Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Virol ; 89(5): 2931-43, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25552710

ABSTRACT

UNLABELLED: 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.


Subject(s)
Biological Transport , Ebolavirus/physiology , Endosomes/virology , Lysosomes/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Virus Internalization , Carrier Proteins/analysis , Cell Line , Endosomes/chemistry , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/chemistry , Membrane Glycoproteins/analysis , Niemann-Pick C1 Protein , Time Factors , Virosomes/metabolism
2.
Sci Transl Med ; 5(190): 190ra79, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23785035

ABSTRACT

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.


Subject(s)
Drug Approval , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/drug therapy , Selective Estrogen Receptor Modulators/therapeutic use , United States Food and Drug Administration , Animals , Cathepsins/metabolism , Chlorocebus aethiops , Clomiphene/pharmacology , Clomiphene/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Ebolavirus/drug effects , Endosomes/drug effects , Endosomes/metabolism , Hemorrhagic Fever, Ebola/virology , Hep G2 Cells , Humans , Hydrogen-Ion Concentration/drug effects , Mice , Mice, Inbred C57BL , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Survival Analysis , Toremifene/pharmacology , Toremifene/therapeutic use , United States , Vero Cells , Virion/drug effects , Virus Internalization/drug effects
3.
PLoS One ; 8(2): e56265, 2013.
Article in English | MEDLINE | ID: mdl-23441171

ABSTRACT

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 (IC(50) 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.


Subject(s)
Carrier Proteins/metabolism , Cations , Ebolavirus/drug effects , Ebolavirus/physiology , Membrane Glycoproteins/metabolism , Surface-Active Agents/pharmacology , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Biosynthetic Pathways/drug effects , Cations/chemistry , Cell Line , Hemorrhagic Fever, Ebola , Humans , Intracellular Signaling Peptides and Proteins , Niemann-Pick C1 Protein , Phenotype , Steroids/biosynthesis , Surface-Active Agents/chemistry
4.
Nat Rev Microbiol ; 10(5): 317-22, 2012 04 11.
Article in English | MEDLINE | ID: mdl-22491356

ABSTRACT

Viruses of the genera Ebolavirus and Marburgvirus are filoviruses that cause haemorrhagic fever in primates, with extremely high fatality rates. Studies have focused on elucidating how these viruses enter host cells, with the aim of developing therapeutics. The ebolavirus glycoprotein has been found to play key parts in all steps of entry. Furthermore, recent studies have identified Niemann-Pick C1 (NPC1), a protein that resides deep in the endocytic pathway, as an important host factor in this process.


Subject(s)
Carrier Proteins/metabolism , Ebolavirus/pathogenicity , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Models, Molecular , Niemann-Pick C1 Protein , Receptors, Virus/metabolism
5.
J Virol ; 86(1): 364-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22031933

ABSTRACT

Cellular entry of Ebola virus (EBOV), a deadly hemorrhagic fever virus, is mediated by the viral glycoprotein (GP). The receptor-binding subunit of GP must be cleaved (by endosomal cathepsins) in order for entry and infection to proceed. Cleavage appears to proceed through 50-kDa and 20-kDa intermediates, ultimately generating a key 19-kDa core. How 19-kDa GP is subsequently triggered to bind membranes and induce fusion remains a mystery. Here we show that 50-kDa GP cannot be triggered to bind to liposomes in response to elevated temperature but that 20-kDa and 19-kDa GP can. Importantly, 19-kDa GP can be triggered at temperatures ∼10°C lower than 20-kDa GP, suggesting that it is the most fusion ready form. Triggering by heat (or urea) occurs only at pH 5, not pH 7.5, and involves the fusion loop, as a fusion loop mutant is defective in liposome binding. We further show that mild reduction (preferentially at low pH) triggers 19-kDa GP to bind to liposomes, with the wild-type protein being triggered to a greater extent than the fusion loop mutant. Moreover, mild reduction inactivates pseudovirion infection, suggesting that reduction can also trigger 19-kDa GP on virus particles. Our results support the hypothesis that priming of EBOV GP, specifically to the 19-kDa core, potentiates GP to undergo subsequent fusion-relevant conformational changes. Our findings also indicate that low pH and an additional endosomal factor (possibly reduction or possibly a process mimicked by reduction) act as fusion triggers.


Subject(s)
Cathepsin L/metabolism , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/enzymology , Membrane Fusion , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Cell Line , Ebolavirus/chemistry , Ebolavirus/genetics , Endosomes/enzymology , Hemorrhagic Fever, Ebola/virology , Humans , Protein Conformation , Protein Processing, Post-Translational , Viral Envelope Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 107(38): 16637-42, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20817853

ABSTRACT

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).


Subject(s)
Ebolavirus/physiology , Ebolavirus/pathogenicity , Lymphocytes/physiology , Lymphocytes/virology , Viral Envelope Proteins/physiology , Virus Internalization , B-Lymphocytes/physiology , B-Lymphocytes/virology , Binding Sites , Cell Adhesion/physiology , Cell Line , Cell Membrane/physiology , Cell Membrane/virology , Host-Pathogen Interactions/physiology , Humans , In Vitro Techniques , Jurkat Cells , Microtubule-Associated Proteins/physiology , Plant Proteins/physiology , Receptors, Virus/physiology , Viral Envelope Proteins/chemistry , trans-Golgi Network/physiology
7.
Proc Natl Acad Sci U S A ; 106(19): 8003-8, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19416892

ABSTRACT

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 alpha(5)beta(1)-integrin, HeLa cells treated with siRNA to alpha(5)-integrin, and mouse beta(1)-integrin knockout fibroblasts, we show that alpha(5)beta(1)-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. Alpha(5)beta(1)-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 alpha(5)beta(1)-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 alpha(5)beta(1)-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.


Subject(s)
Cathepsins/metabolism , Ebolavirus/metabolism , Endosomes/metabolism , Integrin alpha5beta1/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Fibroblasts/metabolism , HeLa Cells , Humans , Integrins/metabolism , Mice , Mice, Knockout , Models, Biological
8.
J Virol ; 83(7): 2883-91, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144707

ABSTRACT

Entry of ebolavirus (EBOV) into cells is mediated by its glycoprotein (GP(1,2)), a class I fusion protein whose structure was recently determined (J. E. Lee et al., Nature 454:177-182, 2008). Here we confirmed two major predictions of the structural analysis, namely, the residues in GP(1) and GP(2) that remain after GP(1,2) is proteolytically primed by endosomal cathepsins for fusion and residues in GP(1) that are critical for binding to host cells. Mass spectroscopic analysis indicated that primed GP(1,2) contains residues 33 to 190 of GP(1) and all residues of GP(2). The location of the receptor binding site was determined by a two-pronged approach. We identified a small receptor binding region (RBR), residues 90 to 149 of GP(1), by comparing the cell binding abilities of four RBR proteins produced in high yield. We characterized the binding properties of the optimal RBR (containing GP(1) residues 57 to 149) and then conducted a mutational analysis to identify critical binding residues. Substitutions at four lysines (K95, K114, K115, and K140) decreased binding and the ability of RBR proteins to inhibit GP(1,2)-mediated infection. K114, K115, and K140 lie in a small region modeled to be located on the top surface of the chalice following proteolytic priming; K95 lies deeper in the chalice bowl. Combined with those of Lee et al., our findings provide structural insight into how GP(1,2) is primed for fusion and define the core of the EBOV RBR (residues 90 to 149 of GP(1)) as a highly conserved region containing a two-stranded beta-sheet, the two intra-GP(1) disulfide bonds, and four critical Lys residues.


Subject(s)
Ebolavirus/physiology , Glycoproteins/chemistry , Viral Fusion Proteins/chemistry , Virus Internalization , Amino Acid Sequence , Amino Acid Substitution/genetics , Binding Sites , Cell Line , DNA Mutational Analysis , Ebolavirus/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism
9.
J Virol ; 82(14): 7238-42, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18448524

ABSTRACT

Ebola virus infects a wide variety of adherent cell types, while nonadherent cells are found to be refractory. To explore this correlation, we compared the ability of pairs of related adherent and nonadherent cells to bind a recombinant Ebola virus receptor binding domain (EboV RBD) and to be infected with Ebola virus glycoprotein (GP)-pseudotyped particles. Both human 293F and THP-1 cells can be propagated as adherent or nonadherent cultures, and in both cases adherent cells were found to be significantly more susceptible to both EboV RBD binding and GP-pseudotyped virus infection than their nonadherent counterparts. Furthermore, with 293F cells the acquisition of EboV RBD binding paralleled cell spreading and did not require new mRNA or protein synthesis.


Subject(s)
Cell Adhesion , Ebolavirus/physiology , Viral Envelope Proteins/metabolism , Virus Attachment , Virus Internalization , Cell Line , Humans , Protein Binding
10.
J Bacteriol ; 188(2): 801-4, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16385070

ABSTRACT

The hemF gene of Rhodobacter sphaeroides 2.4.1 is predicted to code for an oxygen-dependent coproporphyrinogen III oxidase. We found that a HemF- mutant strain is unable to grow under aerobic conditions. We also determined that hemF expression is controlled by oxygen, which is mediated, at least in part, by the response regulatory protein PrrA.


Subject(s)
Bacterial Proteins/genetics , Coproporphyrinogen Oxidase/genetics , Genes, Bacterial/physiology , Rhodobacter sphaeroides/enzymology , Bacterial Proteins/metabolism , Coproporphyrinogen Oxidase/metabolism , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Histidine Kinase , Oxygen , Protein Kinases/genetics , Rhodobacter sphaeroides/genetics , Rhodobacter sphaeroides/growth & development , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...