Network


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

Hotspot


Dive into the research topics where Daryll Vanover is active.

Publication


Featured researches published by Daryll Vanover.


Journal of Virology | 2014

Structural Analysis of Respiratory Syncytial Virus Reveals the Position of M2-1 between the Matrix Protein and the Ribonucleoprotein Complex

Gabriella Kiss; Jens M. Holl; G Williams; Eric Alonas; Daryll Vanover; Aaron W. Lifland; Manasa Gudheti; Ricardo C. Guerrero-Ferreira; Vinod Nair; Barney S. Graham; Philip J. Santangelo; Elizabeth R. Wright

ABSTRACT Respiratory syncytial virus (RSV), a member of the Paramyxoviridae family of nonsegmented, negative-sense, single-stranded RNA genome viruses, is a leading cause of lower respiratory tract infections in infants, young children, and the elderly or immunocompromised. There are many open questions regarding the processes that regulate human RSV (hRSV) assembly and budding. Here, using cryo-electron tomography, we identified virus particles that were spherical, filamentous, and asymmetric in structure, all within the same virus preparation. The three particle morphologies maintained a similar organization of the surface glycoproteins, matrix protein (M), M2-1, and the ribonucleoprotein (RNP). RNP filaments were traced in three dimensions (3D), and their total length was calculated. The measurements revealed the inclusion of multiple full-length genome copies per particle. RNP was associated with the membrane whenever the M layer was present. The amount of M coverage ranged from 24% to 86% in the different morphologies. Using fluorescence light microscopy (fLM), direct stochastic optical reconstruction microscopy (dSTORM), and a proximity ligation assay (PLA), we provide evidence illustrating that M2-1 is located between RNP and M in isolated viral particles. In addition, regular spacing of the M2-1 densities was resolved when hRSV viruses were imaged using Zernike phase contrast (ZPC) cryo-electron tomography. Our studies provide a more complete characterization of the hRSV virion structure and substantiation that M and M2-1 regulate virus organization. IMPORTANCE hRSV is a leading cause of lower respiratory tract infections in infants and young children as well as elderly or immunocompromised individuals. We used cryo-electron tomography and Zernike phase contrast cryo-electron tomography to visualize populations of purified hRSV in 3D. We observed the three distinct morphologies, spherical, filamentous, and asymmetric, which maintained comparable organizational profiles. Depending on the virus morphology examined, the amount of M ranged from 24% to 86%. We complemented the cryo-imaging studies with fluorescence microscopy, dSTORM, and a proximity ligation assay to provide additional evidence that M2-1 is incorporated into viral particles and is positioned between M and RNP. The results highlight the impact of M and M2-1 on the regulation of hRSV organization.


ACS Nano | 2014

Combining Single RNA Sensitive Probes with Subdiffraction-Limited and Live-Cell Imaging Enables the Characterization of Virus Dynamics in Cells

Eric Alonas; Aaron W. Lifland; Manasa Gudheti; Daryll Vanover; Jeenah Jung; Chiara Zurla; Jonathan L. Kirschman; Vincent F. Fiore; Alison M. Douglas; Thomas H. Barker; Elizabeth R. Wright; James E. Crowe; Philip J. Santangelo

The creation of fluorescently labeled viruses is currently limited by the length of imaging observation time (e.g., labeling an envelope protein) and the rescue of viral infectivity (e.g., encoding a GFP protein). Using single molecule sensitive RNA hybridization probes delivered to the cytoplasm of infected cells, we were able to isolate individual, infectious, fluorescently labeled human respiratory syncytial virus virions. This was achieved without affecting viral mRNA expression, viral protein expression, or infectivity. Measurements included the characterization of viral proteins and genomic RNA in a single virion using dSTORM, the development of a GFP fusion assay, and the development of a pulse-chase assay for viral RNA production that allowed for the detection of both initial viral RNA and nascent RNA production at designated times postinfection. Live-cell measurements included imaging and characterization of filamentous virion fusion and the quantification of virus replication within the same cell over an eight-hour period. Using probe-labeled viruses, individual viral particles can be characterized at subdiffraction-limited resolution, and viral infections can be quantified in single cells over an entire cycle of replication. The implication of this development is that MTRIP labeling of viral RNA during virus assembly has the potential to become a general methodology for the labeling and study of many important RNA viruses.


Nano Letters | 2018

A Direct Comparison of in Vitro and in Vivo Nucleic Acid Delivery Mediated by Hundreds of Nanoparticles Reveals a Weak Correlation

Kalina Paunovska; Cory D. Sago; Christopher M. Monaco; William H. Hudson; Marielena Gamboa Castro; Tobi G. Rudoltz; Sujay Kalathoor; Daryll Vanover; Philip J. Santangelo; Rafi Ahmed; Anton V. Bryksin; James E. Dahlman

Endothelial cells and macrophages play active roles in disease and as a result are important targets for nucleic acid therapies. While thousands of chemically distinct lipid nanoparticles (LNPs) can be synthesized to deliver nucleic acids, studying more than a few LNPs in vivo is challenging. As a result, it is difficult to understand how nanoparticles target these cells in vivo. Using high throughput LNP barcoding, we quantified how well LNPs delivered DNA barcodes to endothelial cells and macrophages in vitro, as well as endothelial cells and macrophages isolated from the lung, heart, and bone marrow in vivo. We focused on two fundamental questions in drug delivery. First, does in vitro LNP delivery predict in vivo LNP delivery? By comparing how 281 LNPs delivered barcodes to endothelial cells and macrophages in vitro and in vivo, we found in vitro delivery did not predict in vivo delivery. Second, does LNP delivery change within the microenvironment of a tissue? We quantified how 85 LNPs delivered barcodes to eight splenic cell populations, and found that cell types derived from myeloid progenitors tended to be targeted by similar LNPs, relative to cell types derived from lymphoid progenitors. These data demonstrate that barcoded LNPs can elucidate fundamental questions about in vivo nanoparticle delivery.


Advanced Healthcare Materials | 2017

Arginine-Rich Peptide-Based mRNA Nanocomplexes Efficiently Instigate Cytotoxic T Cell Immunity Dependent on the Amphipathic Organization of the Peptide.

Vimal Kumar Udhayakumar; Ans De Beuckelaer; Joanne McCaffrey; Cian M. McCrudden; Jonathan L. Kirschman; Daryll Vanover; Lien Van Hoecke; Kenny Roose; Kim Deswarte; Bruno G. De Geest; Stefan Lienenklaus; Philip J. Santangelo; Johan Grooten; Helen O. McCarthy; Stefaan De Koker

To date, the mRNA delivery field has been heavily dominated by lipid-based systems. Reports on the use of nonlipid carriers for mRNA delivery in contrast are rare in the context of mRNA vaccination. This paper describes the potential of a cell-penetrating peptide containing the amphipathic RALA motif to deliver antigen-encoding mRNA to the immune system. RALA condenses mRNA into nanocomplexes that display acidic pH-dependent membrane disruptive properties. RALA mRNA nanocomplexes enable mRNA escape from endosomes and thereby allow expression of mRNA inside the dendritic cell cytosol. Strikingly, RALA mRNA nanocomplexes containing pseudouridine and 5-methylcytidine modified mRNA elicit potent cytolytic T cell responses against the antigenic mRNA cargo and show superior efficacy in doing so when compared to RALA mRNA nanocomplexes containing unmodified mRNA. RALAs unique sequence and structural organization are vital to act as mRNA vaccine vehicle, as arginine-rich peptide variants that lack the RALA motif show reduced mRNA complexation, impaired cellular uptake and lose the ability to transfect dendritic cells in vitro and to evoke T cell immunity in vivo.


Nature Communications | 2017

RSV glycoprotein and genomic RNA dynamics reveal filament assembly prior to the plasma membrane

Daryll Vanover; Daisy V. Smith; Emmeline L. Blanchard; Eric Alonas; Jonathan L. Kirschman; Aaron W. Lifland; Chiara Zurla; Philip J. Santangelo

The human respiratory syncytial virus G protein plays an important role in the entry and assembly of filamentous virions. Here, we report the use of fluorescently labeled soybean agglutinin to selectively label the respiratory syncytial virus G protein in living cells without disrupting respiratory syncytial virus infectivity or filament formation and allowing for interrogations of respiratory syncytial virus virion assembly. Using this approach, we discovered that plasma membrane-bound respiratory syncytial virus G rapidly recycles from the membrane via clathrin-mediated endocytosis. This event is then followed by the dynamic formation of filamentous and branched respiratory syncytial virus particles, and assembly with genomic ribonucleoproteins and caveolae-associated vesicles prior to re-insertion into the plasma membrane. We demonstrate that these processes are halted by the disruption of microtubules and inhibition of molecular motors. Collectively, our results show that for respiratory syncytial virus assembly, viral filaments are produced and loaded with genomic RNA prior to insertion into the plasma membrane.Assembly of filamentous RSV particles is incompletely understood due to a lack of techniques suitable for live-cell imaging. Here Vanover et al. use labeled soybean agglutinin to selectively label RSV G protein and show how filamentous RSV assembly, initiated in the cytoplasm, uses G protein recycled from the plasma membrane.


Nucleic Acids Research | 2017

Characterizing exogenous mRNA delivery, trafficking, cytoplasmic release and RNA-protein correlations at the level of single cells

Jonathan L. Kirschman; Sushma Bhosle; Daryll Vanover; Emmeline L. Blanchard; Kristin H. Loomis; Chiara Zurla; Kathryn Murray; Blaine C. Lam; Philip J. Santangelo

Abstract The use of synthetic messenger ribonucleic acid (mRNA) to express specific proteins is a highly promising therapeutic and vaccine approach that avoids many safety issues associated with viral or DNA-based systems. However, in order to optimize mRNA designs and delivery, technology advancements are required to study fundamental mechanisms of mRNA uptake and localization at the single-cell and tissue level. Here, we present a single RNA sensitive fluorescent labeling method which allows us to label and visualize synthetic mRNA without significantly affecting function. This approach enabled single cell characterization of mRNA uptake and release kinetics from endocytic compartments, the measurement of mRNA/protein correlations, and motivated the investigation of mRNA induced cellular stress, all important mechanisms influencing protein production. In addition, we demonstrated this approach can facilitate near-infrared imaging of mRNA localization in vivo and in ex-vivo tissue sections, which will facilitate mRNA trafficking studies in pre-clinical models. Overall, we demonstrate the ability to study fundamental mechanisms necessary to optimize delivery and therapeutic strategies, in order to design the next generation of novel mRNA therapeutics and vaccines.


Bioconjugate Chemistry | 2018

In vitro transcribed messenger RNA vaccines with programmable stimulation of innate immunity

Kristin H. Loomis; Kevin E. Lindsay; Chiara Zurla; Sushma Bhosle; Daryll Vanover; Emmeline L. Blanchard; Jonathan L. Kirschman; Ravi V. Bellamkonda; Philip J. Santangelo

In vitro transcribed (IVT) mRNA is an appealing platform for next generation vaccines, as it can be manufactured rapidly at large scale to meet emerging pathogens. However, its performance as a robust vaccine is strengthened by supplemental immune stimulation, which is typically provided by adjuvant formulations that facilitate delivery and stimulate immune responses. Here, we present a strategy for increasing translation of a model IVT mRNA vaccine while simultaneously modulating its immune-stimulatory properties in a programmable fashion, without relying on delivery vehicle formulations. Substitution of uridine with the modified base N1-methylpseudouridine reduces the intrinsic immune stimulation of the IVT mRNA and enhances antigen translation. Tethering adjuvants to naked IVT mRNA through antisense nucleotides boosts the immunostimulatory properties of adjuvants in vitro, without impairing transgene production or adjuvant activity. In vivo, intramuscular injection of tethered IVT mRNA-TLR7 agonists leads to enhanced local immune responses, and to antigen-specific cell-mediated and humoral responses. We believe this system represents a potential platform compatible with any adjuvant of interest to enable specific programmable stimulation of immune responses.


Methods | 2016

Imaging viral RNA using multiply labeled tetravalent RNA imaging probes in live cells.

Eric Alonas; Daryll Vanover; Emmeline L. Blanchard; Chiara Zurla; Philip J. Santangelo


Biomaterials | 2018

Unifying in vitro and in vivo IVT mRNA expression discrepancies in skeletal muscle via mechanotransduction

Sushma Bhosle; Kristin H. Loomis; Jonathan L. Kirschman; Emmeline L. Blanchard; Daryll Vanover; Chiara Zurla; Damien Habrant; Darin K. Edwards; Patrick Baumhof; Bruno Pitard; Philip J. Santangelo


Archive | 2014

the Matrix Protein and the Ribonucleoprotein Complex

Gabriella Kiss; Jens M. Holl; G Williams; Eric Alonas; Daryll Vanover; W Aaron; Manasa Gudheti; Ricardo C. Guerrero-Ferreira; Vinod Nair; S Barney; Philip J. Santangelo; Elizabeth R. Wright

Collaboration


Dive into the Daryll Vanover's collaboration.

Top Co-Authors

Avatar

Philip J. Santangelo

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Chiara Zurla

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Jonathan L. Kirschman

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Emmeline L. Blanchard

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Eric Alonas

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Aaron W. Lifland

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristin H. Loomis

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Sushma Bhosle

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge