Network


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

Hotspot


Dive into the research topics where Jennifer M. Binning is active.

Publication


Featured researches published by Jennifer M. Binning.


Journal of Virology | 2010

Basic Residues within the Ebolavirus VP35 Protein Are Required for Its Viral Polymerase Cofactor Function

Kathleen C. Prins; Jennifer M. Binning; Reed S. Shabman; Daisy W. Leung; Gaya K. Amarasinghe; Christopher F. Basler

ABSTRACT The ebolavirus (EBOV) VP35 protein binds to double-stranded RNA (dsRNA), inhibits host alpha/beta interferon (IFN-α/β) production, and is an essential component of the viral polymerase complex. Structural studies of the VP35 C-terminal IFN inhibitory domain (IID) identified specific structural features, including a central basic patch and a hydrophobic pocket, that are important for dsRNA binding and IFN inhibition. Several other conserved basic residues bordering the central basic patch and a separate cluster of basic residues, called the first basic patch, were also identified. Functional analysis of alanine substitution mutants indicates that basic residues outside the central basic patch are not required for dsRNA binding or for IFN inhibition. However, minigenome assays, which assess viral RNA polymerase complex function, identified these other basic residues to be critical for viral RNA synthesis. Of these, a subset located within the first basic patch is important for VP35-nucleoprotein (NP) interaction, as evidenced by the inability of alanine substitution mutants to coimmunoprecipitate with NP. Therefore, first basic patch residues are likely critical for replication complex formation through interactions with NP. Coimmunoprecipitation studies further demonstrate that the VP35 IID is sufficient to interact with NP and that dsRNA can modulate VP35 IID interactions with NP. Other basic residue mutations that disrupt the VP35 polymerase cofactor function do not affect interaction with NP or with the amino terminus of the viral polymerase. Collectively, these results highlight the importance of conserved basic residues from the EBOV VP35 C-terminal IID and validate the VP35 IID as a potential therapeutic target.


PLOS Pathogens | 2013

An Upstream Open Reading Frame Modulates Ebola Virus Polymerase Translation and Virus Replication

Reed S. Shabman; Thomas Hoenen; Allison Groseth; Omar J. Jabado; Jennifer M. Binning; Gaya K. Amarasinghe; Heinz Feldmann; Christopher F. Basler

Ebolaviruses, highly lethal zoonotic pathogens, possess longer genomes than most other non-segmented negative-strand RNA viruses due in part to long 5′ and 3′ untranslated regions (UTRs) present in the seven viral transcriptional units. To date, specific functions have not been assigned to these UTRs. With reporter assays, we demonstrated that the Zaire ebolavirus (EBOV) 5′-UTRs lack internal ribosomal entry site function. However, the 5′-UTRs do differentially regulate cap-dependent translation when placed upstream of a GFP reporter gene. Most dramatically, the 5′-UTR derived from the viral polymerase (L) mRNA strongly suppressed translation of GFP compared to a β-actin 5′-UTR. The L 5′-UTR is one of four viral genes to possess upstream AUGs (uAUGs), and ablation of each uAUG enhanced translation of the primary ORF (pORF), most dramatically in the case of the L 5′-UTR. The L uAUG was sufficient to initiate translation, is surrounded by a “weak” Kozak sequence and suppressed pORF translation in a position-dependent manner. Under conditions where eIF2α was phosphorylated, the presence of the uORF maintained translation of the L pORF, indicating that the uORF modulates L translation in response to cellular stress. To directly address the role of the L uAUG in virus replication, a recombinant EBOV was generated in which the L uAUG was mutated to UCG. Strikingly, mutating two nucleotides outside of previously-defined protein coding and cis-acting regulatory sequences attenuated virus growth to titers 10–100-fold lower than a wild-type virus in Vero and A549 cells. The mutant virus also exhibited decreased viral RNA synthesis as early as 6 hours post-infection and enhanced sensitivity to the stress inducer thapsigargin. Cumulatively, these data identify novel mechanisms by which EBOV regulates its polymerase expression, demonstrate their relevance to virus replication and identify a potential therapeutic target.


Biochemistry | 2013

Development of RNA Aptamers Targeting Ebola Virus VP35

Jennifer M. Binning; Tianjiao Wang; Priya Luthra; Reed S. Shabman; Dominika Borek; Gai Liu; Wei Xu; Daisy W. Leung; Christopher F. Basler; Gaya K. Amarasinghe

Viral protein 35 (VP35), encoded by filoviruses, is a multifunctional dsRNA binding protein that plays important roles in viral replication, innate immune evasion, and pathogenesis. The multifunctional nature of these proteins also presents opportunities to develop countermeasures that target distinct functional regions. However, functional validation and the establishment of therapeutic approaches toward such multifunctional proteins, particularly for nonenzymatic targets, are often challenging. Our previous work on filoviral VP35 proteins defined conserved basic residues located within its C-terminal dsRNA binding interferon (IFN) inhibitory domain (IID) as important for VP35 mediated IFN antagonism and viral polymerase cofactor functions. In the current study, we used a combination of structural and functional data to determine regions of Ebola virus (EBOV) VP35 (eVP35) to target for aptamer selection using SELEX. Select aptamers, representing, two distinct classes, were further characterized based on their interaction properties to eVP35 IID. These results revealed that these aptamers bind to distinct regions of eVP35 IID with high affinity (10-50 nM) and specificity. These aptamers can compete with dsRNA for binding to eVP35 and disrupt the eVP35-nucleoprotein (NP) interaction. Consistent with the ability to antagonize the eVP35-NP interaction, select aptamers can inhibit the function of the EBOV polymerase complex reconstituted by the expression of select viral proteins. Taken together, our results support the identification of two aptamers that bind filoviral VP35 proteins with high affinity and specificity and have the capacity to potentially function as filoviral VP35 protein inhibitors.


Journal of Molecular Biology | 2014

In Silico Derived Small Molecules Bind the Filovirus VP35 Protein and Inhibit Its Polymerase Cofactor Activity

Craig S. Brown; Michael S. Lee; Daisy W. Leung; Tianjiao Wang; Wei Xu; Priya Luthra; Manu Anantpadma; Reed S. Shabman; Lisa Melito; Karen S. MacMillan; Dominika Borek; Zbyszek Otwinowski; Parameshwaran Ramanan; Alisha Stubbs; Dayna S. Peterson; Jennifer M. Binning; Marco Tonelli; Mark A. Olson; Robert A. Davey; Joseph M. Ready; Christopher F. Basler; Gaya K. Amarasinghe

The Ebola virus (EBOV) genome only encodes a single viral polypeptide with enzymatic activity, the viral large (L) RNA-dependent RNA polymerase protein. However, currently, there is limited information about the L protein, which has hampered the development of antivirals. Therefore, antifiloviral therapeutic efforts must include additional targets such as protein-protein interfaces. Viral protein 35 (VP35) is multifunctional and plays important roles in viral pathogenesis, including viral mRNA synthesis and replication of the negative-sense RNA viral genome. Previous studies revealed that mutation of key basic residues within the VP35 interferon inhibitory domain (IID) results in significant EBOV attenuation, both in vitro and in vivo. In the current study, we use an experimental pipeline that includes structure-based in silico screening and biochemical and structural characterization, along with medicinal chemistry, to identify and characterize small molecules that target a binding pocket within VP35. NMR mapping experiments and high-resolution x-ray crystal structures show that select small molecules bind to a region of VP35 IID that is important for replication complex formation through interactions with the viral nucleoprotein (NP). We also tested select compounds for their ability to inhibit VP35 IID-NP interactions in vitro as well as VP35 function in a minigenome assay and EBOV replication. These results confirm the ability of compounds identified in this study to inhibit VP35-NP interactions in vitro and to impair viral replication in cell-based assays. These studies provide an initial framework to guide development of antifiloviral compounds against filoviral VP35 proteins.


Meat Science | 2012

Profile of biochemical traits influencing tenderness of muscles from the beef round

Mark J. Anderson; Steven M. Lonergan; C.A. Fedler; Kenneth J. Prusa; Jennifer M. Binning; Elisabeth J. Huff-Lonergan

The objective of this study was to define the biochemical differences that govern tenderness and palatability of economically important muscles from the beef round using cuts with known tenderness differences. At 24h postmortem, the longissimus dorsi (LD), gracillus (GR), adductor (AD), semimembranosus (SM), sartorius (SAR), vastus lateralis (VL), and vastus intermedius (VI) muscles were removed from ten market weight beef cattle. Sensory and biochemical characteristics were determined in each cut and compared with the LD. The GR, SAR and VI had sensory traits similar to the LD while the SM, AD and VL differed. The GR, SAR, AD, and SM all had multiple biochemical characteristics similar to the LD, while the VI and AD had numerous biochemical differences. While no one biochemical characteristic can be used to predict tenderness across all muscles, analysis of the biochemical characteristics revealed that in most beef round cuts postmortem proteolysis provided a good indication of the tenderization occurring during aging.


Frontiers in Microbiology | 2012

Aptamers in Virology: Recent Advances and Challenges

Jennifer M. Binning; Daisy W. Leung; Gaya K. Amarasinghe

Aptamers generated from randomized libraries of nucleic acids have found utility in a wide variety of fields and in the clinic. Aptamers can be used to target both intracellular and extracellular components, including small molecules, proteins, cells, and viruses. With recent technological developments in stringent selection and rapid isolation strategies, it is likely that aptamers will continue to make an impact as useful tools and reagents. Although many recently developed aptamers are intended for use as therapeutic and diagnostic agents, use of aptamers for basic research, including target validation, remains an active area with high potential to impact our understanding of molecular mechanisms and for drug discovery. In this brief review, we will discuss recent aptamer discoveries, their potential role in structural virology, as well as challenges and future prospects.


Cell Reports | 2015

Lineage-Specific Viral Hijacking of Non-Canonical E3 Ubiquitin Ligase Cofactors in the Evolution of Vif Anti-APOBEC3 Activity

Joshua Kane; David J. Stanley; Judd F. Hultquist; Jeffrey R. Johnson; Nicole Mietrach; Jennifer M. Binning; Stefán R. Jónsson; Sarah Barelier; Billy W. Newton; Tasha Johnson; Kathleen Franks-Skiba; Ming Li; William L. Brown; Hörður Ingi Gunnarsson; Adalbjorg Adalbjornsdóttir; J.S. Fraser; Reuben S. Harris; Valgerður Andrésdóttir; John D. Gross; Nevan J. Krogan

HIV-1 encodes the accessory protein Vif, which hijacks a host Cullin-RING ubiquitin ligase (CRL) complex as well as the non-canonical cofactor CBFβ, to antagonize APOBEC3 antiviral proteins. Non-canonical cofactor recruitment to CRL complexes by viral factors, to date, has only been attributed to HIV-1 Vif. To further study this phenomenon, we employed a comparative approach combining proteomic, biochemical, structural, and virological techniques to investigate Vif complexes across the lentivirus genus, including primate (HIV-1 and simian immunodeficiency virus macaque [SIVmac]) and non-primate (FIV, BIV, and MVV) viruses. We find that CBFβ is completely dispensable for the activity of non-primate lentiviral Vif proteins. Furthermore, we find that BIV Vif requires no cofactor and that MVV Vif requires a novel cofactor, cyclophilin A (CYPA), for stable CRL complex formation and anti-APOBEC3 activity. We propose modular conservation of Vif complexes allows for potential exaptation of functions through the acquisition of non-CRL-associated host cofactors while preserving anti-APOBEC3 activity.


Annual Review of Virology | 2017

Making Sense of Multifunctional Proteins: Human Immunodeficiency Virus Type 1 Accessory and Regulatory Proteins and Connections to Transcription

Tyler B. Faust; Jennifer M. Binning; John D. Gross; Alan D. Frankel

Viruses are completely dependent upon cellular machinery to support replication and have therefore developed strategies to co-opt cellular processes to optimize infection and counter host immune defenses. Many viruses, including human immunodeficiency virus type 1 (HIV-1), encode a relatively small number of genes. Viruses with limited genetic content often encode multifunctional proteins that function at multiple stages of the viral replication cycle. In this review, we discuss the functions of HIV-1 regulatory (Tat and Rev) and accessory (Vif, Vpr, Vpu, and Nef) proteins. Each of these proteins has a highly conserved primary activity; however, numerous additional activities have been attributed to these viral proteins. We explore the possibility that HIV-1 proteins leverage their multifunctional nature to alter host transcriptional networks to elicit a diverse set of cellular responses. Although these transcriptional effects appear to benefit the virus, it is not yet clear whether they are strongly selected for during viral evolution or are a ripple effect from the primary function. As our detailed knowledge of these viral proteins improves, we will undoubtedly uncover how the multifunctional nature of these HIV-1 regulatory and accessory proteins, and in particular their transcriptional functions, work to drive viral pathogenesis.


PLOS Pathogens | 2018

Fab-based inhibitors reveal ubiquitin independent functions for HIV Vif neutralization of APOBEC3 restriction factors

Jennifer M. Binning; Amber M. Smith; Judd F. Hultquist; Charles S. Craik; Nathalie Caretta; Melody G. Campbell; Lily Burton; Florencia La Greca; Michael J. McGregor; Hai M. Ta; Koen Bartholomeeusen; B. Matija Peterlin; Nevan J. Krogan; Natalia Sevillano; Yifan Cheng; John D. Gross

The lentiviral protein Viral Infectivity Factor (Vif) counteracts the antiviral effects of host APOBEC3 (A3) proteins and contributes to persistent HIV infection. Vif targets A3 restriction factors for ubiquitination and proteasomal degradation by recruiting them to a multi-protein ubiquitin E3 ligase complex. Here, we describe a degradation-independent mechanism of Vif-mediated antagonism that was revealed through detailed structure-function studies of antibody antigen-binding fragments (Fabs) to the Vif complex. Two Fabs were found to inhibit Vif-mediated A3 neutralization through distinct mechanisms: shielding A3 from ubiquitin transfer and blocking Vif E3 assembly. Combined biochemical, cell biological and structural studies reveal that disruption of Vif E3 assembly inhibited A3 ubiquitination but was not sufficient to restore its packaging into viral particles and antiviral activity. These observations establish that Vif can neutralize A3 family members in a degradation-independent manner. Additionally, this work highlights the potential of Fabs as functional probes, and illuminates how Vif uses a multi-pronged approach involving both degradation dependent and independent mechanisms to suppress A3 innate immunity.


Cell Reports | 2015

An Intrinsically Disordered Peptide from Ebola Virus VP35 Controls Viral RNA Synthesis by Modulating Nucleoprotein-RNA Interactions

Daisy W. Leung; Dominika Borek; Priya Luthra; Jennifer M. Binning; Manu Anantpadma; Gai Liu; Ian B. Harvey; Zhaoming Su; Ariel Endlich-Frazier; Juanli Pan; Reed S. Shabman; Wah Chiu; Robert A. Davey; Zbyszek Otwinowski; Christopher F. Basler; Gaya K. Amarasinghe

Collaboration


Dive into the Jennifer M. Binning's collaboration.

Top Co-Authors

Avatar

Gaya K. Amarasinghe

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daisy W. Leung

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Reed S. Shabman

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Priya Luthra

Georgia State University

View shared research outputs
Top Co-Authors

Avatar

Dominika Borek

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gai Liu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

John D. Gross

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge