Network


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

Hotspot


Dive into the research topics where Ethan C. Settembre is active.

Publication


Featured researches published by Ethan C. Settembre.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Near-atomic resolution using electron cryomicroscopy and single-particle reconstruction

Xing Zhang; Ethan C. Settembre; Chen Xu; Philip R. Dormitzer; Richard Bellamy; Stephen C. Harrison; Nikolaus Grigorieff

Electron cryomicroscopy (cryo-EM) yields images of macromolecular assemblies and their components, from which 3D structures can be determined, by using an image processing method commonly known as “single-particle reconstruction.” During the past two decades, this technique has become an important tool for 3D structure determination, but it generally has not been possible to determine atomic models. In principle, individual molecular images contain high-resolution information contaminated by a much higher level of noise. In practice, it has been unclear whether current averaging methods are adequate to extract this information from the background. We present here a reconstruction, obtained by using recently developed image processing methods, of the rotavirus inner capsid particle (“double-layer particle” or DLP) at a resolution suitable for interpretation by an atomic model. The result establishes single-particle reconstruction as a high-resolution technique. We show by direct comparison that the cryo-EM reconstruction of viral protein 6 (VP6) of the rotavirus DLP is similar in clarity to a 3.8-Å resolution map obtained from x-ray crystallography. At this resolution, most of the amino acid side chains produce recognizable density. The icosahedral symmetry of the particle was an important factor in achieving this resolution in the cryo-EM analysis, but as the size of recordable datasets increases, single-particle reconstruction also is likely to yield structures at comparable resolution from samples of much lower symmetry. This potential has broad implications for structural cell biology.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Structural basis for immunization with postfusion respiratory syncytial virus fusion F glycoprotein (RSV F) to elicit high neutralizing antibody titers

Kurt Swanson; Ethan C. Settembre; Christine A. Shaw; Antu K. Dey; Rino Rappuoli; Christian W. Mandl; Philip R. Dormitzer; Andrea Carfi

Respiratory syncytial virus (RSV), the main cause of infant bronchiolitis, remains a major unmet vaccine need despite more than 40 years of vaccine research. Vaccine candidates based on a chief RSV neutralization antigen, the fusion (F) glycoprotein, have foundered due to problems with stability, purity, reproducibility, and potency. Crystal structures of related parainfluenza F glycoproteins have revealed a large conformational change between the prefusion and postfusion states, suggesting that postfusion F antigens might not efficiently elicit neutralizing antibodies. We have generated a homogeneous, stable, and reproducible postfusion RSV F immunogen that elicits high titers of neutralizing antibodies in immunized animals. The 3.2-Å X-ray crystal structure of this substantially complete RSV F reveals important differences from homology-based structural models. Specifically, the RSV F crystal structure demonstrates the exposure of key neutralizing antibody binding sites on the surface of the postfusion RSV F trimer. This unanticipated structural feature explains the engineered RSV F antigens efficiency as an immunogen. This work illustrates how structural-based antigen design can guide the rational optimization of candidate vaccine antigens.


The EMBO Journal | 2014

Atomic model of an infectious rotavirus particle.

Ethan C. Settembre; James Z. Chen; Philip R. Dormitzer; Nikolaus Grigorieff; Stephen C. Harrison

Non‐enveloped viruses of different types have evolved distinct mechanisms for penetrating a cellular membrane during infection. Rotavirus penetration appears to occur by a process resembling enveloped‐virus fusion: membrane distortion linked to conformational changes in a viral protein. Evidence for such a mechanism comes from crystallographic analyses of fragments of VP4, the rotavirus‐penetration protein, and infectivity analyses of structure‐based VP4 mutants. We describe here the structure of an infectious rotavirus particle determined by electron cryomicroscopy (cryoEM) and single‐particle analysis at about 4.3 Å resolution. The cryoEM image reconstruction permits a nearly complete trace of the VP4 polypeptide chain, including the positions of most side chains. It shows how the two subfragments of VP4 (VP8* and VP5*) retain their association after proteolytic cleavage, reveals multiple structural roles for the β‐barrel domain of VP5*, and specifies interactions of VP4 with other capsid proteins. The virion model allows us to integrate structural and functional information into a coherent mechanism for rotavirus entry.


Science | 2009

Structure of Rotavirus Outer-Layer Protein VP7 Bound with a Neutralizing Fab

Scott T. Aoki; Ethan C. Settembre; Shane D. Trask; Harry B. Greenberg; Stephen C. Harrison; Philip R. Dormitzer

Rotavirus Rumbled Rotavirus infection is the primary cause of severe diarrhea in infants. For the virus to enter cells, a Ca2+-stabilized trimer of the outer layer protein VP7 must be dissociated. Aoki et al. (p. 1444) report the structure of the VP7 trimer in complex with the Fab fragment of a neutralizing monoclonal antibody. Based on the structure and an analysis of positions of neutralization escape mutations, the authors propose that many neutralizing antibodies inhibit cell entry by stabilizing the VP7 trimer even at low calcium concentrations. A disulfide-linked trimer was then produced that is a potential subunit immunogen. Binding of neutralizing antibodies to rotavirus stabilizes coat-protein trimers and blocks cell entry. Rotavirus outer-layer protein VP7 is a principal target of protective antibodies. Removal of free calcium ions (Ca2+) dissociates VP7 trimers into monomers, releasing VP7 from the virion, and initiates penetration-inducing conformational changes in the other outer-layer protein, VP4. We report the crystal structure at 3.4 angstrom resolution of VP7 bound with the Fab fragment of a neutralizing monoclonal antibody. The Fab binds across the outer surface of the intersubunit contact, which contains two Ca2+ sites. Mutations that escape neutralization by other antibodies suggest that the same region bears the epitopes of most neutralizing antibodies. The monovalent Fab is sufficient to neutralize infectivity. We propose that neutralizing antibodies against VP7 act by stabilizing the trimer, thereby inhibiting the uncoating trigger for VP4 rearrangement. A disulfide-linked trimer is a potential subunit immunogen.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Molecular interactions in rotavirus assembly and uncoating seen by high-resolution cryo-EM

James Z. Chen; Ethan C. Settembre; Scott T. Aoki; Xing Zhang; A.Richard Bellamy; Philip R. Dormitzer; Stephen C. Harrison; Nikolaus Grigorieff

Rotaviruses, major causes of childhood gastroenteritis, are nonenveloped, icosahedral particles with double-strand RNA genomes. By the use of electron cryomicroscopy and single-particle reconstruction, we have visualized a rotavirus particle comprising the inner capsid coated with the trimeric outer-layer protein, VP7, at a resolution (4 Å) comparable with that of X-ray crystallography. We have traced the VP7 polypeptide chain, including parts not seen in its X-ray crystal structure. The 3 well-ordered, 30-residue, N-terminal “arms” of each VP7 trimer grip the underlying trimer of VP6, an inner-capsid protein. Structural differences between free and particle-bound VP7 and between free and VP7-coated inner capsids may regulate mRNA transcription and release. The Ca2+-stabilized VP7 intratrimer contact region, which presents important neutralizing epitopes, is unaltered upon capsid binding.


EMBO Reports | 2011

Mechanistic implications for LDL receptor degradation from the PCSK9/LDLR structure at neutral pH.

Paola Lo Surdo; Matthew J. Bottomley; Alessandra Calzetta; Ethan C. Settembre; Agostino Cirillo; Shilpa Pandit; Yan G. Ni; Brian K. Hubbard; Ayesha Sitlani; Andrea Carfi

The protein PCSK9 (proprotein convertase subtilisin/kexin type 9) is a key regulator of low‐density lipoprotein receptor (LDLR) levels and cardiovascular health. We have determined the crystal structure of LDLR bound to PCSK9 at neutral pH. The structure shows LDLR in a new extended conformation. The PCSK9 C‐terminal domain is solvent exposed, enabling cofactor binding, whereas the catalytic domain and prodomain interact with LDLR epidermal growth factor(A) and β‐propeller domains, respectively. Thus, PCSK9 seems to hold LDLR in an extended conformation and to interfere with conformational rearrangements required for LDLR recycling.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Preconfiguration of the antigen-binding site during affinity maturation of a broadly neutralizing influenza virus antibody

Aaron G. Schmidt; Huafeng Xu; Amir R. Khan; T O'Donnell; Surender Khurana; L.R King; J Manischewitz; Hana Golding; Pirada Suphaphiphat; Andrea Carfi; Ethan C. Settembre; Philip R. Dormitzer; Thomas B. Kepler; Ruijun Zhang; Ma Moody; Barton F. Haynes; Hua-Xin Liao; David E. Shaw; Stephen C. Harrison

Affinity maturation refines a naive B-cell response by selecting mutations in antibody variable domains that enhance antigen binding. We describe a B-cell lineage expressing broadly neutralizing influenza virus antibodies derived from a subject immunized with the 2007 trivalent vaccine. The lineage comprises three mature antibodies, the unmutated common ancestor, and a common intermediate. Their heavy-chain complementarity determining region inserts into the conserved receptor-binding pocket of influenza HA. We show by analysis of structures, binding kinetics and long time-scale molecular dynamics simulations that antibody evolution in this lineage has rigidified the initially flexible heavy-chain complementarity determining region by two nearly independent pathways and that this preconfiguration accounts for most of the affinity gain. The results advance our understanding of strategies for developing more broadly effective influenza vaccines.


PLOS Pathogens | 2011

Structure of Herpes Simplex Virus Glycoprotein D Bound to the Human Receptor Nectin-1

Paolo Di Giovine; Ethan C. Settembre; Arjun K. Bhargava; Micah A. Luftig; Huan Lou; Gary H. Cohen; Roselyn J. Eisenberg; Claude Krummenacher; Andrea Carfi

Binding of herpes simplex virus (HSV) glycoprotein D (gD) to a cell surface receptor is required to trigger membrane fusion during entry into host cells. Nectin-1 is a cell adhesion molecule and the main HSV receptor in neurons and epithelial cells. We report the structure of gD bound to nectin-1 determined by x-ray crystallography to 4.0 Å resolution. The structure reveals that the nectin-1 binding site on gD differs from the binding site of the HVEM receptor. A surface on the first Ig-domain of nectin-1, which mediates homophilic interactions of Ig-like cell adhesion molecules, buries an area composed by residues from both the gD N- and C-terminal extensions. Phenylalanine 129, at the tip of the loop connecting β-strands F and G of nectin-1, protrudes into a groove on gD, which is otherwise occupied by C-terminal residues in the unliganded gD and by N-terminal residues in the gD/HVEM complex. Notably, mutation of Phe129 to alanine prevents nectin-1 binding to gD and HSV entry. Together these data are consistent with previous studies showing that gD disrupts the normal nectin-1 homophilic interactions. Furthermore, the structure of the complex supports a model in which gD-receptor binding triggers HSV entry through receptor-mediated displacement of the gD C-terminal region.


Emerging microbes & infections | 2013

Rapidly produced SAM ® vaccine against H7N9 influenza is immunogenic in mice

Armin Hekele; Sylvie Bertholet; Jacob Archer; Daniel G. Gibson; Giuseppe Palladino; Luis A. Brito; Gillis Otten; Michela Brazzoli; Scilla Buccato; Alessandra Bonci; Daniele Casini; Domenico Maione; Zhi-Qing Qi; John Gill; Nicky C. Caiazza; Jun Urano; Bolyn Hubby; George F. Gao; Yuelong Shu; Ennio De Gregorio; Christian W. Mandl; Peter W. Mason; Ethan C. Settembre; Jeffrey B. Ulmer; J. Craig Venter; Philip R. Dormitzer; Rino Rappuoli; Andrew Geall

The timing of vaccine availability is essential for an effective response to pandemic influenza. In 2009, vaccine became available after the disease peak, and this has motivated the development of next generation vaccine technologies for more rapid responses. The SAM® vaccine platform, now in pre-clinical development, is based on a synthetic, self-amplifying mRNA, delivered by a synthetic lipid nanoparticle (LNP). When used to express seasonal influenza hemagglutinin (HA), a SAM vaccine elicited potent immune responses, comparable to those elicited by a licensed influenza subunit vaccine preparation. When the sequences coding for the HA and neuraminidase (NA) genes from the H7N9 influenza outbreak in China were posted on a web-based data sharing system, the combination of rapid and accurate cell-free gene synthesis and SAM vaccine technology allowed the generation of a vaccine candidate in 8 days. Two weeks after the first immunization, mice had measurable hemagglutinin inhibition (HI) and neutralizing antibody titers against the new virus. Two weeks after the second immunization, all mice had HI titers considered protective. If the SAM vaccine platform proves safe, potent, well tolerated and effective in humans, fully synthetic vaccine technologies could provide unparalleled speed of response to stem the initial wave of influenza outbreaks, allowing first availability of a vaccine candidate days after the discovery of a new virus.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Structural and biochemical studies of HCMV gH/gL/gO and Pentamer reveal mutually exclusive cell entry complexes

Claudio Ciferri; Sumana Chandramouli; Danilo Donnarumma; Pavel A. Nikitin; Michael A. Cianfrocco; Rachel Gerrein; Adam L. Feire; Susan W. Barnett; Anders E. Lilja; Rino Rappuoli; Nathalie Norais; Ethan C. Settembre; Andrea Carfi

Significance Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and in fetuses after congenital infection. gH/gL/gO and Pentamer are targets for neutralizing antibodies. We show that gO and UL128/UL130/UL131A bind to the same site on gH/gL through formation of a disulfide bond with gL-Cys144. The alternative use of this binding site by either gO or the ULs may provide a mechanism for cell tropism modulation. Our analysis reveals that gH/gL antigenic sites are conserved among gH/gL, gH/gL/gO, and Pentamer, whereas gH/gL/gO- and Pentamer-specific neutralizing antibody-binding sites are located in the gH/gL N terminus protrusion that contains the gO and the UL subunits. These data support the development of vaccines and antibody therapeutics against HCMV. Human cytomegalovirus (HCMV) is a major cause of morbidity and mortality in transplant patients and the leading viral cause of birth defects after congenital infection. The glycoprotein complexes gH/gL/gO and gH/gL/UL128/UL130/UL131A (Pentamer) are key targets of the human humoral response against HCMV and are required for HCMV entry into fibroblasts and endothelial/epithelial cells, respectively. We expressed and characterized soluble forms of gH/gL, gH/gL/gO, and Pentamer. Mass spectrometry and mutagenesis analysis revealed that gL-Cys144 forms disulfide bonds with gO-Cys351 in gH/gL/gO and with UL128-Cys162 in the Pentamer. Notably, Pentamer harboring the UL128-Cys162Ser/gL-Cys144Ser mutations had impaired syncytia formation and reduced interference of HCMV entry into epithelial cells. Electron microscopy analysis showed that HCMV gH/gL resembles HSV gH/gL and that gO and UL128/UL130/UL131A bind to the same site at the gH/gL N terminus. These data are consistent with gH/gL/gO and Pentamer forming mutually exclusive cell entry complexes and reveal the overall location of gH/gL-, gH/gL/gO-, and Pentamer-specific neutralizing antibody binding sites. Our results provide, to our knowledge, the first structural view of gH/gL/gO and Pentamer supporting the development of vaccines and antibody therapeutics against HCMV.

Collaboration


Dive into the Ethan C. Settembre's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen C. Harrison

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge