Network


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

Hotspot


Dive into the research topics where Philip L. Eisenhauer is active.

Publication


Featured researches published by Philip L. Eisenhauer.


Stem Cells | 2011

Bone Marrow-Derived Mesenchymal Stromal Cells Inhibit Th2-Mediated Allergic Airways Inflammation in Mice†‡§

Meagan Goodwin; Viranuj Sueblinvong; Philip L. Eisenhauer; Nicholas P. Ziats; Laurie W. Leclair; Matthew E. Poynter; Chad Steele; Mercedes Rincon; Daniel J. Weiss

Bone marrow‐derived mesenchymal stromal cells (BMSCs) mitigate inflammation in mouse models of acute lung injury. However, specific mechanisms of BMSC actions on CD4 T lymphocyte‐mediated inflammation in vivo remain poorly understood. Limited data suggests promotion of Th2 phenotype in models of Th1‐mediated diseases. However, whether this might alleviate or worsen Th2‐mediated diseases such as allergic asthma is unknown. To ascertain the effects of systemic administration of BMSCs in a mouse model of Th2‐mediated allergic airways inflammation, ovalbumin (OVA)‐induced allergic airways inflammation was induced in wild‐type C57BL/6 and BALB/c mice as well as in interferon‐γ (IFNγ) receptor null mice. Effects of systemic administration during antigen sensitization of either syngeneic or allogeneic BMSC on airways hyperreactivity, lung inflammation, antigen‐specific CD4 T lymphocytes, and serum immunoglobulins were assessed. Both syngeneic and allogeneic BMSCs inhibited airways hyperreactivity and lung inflammation through a mechanism partly dependent on IFNγ. However, contrary to existing data, BMSCs did not affect antigen‐specific CD4 T lymphocyte proliferation but rather promoted Th1 phenotype in vivo as assessed by both OVA‐specific CD4 T lymphocyte cytokine production and OVA‐specific circulating immunoglobulins. BMSCs treated to prevent release of soluble mediators and a control cell population of primary dermal skin fibroblasts only partly mimicked the BMSC effects and in some cases worsened inflammation. In conclusion, BMSCs inhibit Th2‐mediated allergic airways inflammation by influencing antigen‐specific CD4 T lymphocyte differentiation. Promotion of a Th1 phenotype in antigen‐specific CD4 T lymphocytes by BMSCs is sufficient to inhibit Th2‐mediated allergic airways inflammation through an IFNγ‐dependent process. STEM CELLS 2011;29:1137–1148


Stem Cells | 2013

Endogenous Distal Airway Progenitor Cells, Lung Mechanics, and Disproportionate Lobar Growth Following Long‐Term Postpneumonectomy in Mice

Philip L. Eisenhauer; Benjamin Earle; Roberto Loi; Viranuj Sueblinvong; Meagan Goodwin; Gilman B. Allen; Lennart K. A. Lundblad; Melissa R. Mazan; A. M. Hoffman; Daniel J. Weiss

Using a model of postpneumonectomy (PNY) compensatory lung growth in mice, we previously observed an increase in numbers of a putative endogenous distal airway progenitor cell population (CCSPpos/pro‐SPCpos cells located at bronchoalveolar duct junctions [BADJs]), at 3, 7, and 14 days after pneumonectomy, returning to baseline at 28 days post‐PNY. As the origin of these cells is poorly understood, we evaluated whether bone marrow cells contributed to the pool of these or other cells during prolonged post‐PNY lung regrowth. Naïve and sex‐mismatched chimeric mice underwent left PNY and were evaluated at 1, 2, and 3 months for numbers of BADJ CCSPpos/pro‐SPCpos cells and presence of donor‐derived marrow cells engrafted as airway or alveolar epithelium. Nonchimeric mice were also examined at 12 months after PNY for numbers of BADJ CCSPpos/pro‐SPCpos cells. Notably, the right accessory lobe (RAL) continued to grow disproportionately over 12 months, a novel finding not previously described. Assessment of lung mechanics demonstrated an increase in lung stiffness following PNY, which significantly diminished over 1 year, but remained elevated relative to 1‐year‐old naïve controls. However, the number of CCSPpos/pro‐SPCpos BADJ cells ≥1‐month following PNY was equivalent to that found in naïve controls even after 12 months of continued RAL growth. Notably, no donor bone marrow‐derived cells engrafted as airway or alveolar epithelial cells, including those at the BADJ, up to 3 months after PNY. These studies suggest that lung epithelial cells, including CCSPpos/pro‐SPCpos cells, are not replenished from marrow‐derived cells during post‐PNY lung growth in mice. STEM Cells2013;31:1330–1339


PLOS Pathogens | 2016

The Lymphocytic Choriomeningitis Virus Matrix Protein PPXY Late Domain Drives the Production of Defective Interfering Particles

Christopher M. Ziegler; Philip L. Eisenhauer; Emily A. Bruce; Marion E. Weir; Benjamin R. King; Joseph P. Klaus; Dimitry N. Krementsov; David J. Shirley; Bryan A. Ballif; Jason Botten

Arenaviruses cause severe diseases in humans but establish asymptomatic, lifelong infections in rodent reservoirs. Persistently-infected rodents harbor high levels of defective interfering (DI) particles, which are thought to be important for establishing persistence and mitigating virus-induced cytopathic effect. Little is known about what drives the production of DI particles. We show that neither the PPXY late domain encoded within the lymphocytic choriomeningitis virus (LCMV) matrix protein nor a functional endosomal sorting complex transport (ESCRT) pathway is absolutely required for the generation of standard infectious virus particles. In contrast, DI particle release critically requires the PPXY late domain and is ESCRT-dependent. Additionally, the terminal tyrosine in the PPXY motif is reversibly phosphorylated and our findings indicate that this posttranslational modification may regulate DI particle formation. Thus we have uncovered a new role for the PPXY late domain and a possible mechanism for its regulation.


Journal of Virology | 2017

A Map of the Arenavirus Nucleoprotein-Host Protein Interactome Reveals that Junín Virus Selectively Impairs the Antiviral Activity of Double-Stranded RNA-Activated Protein Kinase (PKR)

Benjamin R. King; Dylan Hershkowitz; Philip L. Eisenhauer; Marion E. Weir; Christopher M. Ziegler; Joanne Russo; Emily A. Bruce; Bryan A. Ballif; Jason Botten

ABSTRACT Arenaviruses are enveloped negative-strand RNA viruses that cause significant human disease. These viruses encode only four proteins to accomplish the viral life cycle, so each arenavirus protein likely plays unappreciated accessory roles during infection. Here we used immunoprecipitation and mass spectrometry to identify human proteins that interact with the nucleoproteins (NPs) of the Old World arenavirus lymphocytic choriomeningitis virus (LCMV) and the New World arenavirus Junín virus (JUNV) strain Candid #1. Bioinformatic analysis of the identified protein partners of NP revealed that host translation appears to be a key biological process engaged during infection. In particular, NP associates with the double-stranded RNA (dsRNA)-activated protein kinase (PKR), a well-characterized antiviral protein that inhibits cap-dependent protein translation initiation via phosphorylation of eIF2α. JUNV infection leads to increased expression of PKR as well as its redistribution to viral replication and transcription factories. Further, phosphorylation of PKR, which is a prerequisite for its ability to phosphorylate eIF2α, is readily induced by JUNV. However, JUNV prevents this pool of activated PKR from phosphorylating eIF2α, even following exposure to the synthetic dsRNA poly(I·C), a potent PKR agonist. This blockade of PKR function is highly specific, as LCMV is unable to similarly inhibit eIF2α phosphorylation. JUNVs ability to antagonize the antiviral activity of PKR appears to be complete, as silencing of PKR expression has no impact on viral propagation. In summary, we provide a detailed map of the host machinery engaged by arenavirus NPs and identify an antiviral pathway that is subverted by JUNV. IMPORTANCE Arenaviruses are important human pathogens for which FDA-approved vaccines do not exist and effective antiviral therapeutics are needed. Design of antiviral treatment options and elucidation of the mechanistic basis of disease pathogenesis will depend on an increased basic understanding of these viruses and, in particular, their interactions with the host cell machinery. Identifying host proteins critical for the viral life cycle and/or pathogenesis represents a useful strategy to uncover new drug targets. This study reveals, for the first time, the extensive human protein interactome of arenavirus nucleoproteins and uncovers a potent antiviral host protein that is neutralized during Junín virus infection. In so doing, it shows further insight into the interplay between the virus and the host innate immune response and provides an important data set for the field.


Journal of General Virology | 2017

Visualization of the lymphocytic choriomeningitis mammarenavirus (LCMV) genome reveals the early endosome as a possible site for genome replication and viral particle pre-assembly

Benjamin R. King; Samuel Kellner; Philip L. Eisenhauer; Emily A. Bruce; Christopher M. Ziegler; Daniel Zenklusen; Jason Botten

We report a fluorescence in situ hybridization (FISH) assay that allows the visualization of lymphocytic choriomeningitis mammarenavirus (LCMV) genomic RNAs in individual cells. We show that viral S segment genomic and antigenomic RNA, along with viral nucleoprotein, colocalize in subcellular structures we presume to be viral replication factories. These viral RNA structures are highly dynamic during acute infection, with the many small foci seen early coalescing into larger perinuclear foci later in infection. These late-forming perinuclear viral RNA aggregates are located near the cellular microtubule organizing centre and colocalize with the early endosomal marker Rab5c and the viral glycoprotein in a proportion of infected cells. We propose that the virus is using the surface of a cellular membrane-bound organelle as a site for the pre-assembly of viral components, including genomic RNA and viral glycoprotein, prior to their transport to the plasma membrane, where new particles will bud.


Journal of Virology | 2017

A proteomic survey of Junín virus interactions with human proteins reveals host factors required for arenavirus replication

Christopher M. Ziegler; Philip L. Eisenhauer; Jamie A. Kelly; Loan N. Dang; Vedran Beganovic; Emily A. Bruce; Benjamin R. King; David J. Shirley; Marion E. Weir; Bryan A. Ballif; Jason Botten

ABSTRACT Arenaviruses are negative-strand, enveloped RNA viruses that cause significant human disease. In particular, Junín mammarenavirus (JUNV) is the etiologic agent of Argentine hemorrhagic fever. At present, little is known about the cellular proteins that the arenavirus matrix protein (Z) hijacks to accomplish its various functions, including driving the process of virus release. Furthermore, there is little knowledge regarding host proteins incorporated into arenavirus particles and their importance for virion function. To address these deficiencies, we used mass spectrometry to identify human proteins that (i) interact with the JUNV matrix protein inside cells or within virus-like particles (VLPs) and/or (ii) are incorporated into bona fide JUNV strain Candid#1 particles. Bioinformatics analyses revealed that multiple classes of human proteins were overrepresented in the data sets, including ribosomal proteins, Ras superfamily proteins, and endosomal sorting complex required for transport (ESCRT) proteins. Several of these proteins were required for the propagation of JUNV (ADP ribosylation factor 1 [ARF1], ATPase, H+ transporting, lysosomal 38-kDa, V0 subunit d1 [ATP6V0D1], and peroxiredoxin 3 [PRDX3]), lymphocytic choriomeningitis mammarenavirus (LCMV) (Rab5c), or both viruses (ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide [ATP5B] and IMP dehydrogenase 2 [IMPDH2]). Furthermore, we show that the release of infectious JUNV particles, but not LCMV particles, requires a functional ESCRT pathway and that ATP5B and IMPDH2 are required for JUNV budding. In summary, we have provided a large-scale map of host machinery that associates with JUNV and identified key human proteins required for its propagation. This data set provides a resource for the field to guide antiviral target discovery and to better understand the biology of the arenavirus matrix protein and the importance of host proteins for virion function. IMPORTANCE Arenaviruses are deadly human pathogens for which there are no U.S. Food and Drug Administration-approved vaccines and only limited treatment options. Little is known about the host proteins that are incorporated into arenavirus particles or that associate with its multifunctional matrix protein. Using Junín mammarenavirus (JUNV), the causative agent of Argentine hemorrhagic fever, as a model organism, we mapped the human proteins that are incorporated into JUNV particles or that associate with the JUNV matrix protein. Functional analysis revealed host machinery that is required for JUNV propagation, including the cellular ESCRT pathway. This study improves our understanding of critical arenavirus-host interactions and provides a data set that will guide future studies to better understand arenavirus pathogenesis and identify novel host proteins that can be therapeutically targeted.


Journal of General Virology | 2016

A novel phosphoserine motif in the LCMV matrix protein Z regulates the release of infectious virus and defective interfering particles

Christopher M. Ziegler; Philip L. Eisenhauer; Emily A. Bruce; Vedran Beganovic; Benjamin R. King; Marion E. Weir; Bryan A. Ballif; Jason Botten

We report that the lymphocytic choriomeningitis virus (LCMV) matrix protein, which drives viral budding, is phosphorylated at serine 41 (S41). A recombinant (r)LCMV bearing a phosphomimetic mutation (S41D) was impaired in infectious and defective interfering (DI) particle release, while a non-phosphorylatable mutant (S41A) was not. The S41D mutant was disproportionately impaired in its ability to release DI particles relative to infectious particles. Thus, DI particle production by LCMV may be dynamically regulated via phosphorylation of S41.


Journal of Immunology | 2018

Bacterial Lipoproteins Constitute the TLR2-Stimulating Activity of Serum Amyloid A

Edward J. Burgess; Laura R. Hoyt; Matthew J. Randall; Madeleine M. Mank; Joseph J. Bivona; Philip L. Eisenhauer; Jason Botten; Bryan A. Ballif; Ying-Wai Lam; Matthew J. Wargo; Jonathan E. Boyson; Jennifer L. Ather; Matthew E. Poynter

Studies comparing endogenous and recombinant serum amyloid A (SAA) have generated conflicting data on the proinflammatory function of these proteins. In exploring this discrepancy, we found that in contrast to commercially sourced recombinant human SAA1 (hSAA1) proteins produced in Escherichia coli, hSAA1 produced from eukaryotic cells did not promote proinflammatory cytokine production from human or mouse cells, induce Th17 differentiation, or stimulate TLR2. Proteomic analysis of E. coli–derived hSAA1 revealed the presence of numerous bacterial proteins, with several being reported or probable lipoproteins. Treatment of hSAA1 with lipoprotein lipase or addition of a lipopeptide to eukaryotic cell–derived hSAA1 inhibited or induced the production of TNF-α from macrophages, respectively. Our results suggest that a function of SAA is in the binding of TLR2-stimulating bacterial proteins, including lipoproteins, and demand that future studies of SAA employ a recombinant protein derived from eukaryotic cells.


American Journal of Respiratory and Critical Care Medicine | 2008

Derivation of lung epithelium from human cord blood-derived mesenchymal stem cells.

Viranuj Sueblinvong; Roberto Loi; Philip L. Eisenhauer; Ira M. Bernstein; Benjamin T. Suratt; Jeffrey L. Spees; Daniel J. Weiss


Cell Host & Microbe | 2013

The Intracellular Cargo Receptor ERGIC-53 Is Required for the Production of Infectious Arenavirus, Coronavirus, and Filovirus Particles

Joseph P. Klaus; Philip L. Eisenhauer; Joanne Russo; Anne B. Mason; Danh Do; Benjamin King; Douglas J. Taatjes; Cromwell Cornillez-Ty; Jonathan E. Boyson; Markus Thali; Chunlei Zheng; Lujian Liao; John R. Yates; Bin Zhang; Bryan A. Ballif; Jason Botten

Collaboration


Dive into the Philip L. Eisenhauer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge