ArticlePDF Available

Small molecule inhibitors reveal Niemann-Pick C1 is essential for Ebola virus infection

Authors:

Abstract and Figures

Ebolavirus (EboV) is a highly pathogenic enveloped virus that causes outbreaks of zoonotic infection in Africa. The clinical symptoms are manifestations of the massive production of pro-inflammatory cytokines in response to infection1 and in many outbreaks, mortality exceeds 75%. The unpredictable onset, ease of transmission, rapid progression of disease, high mortality and lack of effective vaccine or therapy have created a high level of public concern about EboV2. Here we report the identification of a novel benzylpiperazine adamantane diamide-derived compound that inhibits EboV infection. Using mutant cell lines and informative derivatives of the lead compound, we show that the target of the inhibitor is the endosomal membrane protein Niemann-Pick C1 (NPC1). We find that NPC1 is essential for infection, that it binds to the virus glycoprotein (GP), and that the anti-viral compounds interfere with GP binding to NPC1. Combined with the results of previous studies of GP structure and function, our findings support a model of EboV infection in which cleavage of the GP1 subunit by endosomal cathepsin proteases removes heavily glycosylated domains to expose the N-terminal domain, which is a ligand for NPC1 and regulates membrane fusion by the GP2 subunit. Thus, NPC1 is essential for EboV entry and a target for anti-viral therapy.
Content may be subject to copyright.
Small molecule inhibitors reveal Niemann-Pick C1 is essential
for ebolavirus infection
Marceline Côté1,6, John Misasi1,2,6, Tao Ren3,6, Anna Bruchez1,6, Kyungae Lee3, Claire
Marie Filone1,4, Lisa Hensley4, Qi Li1, Daniel Ory5, Kartik Chandran1,7, and James
Cunningham1
1Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA
USA 02115
2Division of Infectious Disease, Department of Medicine, Children's Hospital, Boston, MA, USA
02115
3New England Regional Center of Excellence for Biodefense and Emerging Infectious Diseases,
Harvard Medical School, Boston, MA USA 02115
4United States Army Medical Research Institute of Infectious Diseases, Virology Division,
Frederick, MD USA 21702
5Diabetic Cardiovascular Disease Center, Washington University School of Medicine, Saint Louis,
MO USA 63110
Summary
Ebolavirus (EboV) is a highly pathogenic enveloped virus that causes outbreaks of zoonotic
infection in Africa. The clinical symptoms are manifestations of the massive production of pro-
inflammatory cytokines in response to infection1 and in many outbreaks, mortality exceeds 75%.
The unpredictable onset, ease of transmission, rapid progression of disease, high mortality and
lack of effective vaccine or therapy have created a high level of public concern about EboV2. Here
we report the identification of a novel benzylpiperazine adamantane diamide-derived compound
that inhibits EboV infection. Using mutant cell lines and informative derivatives of the lead
compound, we show that the target of the inhibitor is the endosomal membrane protein Niemann-
Pick C1 (NPC1). We find that NPC1 is essential for infection, that it binds to the virus
glycoprotein (GP), and that the anti-viral compounds interfere with GP binding to NPC1.
Combined with the results of previous studies of GP structure and function, our findings support a
model of EboV infection in which cleavage of the GP1 subunit by endosomal cathepsin proteases
removes heavily glycosylated domains to expose the N-terminal domain3–7, which is a ligand for
Contact information: James Cunningham, MD, 5213 Karp Building, Boston, MA USA 02115 Phone: (617) 355-9058,
jcunningham@rics.bwh.harvard.edu.
6Contributed equally.
7Current address: Department of Microbiology, Albert Einstein College of Medicine, Bronx, NY USA 10461
Author Contributions: MC, JM, TR and AB equally contributed to this work. KC and TR performed the inhibitor screen. KL
synthesized and purified 3.0 analogs and TR tested them. TR, AB, JM, QL and MC carried out infection assays with pseudotyped
viruses. AB performed microscopy. JM purified recombinant glycoprotein. MC and JM designed and performed binding assays. MC
performed immunoprecipitation. DO provided NPC1 constructs, antibodies and CHO cell lines. Ebolavirus infections were performed
in the lab of LH by CF. JC supervised the project and wrote the manuscript. All authors reviewed the manuscript.
Additional experimental procedures are available online at www.nature.com/nature.
Supplementary Information is linked to the online version of the paper at www.nature.com/nature
Author Information: Reprints and permission information is available at www.nature.com/reprints.
Competing financial interests: None.
NIH Public Access
Author Manuscript
Nature. Author manuscript; available in PMC 2012 March 15.
Published in final edited form as:
Nature
. ; 477(7364): 344–348. doi:10.1038/nature10380.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NPC1 and regulates membrane fusion by the GP2 subunit8. Thus, NPC1 is essential for EboV
entry and a target for anti-viral therapy.
To identify chemical probes that target EboV host factors, we screened a library of small
molecules and identified a novel benzylpiperazine adamantane diamide, 3.0, that inhibits
infection of Vero cells by vesicular stomatitis virus particles (VSV) pseudotyped with EboV
Zaire GP, but not with VSV G or Lassa fever virus (LFV) GP (Fig. 1a,b). To verify that 3.0
is a bona fide inhibitor, we measured EboV growth on Vero cells for 96 hours and found it
was reduced by >99% in the presence of 3.0 (Supplementary Fig. 1a). We synthesized and
tested >50 analogs of 3.0 and found that the addition of a (methoxycarbonyl) benzyl group
at the ortho position of the benzene ring (compound 3.47) increased the potency as measured
by a single cycle of EboV GP-dependent infection and efficacy as measured by growth of
EboV on Vero cells (Fig 1a,c,d).
Previous studies revealed that the endosomal protease cathepsin B is essential for EboV
infection because it cleaves the GP1 subunit of GP3,4. To address the possibility that 3.0 and
3.47 target this step, we measured cathepsin B activity in the presence of these compounds
and found no effect in vitro or in cells (data not shown). Moreover, 3.0 and 3.47 inhibited
infection by VSV EboV particles treated with thermolysin, a metalloprotease that faithfully
mimics cathepsin cleavage of the GP1 subunit of GP (Supplementary Fig. 1b)4,9. These
findings demonstrate that cathepsin B is not the target of 3.0 and 3.47.
HeLa cells treated with 3.0 or 3.47 for more than 18 hours developed cytoplasmic vacuoles
that were labeled by cholesterol-avid filipin (Fig. 2a). The induction of filipin-stained
vacuoles by the compounds suggested that they target one or more proteins involved in
regulation of cholesterol uptake in cells. To test this hypothesis, we used mutant cell lines
and cells treated with siRNA to analyze proteins for which loss of activity had been
previously associated with cholesterol accumulation in late endosomes10–12. We found that
EboV GP infection is dependent on the expression of Niemann-Pick C1 (NPC1), but not
Niemann-Pick C2 (NPC2), acid sphingomyelinase (ASM), ALG-2-interacting protein X
(Alix), or oxysterol binding protein 5 (ORP5) (Fig 2b, Supplementary Fig. 2a–c). NPC1 is a
polytopic protein that resides in the limiting membrane of late endosomes and lysosomes
(LE/LY) and mediates distribution of lipoprotein-derived cholesterol in cells10,13. To
analyze the role of NPC1 in infection, we studied Chinese hamster ovary (CHO)-derived
cell lines that differ in expression of NPC1. We found that the titer of a murine leukemia
virus (MLV) vector pseudotyped with EboV GP on wild type CHO cells (CHOwt) exceeded
106 infectious units/ml (Fig. 2c). Importantly, CHO cells lacking NPC1 (CHOnull) were
completely resistant to infection by this virus and infection of these cells was fully restored
when NPC1 was expressed (CHONPC1). Thus, NPC1 expression is essential for EboV
infection.
In CHOnull cells, LE/LY are enlarged and contain excess cholesterol (Supplementary Fig.
3)14. To determine if EboV infection is inhibited by endosome dysfunction secondary to the
absence of NPC1, we studied a well-characterized NPC1 mutant P692S that is defective in
cholesterol uptake and NPC1-dependent membrane trafficking13–15 and found that
expression of NPC1 P692S fully supports infection of CHOnull cells (Fig. 2c). Conversely,
gain-of-function mutants NPC1 L657F and NPC1 D787N14 did not enhance EboV GP
infection. Thus, EboV entry is strictly dependent on NPC1 expression but not NPC1-
dependent cholesterol transport activity. Consistent with the conclusion that NPC1
expression is essential for EboV GP-dependent entry, we found that ebolavirus did not grow
on CHOnull cells (Fig. 2d). In addition, we tested a single round of infection by MLV
particles bearing GPs from the filoviruses EboV Sudan, EboV Côte d'Ivoire, EboV
Bundibugyo, EboV Reston and marburgvirus and found that all are strictly NPC1-dependent
Côté et al. Page 2
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
(Supplementary Fig. 4). Since these viruses are not closely related16, these findings suggest
that the requirement for NPC1 as an entry factor is conserved among viruses in the
Filoviridae family.
Since NPC1 and cathepsin B are both essential host factors, we analyzed their relationship
during infection. In our initial experiment, we compared cathepsin B activity in CHOnull
cells and found it was not significantly different from CHOwt cells (Supplementary Fig. 5).
To determine if NPC1 is required for virus processing by cathepsin B, we tested whether
thermolysin-cleaved particles are dependent on NPC1. As expected, we found that
thermolysin-cleaved particles are infectious and resistant to inactivation of cathepsin B when
NPC1 is present (Fig. 2e). However, thermolysin cleavage did not bypass the barrier to virus
infection of NPC1 deficient cells. Taken together, these findings indicate that cathepsin B
and NPC1 mediate distinct steps in infection.
Previous studies suggest that the product of cathepsin B cleavage of the GP1 subunit of
EboV GP is a ligand for a host factor6,17–20. To test this hypothesis, we performed a series
of experiments measuring binding of EboV GP to LE/LY membranes from CHOnull,
CHONPC1 and CHOP692S cells (Fig. 3a,b left panel). The source of EboV GP is a purified
recombinant protein that is truncated just before the transmembrane domain (EboV GPΔ).
EboV GPΔ is a trimer that is faithfully cleaved by thermolysin (Fig 3b, right panel). We
found that binding of EboV GPΔ to LE/LY membranes is concentration dependent,
saturable, and strictly dependent on both thermolysin cleavage of GP1 and membrane
expression of NPC1 or NPC1 P692S (Fig. 3c and Supplementary Fig. 6a,b). To determine if
cleaved GP binds to NPC1, we performed a co-immunoprecipitation experiment. LE/LY
membranes were incubated with EboV GPΔ and then solubilized in detergent. NPC1 was
recovered from the lysate by immunoprecipitation and the immune complexes were
analyzed for GP1. The findings indicate that cleaved EboV GPΔ binds to NPC1 and that
uncleaved EboV GPΔ does not (Fig. 3d).
Since the small molecules 3.0 and 3.47 inhibit infection of thermolysin-treated VSV EboV
GP particles (Supplementary Fig. 1b) and inhibit cholesterol uptake from LE/LY into cells
(Fig. 2a), both of which require NPC1, this suggests the possibility that these compounds
directly target NPC1. To test this hypothesis, we synthesized the 3.47 derivative 3.98. This
compound has anti-EboV activity and contains two additional functional moieties: an aryl-
azide for photo-affinity labeling of target proteins and an alkyne for click conjugation with
biotin21 (Supplementary Fig. 7). Compound 3.98 was incubated with LE/LY membranes,
activated by UV light and coupled to biotin. NPC1 was then isolated by
immunoprecipitation and analyzed using streptavidin-HRP. The findings show that NPC1 is
cross-linked to 3.98 and that cross-linking is inhibited by the presence of 3.47 but not by the
closely-related analog 3.18, which has weak anti-viral activity (Fig. 4a, Supplementary Fig.
7). In addition, we observed that overexpression of NPC1 conferred resistance to the anti-
viral activity of 3.0 and 3.47 (Supplementary Fig. 8), thus providing additional functional
evidence supporting the conclusion based on the results of the cross-linking experiment
using 3.98 that NPC1 is a direct target of the anti-viral compounds.
The evidence that NPC1 is the target of the 3.0-derived small molecules selected for anti-
EboV suggested that these compounds interfere with binding of cleaved GP to NPC1.
Consistent with this hypothesis, we found that 3.0 and 3.47 inhibited binding of cleaved
EboV GPΔ to NPC1 membranes in a concentration-dependent manner (Fig. 4b).
Importantly, we observed a direct correlation between the potency of 3.47, 3.0, and 3.18 in
inhibiting binding (Fig. 4b, left panel) and in inhibiting EboV infection (Supplementary Fig.
7). We also tested U18666A, a small molecule inhibitor of LE/LY cholesterol transport and
membrane trafficking22,23, and found that it does not inhibit binding of cleaved EboV GP to
Côté et al. Page 3
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NPC1 membranes (Fig. 4b, right panel). These results support the conclusion that the 3.0-
derived compounds inhibit EboV infection by interfering with binding of cleaved GP to
NPC1.
Previous studies show that cleavage of GP by endosomal cathepsin proteases removes
heavily-glycosylated domains in the GP1 subunit and exposes the N-terminal domain3–7. It
has been proposed that binding of this domain to a host factor is essential for
infection6,17–20. The most straight forward interpretation of the findings in this report is that
NPC1 is this host factor. This conclusion is based on the observations that NPC1 is strictly
required for infection, that cleaved GP1 binds to NPC1, and that small molecules that target
NPC1 are potent inhibitors of binding and infection.
Analysis of the EboV GP structure reveals that the residues in the N-terminal domain of
GP1 that mediate binding to NPC1 are interspersed with the residues that make stabilizing
contacts with GP25. This structural feature is consistent with the possibility that binding of
cleaved GP1 to NPC1 relieves the GP1-imposed constraints on GP2 and promotes virus
fusion to the limiting membrane (Fig. 4c). The role of cathepsin proteases in cleavage of
GP1 to expose the NPC1 binding site during EboV infection is analogous to the role of CD4
in inducing a conformational change in gp120 to expose the co-receptor binding site during
human immunodeficiency virus infection8. An alternative possibility is that binding of
protease-cleaved GP1 to NPC1 is an essential step in infection, but virus membrane fusion is
not completed until an additional signal is received, possibly including further cleavage of
GP by cathepsin proteases, as has been proposed3,4,9. These studies provide an example of
how small molecules identified by screening and medicinal chemistry optimization can be
used as molecular probes to analyze virus-host interactions.
Methods Summary
Screening of small molecules was performed at the New England Regional Centers of
Excellence for Biodefense and Emerging Infectious Diseases at Harvard Medical School.
Infection was assayed using VSV pseudotyped viruses encoding GFP or luciferase.
Experiments with native ebolavirus were performed under BSL-4 conditions at the United
States Army Medical Research Institute for Infectious Diseases. Cells were infected with
EboV Zaire-Mayinga GFP and growth was measured by mean fluorescence. EboV GPΔ is
a derivative of EboV GP in which the transmembrane domain has been replaced by a
GCN4-derived trimerization domain followed by a His6 tag for purification. Late
endosomes/lysosomes (LE/LY) were isolated by differential centrifugation and further
purified by Percoll density gradient centrifugation. LE/LY were disrupted by incubation
with methionine methyl ester and coated onto high binding ELISA plates. Following
attachment, unbound LE/LY membranes were removed and plates were blocked. Bound
membranes were incubated with the indicated amounts of native or thermolysin-cleaved
EboV GPΔ protein. Unbound EboV GPΔ protein was removed, membranes were washed
and bound EboV GPΔ protein was recovered in SDS loading buffer and analyzed by
immunoblot using GP1 antiserum. Where applicable, membranes were pre-incubated with
3.0, 3.47, 3.18 or vehicle prior to the addition of EboV GPΔ. To analyze EboV GPΔ
binding to NPC1, LE/LY membranes were dissolved in 10mM CHAPSO and NPC1 was
recovered by immunoprecipitation and the immune complexes were analyzed by
immunoblot probed with EboV GP1 antiserum.
Côté et al. Page 4
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Methods
Cell lines
Vero, 293T, HeLa (ATCC) and human fibroblasts26 (Coriell) were maintained in DMEM
(Invitrogen) supplemented with 5% FetalPlex, 5% FBS (Gemini) or 10% FBS (HeLa,
human fibroblasts). All CHO derived cell lines were grown as previously described14,27. We
have designated the CHO-K1 cell line as CHOwt, CHO-M12 as CHOnull, CHO-wt8 as
CHONPC1, and the CHO-derived cell lines expressing NPC1 mutants as CHO NPC1 P692S,
CHO NPC1 L657F, and CHO NPC1 D787N. CHO/NPC1-1, designated here as CHO
hNPC1, expresses high levels of human NPC127.
Antibodies
Rabbit polyclonal anti-serum was raised against a peptide corresponding to residues 83 to 98
of ebolavirus Zaire Mayinga GP1 (TKRWGFRSGVPPKVVC). Antibodies to NPC1 and V-
ATPase B1/2 were obtained from Abcam and Santa Cruz, respectively.
Expression plasmids
Mucin domain-deleted EboV Zaire Mayinga GP (EboV GP) and VSV G were previously
described3. Plasmids encoding Côte d'Ivoire-Ivory Coast GP, Sudan-Boniface GP, Reston-
Penn. GP and Marburg-Musoke GP were obtained from Anthony Sanchez and the mucin
domain-deleted (ΔMuc) derivatives were created: ZaireΔMuc GP (Δa.a. 309–489), Côte
d'IvoireΔMuc GP (Δa.a. 310–489), SudanΔMuc GP (Δa.a. 309–490), and RestonΔMuc GP
(Δa.a. 310–490). Bundibungyo-Uganda viral RNA was Trizol extracted and PCR used to
generate a construct that expresses a mucin-deleted GP (Δa.a. 309–489). A plasmid
encoding Lassa fever virus GP1 was kindly provided by Gary Nabel. A codon-optimized
sequence encoding GP2 was generated and combined with the GP1 sequence in pCAGGS to
complete a GP expression vector.
Production and purification of pseudotyped virions
VSV-ΔG pseudotyped viruses were created as described previously3. LacZ-encoding
retroviral pseudotypes bearing the designated envelope glycoproteins were prepared as
previously described28.
Thermolysin digestion of EboV GP Virus and EboV GPΔ
Purified EboV GPΔ (50μg/mL) or VSV particles pseudotyped with EboV GP were
incubated at 37°C for 1 hour with the metalloprotease thermolysin (Sigma, 0.2mg/ml) in NT
buffer (10 mM Tris.Cl [pH 7.5], 135 mM NaCl). The reaction was stopped using 500 μM
Phosphoramidon (Sigma) at 4°C. Cleaved EboV GPΔ was stored in phosphate buffered
saline supplemented with 1 mM EDTA, 1 mM PMSF (Sigma) and 1× EDTA-Free Complete
Protease Inhibitor Cocktail (Roche).
Infection assays with pseudotyped virus
VSV pseudotyped viruses expressing GFP were added to cells in serial 10-fold dilutions and
assayed using fluorescence microscopy. An infectious unit (i.u.) is defined as one GFP-
expressing cell within a range where the change in GFP-positive cells is directly
proportional to the virus dilution. For VSV expressing the luciferase reporter, pseudotyped
virus was added to cells and luciferase activity was assayed 6–20 hours post-infection using
the firefly luciferase kit (Promega). Signal was measured in relative luminescence units
(RLU) using an EnVison plate reader (Perkin Elmer). In experiments involving inhibitors,
stock solutions of 3.0 (20mM) and 3.47 (10mM) in DMSO were diluted to a final
concentration of 1% DMSO in media. Inhibitory activity was stable in the media of cultured
Côté et al. Page 5
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
cells for >72hours as assessed using a single cycle entry assay. Infection of target cells with
LacZ-encoding retroviral pseudotypes was performed in the presence of 5 μg/ml polybrene
(Sigma). Seventy-two hours post-infection, cells were stained for LacZ activity and titer was
determined by counting positive foci and expressed as focus forming units (FFU) per ml of
virus.
Ebolavirus infections under BSL-4 conditions
Vero cells or CHO cells were seeded to 96-well plates and exposed to EboV-GFP29. Vero
cells were incubated with 3.0 (40 μM), 3.47 (40 μM), E-64-d (150 μM) or 1% DMSO 90
minutes prior to the addition of virus (moi=0.1). Virus was added to CHO cells at moi of 1
as measured on Vero cells. Virus-encoded GFP fluorescence was determined using a
SpectraMax M5 plate reader (Molecular Devices) at Ex 485nm, Em 515nm, cutoff 495nm at
22.5, 42, 71 and 97 hours post-infection. An additional inhibitor experiment was performed
using 3.0. Vero cells were treated with 3.0 (20μM) or 1% DMSO alone for 4 hours, and then
infected with EBOV Zaire-1995 (moi=0.1). After 1 hour, the virus inoculum was removed,
cells were washed, and fresh media containing 3.0 or DMSO was added. Cell supernatant
was collected at 0, 24, 48, 72, or 91 hours post-infection. RNA was isolated from the
supernatant using Virus RNA Extraction kits (Qiagen) and EboV NP RNA was measured
using a real-time RT-PCR assay30. Virus titer was calculated using a standard curve
obtained using a virus stock of known titer as determined by plaque assay.
Screen for ebolavirus entry inhibitors
Screening of small molecules was performed at the New England Regional Centers of
Excellence for Biodefense and Emerging Infectious Diseases at Harvard Medical School.
Vero cells were seeded in 384-well plates at a density of 5×104 cells per well using a Matrix
WellMate (Thermo Scientific). The ChemBridge3, ChemDiv4, ChemDiv5 and Enamine2
compound libraries were transferred by robotics to the assay plates using stainless steel pin
arrays. The compounds were screened at a constant dilution to achieve a final concentration
between 10μM and 60μM. After incubation for 2 hours at 37 °C, viruses were dispensed into
each well (moi =1) and incubated for an additional 6 hours to allow virus gene expression.
Cells were lysed by addition of Steady-Glo (Promega) and after 10 minutes at room
temperature luminescence was measured using an EnVision plate reader. Each compound
was tested in duplicate. Candidate compounds that inhibited EboV GP infection by more
than 80% were analyzed for potency, selectivity and absence of cytotoxicity (using Cyto-
Tox assay, Promega) and 3.0 (2-((3r,5r,7r)-adamantan-1-yl)-N-(2-(4-benzylpiperazin-1-
yl)-2-oxoethyl)acetamide) was identified. The antiviral activity of the inhibitors was verified
on human cells (HeLa, A549, 293T), mouse embryonic fibroblasts, and Chinese hamster
ovary cells.
Synthesis of 3.0 derivatives
Compound 3.47 (methyl 4-((2-((4-(2-(2-((3r,5r,7r)-adamantan-1-
yl)acetamido)acetyl)piperazin-1-yl)methyl)phenoxy)methyl)benzoate) was prepared via a
multi-step synthesis starting from N-Cbz-piperazine. Thus, coupling of N-Cbz-piperazine
with N-Bocglycine followed by removal of the Boc group under acidic conditions yielded 4-
Cbz-piperazine glycinamide. After acylation of the terminal amine with adamantan-1-acetyl
chloride, the Cbz group was removed by hydrogenolysis to give (1-(adamantan-1-
yl)acetamido)acetyl)piperazine. The piperazine was then benzylated via reductive amination
with 2-(4-methoxycarbonyl)benzyloxybenzaldehyde using sodium triacetoxyborohydride to
provide 3.47. Compound 3.18 was synthesized in a similar fashion. Compound 3.98 was
prepared via a multi-step synthesis as follows. First, 2-hydroxy-5-nitrobenzaldehyde was
alkylated by 4-ethynylbenzyl bromide in the presence of potassium carbonate in DMF.
Resulting benzyloxy aldehyde underwent reductive amination with 2-((3r,5r,7r)-
Côté et al. Page 6
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
adamantan-1-yl)-N-(2-oxo-2-(piperazin-1-yl)ethyl)acetamide using sodium
triacetoxyborohydride. The nitro group was then reduced to aniline (SnCl2), diazotized
(NaNO2), and the diazonium finally converted to azide to yield 3.98. See Supplementary
Information for detailed experimental procedures and characterization data.
Protease inhibitors and protease activity assays
The measurement of cathepsin B activity and the use of the inhibitor CA074 (Sigma) have
been previously described3.
Detection of intracellular cholesterol
Cells were stained with filipin (50 μg/ml, Cayman Chemical) as previously described14.
Images of stained cells were obtained using epifluorescence microscopy (Nikon Eclipse
TE2000U). The images in the supplementary figures were processed using ImageJ software.
Production and purification of EboV GPΔ soluble protein
EboV GPΔ is a derivative of the mucin-deleted EboV Zaire-Mayinga GP in which the
transmembrane domain and C-terminus (a.a. 657–676) has been replaced by a GCN4-
derived trimerization domain (MKQIEDKIEEILSKIYHIENEIARIKKLIGEV) and a His6
tag. The expression plasmid encoding EboV GPΔ was transfected into 293T cells using
lipofectamine2000. Eighteen to twenty-four hours later the culture medium was replaced
with 293SFMII (Invitrogen) supplemented with 1× non-essential amino acids and 2 mM
CaCl2 and harvested daily for four days. Media containing soluble EboV GPΔ was filtered
and PMSF (1 mM)/ 1× EDTA-Free Complete Protease Inhibitor Cocktail was added. EboV
GPΔ was purified by affinity chromatography using Ni-NTA agarose beads (Qiagen),
dialyzed against PBS using a 3kDa dialysis cartridge (Pierce) and stored at 80°C. Purity
and integrity of EboV GPΔ were analyzed by SDS-PAGE.
Membrane Binding Assay
Indicated cells were washed with PBS ×2, scraped in homogenization (HM) buffer (0.25 M
sucrose, 1 mM EDTA, 10 mM Hepes pH7.0), and disrupted with a Dounce homogenizer.
Nuclei and debris were pelleted by centrifugation at 1000 × g for 10 min. The post-nuclear
supernatant was centrifuged at 15000 × g for 30 min at 4°C and the pellet, containing the
LE/LY, was resuspended in a total volume of 0.9ml composed of 20% Percoll (Sigma) and
0.4% BSA (Sigma) in HM and centrifuged at 36000 × g for 30 min at 4°C. Fractions (0.150
ml) were collected from the bottom to the top of the tube and those containing the highest β-
N-acetylglucosamidase activity, as assessed by release of 4-methylumbelliferone from 4-
methylumbellifferyl-N-acetyl-β-D-glucosaminide (Sigma), were pooled and incubated in
20mM methionine methyl-ester (Sigma) for 1 hour at room temperature. Following LE/LY
disruption, 1× EDTA-Free Complete Protease Inhibitor Cocktail and 1 mM PMSF was
added. The amount of purified LE/LY membranes used for the binding assay was
normalized using the activity of the marker β-N-acetylglucosamidase and validated by
immunoblot using V-ATPase B1/2 antibody (Supplemental Fig. 5).
Disrupted LE/LY membranes were coated on high-binding ELISA plates (Corning)
overnight at 4°C. Unbound membranes were removed and wells containing bound
membranes were blocked for 2 hours at room temperature with binding buffer (PBS, 5%
FBS, 1 mM PMSF, 1mM EDTA, 1× Complete Protease Inhibitor Cocktail). The indicated
amount of purified EboV GPΔ, pretreated or not with thermolysin, in binding buffer was
added to each well and incubated for 1 hour at room temperature. Unbound proteins were
removed and wells were washed 3 times with PBS. Membrane bound EboV GPΔ was
solubilized in SDS-loading buffer. Bound and unbound EboV GPΔ were detected by
Côté et al. Page 7
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
immunoblot using the EboV GP1 anti-serum. For binding assays in the presence of
inhibitors, the immobilized membranes were pre-incubated at room temperature with the
inhibitor or vehicle (10% DMSO) in binding buffer. After 30 minutes, thermolysin cleaved
EboV GPΔ was added in the continuous presence of compound and bound and unbound
GP was measured as described above.
Co-Immunoprecipitation
CHOnull and CHOhNPC1 cells were homogenized as described above. The 15000 × g
membrane pellet was resuspended in HM buffer and protein content was measured using the
BCA assay (Pierce). The LE/LY membranes contained in the 15000 × g resuspended pellet
were disrupted by incubation with 20 mM methionine methyl-ester for 1 hour at RT.
Membranes of equal protein content were incubated with indicated amounts of EboV GPΔ,
pre-treated or not with thermolysin, for 1 hour at RT in the presence of Complete Protease
Inhibitor Cocktail (Roche) and incubated for an additional hour on ice before the addition of
membrane lysis buffer (12.5mM CHAPSO, 150mM NaCl, 1mM EDTA, 10mM Tris/HCl
pH7.4) for a final concentration of 10mM CHAPSO. Proteins were solubilized on ice for 20
min and debris was removed by centrifugation at 12000 × g for 10 min at 4°C. The soluble
membrane lysates were incubated with anti-NPC1 antibody for 1 hour at 4°C and then
incubated with Protein A-agarose beads (Sigma) for an additional 4 hours at 4°C. Beads
were then washed 3 times with 8mM CHAPSO, 150 mM NaCl, 1mM EDTA, 10 mM Tris/
HCl pH7.4 and immunoprecipitated product was eluted by incubation in 0.1M glycine pH
3.5 for 5 min at RT. The eluted complex was then neutralized and analyzed by immunoblot
using the indicated antibody.
Photoactivation and click chemistry
Photoactivation and click chemistry were performed as described previously with some
modifications21. Briefly, the 15000× g pellets from homogenized CHOhNPC1 or CHONull
cells were resuspended in PBS and incubated with the indicated concentrations of 3.47, 3.18
or DMSO for 10 minutes at room temperature. Membranes were then incubated with 25μM
of 3.98 for an additional 10 minutes and exposed to ultraviolet light (365 nm) for 1 min on
ice. Proteins were solubilized in lysis buffer (1% Triton X-100, 0.1% NP-40, 20mM HEPES
pH7.4) containing protease inhibitors and 150 μM of biotin-azide (Invitrogen) was added,
followed by 5 mM L-ascorbic acid. The cycloaddition reaction (click chemistry) was
initiated by the addition of 1 mM CuSO4 and samples were incubated for 3 hours at room
temperature. NPC1 was immunoprecipitated and the product was resolved by SDS-PAGE,
transferred to PVDF membrane, and analyzed for conjugation of 3.98 to NPC1 using
streptavidin-HRP (Sigma).
Supplementary Material
Refer to Web version on PubMed Central for supplementary material.
Acknowledgments
We thank Bryden Considine, Anna Nilsson, and Sean Wilkes for assistance, Su Chiang for critical reading of the
manuscript, Gerald Beltz, Nathanael Gray, Sergio Grinstein, Yiannis Iannou, Rodney Infante, Johannes Kornhuber,
Francis Sharom and Sean Whelan for discussion. This work was supported by grants from U54 AI057159, R01
CA104266 to JC, PIDS-Sanofi-Pasteur Fellowship, K12-HD052896 and 5K08AI079381 to JM, 5-T32- HL007623
to AB, and fellowship from Fonds de la Recherche en Santé du Québec to MC. CF was supported by the
Postgraduate Research Participation Program at the U.S. Army Medical Research and Material Command
administered by the Oak Ridge Institute for Science and Education through an interagency agreement between the
U.S. Department of Energy and USAMRMC.
Côté et al. Page 8
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
References
1. Zampieri CA, Sullivan NJ, Nabel GJ. Immunopathology of highly virulent pathogens: insights from
ebola virus. Nat. Immunol. 2007; 8:1–6.
2. Geisbert TW, Jahrling PB. Exotic emerging viral diseases: progress and challenges. Nat. Med. 2004;
10:S110–121. [PubMed: 15577929]
3. Chandran K, Sullivan NJ, Felbor U, Whelan SP, Cunningham JM. Endosomal proteolysis of the
Ebola virus glycoprotein is necessary for infection. Science. 2005; 308:1643–1645. [PubMed:
15831716]
4. Schornberg K, et al. Role of endosomal cathepsins in entry mediated by the Ebola virus
glycoprotein. J. Virol. 2006; 80:4174–4178. [PubMed: 16571833]
5. Lee JE, et al. Structure of the Ebola virus glycoprotein bound to an antibody from a human survivor.
Nature. 2008; 454:177–182. [PubMed: 18615077]
6. Dube D, et al. The primed ebolavirus glycoprotein (19-kilodalton GP1,2): Sequence and residues
critical for host cell binding. J. Virol. 2009; 83:2883–2891. [PubMed: 19144707]
7. Hood CL, et al. Biochemical and structural characterization of cathepsin L-processed Ebola virus
glycoprotein: Implications for viral entry and immunogenicity. J. Virol. 2010; 84:2972–2982.
[PubMed: 20053739]
8. Harrison SC. Viral membrane fusion. Nat. Struct. Mol. Biol. 2008; 15:690–698. [PubMed:
18596815]
9. Wong A, Sandesara R, Mulherkar N, Whelan S, Chandran K. A forward genetic strategy reveals
destabilizing mutations in the ebolavirus glycoprotein that alter its protease dependence during cell
entry. J. Virol. 2010; 84:163–175. [PubMed: 19846533]
10. Kolter T, Sandhoff K. Lysosomal degradation of membrane lipids. FEBS Lett. 2010; 584:1700–
1712. [PubMed: 19836391]
11. Du X, et al. A role for oxysterol-binding protein-related protein 5 in endosomal cholesterol
trafficking. J. Cell Biol. 2011; 192:121–35. [PubMed: 21220512]
12. Chevallier J, et al. Lysobisphosphatidic acid controls endosomal cholesterol levels. J. Biol. Chem.
2008; 283:27871–27880. [PubMed: 18644787]
13. Ko DC, Gordon MD, Jin JY, Scott MP. Dynamic movements of organelles containing Niemann-
Pick C1 protein: NPC1 involvement in late endocytic events. Mol. Biol. Cell. 2001; 12:601–614.
[PubMed: 11251074]
14. Millard EE, et al. The sterol-sensing domain of the Niemann-Pick C1 (NPC1) protein regulates
trafficking of low density lipoprotein cholesterol. J. Biol. Chem. 2005; 280:28581–28590.
[PubMed: 15908696]
15. Ohgami N, et al. Binding between the Niemann-Pick C1 protein and a photoactivatable cholesterol
analog requires a functional sterol-sensing domain. Proc. Natl. Acad. Sci. USA. 2004; 101:12473–
12478. [PubMed: 15314240]
16. Towner JS, et al. Newly discovered ebola virus associated with hemorrhagic fever outbreak in
Uganda. PLoS Pathog. 2008; 4:e1000212. [PubMed: 19023410]
17. Kuhn JH, et al. Conserved receptor-binding domains of Lake Victoria marburgvirus and Zaire
ebolavirus bind a common receptor. J. Biol. Chem. 2006; 281:15951–15958. [PubMed: 16595665]
18. Kaletsky RL, Simmons G, Bates P. Proteolysis of the Ebola virus glycoproteins enhances virus
binding and infectivity. J. Virol. 2007; 81:13378–13384. [PubMed: 17928356]
19. Brindley MA, et al. Ebola virus glycoprotein 1: Identification of residues important for binding and
postbinding events. J. Virol. 2007; 81:7702–7709. [PubMed: 17475648]
20. Dube D, et al. Cell adhesion-dependent membrane trafficking of a binding partner for the
ebolavirus glycoprotein is a determinant of viral entry. Proc. Natl. Acad. Sci. USA. 2010;
107:16637–16642. [PubMed: 20817853]
21. Ban HS, et al. Identification of HSP60 as a primary target of
ocarboranylphenylphenoxyacetanilide, an HIF-1α inhibitor. J. Am. Chem. Soc. 2010; 132:11870–
11871. [PubMed: 20695501]
22. Sobo K, et al. Late endosomal cholesterol accumulation leads to impaired intraendosomal
trafficking. PLoS ONE. 2007; 2:e851. [PubMed: 17786222]
Côté et al. Page 9
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
23. Huynh KK, Gershenzon E, Grinstein S. Cholesterol accumulation by macrophages impairs
phagosome maturation. J. Biol. Chem. 2008; 283:35745–35755. [PubMed: 18955491]
24. Saeed MF, Kolokoltsov AA, Albrecht T, Davey RA. Cellular entry of ebola virus involves uptake
by a macropinocytosis-like mechanism and subsequent trafficking through early and late
endosomes. PLoS Pathogen. 2010; 6:e1001110. [PubMed: 20862315]
25. Nanbo A, et al. Ebolavirus is internalized into host cells via macropinocytosis in a viral
glycoprotein-dependent manner. PLoS Pathogen. 2010; 6:e1001121. [PubMed: 20886108]
26. Gelsthorpe ME, et al. Niemann-Pick type C1 I1061T mutant encodes a functional protein that is
selected for endoplasmic reticulum-associated degradation due to protein misfolding. J Biol Chem.
2008; 283:8229–8236. [PubMed: 18216017]
27. Millard EE, Srivastava K, Traub LM, Schaffer JE, Ory DS. Niemann-pick type C1 (NPC1)
overexpression alters cellular cholesterol homeostasis. J Biol Chem. 2000; 275:38445–38451.
[PubMed: 10964915]
28. Soneoka Y, et al. A transient three-plasmid expression system for the production of high titer
retroviral vectors. Nucleic Acids Res. 1995; 23:628–633. [PubMed: 7899083]
29. Towner JS, et al. Generation of eGFP expressing recombinant Zaire ebolavirus for analysis of early
pathogenesis events and high-throughput antiviral drug screening. Virology. 2005; 332:20–27.
[PubMed: 15661137]
30. Weidmann M, Mühlberger E, Hufert FT. Rapid detection protocol for filoviruses. J Clin Virol.
2004; 30:94–99. [PubMed: 15072761]
Côté et al. Page 10
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Structure and function of ebolavirus entry inhibitors
a, Compounds 3.0 and 3.47.
b,c, Vero cells were cultured in media containing increasing concentrations of 3.0 (b) or
3.47 (c) for 90 minutes prior to the addition of VSV particles encoding luciferase (b) or GFP
(c) and pseudotyped with either EboV GP, VSV G or Lassa fever virus GP (LFV GP). Virus
infection is reported as percent of luminescence units (RLU) or GFP-positive cells relative
to cells exposed to DMSO vehicle alone. Data is mean ± s.d. (n=4) and is representative of 3
experiments.
d, Vero cells were cultured in media containing 3.0 [40 μM], 3.47 [40 μM], vehicle (1%
DMSO) or the cysteine cathepsin protease inhibitor E-64d [150μM] 90 minutes prior to the
Côté et al. Page 11
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
addition of replication competent ebolavirus Zaire-Mayinga encoding GFP (moi = 0.1).
Results are mean relative fluorescence units ± s.e.m. (n=3).
Côté et al. Page 12
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 2. NPC1 is essential for ebolavirus infection
a, HeLa cells were treated with 3.0 (20 μM), 3.47 (1.25 μM) or vehicle for 18 hours, then
fixed and incubated with the cholesterol-avid fluorophore filipin.
b, HeLa cells were transfected with siRNAs targeting ASM, Alix, NPC1, NPC2, and ORP5.
After 72 hours, VSV EboV GP or LFV GP infection of these cells was measured as in Fig
1c. Data is mean ± s.d. (n=3) and is representative of 3 experiments.
c, CHOwt, CHOnull and CHOnull cells stably expressing mouse NPC1 (CHONPC1) or NPC1
mutants L657F, P692S, D787N were exposed to MLV particles encoding LacZ and
pseudotyped with either EboV GP or VSV G. Results are the mean ± s.d. (n=4) and is
representative of 3 experiments.
Côté et al. Page 13
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
d, CHOwt, CHOnull, and CHONPC1 cells were infected with replication competent
ebolavirus Zaire-Mayinga encoding GFP (moi = 1). Results are mean relative fluorescence
units ± s.d. (n=3).
e, CHOwt and CHOnull cells were treated with the cathepsin B inhibitor CA074 (80 μM) or
vehicle. These cells were challenged with VSV G particles or VSV EboV GP particles
treated with thermolysin (EboV GPTHL) or untreated control (EboV GP). Infection was
measured as in Fig 1b. Data is mean ± s.d. (n=9).
Côté et al. Page 14
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 3. Protease-cleaved EboV GP binds to NPC1
a, Schematic diagram of EboV GP1 binding assay used in panel c.
b, (left) LE/LY membranes from CHONPC1, CHOnull and CHO NPC1 P692S cells were
analyzed by immunoblot using antibodies to NPC1 or V-ATPase B1/2. (right) VSV-EboV
GP particles and EboV GPΔ protein were incubated in the presence or absence of
thermolysin (THL) and analyzed by immunoblot for GP1.
c, EboV GPΔ or thermolysin-cleaved EboV GPΔ (0.1, 0.5, or 1.0 μg) was added to LE/
LY membranes purified from CHOnull or CHONPC1 cells. Membrane bound and unbound
GP1 were analyzed by immunoblot.
d,. LE/LY membranes from CHOnull or CHOhNPC1 cells were incubated with EboV GPΔ
or thermolysin-cleaved EboV GPΔ. Following binding, membranes were dissolved in
CHAPSO, NPC1 was precipitated using an NPC1-specific antibody, and the
immunoprecipitate and the input membrane lysate were analyzed by immunoblot for NPC1
(top) or GP1 (bottom). * IgG heavy chain.
Côté et al. Page 15
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 4. NPC1 is a target of the small molecule inhibitors
a, LE/LY membranes from CHOnull or CHOhNPC1 cells were incubated at the indicated
concentrations of 3.47, 3.18 or DMSO (5%) prior to the addition of the photoactivatable
3.98 (25 μM). After incubation, 3.98 was activated by UV light and then conjugated to
biotin. NPC1 was immunoprecipitated and analyzed by immunoblot for conjugation of 3.98
to NPC1 using streptavidin-HRP (top) and recovery of NPC1 (bottom).
b, Thermolysin-cleaved EboV GPΔ protein (1 μg) was added to LE/LY membranes from
CHOnull or CHONPC1 cells in the presence of DMSO (10%) or the indicated concentrations
of 3.47, 3.0, or 3.18 (left panel), and 3.47 or U18666A (U18, right panel). Membrane bound
and unbound GP1 were analyzed by immunoblot.
c, Proposed model of EboV entry. Following EboV uptake and trafficking to late
endosomes24,25, EboV GP is cleaved by cathepsin protease to remove heavily glycosylated
domains (CHO) and expose the putative receptor binding domain (RBD) of GP16,17–19.
Binding of cleaved GP1 to NPC1 is necessary for infection and is blocked by the EboV
inhibitor 3.47.
Côté et al. Page 16
Nature. Author manuscript; available in PMC 2012 March 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... The scientific relevance to test NPC1L1 in SARS-CoV-2 entry was also because multiple other viruses such as Ebola, human immunodeficiency virus, Hepatitis C virus, Dengue, Chikungunya, and Zika virus use NPC1L1 either during cell entry and/or during late stages of virus trafficking [20][21][22][23][24][25][26][27][28]. In the case of the Hepatitis C virus, the NPC1L1 receptor serves as a potential tropism determinant since it is highly expressed in hepatocytes [23,24]. ...
... The scientific relevance to test NPC1L1 in SARS-CoV-2 entry was also because multiple other viruses such as Ebola, human immunodeficiency virus, Hepatitis C virus, Dengue, Chikungunya, and Zika virus use NPC1L1 either during cell entry and/or during late stages of virus trafficking [20][21][22][23][24][25][26][27][28]. In the case of the Hepatitis C virus, the NPC1L1 receptor serves as a potential tropism determinant since it is highly expressed in hepatocytes [23,24]. Interestingly Ebola virus binding to NPC1L1 triggers the priming of the viral glycoprotein into a fusion-competent state resulting in virus cell membrane fusion [22,23]. ...
... In the case of the Hepatitis C virus, the NPC1L1 receptor serves as a potential tropism determinant since it is highly expressed in hepatocytes [23,24]. Interestingly Ebola virus binding to NPC1L1 triggers the priming of the viral glycoprotein into a fusion-competent state resulting in virus cell membrane fusion [22,23]. In fact, the physiological relevance of NPC1L1receptor has been directly connected to an enhanced cholesterol uptake during Dengue virus infection, and the drug ezetimibe inhibits Dengue virus infection in the human hepatoma cell line (Huh-7) by blocking NPC1L1 [27,28]. ...
Article
Full-text available
The entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into human embryonic kidney (HEK293T) cells has been shown to be a cholesterol-rich, lipid raft-dependent process. In this study, we investigated if the presence of a cholesterol uptake receptor Niemann-pick type c1-like1 (NPC1L1) impacts SARS-CoV-2 cell entry. Initially, we utilized reporter-based pseudovirus cell entry assays and a spike (S) glycoprotein-mediated cell-to-cell fusion assay. Using Chinese hamster ovary (CHO-K1) cells, which lack endogenous receptors for SARS-CoV-2 entry, our data showed that the co-expression of NPC1L1 together with the ACE2 receptor synergistically increased SARS-CoV-2 pseudovirus entry even more than the cells expressing ACE-2 receptor alone. Similar results were also found with the HEK293T cells endogenously expressing the ACE2 receptor. Co-cultures of effector cells expressing S glycoprotein together with target cells co-expressing ACE-2 receptor with NPC1L1 significantly promoted quantitative cell-to-cell fusion, including syncytia formation. Finally, we substantiated that an elevated expression of NPC1L1 enhanced entry, whereas the depletion of NPC1L1 resulted in a diminished SARS-CoV-2 entry in HEK293T-ACE2 cells using authentic SARS-CoV-2 virus in contrast to their respective control cells. Collectively, these findings underscore the pivotal role of NPC1L1 in facilitating the cellular entry of SARS-CoV-2. Importance: Niemann-Pick type C1-like1 (NPC1L1) is an endosomal membrane protein that regulates intracellular cholesterol trafficking. This protein has been demonstrated to play a crucial role in the life cycle of several clinically important viruses. Although SARS-CoV-2 exploits cholesterol-rich lipid rafts as part of its viral entry process, the role of NPC1L1 in SARS-CoV-2 entry remains unclear. Our research represents the first-ever demonstration of NPC1L1’s involvement in facilitating SARS-CoV-2 entry. The observed role of NPC1L1 in human kidney cells is not only highly intriguing but also quite relevant. This relevance stems from the fact that NPC1L1 exhibits high expression levels in several organs, including the kidneys, and the fact that kidney damages are reported during severe cases of SARS-CoV-2. These findings may help us understand the new functions and mechanisms of NPC1L1 and could contribute to the identification of new antiviral targets.
... Lastly, small interfering RNAs (siRNAs) can target mRNAs as well as vRNAs or cRNAs, and phosphorodiamidate morpholino oligonucleotides (PMOs) can prevent the translation of viral mRNAs. It is noteworthy that while the majority of strategies targeting the transcription and translation of viral RNA are still in the experimental and developmental stages, the siRNA TKM-Ebola (highlighted in bold) and its nucleoside analogues favipiravir and remdesivir are either in clinical trials or have been used in humans in experiments [8]. ...
Article
Full-text available
Over the preceding 38 years, a few outbreaks have been brought on by the Ebola virus, which produces the Ebola illness. No specific treatment has been approved for EVD. Problem management and supportive care are the cornerstones of treatment. Effective outbreak control requires a multidisciplinary team effort that includes case care, infection prevention and control protocols, contact tracing and surveillance, a high-quality laboratory service, dignified and safe funerals, and social and community mobilization. The 2014 Ebola outbreak started in Africa and swiftly spread to other continents before turning into a pandemic. The illness gained international interest because to its relatively peculiar design, lethality and contagiousness, difficulty in containing its spread, and absence of a reliable treatment. Two medications have received FDA approval to treat EVD. Ebanga is a single monoclonal antibody, whereas Inmazeb is a mixture of three monoclonal antibodies. Individuals utilizing any of the two FDA-approved treatments had a significantly greater overall survival rate. In this article, the known history of the Ebola virus, its mode of infection, epidemiology, lifecycle, and possible treatments are briefly reviewed.
... Genome-wide screening methods have facilitated and accelerated the identification and characterization of host genes involved in infectious diseases. In particular, CRISPR/Cas9-based screens 15,16 and insertional mutagenesis in haploid cell systems 17,18 have enabled the discovery of receptors and intracellular host factors for various virus infections, including Ebola [19][20][21] , Lassa 22,23 and SARS-CoV-2 (refs. 16,21). ...
Article
Full-text available
Climate change and population densities accelerated transmission of highly pathogenic viruses to humans, including the Crimean–Congo haemorrhagic fever virus (CCHFV). Here we report that the Low Density Lipoprotein Receptor (LDLR) is a critical receptor for CCHFV cell entry, playing a vital role in CCHFV infection in cell culture and blood vessel organoids. The interaction between CCHFV and LDLR is highly specific, with other members of the LDLR protein family failing to bind to or neutralize the virus. Biosensor experiments demonstrate that LDLR specifically binds the surface glycoproteins of CCHFV. Importantly, mice lacking LDLR exhibit a delay in CCHFV-induced disease. Furthermore, we identified the presence of Apolipoprotein E (ApoE) on CCHFV particles. Our findings highlight the essential role of LDLR in CCHFV infection, irrespective of ApoE presence, when the virus is produced in tick cells. This discovery holds profound implications for the development of future therapies against CCHFV.
... In fact, many zoonotically transmitted viruses, in particular enveloped viruses including SARS-CoV-2 (Tang et al, 2020), hijack late endosomal proteins to release their viral genome into the host cell. This includes the main cholesterol transporter in LE/Lys, Niemann-Pick type C1 (NPC1), which serves as the entry factor for several filoviruses with Ebola virus using NPC1 in a cholesterolindependent manner (Carette et al, 2011;Cote et al, 2011). In addition, cholesterol accumulation in LE/Lys, using the pharmacological NPC1 inhibitor U18666A, compromised fusion of the influenza lipid envelope with late endosomal membranes (Musiol et al, 2013;Kuhnl et al, 2018;Schloer et al, 2019). ...
Article
Full-text available
The rapid development of vaccines to combat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections has been critical to reduce the severity of COVID-19. However, the continuous emergence of new SARS-CoV-2 subtypes highlights the need to develop additional approaches that oppose viral infections. Targeting host factors that support virus entry, replication, and propagation provide opportunities to lower SARS-CoV-2 infection rates and improve COVID-19 outcome. This includes cellular cholesterol, which is critical for viral spike proteins to capture the host machinery for SARS-CoV-2 cell entry. Once endocytosed, exit of SARS-CoV-2 from the late endosomal/lysosomal compartment occurs in a cholesterol-sensitive manner. In addition, effective release of new viral particles also requires cholesterol. Hence, cholesterol-lowering statins, proprotein convertase subtilisin/kexin type 9 antibodies, and ezetimibe have revealed potential to protect against COVID-19. In addition, pharmacological inhibition of cholesterol exiting late endosomes/lysosomes identified drug candidates, including antifungals, to block SARS-CoV-2 infection. This review describes the multiple roles of cholesterol at the cell surface and endolysosomes for SARS-CoV-2 entry and the potential of drugs targeting cholesterol homeostasis to reduce SARS-CoV-2 infectivity and COVID-19 disease severity.
... The trimeric EBOV glycoprotein (GP) is the sole viral surface protein and it mediates viral infection of host cells (12). Following viral uptake via endocytosis, GP is proteolytically processed in endosomes (13,14), where the mucin-like domain and the glycan cap-two poorly conserved regions (15)-are cleaved to expose its receptor-binding domain, allowing its binding to the intracellular receptor Niemann-Pick C1 (16,17) (Fig. 1A). Subsequently, GP undergoes structural rearrangement to prompt fusion of viral and cellular membranes and transfer of the viral genome into the cytosol (18). ...
... The trimeric EBOV glycoprotein (GP) is the sole viral surface protein and it mediates viral infection of host cells (12). Following viral uptake via endocytosis, GP is proteolytically processed in endosomes (13,14), where the mucin-like domain and the glycan cap-two poorly conserved regions (15)-are cleaved to expose its receptor-binding domain, allowing its binding to the intracellular receptor Niemann-Pick C1 (16,17) (Fig. 1A). Subsequently, GP undergoes structural rearrangement to prompt fusion of viral and cellular membranes and transfer of the viral genome into the cytosol (18). ...
Article
Full-text available
The Ebola virus causes hemorrhagic fever in humans and poses a significant threat to global public health. Although two viral vector vaccines have been approved to prevent Ebola virus disease, they are distributed in the limited ring vaccination setting and only indicated for prevention of infection from orthoebolavirus zairense (EBOV)—one of three orthoebolavirus species that have caused previous outbreaks. Ebola virus glycoprotein GP mediates viral infection and serves as the primary target of neutralizing antibodies. Here, we describe a universal Ebola virus vaccine approach using a structure-guided design of candidates with hyperglycosylation that aims to direct antibody responses away from variable regions and toward conserved epitopes of GP. We first determined the hyperglycosylation landscape on Ebola virus GP and used that to generate hyperglycosylated GP variants with two to four additional glycosylation sites to mask the highly variable glycan cap region. We then created vaccine candidates by displaying wild-type or hyperglycosylated GP variants on ferritin nanoparticles (Fer). Immunization with these antigens elicited potent neutralizing antisera against EBOV in mice. Importantly, we observed consistent cross-neutralizing activity against Bundibugyo virus and Sudan virus from hyperglycosylated GP-Fer with two or three additional glycans. In comparison, elicitation of cross-neutralizing antisera was rare in mice immunized with wild-type GP-Fer. These results demonstrate a potential strategy to develop universal Ebola virus vaccines that confer cross-protective immunity against existing and emerging filovirus species.
Article
Ebola virus glycoprotein (EBOV GP) is one of the most heavily O-glycosylated viral glycoproteins, yet we still lack a fundamental understanding of the structure of its large O-glycosylated mucin-like domain and to what degree the host O-glycosylation capacity influences EBOV replication. Using tandem mass spectrometry, we identified 47 O-glycosites on EBOV GP and found similar glycosylation signatures on virus-like particle- and cell lysate-derived GP. Furthermore, we performed quantitative differential O-glycoproteomics on proteins produced in wild-type HEK293 cells and cell lines ablated for the three key initiators of O-linked glycosylation, GalNAc-T1, -T2, and -T3. The data show that 12 out of the 47 O-glycosylated sites were regulated, predominantly by GalNAc-T1. Using the glycoengineered cell lines for authentic EBOV propagation, we demonstrate the importance of O-linked glycan initiation and elongation for the production of viral particles and the titers of progeny virus. The mapped O-glycan positions and structures allowed to generate molecular dynamics simulations probing the largely unknown spatial arrangements of the mucin-like domain. The data highlight targeting GALNT1 or C1GALT1C1 as a possible way to modulate O-glycan density on EBOV GP for novel vaccine designs and tailored intervention approaches. IMPORTANCE Ebola virus glycoprotein acquires its extensive glycan shield in the host cell, where it is decorated with N-linked glycans and mucin-type O-linked glycans. The latter is initiated by a family of polypeptide GalNAc-transferases that have different preferences for optimal peptide substrates resulting in a spectrum of both very selective and redundant substrates for each isoform. In this work, we map the exact locations of O-glycans on Ebola virus glycoprotein and identify subsets of sites preferentially initiated by one of the three key isoforms of GalNAc-Ts, demonstrating that each enzyme contributes to the glycan shield integrity. We further show that altering host O-glycosylation capacity has detrimental effects on Ebola virus replication, with both isoform-specific initiation and elongation playing a role. The combined structural and functional data highlight glycoengineered cell lines as useful tools for investigating molecular mechanisms imposed by specific glycans and for steering the immune responses in future vaccine designs.
Article
Continually repeating outbreaks of pathogenic viruses necessitate the construction of effective antiviral strategies. Therefore, the development of new specific antiviral drugs in a well-established and efficient manner is crucial. Taking into account the strong ability of viruses to change, therapies with diversified molecular targets must be sought. In addition to the widely explored viral enzyme inhibitor approach, inhibition of protein–protein interactions is a very valuable strategy. In this Perspective, protein–protein interaction inhibitors targeting HIV, SARS-CoV-2, HCV, Ebola, Dengue, and Chikungunya viruses are reviewed and discussed. Antibodies, peptides/peptidomimetics, and small molecules constitute three classes of compounds that have been explored, and each of them has some advantages and disadvantages for drug development.
Article
Full-text available
Most cell types acquire cholesterol by endocytosis of circulating low density lipoprotein, but little is known about the mechanisms of intra-endosomal cholesterol transport and about the primary cause of its aberrant accumulation in the cholesterol storage disorder Niemann-Pick type C (NPC). Here we report that lysobisphosphatidic acid (LBPA), an unconventional phospholipid that is only detected in late endosomes, regulates endosomal cholesterol levels under the control of Alix/AlP1, which is an LBPA-interacting protein involved in sorting into multivesicular endosomes. We find that Alix down-expression decreases both LBPA levels and the lumenal vesicle content of late endosomes. Cellular cholesterol levels are also decreased, presumably because the storage capacity of endosomes is affected and thus cholesterol clearance accelerated. Both lumenal membranes and cholesterol can be restored in Alix knockdown cells by exogenously added LBPA. Conversely, we also find that LBPA becomes limiting upon pathological cholesterol accumulation in NPC cells, because the addition of exogenous LBPA, but not of LBPA isoforms or analogues, partially reverts the NPC phenotype. We conclude that LBPA controls the cholesterol capacity of endosomes.
Article
Full-text available
Oxysterol-binding protein (OSBP) and its related proteins (ORPs) constitute a large and evolutionarily conserved family of lipid-binding proteins that target organelle membranes to mediate sterol signaling and/or transport. Here we characterize ORP5, a tail-anchored ORP protein that localizes to the endoplasmic reticulum. Knocking down ORP5 causes cholesterol accumulation in late endosomes and lysosomes, which is reminiscent of the cholesterol trafficking defect in Niemann Pick C (NPC) fibroblasts. Cholesterol appears to accumulate in the limiting membranes of endosomal compartments in ORP5-depleted cells, whereas depletion of NPC1 or both ORP5 and NPC1 results in luminal accumulation of cholesterol. Moreover, trans-Golgi resident proteins mislocalize to endosomal compartments upon ORP5 depletion, which depends on a functional NPC1. Our results establish the first link between NPC1 and a cytoplasmic sterol carrier, and suggest that ORP5 may cooperate with NPC1 to mediate the exit of cholesterol from endosomes/lysosomes.
Article
Full-text available
Ebolavirus (EBOV) is an enveloped, single-stranded, negative-sense RNA virus that causes severe hemorrhagic fever with mortality rates of up to 90% in humans and nonhuman primates. Previous studies suggest roles for clathrin- or caveolae-mediated endocytosis in EBOV entry; however, ebolavirus virions are long, filamentous particles that are larger than the plasma membrane invaginations that characterize clathrin- or caveolae-mediated endocytosis. The mechanism of EBOV entry remains, therefore, poorly understood. To better understand Ebolavirus entry, we carried out internalization studies with fluorescently labeled, biologically contained Ebolavirus and Ebolavirus-like particles (Ebola VLPs), both of which resemble authentic Ebolavirus in their morphology. We examined the mechanism of Ebolavirus internalization by real-time analysis of these fluorescently labeled Ebolavirus particles and found that their internalization was independent of clathrin- or caveolae-mediated endocytosis, but that they co-localized with sorting nexin (SNX) 5, a marker of macropinocytosis-specific endosomes (macropinosomes). Moreover, the internalization of Ebolavirus virions accelerated the uptake of a macropinocytosis-specific cargo, was associated with plasma membrane ruffling, and was dependent on cellular GTPases and kinases involved in macropinocytosis. A pseudotyped vesicular stomatitis virus possessing the Ebolavirus glycoprotein (GP) also co-localized with SNX5 and its internalization and infectivity were affected by macropinocytosis inhibitors. Taken together, our data suggest that Ebolavirus is internalized into cells by stimulating macropinocytosis in a GP-dependent manner. These findings provide new insights into the lifecycle of Ebolavirus and may aid in the development of therapeutics for Ebolavirus infection.
Article
Full-text available
Zaire ebolavirus (ZEBOV), a highly pathogenic zoonotic virus, poses serious public health, ecological and potential bioterrorism threats. Currently no specific therapy or vaccine is available. Virus entry is an attractive target for therapeutic intervention. However, current knowledge of the ZEBOV entry mechanism is limited. While it is known that ZEBOV enters cells through endocytosis, which of the cellular endocytic mechanisms used remains unclear. Previous studies have produced differing outcomes, indicating potential involvement of multiple routes but many of these studies were performed using noninfectious surrogate systems such as pseudotyped retroviral particles, which may not accurately recapitulate the entry characteristics of the morphologically distinct wild type virus. Here we used replication-competent infectious ZEBOV as well as morphologically similar virus-like particles in specific infection and entry assays to demonstrate that in HEK293T and Vero cells internalization of ZEBOV is independent of clathrin, caveolae, and dynamin. Instead the uptake mechanism has features of macropinocytosis. The binding of virus to cells appears to directly stimulate fluid phase uptake as well as localized actin polymerization. Inhibition of key regulators of macropinocytosis including Pak1 and CtBP/BARS as well as treatment with the drug EIPA, which affects macropinosome formation, resulted in significant reduction in ZEBOV entry and infection. It is also shown that following internalization, the virus enters the endolysosomal pathway and is trafficked through early and late endosomes, but the exact site of membrane fusion and nucleocapsid penetration in the cytoplasm remains unclear. This study identifies the route for ZEBOV entry and identifies the key cellular factors required for the uptake of this filamentous virus. The findings greatly expand our understanding of the ZEBOV entry mechanism that can be applied to development of new therapeutics as well as provide potential insight into the trafficking and entry mechanism of other filoviruses.
Article
Full-text available
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).
Article
Full-text available
Ebola virus (EBOV) cellular attachment and entry is initiated by the envelope glycoprotein (GP) on the virion surface. Entry of this virus is pH dependent and associated with the cleavage of GP by proteases, including cathepsin L (CatL) and/or CatB, in the endosome or cell membrane. Here, we characterize the product of CatL cleavage of Zaire EBOV GP (ZEBOV-GP) and evaluate its relevance to entry. A stabilized recombinant form of the EBOV GP trimer was generated using a trimerization domain linked to a cleavable histidine tag. This trimer was purified to homogeneity and cleaved with CatL. Characterization of the trimeric product by N-terminal sequencing and mass spectrometry revealed three cleavage fragments, with masses of 23, 19, and 4 kDa. Structure-assisted modeling of the cathepsin L-cleaved ZEBOV-GP revealed that cleavage removes a glycosylated glycan cap and mucin-like domain (MUC domain) and exposes the conserved core residues implicated in receptor binding. The CatL-cleaved ZEBOV-GP intermediate bound with high affinity to a neutralizing antibody, KZ52, and also elicited neutralizing antibodies, supporting the notion that the processed intermediate is required for viral entry. Together, these data suggest that CatL cleavage of EBOV GP exposes its receptor-binding domain, thereby facilitating access to a putative cellular receptor in steps that lead to membrane fusion.
Article
Full-text available
Ebolavirus (EBOV) entry into cells requires proteolytic disassembly of the viral glycoprotein, GP. This proteolytic processing, unusually extensive for an enveloped virus entry protein, is mediated by cysteine cathepsins, a family of endosomal/lysosomal proteases. Previous work has shown that cleavage of GP by cathepsin B (CatB) is specifically required to generate a critical entry intermediate. The functions of this intermediate are not well understood. We used a forward genetic strategy to investigate this CatB-dependent step. Specifically, we generated a replication-competent recombinant vesicular stomatitis virus bearing EBOV GP as its sole entry glycoprotein and used it to select viral mutants resistant to a CatB inhibitor. We obtained mutations at six amino acid positions in GP that independently confer complete resistance. All of the mutations reside at or near the GP1-GP2 intersubunit interface in the membrane-proximal base of the prefusion GP trimer. This region forms a part of the "clamp" that holds the fusion subunit GP2 in its metastable prefusion conformation. Biochemical studies suggest that most of the mutations confer CatB independence not by altering specific cleavage sites in GP but rather by inducing conformational rearrangements in the prefusion GP trimer that dramatically enhance its susceptibility to proteolysis. The remaining mutants did not show the preceding behavior, indicating the existence of multiple mechanisms for acquiring CatB independence during entry. Altogether, our findings suggest that CatB cleavage is required to facilitate the triggering of viral membrane fusion by destabilizing the prefusion conformation of EBOV GP.
Article
The constitutive degradation of membrane components takes place in the acidic compartments of a cell, the endosomes and lysosomes. Sites of lipid degradation are intralysosomal membranes that are formed in endosomes, where the lipid composition is adjusted for degradation. Cholesterol is sorted out of the inner membranes, their content in bis(monoacylglycero)phosphate increases, and, most likely, sphingomyelin is degraded to ceramide. Together with endosomal and lysosomal lipid-binding proteins, the Niemann-Pick disease, type C2-protein, the GM2-activator, and the saposins sap-A, -B, -C, and -D, a suitable membrane lipid composition is required for degradation of complex lipids by hydrolytic enzymes.
Article
We succeeded in the design and synthesis of multifunctional chemical probes of the HIF-1alpha inhibitor carboranylphenoxyacetanilide (1) that combine photoaffinity labeling and click reaction to identify the target protein. HSP60 was identified as a primary target protein of 1 using the chemical probes 2 and 3. Furthermore, HSP60 inhibitor 4 suppressed hypoxia-induced HIF activation, indicating that HSP60 affects HIF-1alpha accumulation directly or indirectly.