Network


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

Hotspot


Dive into the research topics where Altaira D. Dearborn is active.

Publication


Featured researches published by Altaira D. Dearborn.


EMBO Reports | 2007

The carboxy-terminal coiled-coil of the RNA polymerase β′-subunit is the main binding site for Gre factors

Marina N. Vassylyeva; Vladimir Svetlov; Altaira D. Dearborn; Sergiy Klyuyev; Irina Artsimovitch; Dmitry G. Vassylyev

Bacterial Gre transcript cleavage factors stimulate the intrinsic endonucleolytic activity of RNA polymerase (RNAP) to rescue stalled transcription complexes. They bind to RNAP and extend their coiled‐coil (CC) domains to the catalytic centre through the secondary channel. Three existing models for the Gre–RNAP complex postulate congruent mechanisms of Gre‐assisted catalysis, while offering conflicting views of the Gre–RNAP interactions. Here, we report the GreB structure of Escherichia coli. The GreB monomers form a triangle with the tip of the amino‐terminal CC of one molecule trapped within the hydrophobic cavity of the carboxy‐terminal domain of a second molecule. This arrangement suggests an analogous model for recruitment to RNAP. Indeed, the β′‐subunit CC located at the rim of the secondary channel has conserved hydrophobic residues at its tip. We show that substitutions of these residues and those in the GreB C‐terminal domain cavity confer defects in GreB activity and binding to RNAP, and present a plausible model for the RNAP–GreB complex.


Journal of Biological Chemistry | 2016

α-Synuclein Amyloid Fibrils with Two Entwined, Asymmetrically Associated, Protofibrils

Altaira D. Dearborn; Joseph S. Wall; Naiqian Cheng; J. Bernard Heymann; Andrey V. Kajava; Jobin Varkey; Ralf Langen; Alasdair C. Steven

Parkinson disease and other progressive neurodegenerative conditions are characterized by the intracerebral presence of Lewy bodies, containing amyloid fibrils of α-synuclein. We used cryo-electron microscopy and scanning transmission electron microscopy (STEM) to study in vitro-assembled fibrils. These fibrils are highly polymorphic. Focusing on twisting fibrils with an inter-crossover spacing of 77 nm, our reconstructions showed them to consist of paired protofibrils. STEM mass per length data gave one subunit per 0.47 nm axial rise per protofibril, consistent with a superpleated β-structure. The STEM images show two thread-like densities running along each of these fibrils, which we interpret as ladders of metal ions. These threads confirmed the two-protofibril architecture of the 77-nm twisting fibrils and allowed us to identify this morphotype in STEM micrographs. Some other, but not all, fibril morphotypes also exhibit dense threads, implying that they also present a putative metal binding site. We propose a molecular model for the protofibril and suggest that polymorphic variant fibrils have different numbers of protofibrils that are associated differently.


Virology | 2012

The roles of SaPI1 proteins gp7 (CpmA) and gp6 (CpmB) in capsid size determination and helper phage interference.

Priyadarshan K. Damle; Erin A. Wall; Michael S. Spilman; Altaira D. Dearborn; Geeta Ram; Richard P. Novick; Terje Dokland; Gail E. Christie

SaPIs are molecular pirates that exploit helper bacteriophages for their own high frequency mobilization. One striking feature of helper exploitation by SaPIs is redirection of the phage capsid assembly pathway to produce smaller phage-like particles with T=4 icosahedral symmetry rather than T=7 bacteriophage capsids. Small capsids can accommodate the SaPI genome but not that of the helper phage, leading to interference with helper propagation. Previous studies identified two proteins encoded by the prototype element SaPI1, gp6 and gp7, in SaPI1 procapsids but not in mature SaPI1 particles. Dimers of gp6 form an internal scaffold, aiding fidelity of small capsid assembly. Here we show that both SaPI1 gp6 (CpmB) and gp7 (CpmA) are necessary and sufficient to direct small capsid formation. Surprisingly, failure to form small capsids did not restore wild-type levels of helper phage growth, suggesting an additional role for these SaPI1 proteins in phage interference.


Journal of Molecular Biology | 2011

A conformational switch involved in maturation of Staphylococcus aureus bacteriophage 80α capsids.

Michael S. Spilman; Altaira D. Dearborn; Jenny R. Chang; Priyadarshan K. Damle; Gail E. Christie; Terje Dokland

Bacteriophages are involved in many aspects of the spread and establishment of virulence factors in Staphylococcus aureus, including the mobilization of genetic elements known as S. aureus pathogenicity islands (SaPIs), which carry genes for superantigen toxins and other virulence factors. SaPIs are packaged into phage-like transducing particles using proteins supplied by the helper phage. We have used cryo-electron microscopy and icosahedral reconstruction to determine the structures of the procapsid and the mature capsid of 80α, a bacteriophage that can mobilize several different SaPIs. The 80α capsid has T=7 icosahedral symmetry with the capsid protein organized into pentameric and hexameric clusters that interact via prominent trimeric densities. The 80α capsid protein was modeled based on the capsid protein fold of bacteriophage HK97 and fitted into the 80α reconstructions. The models show that the trivalent interactions are mediated primarily by a 22-residue β hairpin structure called the P loop that is not found in HK97. Capsid expansion is associated with a conformational switch in the spine helix that is propagated throughout the subunit, unlike the domain rotation mechanism in phage HK97 or P22.


Journal of Structural Biology | 2012

Structure and size determination of bacteriophage P2 and P4 procapsids: Function of size responsiveness mutations

Altaira D. Dearborn; Pasi Laurinmäki; Preethi Chandramouli; Cynthia M. Rodenburg; Sifang Wang; Sarah J. Butcher; Terje Dokland

Bacteriophage P4 is dependent on structural proteins supplied by a helper phage, P2, to assemble infectious virions. Bacteriophage P2 normally forms an icosahedral capsid with T=7 symmetry from the gpN capsid protein, the gpO scaffolding protein and the gpQ portal protein. In the presence of P4, however, the same structural proteins are assembled into a smaller capsid with T=4 symmetry. This size determination is effected by the P4-encoded protein Sid, which forms an external scaffold around the small P4 procapsids. Size responsiveness (sir) mutants in gpN fail to assemble small capsids even in the presence of Sid. We have produced large and small procapsids by co-expression of gpN with gpO and Sid, respectively, and applied cryo-electron microscopy and three-dimensional reconstruction methods to visualize these procapsids. gpN has an HK97-like fold and interacts with Sid in an exposed loop where the sir mutations are clustered. The T=7 lattice of P2 has dextro handedness, unlike the laevo lattices of other phages with this fold observed so far.


Bacteriophage | 2012

Mobilization of pathogenicity islands by Staphylococcus aureus strain Newman bacteriophages.

Altaira D. Dearborn; Terje Dokland

Staphylococcus aureus pathogenicity islands (SaPIs) are mobile genetic elements that encode virulence factors and depend on helper phages for their mobilization. Such mobilization is specific and depends on the ability of a phage protein to inactivate the SaPI repressor Stl. Phage 80α can mobilize several SaPIs, including SaPI1 and SaPIbov1, via its Sri and Dut proteins, respectively. In many cases, the capsids formed in the presence of the SaPI are smaller than those normally produced by the phage. Two SaPI-encoded proteins, CpmA and CpmB, are involved in this size determination process. S. aureus strain Newman contains four prophages, named φNM1 through φNM4. Phages φNM1 and φNM2 are very similar to phage 80α in the structural genes, and encode almost identical Sri proteins, while their Dut proteins are highly divergent. We show that φNM1 and φNM2 are able to mobilize both SaPI1 and SaPIbov1 and yield infectious transducing particles. The majority of the capsids formed in all cases are small, showing that both SaPIs can redirect the capsid size of both φNM1 and φNM2.


eLife | 2017

Competing scaffolding proteins determine capsid size during mobilization of Staphylococcus aureus pathogenicity islands

Altaira D. Dearborn; Erin A. Wall; James L. Kizziah; Laura Klenow; Laura K. Parker; Keith A. Manning; Michael S. Spilman; John M. Spear; Gail E. Christie; Terje Dokland

Staphylococcus aureus pathogenicity islands (SaPIs), such as SaPI1, exploit specific helper bacteriophages, like 80α, for their high frequency mobilization, a process termed ‘molecular piracy’. SaPI1 redirects the helper’s assembly pathway to form small capsids that can only accommodate the smaller SaPI1 genome, but not a complete phage genome. SaPI1 encodes two proteins, CpmA and CpmB, that are responsible for this size redirection. We have determined the structures of the 80α and SaPI1 procapsids to near-atomic resolution by cryo-electron microscopy, and show that CpmB competes with the 80α scaffolding protein (SP) for a binding site on the capsid protein (CP), and works by altering the angle between capsomers. We probed these interactions genetically and identified second-site suppressors of lethal mutations in SP. Our structures show, for the first time, the detailed interactions between SP and CP in a bacteriophage, providing unique insights into macromolecular assembly processes.


Journal of Biological Chemistry | 2017

Chimeric Rabbit/Human Fab Antibodies Against the Hepatitis B e-antigen and Their Potential Applications in Assay, Characterization and Therapy

Xiaolei Zhuang; Norman R. Watts; Ira Palmer; Joshua D. Kaufman; Altaira D. Dearborn; Joni L. Trenbeath; Elif Eren; Alasdair C. Steven; Christoph Rader; Paul T. Wingfield

Hepatitis B virus (HBV) infection afflicts millions worldwide, causing cirrhosis and liver cancer. HBV e-antigen (HBeAg), a clinical marker for disease severity, is a soluble variant of the viral capsid protein. HBeAg is not required for viral replication but is implicated in establishing immune tolerance and chronic infection. The structure of recombinant e-antigen (rHBeAg) was recently determined, yet to date, the exact nature and quantitation of HBeAg still remain uncertain. Here, to further characterize HBeAg, we used phage display to produce a panel of chimeric rabbit/human monoclonal antibody fragments (both Fab and scFv) against rHBeAg. Several of the Fab/scFv, expressed in Escherichia coli, had unprecedentedly high binding affinities (Kd ∼10−12 m) and high specificity. We used Fab/scFv in the context of an enzyme-linked immunosorbent assay (ELISA) for HBeAg quantification, which we compared with commercially available kits and verified with seroconversion panels, the WHO HBeAg standard, rHBeAg, and patient plasma samples. We found that the specificity and sensitivity are superior to those of existing commercial assays. To identify potential fine differences between rHBeAg and HBeAg, we used these Fabs in microscale immunoaffinity chromatography to purify HBeAg from individual patient plasmas. Western blotting and MS results indicated that rHBeAg and HBeAg are essentially structurally identical, although HBeAg from different patients exhibits minor carboxyl-terminal heterogeneity. We discuss several potential applications for the humanized Fab/scFv.


Biochemistry | 2017

Expression and Purification of ZASP Subdomains and Clinically Important Isoforms: High-Affinity Binding to G-Actin

Norman R. Watts; Xiaolei Zhuang; Joshua D. Kaufman; Ira Palmer; Altaira D. Dearborn; Stephen Coscia; Yotam Blech-Hermoni; Caterina Alfano; Annalisa Pastore; Ami Mankodi; Paul T. Wingfield

Z-disc-associated, alternatively spliced, PDZ motif-containing protein (ZASP) is a principal component of the sarcomere. The three prevalent isoforms of ZASP in skeletal muscle are generated by alternative splicing of exons 9 and 10. The long isoforms, either having (ZASP-L) or lacking exon 10 (ZASP-LΔex10), include an N-terminal PDZ domain, an actin-binding region (ABR) with a conserved motif (ZM), and three C-terminal LIM domains. The short isoform (ZASP-S) lacks the LIM domains. Mutations, A147T and A165V, within the ZM of ZASP-LΔex10 cause myofibrillar myopathy, but the mechanism is unknown. We have prepared these proteins, their ABR, and the respective mutant variants in recombinant form, characterized them biophysically, and analyzed their actin-binding properties by surface plasmon resonance and electron microscopy. All the proteins were physically homogeneous and monomeric and had circular dichroic spectra consistent with partially folded conformations. Comparison of the NMR HSQC spectra of ZASP-S and the PDZ domain showed that the ABR is unstructured. ZASP-S and its mutant variants and ZASP-LΔex10 all bound to immobilized G-actin with high affinity (Kd ≈ 10-8 to 10-9 M). Constructs of the isolated actin-binding region missing exon 10 (ABRΔ10) bound with lower affinity (Kd ≈ 10-7 M), but those retaining exon 10 (ABR+10) did so only weakly (Kd ≈ 10-5 M). ZASP-S, and the ABRΔ10, also induced F-actin and array formation, even in conditions of low ionic strength and in the absence of KCl and Mg2+ ions. Interestingly, the ZM mutations A147T and A165V did not affect any of the results described above.


Structure | 2018

Structures of Hepatitis B Virus Core- and e-Antigen Immune Complexes Suggest Multi-point Inhibition.

Elif Eren; Norman R. Watts; Altaira D. Dearborn; Ira Palmer; Joshua D. Kaufman; Alasdair C. Steven; Paul T. Wingfield

Hepatitis B virus (HBV) is the leading cause of liver disease worldwide. While an adequate vaccine is available, current treatment options are limited, not highly effective, and associated with adverse effects, encouraging the development of alternative therapeutics. The HBV core gene encodes two different proteins: core, which forms the viral nucleocapsid, and pre-core, which serves as an immune modulator with multiple points of action. The two proteins mostly have the same sequence, although they differ at their N and C termini and in their dimeric arrangements. Previously, we engineered two human-framework antibody fragments (Fab/scFv) with nano- to picomolar affinities for both proteins. Here, by means of X-ray crystallography, analytical ultracentrifugation, and electron microscopy, we demonstrate that the antibodies have non-overlapping epitopes and effectively block biologically important assemblies of both proteins. These properties, together with the anticipated high tolerability and long half-lives of the antibodies, make them promising therapeutics.

Collaboration


Dive into the Altaira D. Dearborn's collaboration.

Top Co-Authors

Avatar

Terje Dokland

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Gail E. Christie

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Alasdair C. Steven

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Michael S. Spilman

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Erin A. Wall

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Priyadarshan K. Damle

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Ira Palmer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

James L. Kizziah

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Jenny R. Chang

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Joshua D. Kaufman

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge