Network


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

Hotspot


Dive into the research topics where Charles J. Shoemaker is active.

Publication


Featured researches published by Charles J. Shoemaker.


Science Translational Medicine | 2013

FDA-approved selective estrogen receptor modulators inhibit Ebola virus infection.

Lisa M. Johansen; Jennifer M. Brannan; Sue E. Delos; Charles J. Shoemaker; Andrea Stossel; Calli Lear; Benjamin G. Hoffstrom; Lisa Evans DeWald; Kathryn L. Schornberg; Corinne Scully; Joseph Lehar; Lisa E. Hensley; Judith M. White; Gene G. Olinger

Clomiphene and toremifene inhibit Ebola virus infection. Fertile Strategy for Ebola Infection Perhaps no virus has grasped the public’s imagination like Ebola virus. Although infection is rare, the threat from bioweapons and sporadic outbreaks is the stuff of nightmares. Our inability to treat infected individuals or even to prevent infection with therapeutics raises the stakes. Now, Johansen et al. have found that FDA-approved selective estrogen receptor modulators (SERMs) could potentially be repurposed to treat Ebola virus infection. The authors performed an in vitro screen to identify classes of compounds with antiviral activity against Zaire ebolavirus (EBOV). They found that SERMs, which have many uses that range from fertility treatments to breast cancer therapy, could inhibit EBOV infection both in vitro and in a mouse model. Somewhat surprisingly, this effect was not through on-target interactions with the estrogen receptor—the inhibition was still present in cells that lacked estrogen receptor expression. Instead, the compounds likely act late in viral entry, preventing viral fusion. These data support the off-target testing of SERMs for Ebola virus infection and suggest that screens of FDA-approved drugs to treat infectious diseases could yield fertile results. Ebola viruses remain a substantial threat to both civilian and military populations as bioweapons, during sporadic outbreaks, and from the possibility of accidental importation from endemic regions by infected individuals. Currently, no approved therapeutics exist to treat or prevent infection by Ebola viruses. Therefore, we performed an in vitro screen of Food and Drug Administration (FDA)– and ex–US-approved drugs and selected molecular probes to identify drugs with antiviral activity against the type species Zaire ebolavirus (EBOV). From this screen, we identified a set of selective estrogen receptor modulators (SERMs), including clomiphene and toremifene, which act as potent inhibitors of EBOV infection. Anti-EBOV activity was confirmed for both of these SERMs in an in vivo mouse infection model. This anti-EBOV activity occurred even in the absence of detectable estrogen receptor expression, and both SERMs inhibited virus entry after internalization, suggesting that clomiphene and toremifene are not working through classical pathways associated with the estrogen receptor. Instead, the response appeared to be an off-target effect where the compounds interfere with a step late in viral entry and likely affect the triggering of fusion. These data support the screening of readily available approved drugs to identify therapeutics for the Ebola viruses and other infectious diseases. The SERM compounds described in this report are an immediately actionable class of approved drugs that can be repurposed for treatment of filovirus infections.


PLOS ONE | 2013

Multiple Cationic Amphiphiles Induce a Niemann-Pick C Phenotype and Inhibit Ebola Virus Entry and Infection

Charles J. Shoemaker; Kathryn L. Schornberg; Sue E. Delos; Corinne Scully; Hassan Pajouhesh; Gene G. Olinger; Lisa M. Johansen; Judith M. White

Ebola virus (EBOV) is an enveloped RNA virus that causes hemorrhagic fever in humans and non-human primates. Infection requires internalization from the cell surface and trafficking to a late endocytic compartment, where viral fusion occurs, providing a conduit for the viral genome to enter the cytoplasm and initiate replication. In a concurrent study, we identified clomiphene as a potent inhibitor of EBOV entry. Here, we screened eleven inhibitors that target the same biosynthetic pathway as clomiphene. From this screen we identified six compounds, including U18666A, that block EBOV infection (IC50 1.6 to 8.0 µM) at a late stage of entry. Intriguingly, all six are cationic amphiphiles that share additional chemical features. U18666A induces phenotypes, including cholesterol accumulation in endosomes, associated with defects in Niemann–Pick C1 protein (NPC1), a late endosomal and lysosomal protein required for EBOV entry. We tested and found that all six EBOV entry inhibitors from our screen induced cholesterol accumulation. We further showed that higher concentrations of cationic amphiphiles are required to inhibit EBOV entry into cells that overexpress NPC1 than parental cells, supporting the contention that they inhibit EBOV entry in an NPC1-dependent manner. A previously reported inhibitor, compound 3.47, inhibits EBOV entry by blocking binding of the EBOV glycoprotein to NPC1. None of the cationic amphiphiles tested had this effect. Hence, multiple cationic amphiphiles (including several FDA approved agents) inhibit EBOV entry in an NPC1-dependent fashion, but by a mechanism distinct from that of compound 3.47. Our findings suggest that there are minimally two ways of perturbing NPC1-dependent pathways that can block EBOV entry, increasing the attractiveness of NPC1 as an anti-filoviral therapeutic target.


Science Translational Medicine | 2015

A screen of approved drugs and molecular probes identifies therapeutics with anti–Ebola virus activity

Lisa M. Johansen; Lisa Evans DeWald; Charles J. Shoemaker; Benjamin G. Hoffstrom; Calli M. Lear-Rooney; Andrea Stossel; Elizabeth A. Nelson; Sue E. Delos; James A. Simmons; Jill M. Grenier; Laura T. Pierce; Hassan Pajouhesh; Joseph Lehar; Lisa E. Hensley; Pamela J. Glass; Judith M. White; Gene G. Olinger

Several FDA-approved drugs, including bepridil and sertraline, may be therapeutics against filovirus infections. Drug repurposing for Ebola virus The recent Ebola virus outbreak has highlighted the lack of therapies for filovirus infection. FDA-approved drugs serve as a source of medications with a proven safety record. Johansen et al. have now screened about 2600 FDA-approved drugs and molecular probes and found 80 that have some efficacy against Zaire ebolavirus in vitro. These drugs were mechanistically diverse, and several drugs, including a calcium channel blocker and an antidepressant, could protect against infection in a mouse model. Although new therapy development is ongoing, these repurposed drugs can rapidly move to human testing and may serve on the frontline against Ebolavirus infection. Currently, no approved therapeutics exist to treat or prevent infections induced by Ebola viruses, and recent events have demonstrated an urgent need for rapid discovery of new treatments. Repurposing approved drugs for emerging infections remains a critical resource for potential antiviral therapies. We tested ~2600 approved drugs and molecular probes in an in vitro infection assay using the type species, Zaire ebolavirus. Selective antiviral activity was found for 80 U.S. Food and Drug Administration–approved drugs spanning multiple mechanistic classes, including selective estrogen receptor modulators, antihistamines, calcium channel blockers, and antidepressants. Results using an in vivo murine Ebola virus infection model confirmed the protective ability of several drugs, such as bepridil and sertraline. Viral entry assays indicated that most of these antiviral drugs block a late stage of viral entry. By nature of their approved status, these drugs have the potential to be rapidly advanced to clinical settings and used as therapeutic countermeasures for Ebola virus infections.


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

α5β1-Integrin controls ebolavirus entry by regulating endosomal cathepsins

Kathryn L. Schornberg; Charles J. Shoemaker; Derek Dube; Michelle Y. Abshire; Sue E. Delos; Amy H. Bouton; Judith M. White

Integrins are involved in the binding and internalization of both enveloped and nonenveloped viruses. By using 3 distinct cell systems—CHO cells lacking expression of α5β1-integrin, HeLa cells treated with siRNA to α5-integrin, and mouse β1-integrin knockout fibroblasts, we show that α5β1-integrin is required for efficient infection by pseudovirions bearing the ebolavirus glycoprotein (GP). These integrins are necessary for viral entry but not for binding or internalization. Given the need for endosomal cathepsins B and L (CatB and CatL) to prime GPs for fusion, we investigated the status of CatB and CatL in integrin-positive and integrin-negative cell lines. α5β1-Integrin-deficient cells lacked the double-chain (DC) forms of CatB and CatL, and this correlated with decreased CatL activity in integrin-negative CHO cells. These data indicate that α5β1-integrin-negative cells may be refractory to infection by GP pseudovirions because they lack the necessary priming machinery (the double-chain forms of CatB and CatL). In support of this model, we show that GP pseudovirions that have been preprimed in vitro to generate the 19-kDa form of GP overcome the requirement for α5β1-integrin for infection. These results provide further support for the requirement for endosomal cathepsins for ebolavirus infection, identify the DC forms of these cathepsins as previously unrecognized factors that contribute to cell tropism of this virus, and reveal a previously undescribed role for integrins during viral entry as regulators of endosomal cathepsins, which are required to prime the entry proteins of ebolavirus and other pathogenic viruses.


Journal of Virology | 2015

Ebola Virus and Severe Acute Respiratory Syndrome Coronavirus Display Late Cell Entry Kinetics: Evidence that Transport to NPC1+ Endolysosomes Is a Rate-Defining Step

Rebecca M. Mingo; James A. Simmons; Charles J. Shoemaker; Elizabeth A. Nelson; Kathryn L. Schornberg; Ryan S. D'Souza; James E. Casanova; Judith M. White

ABSTRACT Ebola virus (EBOV) causes hemorrhagic fevers with high mortality rates. During cellular entry, the virus is internalized by macropinocytosis and trafficked through endosomes until fusion between the viral and an endosomal membrane is triggered, releasing the RNA genome into the cytoplasm. We found that while macropinocytotic uptake of filamentous EBOV viruslike particles (VLPs) expressing the EBOV glycoprotein (GP) occurs relatively quickly, VLPs only begin to enter the cytoplasm after a 30-min lag, considerably later than particles bearing the influenza hemagglutinin or GP from lymphocytic choriomeningitis virus, which enter through late endosomes (LE). For EBOV, the long lag is not due to the large size or unusual shape of EBOV filaments, the need to prime EBOV GP to the 19-kDa receptor-binding species, or a need for unusually low endosomal pH. In contrast, since we observed that EBOV entry occurs upon arrival in Niemann-Pick C1 (NPC1)-positive endolysosomes (LE/Lys), we propose that trafficking to LE/Lys is a key rate-defining step. Additional experiments revealed, unexpectedly, that severe acute respiratory syndrome (SARS) S-mediated entry also begins only after a 30-min lag. Furthermore, although SARS does not require NPC1 for entry, SARS entry also begins after colocalization with NPC1. Since the only endosomal requirement for SARS entry is cathepsin L activity, we tested and provide evidence that NPC1+ LE/Lys have higher cathepsin L activity than LE, with no detectable activity in earlier endosomes. Our findings suggest that both EBOV and SARS traffic deep into the endocytic pathway for entry and that they do so to access higher cathepsin activity. IMPORTANCE Ebola virus is a hemorrhagic fever virus that causes high fatality rates when it spreads from zoonotic vectors into the human population. Infection by severe acute respiratory syndrome coronavirus (SARS-CoV) causes severe respiratory distress in infected patients. A devastating outbreak of EBOV occurred in West Africa in 2014, and there was a significant outbreak of SARS in 2003. No effective vaccine or treatment has yet been approved for either virus. We present evidence that both viruses traffic late into the endocytic pathway, to NPC1+ LE/Lys, in order to enter host cells, and that they do so to access high levels of cathepsin activity, which both viruses use in their fusion-triggering mechanisms. This unexpected similarity suggests an unexplored vulnerability, trafficking to NPC1+ LE/Lys, as a therapeutic target for SARS and EBOV.


Journal of Virology | 2014

Spatial Localization of the Ebola Virus Glycoprotein Mucin-Like Domain Determined by Cryo-Electron Tomography

Erin E. H. Tran; James A. Simmons; Alberto Bartesaghi; Charles J. Shoemaker; Elizabeth A. Nelson; Judith M. White; Sriram Subramaniam

ABSTRACT The Ebola virus glycoprotein mucin-like domain (MLD) is implicated in Ebola virus cell entry and immune evasion. Using cryo-electron tomography of Ebola virus-like particles, we determined a three-dimensional structure for the full-length glycoprotein in a near-native state and compared it to that of a glycoprotein lacking the MLD. Our results, which show that the MLD is located at the apex and the sides of each glycoprotein monomer, provide a structural template for analysis of MLD function.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Cell adhesion-dependent membrane trafficking of a binding partner for the ebolavirus glycoprotein is a determinant of viral entry.

Derek Dube; Kathryn L. Schornberg; Charles J. Shoemaker; Sue E. Delos; Tzanko S. Stantchev; Kathleen A. Clouse; Christopher C. Broder; Judith M. White

Ebolavirus is a hemorrhagic fever virus associated with high mortality. Although much has been learned about the viral lifecycle and pathogenesis, many questions remain about virus entry. We recently showed that binding of the receptor binding region (RBR) of the ebolavirus glycoprotein (GP) and infection by GP pseudovirions increase on cell adhesion independently of mRNA or protein synthesis. One model to explain these observations is that, on cell adhesion, an RBR binding partner translocates from an intracellular vesicle to the cell surface. Here, we provide evidence for this model by showing that suspension 293F cells contain an RBR binding site within a membrane-bound compartment associated with the trans-Golgi network and microtubule-organizing center. Consistently, trafficking of the RBR binding partner to the cell surface depends on microtubules, and the RBR binding partner is internalized when adherent cells are placed in suspension. Based on these observations, we reexamined the claim that lymphocytes, which are critical for ebolavirus pathogenesis, are refractory to infection because they lack an RBR binding partner. We found that both cultured and primary human lymphocytes (in suspension) contain an intracellular pool of an RBR binding partner. Moreover, we identified two adherent primate lymphocytic cell lines that bind RBR at their surface and strikingly, support GP-mediated entry and infection. In summary, our results reveal a mode of determining viral entry by a membrane-trafficking event that translocates an RBR binding partner to the cell surface, and they suggest that this process may be operative in cells important for ebolavirus pathogenesis (e.g., lymphocytes and macrophages).


Journal of Virology | 2016

Mapping of Ebolavirus Neutralization by Monoclonal Antibodies in the ZMapp Cocktail Using Cryo-Electron Tomography and Studies of Cellular Entry

Erin E. H. Tran; Elizabeth A. Nelson; Pranay Bonagiri; James A. Simmons; Charles J. Shoemaker; Connie S. Schmaljohn; Gary P. Kobinger; Larry Zeitlin; Sriram Subramaniam; Judith M. White

ABSTRACT ZMapp, a cocktail of three monoclonal antibodies (MAbs; c2G4, c4G7, and c13C6) against the ebolavirus (EBOV) glycoprotein (GP), shows promise for combatting outbreaks of EBOV, as occurred in West Africa in 2014. Prior studies showed that Fabs from these MAbs bind a soluble EBOV GP ectodomain and that MAbs c2G4 and c4G7, but not c13C6, neutralize infections in cell cultures. Using cryo-electron tomography, we extended these findings by characterizing the structures of c2G4, c4G7, and c13C6 IgGs bound to native, full-length GP from the West African 2014 isolate embedded in filamentous viruslike particles (VLPs). As with the isolated ectodomain, c13C6 bound to the glycan cap, whereas c2G4 and c4G7 bound to the base region of membrane-bound GP. The tomographic data suggest that all three MAbs bind with high occupancy and that the base-binding antibodies can potentially bridge neighboring GP spikes. Functional studies indicated that c2G4 and c4G7, but not c13C6, competitively inhibit entry of VLPs bearing EBOV GP into the host cell cytoplasm, without blocking trafficking of VLPs to NPC1+ endolysosomes, where EBOV fuses. Moreover, c2G4 and c4G7 bind to and can block entry mediated by the primed (19-kDa) form of GP without impeding binding of the C-loop of NPC1, the endolysosomal receptor for EBOV. The most likely mode of action of c2G4 and c4G7 is therefore by inhibiting conformational changes in primed, NPC1-bound GP that initiate fusion between the viral and target membranes, similar to the action of certain broadly neutralizing antibodies against influenza hemagglutinin and HIV Env. IMPORTANCE The recent West African outbreak of ebolavirus caused the deaths of more than 11,000 individuals. Hence, there is an urgent need to be prepared with vaccines and therapeutics for similar future disasters. ZMapp, a cocktail of three MAbs directed against the ebolavirus glycoprotein, is a promising anti-ebolavirus therapeutic. Using cryo-electron tomography, we provide structural information on how each of the MAbs in this cocktail binds to the ebolavirus glycoprotein as it is displayed—embedded in the membrane and present at high density—on filamentous viruslike particles that recapitulate the surface structure and entry functions of ebolavirus. Moreover, after confirming that two of the MAbs bind to the same region in the base of the glycoprotein, we show that they competitively block the entry function of the glycoprotein and that they can do so after the glycoprotein is proteolytically primed and bound to its intracellular receptor, Niemann-Pick C1. These findings should inform future developments of ebolavirus therapeutics.


Archives of Microbiology | 2011

Two putative histidine kinases are required for cyst formation in Rhodospirillum Centenum

Neena Din; Charles J. Shoemaker; Kent L. Akin; Christopher Frederick; Terry H. Bird

The photosynthetic bacterium, Rhodospirillum centenum, has a flexible life cycle that permits it to survive starvation as dormant cyst cells. Previous studies have identified some of the key regulators for encystment and demonstrated that the control of development is intricate. This complexity may arise from the need to integrate several environmental signals to mediate a switch from one mode of energy metabolism to another and to ensure that a transition to dormancy is initiated only when necessary. We searched for additional regulators of development by screening for encystment deficient strains after subjecting wild type R. centenum to mini-Tn5 mutagenesis. Analysis of “hypo-cyst” strains led to the identification of two genes that encode putative hybrid histidine kinases (cyd1 and cyd2). Cells with deletions of either gene fail to form cysts under conditions that normally induce development. Furthermore, the deletion strains exhibit altered swarming behavior suggesting that Cyd1 and Cyd2 affect behaviors utilized when the organism is attached to a substrate.


The Journal of Infectious Diseases | 2018

The Genetic Adjuvants Interleukin-12 and Granulocyte-Macrophage Colony Stimulating Factor Enhance the Immunogenicity of an Ebola Virus Deoxyribonucleic Acid Vaccine in Mice

John J. Suschak; Kenneth Bagley; Charles J. Shoemaker; Carolyn Six; Steven A. Kwilas; Lesley C. Dupuy; Connie S. Schmaljohn

In previous studies, we showed that deoxyribonucleic acid (DNA) vaccines expressing codon-optimized filovirus envelope glycoprotein genes protect mice and nonhuman primates from viral challenge when delivered by intramuscular (IM) electroporation (EP). To determine whether we could achieve equivalent immunogenicity and protective efficacy by a simplified delivery method, we generated DNA vaccine plasmids expressing genetic adjuvants to potentiate immune responses. We tested the Th1-inducing cytokine interleukin-12 and the granulocyte growth factor granulocyte-macrophage colony stimulating factor, both of which have demonstrated significant adjuvant effect when included in clinical DNA vaccine formulations. In addition, because interferon (IFN)-αβ is required for DNA vaccine-induced immunity, we tested inclusion of a potent stimulator of the IFN-αβ pathway. Our data suggest that IM vaccination of mice with plasmid DNA encoding genetic adjuvants enhances vaccine immunogenicity, resulting in increased anti-Ebola virus (EBOV) immunoglobulin G and T-cell responses. Codelivery of genetic adjuvants also improved EBOV neutralizing capability compared with vaccine alone. Finally, IM vaccination with plasmid EBOV and genetic adjuvants provided complete protection against EBOV challenge. Overall, our data suggest that codelivery of genetic adjuvants with filovirus DNA vaccines using IM delivery can provide comparable efficacy to the same DNA vaccines when delivered using IM-EP devices.

Collaboration


Dive into the Charles J. Shoemaker's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gene G. Olinger

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Connie S. Schmaljohn

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Corinne Scully

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carolyn Six

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Researchain Logo
Decentralizing Knowledge