Network


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

Hotspot


Dive into the research topics where Florence M. Brunel is active.

Publication


Featured researches published by Florence M. Brunel.


Journal of Virology | 2007

An Affinity-Enhanced Neutralizing Antibody against the Membrane-Proximal External Region of Human Immunodeficiency Virus Type 1 gp41 Recognizes an Epitope between Those of 2F5 and 4E10

Josh D. Nelson; Florence M. Brunel; Richard Jensen; Emma T. Crooks; Rosa M.F. Cardoso; Meng Wang; Ann J. Hessell; Ian A. Wilson; James M. Binley; Philip E. Dawson; Dennis R. Burton; Michael B. Zwick

ABSTRACT The membrane-proximal external region (MPER) of human immunodeficiency virus type 1 (HIV-1) gp41 bears the epitopes of two broadly neutralizing antibodies (Abs), 2F5 and 4E10, making it a target for vaccine design. A third Ab, Fab Z13, had previously been mapped to an epitope that overlaps those of 2F5 and 4E10 but only weakly neutralizes a limited set of primary isolates. Here, libraries of Fab Z13 variants displayed on phage were engineered and affinity selected against an MPER peptide and recombinant gp41. A high-affinity variant, designated Z13e1, was isolated and found to be ∼100-fold improved over the parental Fab not only in binding affinity for the MPER antigens but also in neutralization potency against sensitive HIV-1. Alanine scanning of MPER residues 664 to 680 revealed that N671 and D674 are crucial for peptide recognition as well as for the neutralization of HIV-1 by Z13e1. Ab competition studies and truncation of MPER peptides indicate that Z13e1 binds with high affinity to an epitope between and overlapping with those of 2F5 and 4E10, with the minimal peptide epitope WASLWNWFDITN. Still, Z13e1 remained about an order of magnitude less potent than 4E10 against several isolates of pseudotyped HIV-1. The sum of our molecular analyses with Z13e1 suggests that the segment on the MPER of gp41 between the 2F5 and 4E10 epitopes is exposed on the functional envelope trimer but that access to the specific Z13e1 epitope within this segment is limited. Thus, the ability of MPER-bearing immunogens to elicit potent HIV-1-neutralizing Abs may depend in part on recapitulating the particular constraints that the functional envelope trimer imposes on the segment of the MPER to which Z13e1 binds.


Journal of Virology | 2006

Structure-Function Analysis of the Epitope for 4E10, a Broadly Neutralizing Human Immunodeficiency Virus Type 1 Antibody

Florence M. Brunel; Michael B. Zwick; Rosa M.F. Cardoso; Josh D. Nelson; Ian A. Wilson; Dennis R. Burton; Philip E. Dawson

ABSTRACT The human immunodeficiency virus type 1 (HIV-1) neutralizing antibody 4E10 binds to a linear, highly conserved epitope within the membrane-proximal external region of the HIV-1 envelope glycoprotein gp41. We have delineated the peptide epitope of the broadly neutralizing 4E10 antibody to gp41 residues 671 to 683, using peptides with different lengths encompassing the previously suggested core epitope (NWFDIT). Peptide binding to the 4E10 antibody was assessed by competition enzyme-linked immunosorbent assay, and the Kd values of selected peptides were determined using surface plasmon resonance. An Ala scan of the epitope indicated that several residues, W672, F673, and T676, are essential (>1,000-fold decrease in binding upon replacement with alanine) for 4E10 recognition. In addition, five other residues, N671, D674, I675, W680, and L679, make significant contributions to 4E10 binding. In general, the Ala scan results agree well with the recently reported crystal structure of 4E10 in complex with a 13-mer peptide and with our circular dichroism analyses. Neutralization competition assays confirmed that the peptide NWFDITNWLWYIKKKK-NH2 could effectively inhibit 4E10 neutralization. Finally, to limit the conformational flexibility of the peptides, helix-promoting 2-aminoisobutyric acid residues and helix-inducing tethers were incorporated. Several peptides have significantly improved affinity (>1,000-fold) over the starting peptide and, when used as immunogens, may be more likely to elicit 4E10-like neutralizing antibodies. Hence, this study represents the first stage toward iterative development of a vaccine based on the 4E10 epitope.


Nano Letters | 2010

Hydrazone ligation strategy to assemble multifunctional viral nanoparticles for cell imaging and tumor targeting

Florence M. Brunel; John D. Lewis; Giuseppe Destito; Nicole F. Steinmetz; Marianne Manchester; Heidi Stuhlmann; Philip E. Dawson

Multivalent nanoparticle platforms are attractive for biomedical applications because of their improved target specificity, sensitivity, and solubility. However, their controlled assembly remains a considerable challenge. An efficient hydrazone ligation chemistry was applied to the assembly of Cowpea mosaic virus (CPMV) nanoparticles with individually tunable levels of a VEGFR-1 ligand and a fluorescent PEGylated peptide. The nanoparticles recognized VEGFR-1 on endothelial cell lines and VEGFR1-expressing tumor xenografts in mice, validating targeted CPMV as a nanoparticle platform in vivo.


Journal of Virology | 2009

A Conformational Switch in Human Immunodeficiency Virus gp41 Revealed by the Structures of Overlapping Epitopes Recognized by Neutralizing Antibodies

Robert Pejchal; Johannes S. Gach; Florence M. Brunel; Rosa M.F. Cardoso; Robyn L. Stanfield; Philip E. Dawson; Dennis R. Burton; Michael B. Zwick; Ian A. Wilson

ABSTRACT The membrane-proximal external region (MPER) of the human immunodeficiency virus (HIV) envelope glycoprotein (gp41) is critical for viral fusion and infectivity and is the target of three of the five known broadly neutralizing HIV type 1 (HIV-1) antibodies, 2F5, Z13, and 4E10. Here, we report the crystal structure of the Fab fragment of Z13e1, an affinity-enhanced variant of monoclonal antibody Z13, in complex with a 12-residue peptide corresponding to the core epitope (W670NWFDITN677) at 1.8-Å resolution. The bound peptide adopts an S-shaped conformation composed of two tandem, perpendicular helical turns. This conformation differs strikingly from the α-helical structure adopted by an overlapping MPER peptide bound to 4E10. Z13e1 binds to an elbow in the MPER at the membrane interface, making relatively few interactions with conserved aromatics (Trp672 and Phe673) that are critical for 4E10 recognition. The comparison of the Z13e1 and 4E10 epitope structures reveals a conformational switch such that neutralization can occur by the recognition of the different conformations and faces of the largely amphipathic MPER. The Z13e1 structure provides significant new insights into the dynamic nature of the MPER, which likely is critical for membrane fusion, and it has significant implications for mechanisms of HIV-1 neutralization by MPER antibodies and for the design of HIV-1 immunogens.


Analytical Chemistry | 2009

Multiplex Charge-Transfer Interactions between Quantum Dots and Peptide-Bridged Ruthenium Complexes

Igor L. Medintz; Dorothy Farrell; Kimihiro Susumu; Scott A. Trammell; Jeffrey R. Deschamps; Florence M. Brunel; Philip E. Dawson; Hedi Mattoussi

Simultaneous detection of multiple independent fluorescent signals or signal multiplexing has the potential to significantly improve bioassay throughput and to allow visualization of concurrent cellular events. Applications based on signal multiplexing, however, remain hard to achieve in practice due to difficulties in both implementing hardware and the photophysical liabilities associated with available organic dye and protein fluorophores. Here, we used charge-transfer interactions between luminescent semiconductor quantum dots (QDs) and proximal redox complexes to demonstrate controlled quenching of QD photoemission in a multiplexed format. In particular, we show that, because of the ability of the Ru complex to effectively interact with CdSe-ZnS QDs emitting over a broad window of the optical spectrum, higher orders of multiplexed quenching can be achieved in a relatively facile manner. Polyhistidine-appended peptides were site-specifically labeled with a redox-active ruthenium (Ru) phenanthroline complex and self-assembled onto QDs, resulting in controlled quenching of the QD emission. Different QD colors either alone or coupled to Ru-phen-peptide were then mixed together and optically interrogated. Composite spectra collected from mixtures ranging from four up to eight distinct QD colors were deconvoluted, and the individual QD photoluminescence (PL) loss due to charge transfer was quantified. The current multiplexing modality provides a simpler format for exploiting the narrow, size-tunable QD emissions than that offered by resonance energy transfer; for the latter, higher orders of multiplexing are limited by spectral overlap requirements.


Virology | 2008

Antibody Elicited Against the gp41 N-Heptad Repeat (NHR) Coiled-Coil Can Neutralize HIV-1 with Modest Potency but Non-neutralizing Antibodies Also Bind to NHR Mimetics

Josh D. Nelson; Heather Kinkead; Florence M. Brunel; Dan P. Leaman; Richard Jensen; John M. Louis; Toshiaki Maruyama; Carole A. Bewley; Katherine S. Bowdish; G. Marius Clore; Philip E. Dawson; Shana Frederickson; Rose G. Mage; Douglas D. Richman; Dennis R. Burton; Michael B. Zwick

Following CD4 receptor binding to the HIV-1 envelope spike (Env), the conserved N-heptad repeat (NHR) region of gp41 forms a coiled-coil that is a precursor to the fusion reaction. Although it has been a target of drug and vaccine design, there are few monoclonal antibody (mAb) tools with which to probe the antigenicity and immunogenicity specifically of the NHR coiled-coil. Here, we have rescued HIV-1-neutralizing anti-NHR mAbs from immune phage display libraries that were prepared (i) from b9 rabbits immunized with a previously described mimetic of the NHR coiled-coil, N35(CCG)-N13, and (ii) from an HIV-1 infected individual. We describe a rabbit single-chain Fv fragment (scFv), 8K8, and a human Fab, DN9, which specifically recognize NHR coiled-coils that are unoccupied by peptide corresponding to the C-heptad repeat or CHR region of gp41 (e.g. C34). The epitopes of 8K8 and DN9 were found to partially overlap with that of a previously described anti-NHR mAb, IgG D5; however, 8K8 and DN9 were much more specific than D5 for unoccupied NHR trimers. The mAbs, including a whole IgG 8K8 molecule, neutralized primary HIV-1 of clades B and C in a pseudotyped virus assay with comparable, albeit relatively modest potency. Finally, a human Fab T3 and a rabbit serum (both non-neutralizing) were able to block binding of D5 and 8K8 to a gp41 NHR mimetic, respectively, but not the neutralizing activity of these mAbs. We conclude from these results that NHR coiled-coil analogs of HIV-1 gp41 elicit many Abs during natural infection and through immunization, but that due to limited accessibility to the corresponding region on fusogenic gp41 few can neutralize. Caution is therefore required in targeting the NHR for vaccine design. Nevertheless, the mAb panel may be useful as tools for elucidating access restrictions to the NHR of gp41 and in designing potential improvements to mimetics of receptor-activated Env.


Chemical Communications | 2005

Synthesis of constrained helical peptides by thioether ligation: application to analogs of gp41.

Florence M. Brunel; Philip E. Dawson

We present a straightforward and high yielding method to synthesize constrained helical peptides via thioether ligation; this method represents an attractive alternative to the formation of lactam bridge constraints to induce helicity in peptides.


Proceedings of SPIE | 2008

Specific cellular delivery and intracellular fate of quantum dot-peptide and quantum dot-polymer nanoassemblies

James B. Delehanty; Christopher E. Bradburne; Igor L. Medintz; Dorothy Farrell; Thomas Pons; Florence M. Brunel; Philip E. Dawson; Hedi Mattoussi

Luminescent semiconductor quantum dots (QDs) possess several unique optical and spectroscopic properties that are of great interest and promise in biology. These properties suggest that QDs will be integral to the development of the next generation of biosensors capable of detecting molecular processes in both living and fixed cells. We are developing robust and facile delivery schemes for the selective intracellular delivery of QD-based nanoassemblies. These schemes are based upon the self-assembly and subsequent cellular uptake of QD-peptide and QD-polymer bioconjugates. The QD-peptide structures are generated by the self-assembly of the peptide onto CdSe-ZnS core-shell QDs via metal ion coordination between the peptides polyhistidine motif and the Zn-rich QD shell. The polymer-based QD assemblies are formed via the electrostatic interaction of aqueous cationic liposomes with available carboxylate moieties on the QD surface ligands. Cellular delivery experiments utilizing both delivery schemes will be presented. The advantages and disadvantages of each approach will be discussed, including the intracellular fate and stability of the QD-nanoassemblies.


Proceedings of SPIE | 2007

Selective cellular delivery of self-assembled quantum dot-peptide bioconjugates

James B. Delehanty; Igor L. Medintz; Thomas Pons; Philip E. Dawson; Florence M. Brunel; Hedi Mattoussi

We demonstrate the selective delivery of self-assembled luminescent semiconductor quantum dot (QD)-peptide bioconjugates into several eukaryotic cell lines. A 23-mer hetero-bifunctional peptide bearing a positively-charged oligoarginine domain and a terminal polyhistidine tract was synthesized and used to mediate the cellular internalization of the QD-bioconjugates. The polyhistidine tract allows the peptide to self-assemble onto the QD surface via metal-ion coordination while the oligoarginine domain mediates the specific uptake of the QD-bioconjugates via electrostatic interactions with cell surface receptors. In both HEK 293T/17 and COS-1 cells, this peptide-mediated delivery is concentration-dependent in terms of both the QD concentration and the peptide:QD ratio. Intracellularly, the QD signal is punctate in appearance and some, but not all, of the QDs are located within recycling endosomes as evidenced by their colocalization with transferrin. In both cell lines, the QD-bioconjugates elicit minimal cytotoxicity within the timeframe required for adequate cellular uptake. The specificity of this delivery strategy is demonstrated by performing a multicolor QD labeling, wherein the presence or absence of the peptide on the QD surface controls cellular uptake.


Biomedical optics | 2006

Quantum dot based nanosensors designed for proteolytic monitoring

Igor L. Medintz; Aaron R. Clapp; Florence M. Brunel; Ellen R. Goldman; Eddie L. Chang; Phillip E. Dawson; Hedi Mattoussi

We have previously assembled QD-based fluorescence resonance energy transfer (FRET) sensors specific for the sugar nutrient maltose and the explosive TNT. These sensors utilize several inherent benefits of QDs as FRET donors. In this report, we show that QD-FRET based sensors can also function in the monitoring of proteolytic enzyme activity. We utilize a QD with multiple dye-labeled proteins attached to the surface as a substrate for a prototypical protease. We then demonstrate how this strategy can be extended to detect protease activity by utilizing a dye-labeled peptide attached to the QD as a proteolytic substrate. Self-assembly of the peptide-dye on the QD brings the dye in close proximity to the QD and result in efficient FRET. Addition of a proteolytic enzyme that specifically recognizes and cleaves the peptide alters the FRET signature of the sensor in a concentration-dependent manner. Both qualitative and quantitative data can be derived from these sensors. The potential benefits of this type of QD sensing strategy are discussed.

Collaboration


Dive into the Florence M. Brunel's collaboration.

Top Co-Authors

Avatar

Philip E. Dawson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Hedi Mattoussi

Florida State University

View shared research outputs
Top Co-Authors

Avatar

Igor L. Medintz

United States Naval Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Dennis R. Burton

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Michael B. Zwick

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ian A. Wilson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Thomas Pons

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

James B. Delehanty

United States Naval Research Laboratory

View shared research outputs
Top Co-Authors

Avatar

Rosa M.F. Cardoso

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dorothy Farrell

United States Naval Research Laboratory

View shared research outputs
Researchain Logo
Decentralizing Knowledge