokine therapies. Here we report three crystal structures: the murine antibody 125-2H Fab fragment bound to human IL-18, at 1.5 Å resolution; the 125-2H Fab (2.3 Å); and the ABT-325 Fab (1.5 Å). These structures, along with human/mouse IL-18 chimera binding data, allow us to make three key observations relevant to the biology and antigenic recognition of IL-18 and related cytokines. First, several IL-18 residues shift dramatically (>10 Å) upon binding 125-2H, compared with unbound IL-18 (Kato, Z., Jee, J., Shikano, H., Mishima, M., Ohki, I., Ohnishi, H., Li, A., Hashimoto, K., Matsukuma, E., Omoya, K., Yamamoto, Y., Yoneda, T., Hara, T., Kondo, N., and Shirakawa, M. (2003) Nat. Struct. Biol. 10, 966–971). IL-18 thus exhibits plasticity that may be common to its interactions with other receptors. Related cytokines may exhibit similar plasticity. Second, ABT-325 and 125-2H differ significantly in combining site character and architecture, thus explaining their ability to bind IL-18 simultaneously at distinct epitopes. These data allow us to define the likely ABT-325 epitope and thereby explain the distinct neutralizing mechanisms of both antibodies. Third, given the high 125-2H potency, 10 well ordered water molecules are trapped upon complex formation in a cavity between two IL-18 loops and all six 125-2H complementarity-determining regions. Thus, counterintuitively, tight and specific antibody binding may in some cases be water-mediated.


Journal of Medicinal Chemistry | 2015

Discovery of selective and orally bioavailable protein kinase Cθ (PKCθ) inhibitors from a fragment hit.

Dawn M. George; Eric C. Breinlinger; Michael M. Friedman; Yang Zhang; Jianfei Wang; Maria A. Argiriadi; Pratima Bansal-Pakala; Martine Barth; David B. Duignan; Prisca Honore; QingYu Lang; Scott W. Mittelstadt; Dominique Potin; Lian Rundell; Jeremy John Edmunds

Protein kinase Cθ (PKCθ) regulates a key step in the activation of T cells. On the basis of its mechanism of action, inhibition of this kinase is hypothesized to serve as an effective therapy for autoimmune diseases such as rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and psoriasis. Herein, the discovery of a small molecule PKCθ inhibitor is described, starting from a fragment hit 1 and advancing to compound 41 through the use of structure-based drug design. Compound 41 demonstrates excellent in vitro activity, good oral pharmacokinetics, and efficacy in both an acute in vivo mechanistic model and a chronic in vivo disease model but suffers from tolerability issues upon chronic dosing.


Bioorganic & Medicinal Chemistry Letters | 2010

2,4-Diaminopyrimidine MK2 inhibitors. Part I: Observation of an unexpected inhibitor binding mode.

Maria A. Argiriadi; Anna M. Ericsson; Christopher M. Harris; David Banach; David W. Borhani; David J. Calderwood; Megan Demers; Jennifer DiMauro; Richard W. Dixon; Jennifer Hardman; Silvia Kwak; Biqin Li; John A. Mankovich; Douglas Marcotte; Kelly D. Mullen; Baofu Ni; M. Pietras; Ramkrishna Sadhukhan; Silvino Sousa; Medha J. Tomlinson; Lu Wang; Tao Xiang; Robert V. Talanian

MK2 is a Ser/Thr kinase of significant interest as an anti-inflammatory drug discovery target. Here we describe the development of in vitro tools for the identification and characterization of MK2 inhibitors, including validation of inhibitor interactions with the crystallography construct and determination of the unique binding mode of 2,4-diaminopyrimidine inhibitors in the MK2 active site. Use of these tools in the optimization of a potent and selective inhibitor lead series is described in the accompanying Letter.


Bioorganic & Medicinal Chemistry Letters | 2010

2,4-Diaminopyrimidine MK2 inhibitors. Part II: Structure-based inhibitor optimization

Christopher M. Harris; Anna M. Ericsson; Maria A. Argiriadi; Claude Barberis; David W. Borhani; Andrew Burchat; David J. Calderwood; George A. Cunha; Richard W. Dixon; Kristine E. Frank; Eric F. Johnson; Joanne Kamens; Silvia Kwak; Biqin Li; Kelly D. Mullen; Denise C. Perron; Lu Wang; Neil Wishart; Xiaoyun Wu; Xiaolei Zhang; Tami R. Zmetra; Robert V. Talanian

We describe structure-based optimization of a series of novel 2,4-diaminopyrimidine MK2 inhibitors. Co-crystal structures (see accompanying Letter) demonstrated a unique inhibitor binding mode. Resulting inhibitors had IC(50) values as low as 19nM and moderate selectivity against a kinase panel. Compounds 15, 31a, and 31b inhibit TNFalpha production in peripheral human monocytes.


Journal of Medicinal Chemistry | 2015

Optimized protein kinase Cθ (PKCθ) inhibitors reveal only modest anti-inflammatory efficacy in a rodent model of arthritis.

Dawn M. George; Eric C. Breinlinger; Maria A. Argiriadi; Yang Zhang; Jianfei Wang; Pratima Bansal-Pakala; David B. Duignan; Prisca Honore; QingYu Lang; Scott W. Mittelstadt; Lian Rundell; Annette Schwartz; Jiakang Sun; Jeremy John Edmunds

We previously demonstrated that selective inhibition of protein kinase Cθ (PKCθ) with triazinone 1 resulted in dose-dependent reduction of paw swelling in a mouse model of arthritis.1,2 However, a high concentration was required for efficacy, thus providing only a minimal safety window. Herein we describe a strategy to deliver safer compounds based on the hypothesis that optimization of potency in concert with good oral pharmacokinetic (PK) properties would enable in vivo efficacy at reduced exposures, resulting in an improved safety window. Ultimately, transformation of 1 yielded analogues that demonstrated excellent potency and PK properties and fully inhibited IL-2 production in an acute model. In spite of good exposure, twice-a-day treatment with 17l in the glucose-6-phosphate isomerase chronic in vivo mouse model of arthritis yielded only moderate efficacy. On the basis of the exposure achieved, we conclude that PKCθ inhibition alone is insufficient for complete efficacy in this rodent arthritis model.


BMC Structural Biology | 2012

Enabling structure-based drug design of Tyk2 through co-crystallization with a stabilizing aminoindazole inhibitor

Maria A. Argiriadi; Eric R. Goedken; David Banach; David W. Borhani; Andrew Burchat; Richard W. Dixon; Doug Marcotte; Gary T. Overmeyer; Valerie L. Pivorunas; Ramkrishna Sadhukhan; Silvino Sousa; Nigel StJohn Moore; Medha J. Tomlinson; Jeffrey W. Voss; Lu Wang; Neil Wishart; Kevin R. Woller; Robert V. Talanian

BackgroundStructure-based drug design (SBDD) can accelerate inhibitor lead design and optimization, and efficient methods including protein purification, characterization, crystallization, and high-resolution diffraction are all needed for rapid, iterative structure determination. Janus kinases are important targets that are amenable to structure-based drug design. Here we present the first mouse Tyk2 crystal structures, which are complexed to 3-aminoindazole compounds.ResultsA comprehensive construct design effort included N- and C-terminal variations, kinase-inactive mutations, and multiple species orthologs. High-throughput cloning and expression methods were coupled with an abbreviated purification protocol to optimize protein solubility and stability. In total, 50 Tyk2 constructs were generated. Many displayed poor expression, inadequate solubility, or incomplete affinity tag processing. One kinase-inactive murine Tyk2 construct, complexed with an ATP-competitive 3-aminoindazole inhibitor, provided crystals that diffracted to 2.5–2.6 Å resolution. This structure revealed initial “hot-spot” regions for SBDD, and provided a robust platform for ligand soaking experiments. Compared to previously reported human Tyk2 inhibitor crystal structures (Chrencik et al. (2010) J Mol Biol 400:413), our structures revealed a key difference in the glycine-rich loop conformation that is induced by the inhibitor. Ligand binding also conferred resistance to proteolytic degradation by thermolysin. As crystals could not be obtained with the unliganded enzyme, this enhanced stability is likely important for successful crystallization and inhibitor soaking methods.ConclusionsPractical criteria for construct performance and prioritization, the optimization of purification protocols to enhance protein yields and stability, and use of high-throughput construct exploration enable structure determination methods early in the drug discovery process. Additionally, specific ligands stabilize Tyk2 protein and may thereby enable crystallization.


Bioorganic & Medicinal Chemistry Letters | 2016

Creation of a S1P Lyase bacterial surrogate for structure-based drug design.

Maria A. Argiriadi; David Banach; Elzbieta Radziejewska; Susan Marchie; Jennifer DiMauro; Jurgen Dinges; Eric Dominguez; Charles W. Hutchins; Russell A. Judge; Kara Queeney; Grier A. Wallace; Christopher M. Harris

S1P Lyase (SPL) has been described as a drug target in the treatment of autoimmune diseases. It plays an important role in maintaining intracellular levels of S1P thereby affecting T cell egress from lymphoid tissues. Several groups have already published approaches to inhibit S1P Lyase with small molecules, which in turn increase endogenous S1P concentrations resulting in immunosuppression. The use of structural biology has previously aided SPL inhibitor design. Novel construct design is at times necessary to provide a reagent for protein crystallography. Here we present a chimeric bacterial protein scaffold used for protein X-ray structures in the presence of early small molecule inhibitors. Mutations were introduced to the bacterial SPL from Symbiobacterium thermophilum which mimic the human enzyme. As a result, two mutant StSPL crystal structures resolved to 2.8Å and 2.2Å resolutions were solved and provide initial structural hypotheses for an isoxazole chemical series, whose optimization is discussed in the accompanying paper.


Bioorganic & Medicinal Chemistry Letters | 2016

Hit-to-lead evaluation of a novel class of sphingosine 1-phosphate lyase inhibitors

Jurgen Dinges; Christopher M. Harris; Grier A. Wallace; Maria A. Argiriadi; Kara Queeney; Denise C. Perron; Eric Dominguez; Tegest Kebede; Kelly E. Desino; Hetal Patel; Anil Vasudevan

Inhibition of sphingosine-1-phosphate lyase has recently been proposed as a potential treatment option for inflammatory disorders such as multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease. In this report we describe our hit-to-lead evaluation of the isoxazolecarboxamide 6, a high-throughput screening hit (in vitro IC50=1.0 μM, cell IC50=1.8 μM), as a novel S1P lyase inhibitor. We were able to establish basic structure-activity relationships around 6 and succeeded in obtaining X-ray structural information which enabled structure-based design. With the discovery of 28, enzyme activity was quickly improved to IC50=120 nM and cell potency to IC50=230 nM. The main liability in the established isoxazolecarboxamide hit series was determined to be metabolic stability. In particular we identified that future lead-optimization efforts to overcome this problem should focus on blocking the N-dealkylation on the secondary amine.

Collaboration


Dive into the Maria A. Argiriadi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David W. Borhani

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge