Network


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

Hotspot


Dive into the research topics where Andrew B. Herr is active.

Publication


Featured researches published by Andrew B. Herr.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Structural basis for concerted recruitment and activation of IRF-3 by innate immune adaptor proteins

Baoyu Zhao; Chang Shu; Xinsheng Gao; Banumathi Sankaran; Fenglei Du; Catherine L. Shelton; Andrew B. Herr; Jun-Yuan Ji; Pingwei Li

Significance Type I IFNs are key cytokines involved in antiviral immunity. A number of innate sensing pathways regulate the induction of type I IFNs. These pathways converge at the activation of the transcription factor IRF-3 (IFN regulatory factor 3). Three different adaptors mediate the recruitment of IRF-3 using a conserved structural motif. In this study, we determined the molecular mechanisms by which these adaptors recruit IRF-3 upon phosphorylation, the mechanism of IRF-3 activation, and how rotavirus subverts these signaling mechanisms to evade innate immune surveillance. These results provide critical insights into the molecular basis of innate immunity against microbial and viral infections. Type I IFNs are key cytokines mediating innate antiviral immunity. cGMP-AMP synthase, ritinoic acid-inducible protein 1 (RIG-I)–like receptors, and Toll-like receptors recognize microbial double-stranded (ds)DNA, dsRNA, and LPS to induce the expression of type I IFNs. These signaling pathways converge at the recruitment and activation of the transcription factor IRF-3 (IFN regulatory factor 3). The adaptor proteins STING (stimulator of IFN genes), MAVS (mitochondrial antiviral signaling), and TRIF (TIR domain-containing adaptor inducing IFN-β) mediate the recruitment of IRF-3 through a conserved pLxIS motif. Here we show that the pLxIS motif of phosphorylated STING, MAVS, and TRIF binds to IRF-3 in a similar manner, whereas residues upstream of the motif confer specificity. The structure of the IRF-3 phosphomimetic mutant S386/396E bound to the cAMP response element binding protein (CREB)-binding protein reveals that the pLxIS motif also mediates IRF-3 dimerization and activation. Moreover, rotavirus NSP1 (nonstructural protein 1) employs a pLxIS motif to target IRF-3 for degradation, but phosphorylation of NSP1 is not required for its activity. These results suggest a concerted mechanism for the recruitment and activation of IRF-3 that can be subverted by viral proteins to evade innate immune responses.


Blood Advances | 2017

Fibrin and D-dimer bind to monomeric GPVI

Marie-Blanche Onselaer; Alexander T. Hardy; Clare Wilson; Ximena Sánchez; Amir K. Babar; Jeanette L. C. Miller; Callum N. Watson; Stephanie Watson; Arkadiusz Mieczyslaw Bonna; Helen Philippou; Andrew B. Herr; Diego Mezzano; Robert A. S. Ariëns; Steve P. Watson

Fibrin has recently been shown to activate platelets through the immunoglobulin receptor glycoprotein VI (GPVI). In the present study, we show that spreading of human platelets on fibrin is abolished in patients deficient in GPVI, confirming that fibrin activates human platelets through the immunoglobulin receptor. Using a series of proteolytic fragments, we show that D-dimer, but not the E fragment of fibrin, binds to GPVI and that immobilized D-dimer induces platelet spreading through activation of Src and Syk tyrosine kinases. In contrast, when platelets are activated in suspension, soluble D-dimer inhibits platelet aggregation induced by fibrin and collagen, but not by a collagen-related peptide composed of a repeat GPO sequence or by thrombin. Using surface plasmon resonance, we demonstrate that fibrin binds selectively to monomeric GPVI with a KD of 302 nM, in contrast to collagen, which binds primarily to dimeric GPVI. These results establish GPVI as the major signaling receptor for fibrin in human platelets and provide evidence that fibrin binds to a distinct configuration of GPVI. This indicates that it may be possible to develop agents that selectively block the interaction of fibrin but not collagen with the immunoglobulin receptor. Such agents are required to establish whether selective targeting of either interaction has the potential to lead to development of an antithrombotic agent with a reduced effect on bleeding relative to current antiplatelet drugs.


Blood | 2016

Structural basis for collagen recognition by the immune receptor OSCAR.

Long Zhou; Jennifer M. Hinerman; Michal Blaszczyk; Jeanette L. C. Miller; Deborah G. Conrady; Alexander D. Barrow; Dimitri Y. Chirgadze; Dominique Bihan; Richard W. Farndale; Andrew B. Herr

The osteoclast-associated receptor (OSCAR) is a collagen-binding immune receptor with important roles in dendritic cell maturation and activation of inflammatory monocytes as well as in osteoclastogenesis. The crystal structure of the OSCAR ectodomain is presented, both free and in complex with a consensus triple-helical peptide (THP). The structures revealed a collagen-binding site in each immunoglobulin-like domain (D1 and D2). The THP binds near a predicted collagen-binding groove in D1, but a more extensive interaction with D2 is facilitated by the unusually wide D1-D2 interdomain angle in OSCAR. Direct binding assays, combined with site-directed mutagenesis, confirm that the primary collagen-binding site in OSCAR resides in D2, in marked contrast to the related collagen receptors, glycoprotein VI (GPVI) and leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1). Monomeric OSCAR D1D2 binds to the consensus THP with a KD of 28 µM measured in solution, but shows a higher affinity (KD 1.5 μM) when binding to a solid-phase THP, most likely due to an avidity effect. These data suggest a 2-stage model for the interaction of OSCAR with a collagen fibril, with transient, low-affinity interactions initiated by the membrane-distal D1, followed by firm adhesion to the primary binding site in D2.


Methods in Enzymology | 2015

Elucidating Complicated Assembling Systems in Biology Using Size-and-Shape Analysis of Sedimentation Velocity Data.

Catherine T. Chaton; Andrew B. Herr

Sedimentation velocity analytical ultracentrifugation (SV-AUC) has seen a resurgence in popularity as a technique for characterizing macromolecules and complexes in solution. SV-AUC is a particularly powerful tool for studying protein conformation, complex stoichiometry, and interacting systems in general. Deconvoluting velocity data to determine a sedimentation coefficient distribution c(s) allows for the study of either individual proteins or multicomponent mixtures. The standard c(s) approach estimates molar masses of the sedimenting species based on determination of the frictional ratio (f/f0) from boundary shapes. The frictional ratio in this case is a weight-averaged parameter, which can lead to distortion of mass estimates and loss of information when attempting to analyze mixtures of macromolecules with different shapes. A two-dimensional extension of the c(s) analysis approach provides size-and-shape distributions that describe the data in terms of a sedimentation coefficient and frictional ratio grid. This allows for better resolution of species with very distinct shapes that may co-sediment and provides better molar mass determinations for multicomponent mixtures. An example case is illustrated using globular and nonglobular proteins of different masses with nearly identical sedimentation coefficients that could only be resolved using the size-and-shape distribution. Other applications of this analytical approach to complex biological systems are presented, focusing on proteins involved in the innate immune response to cytosolic microbial DNA.


Journal of Molecular Biology | 2017

The Proline/Glycine-Rich Region of the Biofilm Adhesion Protein Aap Forms an Extended Stalk that Resists Compaction

Alexander E. Yarawsky; Lance R. English; Steven T. Whitten; Andrew B. Herr

Staphylococcus epidermidis is one of the primary bacterial species responsible for healthcare-associated infections. The most significant virulence factor for S. epidermidis is its ability to form a biofilm, which renders the bacteria highly resistant to host immune responses and antibiotic action. Intercellular adhesion within the biofilm is mediated by the accumulation-associated protein (Aap), a cell wall-anchored protein that self-assembles in a zinc-dependent manner. The C-terminal portion of Aap contains a 135-aa-long, proline/glycine-rich region (PGR) that has not yet been characterized. The region contains a set of 18 nearly identical AEPGKP repeats. Analysis of the PGR using biophysical techniques demonstrated the region is a highly extended, intrinsically disordered polypeptide with unusually high polyproline type II helix propensity. In contrast to many intrinsically disordered polypeptides, there was a minimal temperature dependence of the global conformational state of PGR in solution as measured by analytical ultracentrifugation and dynamic light scattering. Furthermore, PGR was resistant to conformational collapse or α-helix formation upon the addition of the osmolyte trimethylamine N-oxide or the cosolvent 2,2,2-trifluoroethanol. Collectively, these results suggest PGR functions as a resilient, extended stalk that projects the rest of Aap outward from the bacterial cell wall, promoting intercellular adhesion between cells in the biofilm. This work sheds light on regions of low complexity often found near the attachment point of bacterial cell wall-anchored proteins.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2017

Charming the Snake: Venom-Derived Peptides Show Surprising Efficacy as Glycoprotein VI–Targeting Antithrombotic Agents

Andrew B. Herr

Snake venoms are potent cocktails containing a diverse array of proteins that typically target neuromuscular junctions or the hemostatic system of the snake’s prey. Many of the venom proteins that target hemostasis adopt a C-type lectinlike fold and are therefore termed snaclecs (ie, snake venom C-type lectins). Despite the structural similarity to functional lectins, the snaclecs do not exhibit carbohydrateor calciumbinding properties. Instead, the typical calcium-stabilized carbohydrate-binding loop found in functional lectins adopts a highly extended loop that forms a domain swap between α and β subunits in the snaclecs, forming an αβ heterodimer that interacts with critical proteins that control hemostasis. The resulting effect can be proor anticoagulant (eg, snaclecs that target factor IX, factor X, prothrombin, or α-thrombin) or proor antithrombotic (eg, snaclecs that target platelet receptors, such as glycoprotein Ibα, glycoprotein Ia/IIa, or GPVI [glycoprotein VI]). The structures of several snaclecs have been determined, revealing conserved features of the fold: each forms a disulfide-linked heterodimeric subunit of α and β chains that may comprise the mature protein (eg, factor X–binding protein or EMS16) or may further assemble into higher order structures, such as the (αβ) 4 rings of convulxin or flavocetin. Several structures have been reported for complexes between snaclecs and their targets, revealing that a concave surface between the α and β subunits interacts with the target protein to form an extensive binding site.


Biochemical Journal | 2017

Functional consequences of B-repeat sequence variation in the staphylococcal biofilm protein Aap: deciphering the assembly code

Catherine L. Shelton; Deborah G. Conrady; Andrew B. Herr

Staphylococcus epidermidis is an opportunistic pathogen that can form robust biofilms that render the bacteria resistant to antibiotic action and immune responses. Intercellular adhesion in S. epidermidis biofilms is mediated by the cell wall-associated accumulation-associated protein (Aap), via zinc-mediated self-assembly of its B-repeat region. This region contains up to 17 nearly identical sequence repeats, with each repeat assumed to be functionally equivalent. However, Aap B-repeats exist as two subtypes, defined by a cluster of consensus or variant amino acids. These variable residues are positioned near the zinc-binding (and dimerization) site and the stability determinant for the B-repeat fold. We have characterized four B-repeat constructs to assess the functional relevance of the two Aap B-repeat subtypes. Analytical ultracentrifugation experiments demonstrated that constructs with the variant sequence show reduced or absent Zn2+-induced dimerization. Likewise, circular dichroism thermal denaturation experiments showed that the variant sequence could significantly stabilize the fold, depending on its location within the construct. Crystal structures of three of the constructs revealed that the side chains from the variant sequence form an extensive bonding network that can stabilize the fold. Furthermore, altered distribution of charged residues between consensus and variant sequences changes the electrostatic potential in the vicinity of the Zn2+-binding site, providing a mechanistic explanation for the loss of zinc-induced dimerization in the variant constructs. These data suggest an assembly code that defines preferred oligomerization modes of the B-repeat region of Aap and a slip-grip model for initial contact followed by firm intercellular adhesion during biofilm formation.


Journal of Biological Chemistry | 2018

Evolution of an allosteric “off switch” in apoptotic caspases

Andrew B. Herr

Caspase-3 is well known as the “executioner” whose activation commits the cell to an apoptotic fate, but low levels of caspase-3 activity also play key roles in development. A new study explains how cells can balance these functions, using biophysical, structural, and computational approaches to demonstrate the mechanism by which phosphorylation of conserved sites on a distal surface loop reduces or abolishes catalytic activity. These results provide new insights into allosteric regulation mechanisms and offer new opportunities for development of caspase-3 modulators.


Protein Science | 2017

Defining the metal specificity of a multifunctional biofilm adhesion protein

Catherine T. Chaton; Andrew B. Herr

The accumulation associated protein (Aap) of Staphylococcus epidermidis mediates intercellular adhesion events necessary for biofilm growth. This process depends upon Zn2+‐induced self‐assembly of G5 domains within the B‐repeat region of the protein, forming anti‐parallel, intertwined protein “ropes” between cells. Pleomorphism in the Zn2+‐coordinating residues was observed in previously solved crystal structures, suggesting that the metal binding site might accommodate other transition metals and thereby support dimerization. By use of carefully selected buffer systems and a specialized approach to analyze sedimentation velocity analytical ultracentrifugation data, we were able to analyze low‐affinity metal binding events in solution. Our data show that both Zn2+ and Cu2+ support B‐repeat assembly, whereas Mn2+, Co2+, and Ni2+ bind to Aap but do not support self‐association. As the number of G5 domains are increased in longer B‐repeat constructs, the total concentration of metal required for dimerization decreases and the transition between monomer and dimer becomes more abrupt. These characteristics allow Aap to function as an environmental sensor that regulates biofilm formation in response to local concentrations of Zn2+ and Cu2+, both of which are implicated in immune cell activity.


Current Allergy and Asthma Reports | 2017

Staphylococcal Biofilms in Atopic Dermatitis

Tammy Gonzalez; Jocelyn M. Biagini Myers; Andrew B. Herr; Gurjit K. Khurana Hershey

Purpose of ReviewAtopic dermatitis (AD) is a chronic, relapsing inflammatory skin disorder that is a major public health burden worldwide. AD lesions are often colonized by Staphylococcus aureus and Staphylococcus epidermidis. An important aspect of Staphylococcus spp. is their propensity to form biofilms, adhesive surface-attached colonies that become highly resistant to antibiotics and immune responses, and recent studies have found that clinical isolates colonizing AD skin are often biofilm-positive. Biofilm formation results in complex bacterial communities that have unique effects on keratinocytes and host immunity. This review will summarize recent studies exploring the role of staphyloccocal biofilms in atopic dermatitis and the implications for treatment.Recent FindingsRecent studies suggest an important role for biofilms in the pathogenesis of numerous dermatologic diseases including AD. S. aureus biofilms have been found to colonize the eccrine ducts of AD skin, and these biofilms influence secretion of keratinocyte cytokines and trigger differentiation and apoptosis of keratinocytes. These activities may act to disrupt barrier function and promote disease pathogenesis as well as allergen sensitization.SummaryFormation of biofilm is a successful strategy that protects the bacteria from environmental danger, antibiotics, and phagocytosis, enabling chronic persistence in the host. An increasing number of S. aureus skin isolates are resistant to conventional antibiotics, and staphylococcal biofilm communities are prevalent on the skin of individuals with AD. Staphylococcal colonization of the skin impacts skin barrier function and plays multiple important roles in AD pathogenesis.

Collaboration


Dive into the Andrew B. Herr's collaboration.

Top Co-Authors

Avatar

Deborah G. Conrady

University of Cincinnati Academic Health Center

View shared research outputs
Top Co-Authors

Avatar

Tammy Gonzalez

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jeanette L. C. Miller

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kirby R. Lattwein

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Klazina Kooiman

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge