Network


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

Hotspot


Dive into the research topics where Apostolia Tzekou is active.

Publication


Featured researches published by Apostolia Tzekou.


Developmental Cell | 2011

Complement Fragment C3a Controls Mutual Cell Attraction during Collective Cell Migration

Carlos Carmona-Fontaine; Eric Theveneau; Apostolia Tzekou; Masazumi Tada; Mae Woods; Karen M. Page; Madeline Parsons; John D. Lambris; Roberto Mayor

Summary Collective cell migration is a mode of movement crucial for morphogenesis and cancer metastasis. However, little is known about how migratory cells coordinate collectively. Here we show that mutual cell-cell attraction (named here coattraction) is required to maintain cohesive clusters of migrating mesenchymal cells. Coattraction can counterbalance the natural tendency of cells to disperse via mechanisms such as contact inhibition and epithelial-to-mesenchymal transition. Neural crest cells are coattracted via the complement fragment C3a and its receptor C3aR, revealing an unexpected role of complement proteins in early vertebrate development. Loss of coattraction disrupts collective and coordinated movements of these cells. We propose that coattraction and contact inhibition act in concert to allow cell collectives to self-organize and respond efficiently to external signals, such as chemoattractants and repellents.


Nature Immunology | 2009

Structural and functional implications of the alternative complement pathway C3 convertase stabilized by a staphylococcal inhibitor

Suzan H.M. Rooijakkers; Jin Wu; Maartje Ruyken; Robert van Domselaar; Karel L. Planken; Apostolia Tzekou; Daniel Ricklin; John D. Lambris; Bert J. C. Janssen; Jos A. G. van Strijp; Piet Gros

Activation of the complement system generates potent chemoattractants and leads to the opsonization of cells for immune clearance. Short-lived protease complexes cleave complement component C3 into anaphylatoxin C3a and opsonin C3b. Here we report the crystal structure of the C3 convertase formed by C3b and the protease fragment Bb, which was stabilized by the bacterial immune-evasion protein SCIN. The data suggest that the proteolytic specificity and activity depend on the formation of dimers of C3 with C3b of the convertase. SCIN blocked the formation of a productive enzyme-substrate complex. Irreversible dissociation of the complex of C3b and Bb is crucial to complement regulation and was determined by slow binding kinetics of the Mg2+-adhesion site in Bb. Understanding the mechanistic basis of the central complement-activation step and microbial immune evasion strategies targeting this step will aid in the development of complement therapeutics.


Science | 2010

Structures of C3B in Complex with Factors B and D Give Insight Into Complement Convertase Formation.

Federico Forneris; Daniel Ricklin; Jin Wu; Apostolia Tzekou; Rachel S. Wallace; John D. Lambris; Piet Gros

A Safety Catch on Immune Response The complement system is an integral part of the innate immune system. When triggered, it initiates a cascade that marks intruders for elimination and stimulates immune responses. The key amplification step is cleavage of a complex comprising the complement fragment C3b and factor B (C3bB) by factor D (FD). Forneris et al. (p. 1816) now describe the crystal structure of C3bB and its complex with FD. The structures support a mechanism in which membrane-bound C3b stabilizes an open form of factor B (FB) that has its scissile bond accessible. FD binds through a site distant from its catalytic center to the open form of FB, which activates FD. The two conformational equilibria represent a double safety-catch that would allow tight regulation of this immune response pathway. A double-safety–catch mechanism controls amplification of the complement cascade during immune responses. Activation of the complement cascade induces inflammatory responses and marks cells for immune clearance. In the central complement-amplification step, a complex consisting of surface-bound C3b and factor B is cleaved by factor D to generate active convertases on targeted surfaces. We present crystal structures of the pro-convertase C3bB at 4 angstrom resolution and its complex with factor D at 3.5 angstrom resolution. Our data show how factor B binding to C3b forms an open “activation” state of C3bB. Factor D specifically binds the open conformation of factor B through a site distant from the catalytic center and is activated by the substrate, which displaces factor D’s self-inhibitory loop. This concerted proteolytic mechanism, which is cofactor-dependent and substrate-induced, restricts complement amplification to C3b-tagged target cells.


Journal of Immunology | 2011

The C5a Receptor Impairs IL-12–Dependent Clearance of Porphyromonas gingivalis and Is Required for Induction of Periodontal Bone Loss

Shuang Liang; Jennifer L. Krauss; Hisanori Domon; Megan L. McIntosh; Kavita B. Hosur; Fenge Li; Apostolia Tzekou; John D. Lambris; George Hajishengallis

The C5a anaphylatoxin receptor (C5aR; CD88) is activated as part of the complement cascade and exerts important inflammatory, antimicrobial, and regulatory functions, at least in part, via crosstalk with TLRs. However, the periodontal pathogen Porphyromonas gingivalis can control C5aR activation by generating C5a through its own C5 convertase-like enzymatic activity. In this paper, we show that P. gingivalis uses this mechanism to proactively and selectively inhibit TLR2-induced IL-12p70, whereas the same pathogen-instigated C5aR-TLR2 crosstalk upregulates other inflammatory and bone-resorptive cytokines (IL-1β, IL-6, and TNF-α). In vivo, the ability of P. gingivalis to manipulate TLR2 activation via the C5a-C5aR axis allowed it to escape IL-12p70–dependent immune clearance and to cause inflammatory bone loss in a murine model of experimental periodontitis. In the latter regard, C5aR-deficient or TLR2-deficient mice were both resistant to periodontal bone loss, in stark contrast with wild-type control mice, which is consistent with the interdependent interactions of C5aR and TLR2 in P. gingivalis immune evasion and induction of bone-resorptive cytokines. In conclusion, P. gingivalis targets C5aR to promote its adaptive fitness and cause periodontal disease. Given the current availability of safe and effective C5aR antagonists, pharmacological blockade of C5aR could act therapeutically in human periodontitis and reduce associated systemic risks.


Immunobiology | 2013

New analogs of the clinical complement inhibitor compstatin with subnanomolar affinity and enhanced pharmacokinetic properties

Daniel Ricklin; Hongjun Bai; Hui Chen; Edimara S. Reis; Mateusz Maciejewski; Apostolia Tzekou; Robert A. DeAngelis; Ranillo R.G. Resuello; Florea Lupu; Paul N. Barlow; John D. Lambris

Therapeutic modulation of the complement system has become increasingly important in line with the growing recognition of the role of complement in numerous diseases. Compstatin, a peptidic inhibitor that acts at the central level of the complement cascade, is currently in clinical evaluation but routes to improve its efficacy have not yet been fully explored. Here, we report improvements in both the inhibitory potency and pharmacokinetic parameters of compstatin that broaden its clinical applications. Selective modification of the compstatin N-terminus with non-proteinogenic amino acids resulted in the first analogue with subnanomolar binding affinity (KD=0.5nM) and other similarly potent derivatives with improved solubility in clinically relevant solvents. Detailed structure-activity relationship studies based on biophysical and computational methods revealed key structural determinants for the observed improvements. Importantly, pharmacokinetic evaluation in non-human primates revealed target-driven elimination kinetics with plasma half-life values exceeding expectations for peptidic drugs (close to 12h). This successful optimization strategy is expected to pave the way for systemic administration of compstatin in a range of clinical conditions.


Journal of Immunology | 2011

Protection of Nonself Surfaces from Complement Attack by Factor H-Binding Peptides: Implications for Therapeutic Medicine

You Qiang Wu; Georgia Sfyroera; Apostolia Tzekou; Brian K. Kay; Bo Nilsson; Kristina Nilsson Ekdahl; Daniel Ricklin; John D. Lambris

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5–18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Allosteric inhibition of complement function by a staphylococcal immune evasion protein

Hui Chen; Daniel Ricklin; Michal Hammel; Brandon L. Garcia; William J. McWhorter; Georgia Sfyroera; You-Qiang Wu; Apostolia Tzekou; Sheng Li; Brian V. Geisbrecht; Virgil L. Woods; John D. Lambris

The complement system is a major target of immune evasion by Staphylococcus aureus. Although many evasion proteins have been described, little is known about their molecular mechanisms of action. Here we demonstrate that the extracellular fibrinogen-binding protein (Efb) from S. aureus acts as an allosteric inhibitor by inducing conformational changes in complement fragment C3b that propagate across several domains and influence functional regions far distant from the Efb binding site. Most notably, the inhibitor impaired the interaction of C3b with complement factor B and, consequently, formation of the active C3 convertase. As this enzyme complex is critical for both activation and amplification of the complement response, its allosteric inhibition likely represents a fundamental contribution to the overall immune evasion strategy of S. aureus.


ACS Chemical Biology | 2011

Synthesis and Activity of Thioether-Containing Analogues of the Complement Inhibitor Compstatin

Patrick J. Knerr; Apostolia Tzekou; Daniel Ricklin; Hui Chen; Wilfred A. van der Donk; John D. Lambris

Disulfide bonds are essential for the structural stability and biological activity of many bioactive peptides. However, these bonds are labile to reducing agents, which can limit the therapeutic utility of such peptides. Substitution of a disulfide bond with a reduction-resistant cystathionine bridge is an attractive means of improving stability while imposing minimal structural perturbation to the peptide. We have applied this approach to the therapeutic complement inhibitor compstatin, a disulfide-containing peptide currently in clinical trials for age-related macular degeneration, in an effort to maintain its potent activity while improving its biological stability. Thioether-containing compstatin analogues were produced via solid-phase peptide synthesis utilizing orthogonally protected cystathionine amino acid building blocks and solid-supported peptide cyclization. Overall, the affinity of these analogues for their biological target and potent inhibition of complement activation were largely maintained when compared to those of the parent disulfide-containing peptides. Thus, the improved stability to reduction conferred by the thioether bond makes this new class of compstatin peptides a promising alternative for therapeutic applications. Additionally, the versatility of this synthesis allows for exploration of disulfide-to-thioether substitution in a variety of other therapeutic peptides.


Nature Communications | 2013

Complement anaphylatoxin C3a is a potent inducer of embryonic chick retina regeneration

Tracy Haynes; Agustin Luz-Madrigal; Edimara S. Reis; Nancy P. Echeverri Ruiz; Erika Grajales-Esquivel; Apostolia Tzekou; Panagiotis A. Tsonis; John D. Lambris; Katia Del Rio-Tsonis

Identifying the initiation signals for tissue regeneration in vertebrates is one of the major challenges in regenerative biology. Much of the research thus far has indicated that certain growth factors have key roles. Here we show that complement fragment C3a is sufficient to induce complete regeneration of the embryonic chick retina from stem/progenitor cells present in the eye, independent of fibroblast growth factor receptor signaling. Instead, C3a induces retina regeneration via STAT3 activation, which in turn activates the injury- and inflammation-responsive factors, IL-6, IL-8 and TNF-α. This activation sets forth regulation of Wnt2b, Six3 and Sox2, genes associated with retina stem and progenitor cells. Thus, our results establish a mechanism for retina regeneration based on injury and inflammation signals. Furthermore, our results indicate a unique function for complement anaphylatoxins that implicate these molecules in the induction and complete regeneration of the retina, opening new avenues of experimentation in the field.


Journal of Immunology | 2009

A Molecular Insight into Complement Evasion by the Staphylococcal Complement Inhibitor Protein Family

Daniel Ricklin; Apostolia Tzekou; Brandon L. Garcia; Michal Hammel; William J. McWhorter; Georgia Sfyroera; You-Qiang Wu; V. Michael Holers; Andrew P. Herbert; Paul N. Barlow; Brian V. Geisbrecht; John D. Lambris

Staphylococcus aureus possesses an impressive arsenal of complement evasion proteins that help the bacterium escape attack of the immune system. The staphylococcal complement inhibitor (SCIN) protein exhibits a particularly high potency and was previously shown to block complement by acting at the level of the C3 convertases. However, many details about the exact binding and inhibitory mechanism remained unclear. In this study, we demonstrate that SCIN directly binds with nanomolar affinity to a functionally important area of C3b that lies near the C terminus of its β-chain. Direct competition of SCIN with factor B for C3b slightly decreased the formation of surface-bound convertase. However, the main inhibitory effect can be attributed to an entrapment of the assembled convertase in an inactive state. Whereas native C3 is still able to bind to the blocked convertase, no generation and deposition of C3b could be detected in the presence of SCIN. Furthermore, SCIN strongly competes with the binding of factor H to C3b and influences its regulatory activities: the SCIN-stabilized convertase was essentially insensitive to decay acceleration by factor H and the factor I- and H-mediated conversion of surface-bound C3b to iC3b was significantly reduced. By targeting a key area on C3b, SCIN is able to block several essential functions within the alternative pathway, which explains the high potency of the inhibitor. Our findings provide an important insight into complement evasion strategies by S. aureus and may act as a base for further functional studies.

Collaboration


Dive into the Apostolia Tzekou's collaboration.

Top Co-Authors

Avatar

John D. Lambris

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Daniel Ricklin

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Georgia Sfyroera

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Chen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

William J. McWhorter

University of Missouri–Kansas City

View shared research outputs
Top Co-Authors

Avatar

Edimara S. Reis

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

You-Qiang Wu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge