Structural Biology
Research Article
29 September 2023

Single-molecule-binding studies of antivirals targeting the hepatitis C virus core protein

ABSTRACT

The hepatitis C virus (HCV) core protein (HCVcp) is the most highly conserved protein encoded by the HCV genome, and its N-terminal domain (NTDHCVcp) plays a crucial role in nucleocapsid assembly. Together, these two features make it an attractive target for antiviral therapeutics. However, NTDHCVcp is intrinsically disordered, leading to a high degree of conformational heterogeneity, and given its essential role in nucleocapsid assembly, it also tends to oligomerize at high concentrations, both of which make it difficult to characterize heterotypic intermolecular interactions between monomeric NTDHCVcp and potential therapeutics using conventional structure-based approaches. Here, we use single-molecule FRET spectroscopy to overcome these challenges and study the structural and energetic aspects of binding interactions involving different viral genotypes of NTDHCVcp and antiviral therapeutics based on antibodies, aptamers, peptides, and small molecules. Our findings highlight distinct binding mechanisms associated with these molecular interactions. For example, binding of high-affinity antibodies does not perturb the end-to-end distance of NTDHCVcp and is weakly impeded by electrostatic repulsions between similarly charged residues. Conversely, the low-nanomolar equilibrium dissociation constants of antiviral DNA aptamers arise from strong attractive electrostatic interactions that greatly decrease the end-to-end distance of NTDHCVcp. Furthermore, low-affinity antiviral peptides promote oligomerization of NTDHCVcp. Finally, the small-molecule antiviral compound we studied does not appear to affect any of our experimental observables, suggesting that binding may not alter the conformational properties of NTDHCVcp.

IMPORTANCE

The hepatitis C virus is associated with nearly 300,000 deaths annually. At the core of the virus is an RNA-protein complex called the nucleocapsid, which consists of the viral genome and many copies of the core protein. Because the assembly of the nucleocapsid is a critical step in viral replication, a considerable amount of effort has been devoted to identifying antiviral therapeutics that can bind to the core protein and disrupt assembly. Although several candidates have been identified, little is known about how they interact with the core protein or how those interactions alter the structure and thus the function of this viral protein. Our work biochemically characterizes several of these binding interactions, highlighting both similarities and differences as well as strengths and weaknesses. These insights bolster the notion that this viral protein is a viable target for novel therapeutics and will help to guide future developments of these candidate antivirals.

INTRODUCTION

The hepatitis C virus (HCV) is a widespread pathogen infecting nearly 2% of the world’s population and is one of the primary causes of cirrhosis and hepatocellular carcinoma (1, 2). HCV forms nanoscopic lipid-enveloped infectious particles that contain a positive sense, single-stranded RNA genome comprised of about 9,600 nucleotides. The genome has a single open reading frame that encodes for a polyprotein of about 3,000 amino acids (Fig. 1A) that is later cleaved by both viral and host proteases to form three structural and seven non-structural (NS) proteins associated with HCV (3, 4). Several promising antiviral therapeutics have been developed to inhibit the function of NS proteins, including NS3, NS4A, NS5A, and NS5B (5). However, due to the potential emergence of resistance-associated mutations within these proteins, currently available antiviral therapeutics are routinely administered together as part of a combination therapy (5 9).
Fig 1
Fig 1 Structural organization of the HCV core protein. (A) The single open reading frame of the HCV genome translates into a polyprotein that is later cleaved into 10 HCV proteins. The first, and most conserved, is the HCV core protein (HCVcp), which has two domains. (B) AlphaFold (10) predicted structures are shown for the two variants of the N-terminal domain of HCVcp (NTDHCVcp) used in this study. The individual amino acid sequences (Fig. S1) were derived from either genotype 1a (cp1a) and genotype 2a (cp2a) HCV isolates, respectively. Site-specific cysteine mutations are present in both cp1a (S2C, C98S, and S130C) and cp2a (S2C) for thiol-based bioconjugation of fluorophores and denoted by stars located near the spherical representation of the cysteine residues. Positively charged residues (K and R) are colored blue, and negatively charged residues (D and E) are colored red. (C) Numerical scores of AlphaFold’s predicted local distance difference test (pLDDT) (11, 12), scaled from 0 to 100, provide a metric to assess the overall confidence of the structure predictions for cp1a (cyan) and cp2a (magenta). Values between 50 and 70 correspond to low confidence, whereas those <50 correspond to very low confidence. Previously, low pLDDT values have also been shown to correlate with structural disorder within proteins (13, 14).
The most evolutionary conserved protein encoded by the HCV genome is the core protein (HCVcp), which has an average per-residue conservation rate of >0.95 across all of the genetic variants of HCV (Fig. 1A). This structural protein is derived from the first 191 amino acids of the HCV polyprotein (15) and contains two domains (Fig. 1A). The more highly conserved N-terminal domain of HCVcp (NTDHCVcp) contains several clusters of positively charged residues (Fig. 1B; Fig. S1) that help it remain intrinsically disordered (Fig. 1C) as a monomer under physiological conditions (16 18). Attractive electrostatic interactions between these positively charged residues and the negatively charged phosphates in RNA facilitate condensation of the viral genome into a small (30–40 nm diameter) pseudospherical nucleocapsid particle, which resides at the core of the virus particle (19 21). Given the high degree of sequence conservation and its essential role in nucleocapsid assembly, many view NTDHCVcp as an attractive target for novel pangenotypic antivirals. As such, research efforts over the past decade have identified several different classes of prospective antiviral therapeutics that have the potential to bind to NTDHCVcp and disrupt its critical function (22 28).
Single-stranded nucleic acid aptamers are one such class of antiviral therapeutics that are capable of interacting with (and potentially disrupting the function of) their biomolecular targets (29, 30). They can arise naturally or synthetically via an artificial selection process called systematic evolution of ligands by exponential enrichment (SELEX) (31). Natural RNA-based aptamers targeting HIV RNA-binding proteins [rev (32) and tat (33)] have been used in clinical studies to inhibit HIV replication. A synthetic aptamer, pegaptanib, targeting vascular endothelial growth factors has already been approved by the US Food and Drug Administration (FDA) to treat macular degeneration (34 36). In a previous study, SELEX was used to identify seven HCVcp-binding DNA aptamers, and ELISA-based assays were used to confirm that the selected aptamers bound specifically to HCVcp (22). Additionally, naïve Huh-7.5 cells treated with sub-micromolar concentrations of aptamers for 72 hours were less effective at producing infectious viral particles in a focus-forming assay when compared to cells treated with a library of random nucleotide sequences of identical length (22). Combined, these findings suggest that antiviral aptamers can bind to HCVcp and impair the production of infectious viral particles.
Antigen-binding proteins are another class of antivirals with therapeutic potential. Recently, three antibodies were isolated from genotype 1a (H77) HCV-immunized chimpanzees and mice that specifically neutralize three regions of the HCV envelope glycoproteins (E1/E2) (37). The antibody raised against the most highly conserved region of E2 was able to cross neutralize other viral genotypes. These results show that targeting the most conserved regions of HCV proteins is beneficial for the development of pangenotypic antiviral therapeutics. Commercial monoclonal antibodies have also been raised against HCVcp and have been used for a wide range of immunochemical applications (38 40). However, the large size of these antibodies limits their therapeutic utility in sub-cellular compartments. Fortunately, single-chain variable fragment (scFv) antibodies are engineered antigen-binding proteins that link together the heavy and light variable domains of a traditional antibody. Given their much smaller size scFv antibodies can more easily cross biological membranes and bind to intracellular antigens. Previously, other labs have used phage display to generate novel scFv antibodies capable of binding to HCVcp (24, 41). In co-transfection assays, this scFv antibody was shown to deplete and/or sequester intracellular HCVcp, as well as slow down the HCVcp-induced cellular proliferation associated with hepatocellular carcinoma (24). Combined, these efforts indicate that proteins containing epitopes for HCVcp can potentially function as antiviral therapeutics.
Peptides are a third class of therapeutic agents with antiviral activity arising from their ability to mimic the natural ligands of protein-protein interactions. They are highly selective, effective, and biologically well-tolerated, all while being relatively easy to synthesize (42). Several studies have used antiviral peptides to disrupt the function of viral nucleocapsid proteins and thus inhibit viral assembly (25, 43 45). Such an approach has also been used for NTDHCVcp, where antiviral peptides have been generated based on sequences of amino acids within the protein that are thought to participate in homotypic interactions within the assembled nucleocapsid particle. One such example is a set of peptides derived from the NTDHCVcp of a genotype 1a (H77) HCV isolate (45). These short antiviral peptides were able to inhibit oligomerization of NTDHCVcp in vitro and decrease the proliferation of a genotype 2a (JFH-1) HCV isolate in naïve Huh-7.5 cells (45). This result demonstrates that short NTDHCVcp-derived peptides can have antiviral activity.
Pharmaceutically active small molecules are a fourth class of antiviral therapeutics. Broadly speaking, these compounds constitute 90% of the current drug market (46). A number of small molecules are currently being used in humans as antivirals to treat infections associated with HBV, EBOV, SARS-CoV-2, HIV, and MPV (47 51). Additionally, several small molecules called direct-acting antivirals are FDA-approved to treat HCV (52 55). However, all of these target NS proteins, and because of genotypic variations and resistance-associated mutations, they are often combined together to increase their effectiveness. While screening a small-molecule library, researchers identified several indoline alkaloid-based compounds that bind to NTDHCVcp and disrupt oligomerization (56). These compounds were further derivatized as a part of a structure-activity-relationship study, resulting in a promising low-cytotoxicity compound (C20) that was found to inhibit oligomerization of NTDHCVcp and decrease the viral RNA copy number in Huh-7.5 cells infected with a genotype 2a (J6/JFH-1) HCV isolate (43). This suggests that small molecules that bind to NTDHCVcp can impair critical aspects of viral replication.
Although therapeutics in each of these four classes have been shown to interact with NTDHCVcp in a manner that may result in antiviral activity, little is known about the actual binding mechanisms governing these interactions. Additionally, it is not clear if/how the formation of these binding interaction alters the structure, and thus function, of the intrinsically disordered NTDHCVcp. Therefore, we set out to study binding interactions involving different viral genotypes of NTDHCVcp and representative members of the four classes of antiviral therapeutics discussed above.
Unfortunately, the intrinsically disordered nature of NTDHCVcp coincides with a high degree of conformational heterogeneity, making it challenging to study biochemical interactions between NTDHCVcp and potential antivirals. Many of these challenges are associated with classical structure-based approaches because they generally require conformationally homogeneous ensembles of molecules at high concentrations where HCVcp is prone to oligomerize/aggregate (57, 58). To overcome these limitations, we used Förster Resonance Energy Transfer (FRET) spectroscopy to study the structural and energetic aspects of these binding interactions at the single-molecule level, allowing us to avoid complications arising from both ensemble-averaging and self-assembly. With FRET, the efficiency of energy transfer from an energetically excited donor fluorophore to a proximal acceptor fluorophore strongly depends on the distance between them (59, 60). For this reason, it is often used as a spectroscopic ruler to measure nanometer distances (2–10 nm) in biological macromolecules.
Technological advances over the last two decades have made it possible to monitor energy transfer between individual fluorophores. Over this time, single-molecule FRET (smFRET) has proven to be an excellent technique to study IDPs in part because it is possible to study the structural, energetic, and dynamic properties of individual subpopulations within heterogenous ensembles under dilute conditions at equilibrium (18, 61 64). Using this spectroscopic approach, we have been able to gain insight into the breadth of binding mechanisms governing the interactions between fluorescently labeled NTDHCVcp and these prospective antiviral therapeutics; in the process we have also uncovered a few pros and cons associated with any potential future applications in which they may be involved.

MATERIALS AND METHODS

Chemicals and reagents

All solutions and reagents were prepared using ultrapure (18.2 MΩ cm) water. Na2HPO4, NaH2PO4, K2HPO4, KH2PO4, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES), ethylenediaminetetraacetic acid (EDTA), tris(2-carboxyethyl) phosphine (TCEP), trifluoroacetic acid (TFA), dimethyl sulfoxide (DMSO), isopropyl β-D-1-thiogalactopyranoside (IPTG), TWEEN20, and guanidinium chloride (GdmCl) were purchased from Sigma-Aldrich (St. Louis, MO). NaCl, tris(hydroxymethyl)aminomethane (Tris), and NaOH were purchased from Thermo Fisher Scientific (Fair Lawn, NJ). 200 proof Ethyl alcohol (EtOH), acetonitrile (ACN), and dithiothreitol (DTT) were purchased from Decon Labs (King of Prussia, PA), Supelco (Bellefonte, PA), and bioWORLD (Dublin, OH), respectively.

HCV polyprotein sequence conservation analysis

To explore the degree of sequence conservation among the 10 HCV proteins, all 220,667 amino acid sequence entries related to Hepacivirus C (Taxon ID: 11103) were downloaded from the UniProt Knowledgebase. Then, a suite of command line tools (65) was used to align the protein sequences using a local installation of NCBI-BLAST. First, a local database was created using the 220,667 protein sequences. Next, the 3,017 amino acid polyprotein derived from a genotype 6a (ZS202) HCV isolate (Accession: AHH29575.1) was used as a query sequence to generate an alignment of nearly every other entry in our local database. Finally, the sequence conservation for each protein (Fig. 1A) was calculated by averaging the conservation rate for each amino acid associated with each of the 10 distinct proteins of HCV.

Expression, purification, and labeling of NTDHCVcp variants

The NTDHCVcp amino acid sequences used in this study (Fig. S1) are minimally modified, dual-cysteine variants associated with either a genotype 1a (PE8) HCV isolate [Accession: CAE46584.1 (66)] or a genotype 2a (J6/JFH-1) HCV isolate [Accession: BAB32872.1 (67)], where the two cystine residues are used for the bioconjugation of donor and acceptor fluorophores for FRET-based studies. The 136 amino acid sequence of the genotype 1a variant used in our study (cp1a) differed from the first 133 amino acids of the PE8 viral isolate at three positions (S2C, C98M, S130C) and contains three additional residues (GPG) at N-terminus resulting from recombinant expression. The 131 amino acid sequence of the genotype 2a construct used in our study (cp2a) also contained three additional residues at the N-terminus, with the only other difference from the first 128 amino acids of the J6/JFH-1 viral isolate being an S2C mutation, as there is naturally a cysteine at position 128. Importantly, the locations of the two cysteine residues in cp2a (i.e., C2 and C128) closely mimic the location of those in cp1a (i.e., C2 and C130), allowing us to make meaningful comparisons between the two in our FRET-based studies.
The expression, purification, and labeling of cp1a have all been described in detail elsewhere (18). Those procedures were used to guide the expression, purification, and labeling of cp2a, which are described in detail here. Competent BL21(DE3) cells (Thermo Scientific, Waltham, MA) were transfected with a pET47b(+) plasmid (Celtek Genes, Franklin, TN) whose codons were optimized for the expression of a recombinant protein consisting of an N-terminal hexahistidine affinity tag adjacent to a HRV 3C protease cleavage site followed by the NTDHCVcp amino acid sequence of interest. An Erlenmeyer flask containing 2 L of lysogeny broth and Kanamycin at a concentration of 100 µg mL−1 was inoculated with a 30-mL overnight culture of transfected cells. After incubating for 2 hours at 37°C, protein expression was induced by adding IPTG into the broth at a final concentration of 1 mM. Cells were grown for 3 hours to allow for the expression of the recombinant protein. Next, the broth was centrifuged at 4°C for 15 minutes at 4,000 rpm to pellet the cells. The pellet was resuspended in a lysis buffer (50 mM Tris-HCl, 10 mM imidazole, 6 M GdmCl, 10 mM NaH2PO4, 5 mM TCEP, pH 8.0). The resulting milieu was centrifuged at 19,000 rpm for 30 minutes to pellet insoluble cellular debris. The supernatant, which contained hexahistidine-tagged recombinant protein, was allowed to flow through the Probond nickel-chelating resin (Novex, Carlsbad, CA) of an immobilized-metal ion affinity chromatography (IMAC) column pre-equilibrated with lysis buffer. The nickel-bound recombinant protein was then eluted from the column with elution buffer (50 mM Tris-HCl, 250 mM imidazole, 6 M GdmCl, 260 mM NaH2PO4, 5 mM TCEP, pH 8.0). The eluate containing the hexahistidine-tagged recombinant protein was dialyzed against 2 L cleavage buffer (50 mM Tris-HCl, 150 mM NaCl, pH 7.5) at 4°C. After overnight dialysis, urea was added to a final concentration of 400 mM to solubilize the protein precipitate, and then, HRV 3C protease (Millipore Sigma, Burlington, MA) was added to a final concentration of 0.6 µM. This solution was again dialyzed against a fresh 2 L of cleavage buffer overnight at 4°C to allow ample time for the enzyme to cleave the N-terminal hexahistidine tag from the recombinant protein and the solutions to osmotically equilibrate. The cleavage product, cp2a, was purified from the hexahistidine tag using second round of IMAC. The mass of the purified cp2a (14,655.80 Da) was confirmed with the assistance of the mass spectrometry core facility at the University of Kansas (Fig. S2).
The strategy for labeling cp2a was based on a general approach for single-molecule FRET studies (68). First, cp2a was incubated in 10 mM DTT to ensure that all cystines were reduced. Progress of the reduction reaction was monitored via a high-performance liquid chromatography (HPLC) system (Infinity II 1260; Agilent Technologies, Inc., Santa Clara, CA) equipped with an analytical reversed-phase column (Reprosil Gold C18; Dr. Maisch, Ammerbuch-Entringen, Germany) using a 0.1% TFA water-acetonitrile gradient. The reduced form of cp2a was separated from DTT and purified using HPLC, and then lyophilized to remove the solvent. The lyophilized cp2a was then resuspended with a denaturing labeling buffer (6 M GdmCl, 0.1 M potassium phosphate, pH 7.3) to achieve a protein concentration of 50 µM to minimize disulfide bond formation. The thiol-reactive donor (Cy3B-maleimide) and acceptor (CF660R-maleimide) fluorophores were suspended in DMSO, sonicated for 15 minutes to break up aggregates, and added to the resuspended cp2a to achieve a final concentration of 50 µM, resulting in slight excess of the two fluorophores. The reaction mixture was vortexed and incubated overnight at 4°C in the dark. Afterward, unreacted fluorophores were removed by passing the labeling reaction through a desalting column (Zeba Spin; Thermo Scientific) pre-equilibrated with the denaturing labeling buffer. The FRET-labeled permutations of the labeled proteins (i.e., donor-acceptor and acceptor-donor) were partially purified using HPLC. However, due to small differences in elution times, complete separation from the other labeling permutations (e.g., donor-donor and acceptor-acceptor) was not possible. Nevertheless, downstream data analyses (see below) are used to filter out these unwanted species. Finally, the resulting fluorescently labeled proteins were lyophilized and stored at −80°C.

Prospective antivirals: aptamers, antibodies, peptides, and small molecules

The Cnew antiviral aptamer and a library of random 44-nucleotide long sequences (Fig. S1) were purchased from Integrated DNA Technologies (Coralville, IA). A monoclonal antibody (ab2740) with an epitope that reportedly recognizes amino acids 21–40 in NTDHCVcp was purchased from Abcam (Cambridge, United Kingdom). A set of four antiviral peptides (SL173, SL174, SL175, and SL571) with amino acid sequences (Fig. S1) derived from the NTDHCVcp of a genotype 1a (H77) HCV isolate were chemically synthesized by the Synthetic Chemical Biology core facility at the University of Kansas. The same core facility also chemically synthesized C20 (Fig. S1), a small-molecule antiviral compound that reportedly inhibits oligomerization of NTDHCVcp (43). The molecular mass of Cnew, SL173, SL174, SL175, SL571, and C20 was confirmed using mass spectrometry.

Sample preparation

All single-molecule measurements were conducted on samples consisting of 50 µL of aqueous solution within a single well of a 15-well 3D μ-slide (ibidi GmBH, Grafelfing, Germany). These solutions consisted of labeled NTDHCVcp at a concentration of roughly 100 pM (to ensure single-molecule detection) in measurement buffer (25 mM HEPES, 12.5 mM NaOH, 0.5 mM TWEEN20, and 2 nM unlabeled NTDHCVcp) containing varying concentrations of NaCl and antivirals depending on the particular measurement. The use of unlabeled NTDHCVcp and TWEEN20 in the measurement buffer helps minimize unwanted interactions between labeled NTDHCVcp and the wells of the µ-slide.

Single-molecule fluorescence microscopy

All single-molecule measurements were performed on a single-molecule instrument (Fig. 2A) consisting of a modified confocal fluorescence microscope (MicroTime200; PicoQuant, Berlin, Germany) equipped with two dual-mode diode lasers with output wavelengths of 515 nm (Quixx 515–80PS; Omicron, Rodgau, Germany) and 642 nm (Quixx 642–140PS; Omicron). The output from the two excitation sources was each adjusted to 90 µW while being digitally modulated by a function generator (577–4C; Berkeley Nucleonics Corporation, San Rafael, CA). Most experiments were conducted with the lasers in continuous-wave mode and the function generator set to a repetition rate of 20 kHz to achieve the so-called alternating laser excitation (ALEX) configuration (69). However, measurements of antibody-containing samples were made with the lasers in pulsed mode and the function generator set to a repetition rate of 20 MHz to achieve the so-called pulsed interleaved excitation (PIE) configuration (70), enabling accurate measurements of the sub-millisecond translational diffusion of NTDHCVcp. In either configuration, the excitation light passed through a multi-band dichroic mirror (ZT532/640rpc; Chroma, Bellows Falls, VT) prior to being focused into the sample via a 1.2 NA, 60×, water immersion objective (UPlanSAPO; Olympus, Tokyo, Japan).
Fig 2
Fig 2 Schematic diagram of the single-molecule confocal fluorescence microscope. (A) The two diode lasers (515 nm and 642 nm) are directed to the water immersion objective using a multi-band dichroic mirror. The fluorescence emitted from the sample is collected via the same objective. The excitation and emission are separated by the multi-band dichroic mirror. The emitted light is then focused through a 100-µm confocal pinhole to remove out-of-plane fluorescence. Next, the emitted light is separated based first on the polarization and then the wavelength. Finally, the spatially separated emission is focused on one-of-four avalanche photodiode (APD) detectors. (B) A cartoon representation of confocal volume and a fluorescently labeled cp1a molecule. The small confocal volume and dilute concentrations of labeled cp1a molecules (100 pM) ensure that the bursts of fluorescence arise from single molecules diffusing through the confocal volume.
Given extremely dilute concentrations of fluorescent species in the sample, the focus is expected to be devoid of fluorescent molecules >99% of the time. However, when a fluorescently labeled NTDHCVcp molecule in the aqueous sample diffuses through the diffraction-limited focus (Fig. 2B), the fluorophores attached to it will repeatedly absorb photons from the alternating excitation sources and emit a burst of fluorescence photons. Dozens of these photons are collected via the microscope objective before being directed back through the multi-band dichroic mirror toward both a 100-µm pinhole to remove out-of-plane photons and toward a polarizing beam splitter (BS cube polarizer; PicoQuant), which spatially separate the emitted photons based on polarization. Next, the two streams of photons each impinge on a long-pass dichroic (635LPXR; Chroma) to spatially separate them again, this time based on wavelength. The resulting four photon streams are then focused on to one-of-four single-photon avalanche photodiode (APD) detectors (SPCM-AQRH-14-TR; Excelitas Technologies, Waltham, MA). Finally, the arrival time of each detected photon is recorded and processed via a time-correlated single-photon counting (TCSPC) module (HydraHarp 400; PicoQuant) with a temporal resolution of either 16 ps (PIE) or 16,384 ps (ALEX) and saved to a .ptu file. All single-molecule measurements were performed at 23°C for either 10 minutes (ALEX) or 20 minutes (PIE).

Single-molecule analyses

Photon detection events saved to the .ptu data files were analyzed in Mathematica using Fretica (71), a user-extendable toolbox for analyzing single-molecule fluorescence data.

Anomalous diffusion analysis

First, photons were placed into consecutive one-second time bins based on their arrival times. Photons in one-second time bins that contained more photons than the mean photon count plus three times the standard deviation were considered to arise from large, slowly diffusing, fluorescent particles (e.g., oligomers or aggregates) and not small, quickly diffusing, fluorescently labeled NTDHCVcp molecules. Photons associated with these anomalous events were excluded from downstream single-molecule analyses.

Single-molecule transfer efficiency analysis

To identify bursts of fluorescence arising from single molecules, photons were placed into consecutive two-millisecond time bins based on their arrival times. For each bin, the number of photons associated with each of the four detectors was corrected to account for: (i) different excitation and detection efficiencies of the two fluorophores, (ii) detector cross talk, and (iii) direct excitation of the acceptor fluorophore by the 515 nm laser (72). Photons in two-millisecond time bins containing between 20 and 500 total photons were considered to arise from single molecules diffusing through the focused laser beams. Correspondingly, the time bins associated with these single-molecule observations were labeled as bursts, and all other time bins were labeled as background (Fig. 3A). The photon counts on all four detectors in each bin were background corrected, and then the labels of each bin (burst or background) were updated; this process was repeated until the number of burst and background bins in a measurement remained unchanged from one iteration to the next. Only photons associated with bursts were used in downstream data analyses.
Fig 3
Fig 3 Overview of single-molecule data analyses. (A) Representative fluorescence time trace recorded in the ALEX configuration for cp1a under standard experimental conditions. Time bins with dark green and red fill correspond to photons on the donor and acceptor detectors, respectively. Time bins with bright green and red strokes represent 515 nm and 642 nm excitation, respectively. A window of 20–500 total photons is used to differentiate between bursts (opaque) and background (transparent) time bins. (B) Representative transfer efficiency histogram for cp1a under standard experimental conditions. (C) Representative fluorescence time trace recorded in the PIE configuration for cp1a under standard experimental conditions. (D) Autocorrelation analysis of acceptor photons arising from 642 nm excitation. The resulting correlation function G( τ ) is fit (dashed gray line) to equation 4, which describes translational diffusion through a three-dimensional Gaussian volume. The average diffusion time, ⟨τD⟩, is extracted as a fitted parameter.
The arrival time of every photon within a burst is used to determine which of the two alternating excitation sources (515 nm or 642 nm) was active during or immediately prior to the detected photon (Fig. 3A). This allowed us to calculate the total number of photons in a burst arising from either direct excitation of the donor at 515 nm ( N515Tot ) or direct excitation of the acceptor at 642 nm ( N642Tot ), as well as the fluorophore stoichiometry ( S ) of each burst (equation 1).
S=N515TotN515Tot + N642Tot
(1)
In this way, bursts with S < 0.35 are considered to arise from molecules containing only active acceptor fluorophores, and bursts with S > 0.80 are considered to arise from molecules containing only active donor fluorophores. These donor-only and acceptor-only bursts can arise from, for example, inefficiencies in fluorophore incorporation. Although they make roughly half of the fluorescently labeled species in solution, they are not considered in any downstream analyses (Fig. S3). Additionally, the remaining FRET-active bursts (i.e., 0.35 < S < 0.80) are further refined by removing those bursts with (i) substantial asymmetries in their mean photon arrival times to avoid the undesirable effects associated with conformational exchange, fluorophore bleaching, and triplet blinking and (ii) fewer than 50 total photons (after corrections) to limit the effect of shot noise on subsequent calculations.
The transfer efficiency ( E ) values for the remaining so-called high-quality bursts were calculated using equation 2, where N515Acc and N515Don are the number of acceptor and donor photons resulting from 515 nm excitation:
E=N515AccN515Acc + N515Don
(2)
Transfer efficiency histograms were used to visually display the results from all bursts in a measurement (Fig. 3B). To quantify the results of each measurement, the average transfer efficiency, E , was calculated using equation 3, where # is the total number of high-quality bursts associated with that measurement.
E=1#i=1#Ei
(3)

Translational diffusion analysis

Measurements were performed in the PIE configuration, and then, fluorescence correlation spectroscopy (FCS) analyses were performed to monitor the diffusive behavior of our fluorescently labeled single molecules (Fig. 3C). To do this, we used acceptor photons resulting from direct excitation of the acceptor fluorophore by the 642 nm laser (i.e., N642Acc ) after first removing any photons associated with anomalous diffusion but prior to identifying burst and background bins. The inter-photon times associated with these photons were used to calculate the autocorrelation function (73) for lag times ranging from τ = 10-5.5 s to τ = 100 s (Fig. 3D). The autocorrelation functions were then fit to an analytical model for diffusion (equation 4) where the diffraction-limited focus of the laser beam is approximated by a three-dimensional Gaussian volume (74).
G(τ)=1+x1(1+ττD)1(1+p2ττD)12
(4)
Here, x is the average number of molecules in the focus, ⟨τD⟩ is the average translational diffusion time through the focus, and p is a structural parameter that characterizes the shape of the focus and is defined by the ratio of semi-minor axis to the semi-major axis of the three-dimensional Gaussian volume , which for our optical systems is well approximated by P = 1/7.

Binding affinity analysis

To quantify the binding affinity of NTDHCVcp for the antiviral aptamers and antibodies used in this study, we fit plots of our experimental observables vs antiviral concentration to a quadratic form of the single-site binding equation (equation 5).
y=(finalinitial)([Pt] + [Lt] + appKd) ± ([Pt]+[Lt] + appKd)2  4[Pt][Lt]2[Pt]+initial
(5)
Here, y is the experimental observables (either E for aptamers or ⟨τD⟩ for antibodies), final and initial are the asymptotic values associated with the experimental observable, Pt is the total concentration of NTDHCVcp, Lt is the total antiviral concentration, and appKd is the apparent dissociation constant.
To determine the concentration at which the antiviral peptides were able to induce half-maximal oligomerization of NTDHCVcp ( EC50 ), we fit plots of the relative number of high-quality bursts ( BN ) vs peptide concentration to a hill-type binding equation (equation 6):
BN=[Lt]n[Lt]n + EC50n
(6)
Here, BN is defined as the ratio of the number of high-quality bursts in the presence (# +) and absence (# -) of antiviral peptides (i.e., BN = # +/# -), Lt is the total concentration of antiviral peptide, and n is a cooperativity coefficient.

RESULTS

HCVcp is the most conserved of all 10 HCV proteins, and its N-terminal domain (NTDHCVcp) plays a crucial role in the condensation of genomic RNA to form nucleocapsid particles (19 21), making it an attractive target for antiviral treatments. As such, past research efforts have identified several classes of antiviral therapeutics that bind to NTDHCVcp and inhibit viral replication (22, 23, 43 45, 56, 75 77). However, little is known about the binding mechanisms governing these interactions. Here, we use single-molecule FRET spectroscopy to characterize antiviral binding interactions involving different variants of NTDHCVcp and representatives from several classes of prospective antiviral therapeutics. The two NTDHCVcp variants studied here are derived from genotype 1a and 2a HCV isolates. Their amino acid sequences are quite similar for the first 65 residues, but the remaining C-terminal residues are rather divergent (Fig. S1), which may alter antiviral binding.

NTDHCVcp in the absence of antivirals

First, it is of course critical to establish a baseline for NTDHCVcp in the absence of any antivirals. To do this, we prepared aqueous samples containing fluorescently labeled variants derived from either genotype 1a (cp1a) or 2a (cp2a) HCV isolates at a concentration of roughly 100 pM under our standard experimental conditions (measurement buffer with 100 mM NaCl). When single FRET-labeled NTDHCVcp molecules pass through the confocal volume, a burst of several dozen fluorescence photons is generated that lasts about 1 ms. The photons associated with each burst are then used to calculate the transfer efficiency, E , of each burst, which, after the duration of the entire measurement, are compiled together in a so-called transfer efficiency histogram and analyzed to determine the mean transfer efficiency, E (Fig. S4). Using this approach and averaging over several repeated measurements, we found E = 0.33 ± 0.02 and E = 0.38 ± 0.02 for cp1a and cp2a, respectively.
Next, to demonstrate our ability to resolve conformational changes in NTDHCVcp that occur after adjusting experimental conditions, we first adjusted the NaCl concentration of the samples. Increasing the NaCl concentration systematically shifts the transfer efficiency histograms to higher E values (Fig. S5), whereas the opposite occurs in solution with lower concentrations of NaCl. Given the high net positive charge of cp1a (+22) and cp2a (+23), it is not surprising that both proteins become more compact at high NaCl concentrations due to the additional charge screening provided by the counterions in solution. Indeed, similar observations have been made in studies of other positively charged, intrinsically disordered proteins (78 83) including HCVcp (18, 76). Furthermore, these findings demonstrate that single-molecule transfer efficiency measurements can be used to detect solute-induced conformational changes within NTDHCVcp.

Antiviral aptamers bind to and compact NTDHCVcp

Previously, SELEX was used to identify several DNA aptamers that bind to HCVcp and exhibit antiviral activity in cell-culture systems (22). Here, we studied the interactions between NTDHCVcp and one such aptamer, Cnew, which is 44-nucleotides long (Fig. S1). This was accomplished by performing single-molecule transfer efficiency measurements of fluorescently labeled cp1a under standard experimental conditions with increasing concentrations of unlabeled Cnew aptamer. Analysis of this Cnew titration revealed a systematic decrease of the unbound population at E = 0.324 ± 0.004 and a corresponding increase of a bound population at E = 0.551 ± 0.003, suggesting that the intrinsically disordered cp1a becomes significantly more compact after binding to Cnew at low-nanomolar concentrations (Fig. S6A).
Given that Cnew is highly negatively charged and NTDHCVcp is highly positively charged, we expected the interactions between these two binding partners to strongly depend on electrostatics. Therefore, we carried out analogous Cnew titrations at higher NaCl concentrations to increase the extent of charge screening between these oppositely charged polymers (Fig. 4; Fig. S6). All of these Cnew titrations showed the same qualitative behavior regardless of NaCl concentration—the E value increases at lower concentrations of Cnew before it asymptotically plateaus at higher Cnew concentrations (Fig. 4A).
Fig 4
Fig 4 Binding of antiviral aptamer to cp1a. Binding titration plots showing the E for cp1a at different (A) Cnew (B) and Rand oligo concentrations for solutions containing increasing concentrations of NaCl. Oligo titration curves were analyzed to obtain the apparent dissociation constant ( appKd ). (C) Values for appKd increase with the increase in NaCl concentrations for the Cnew and Rand oligos. Under all conditions, the Cnew antiviral aptamer binds with a tighter binding affinity than Rand.
Consistent with our observations in the absence of antivirals (Fig. S5), we see that increasing NaCl concentration in the absence of Cnew increases the E of free NTDHCVcp (Fig. 4A), indicating that the molecular dimensions of this intrinsically disordered protein are likely modulated by repulsive intramolecular electrostatic interactions. Conversely, we observed that at saturating concentrations of Cnew, the E of the bound cp1a-Cnew complex decreases with increasing NaCl concentration (Fig. 4A), suggesting that the molecular dimensions of cp1a within the nucleoprotein complex are highly dependent on attractive intermolecular electrostatic interactions.
To quantify the binding affinity of the NTDHCVcp-aptamer interaction, we calculated the apparent dissociation constant ( appKd ) for each of the complete Cnew titrations (Fig. 4.A) by fitting our E vs Cnew concentration data to a quadratic form of the single-site binding equation (equation 4). Under standard experimental conditions, the appKd for the cp1a-Cnew complex was much lower than the total NTDHCVcp concentration in the solution ( appKd ≪ 2 nM), and therefore, it could not be accurately quantified using our fitting routine. However, as the NaCl concentration increased, the appKd for the cp1a-Cnew complex also increased from appKd = 0.160.06+0.10 nM at 150 mM NaCl to appKd = 160100+260 nM at 400 mM NaCl. These findings suggest that attractive intermolecular electrostatic interactions play a major role in the binding of positively charged cp1a to the negatively charged antiviral aptamer, Cnew. From an electrostatic perspective, one can imagine that the decrease in binding affinity at elevated NaCl concentration could arise from counterions in solution screening and weakening of the attractive electrostatic forces between oppositely charged functional groups in cp1a and Cnew. A quantitative linkage analysis of the NaCl-dependent binding affinity data (84) suggests that the association of these two biomolecules gives rise to the release of approximately eight counterions, as indicated by the slope, Γ, of a double logarithmic plot of appKd vs NaCl concentration (Fig. 4C, solid line). From a thermodynamic perspective, this indicates that releasing counterions to the bulk solution becomes increasingly less favorable as the bulk counterion concentration increases, yielding a positive increase in the binding free energy change.
Next, we sought to assess the specificity of the binding interaction between cp1a and Cnew. Therefore, we performed an entire collection of single-molecule measurements wherein the Cnew aptamer was replaced with Rand, a chemically synthesized library of 44-nucleotide long oligos with random sequences. The results of the measurements with Rand (Fig. 4B) are quite similar to what was observed with Cnew (Fig. 4A), except that the transition midpoints and the corresponding appKd values were systematically shifted to higher concentrations. This observation was also present at higher NaCl concentrations with more electrostatic screening. Furthermore, quantification of the NaCl-dependent binding affinity data suggests that the cp1a-Rand binding interactions also release approximately eight ions (Fig. 4C, dashed line). Taken together, the above findings indicate that cp1a can promiscuously bind to a range of nucleic acid sequences via electrostatic interactions but that the SELEX-derived sequence associated with the Cnew aptamer binds more than an order of magnitude more tightly under conditions with near-physiological concentrations of monovalent ions.
Finally, to assess the applicability of these findings to other viral genotypes, an analogous suite of experiments was conducted using a NTDHCVcp variant (cp2a) derived from a genotype 2a HCV isolate. Under standard experimental conditions, we were unable to generate a complete series of single-molecule transfer efficiency histograms for cp2a because the addition of low-nanomolar concentrations of Cnew resulted in the formation of large, slowly diffusing, fluorescent particles and a corresponding reduction in the relative number of high-quality bursts, BN (Fig. 5). Nevertheless, at higher Cnew concentrations, values of BN returned to typical values near unity, and we were again able to generate transfer efficiency histograms, this time with an E = 0.616 ± 0.014, suggesting that, like cp1a, cp2a also becomes compact after binding to Cnew.
Fig 5
Fig 5 Relative number of high-quality bursts associated with NTD HCVcp. Heat map of BN values for different NaCl and oligo concentrations for the binding interactions involving cp1a and either (A) Cnew or (B) Rand as well as binding interactions involving cp2a and either (C) Cnew or (D) Rand. Measurements with BN < 0.4 (hashed tiles) were considered to be of insufficient quality for downstream single-molecule analyses. In all cases, decreases in BN occurred concurrently with an increase in the formation of large, slowly diffusing, fluorescent particles. White tiles correspond to conditions with no data (N.D.).
Fortunately, complications arising from the formation of large, slowly diffusing, fluorescent particles were less severe at higher NaCl concentrations, resulting in larger values of BN (Fig. 5). A collective analysis of all measurements where BN > 0.4 generated well-populated transfer efficiency histograms for cp2a in both the Cnew- and Rand-binding titrations with NaCl concentrations ranging from 225 mM to 400 mM (Fig. S7). For all binding titrations carried out over this range, the E values for cp2a first increase and then plateau asymptotically at high oligo concentrations (Fig. S7). A quantitative analysis of the NaCl-dependent binding affinity data reveals that as the NaCl concentration increases, the appKd values for both Cnew and Rand systematically increase in a manner that is consistent with the release of approximately 11 counterions and 6 counterions, respectively (Fig. S7). All told, this suite of experimental evidence suggests the NTDHCVcp-aptamer binding interactions involving cp2a are incredibly similar to those involving cp1a. Nevertheless, we observed one stark difference between the two genotypic variants of NTDHCVcp, which is that cp2a and Cnew can form higher-order structures resulting in the appearance of large, slowly diffusing, fluorescent particles and an overall decrease in BN (Fig. 5).

Antibody binding does not change the conformation of NTDHCVcp

Next, we shift our attention to a second class of prospective antiviral therapeutics that can bind to HCVcp and impair viral replication, specifically, antigen-binding proteins (e.g., scFv) (24). To do so, we again carried out single-molecule fluorescence studies of the binding interactions between NTDHCVcp and a simple, commercially available, mouse monoclonal IgG1 antibody (ab2740) raised against the core protein of a genotype 1b HCV isolate.
Under standard experimental conditions, the addition of ab2740 resulted in little-to-no change in the E of fluorescently labeled cp1a across the entire binding titration (Fig. 6A), suggesting that either ab2740 doesn’t bind to cp1a under our experimental condition or that binding does not appreciably change the conformation of cp1a. Given that the IgG1 antibody (150 kDa) is much larger than NTDHCVcp (15 kDa), the diffusive properties of fluorescently labeled cp1a should change significantly when bound to ab2740. Therefore, we use fluorescence correlation spectroscopy (FCS) analyses to calculate the average diffusion time, τD , of cp1a through our sub-femtoliter diffraction-limited confocal volume. In the absence of ab2740 τD = 449 ± 6 µs. Upon increasing the concentration of ab2740 in solution, the values of τD first increase and then plateau asymptotically at τD = 856 ± 6 µs (Fig. 6B). The binding affinity of cp1a for ab2740 was then quantified by fitting the antibody-dependent τD of cp1a (Fig. 6C) to our single-site binding equation (equation 4), which resulted in an appKd = 3.70.6+0.7 nM. This result indicates that cp1a does indeed bind tightly to ab2740 and that binding must not substantially alter the conformational ensemble of the intrinsically disordered NTDHCVcp.
Fig 6
Fig 6 Binding of antibodies to NTD HCVcp. (A) Antibody-binding titrations do not alter the E of cp1a. Although E increases with increasing NaCl concentration (due to compaction of free cp1a), it remains the same at different concentrations of ab2740. (B) The average diffusion time, τD , of cp1a increases upon adding antibody to solution, consistent with the formation of a cp1a-ab2740 complex. (C) Apparent dissociation constants ( appKd ) for the cp1a-antibody (cyan) and cp2a-antibody (purple) binding interaction at different NaCl concentrations. The values of appKd for the cp1a-antibody interaction depend weakly on NaCl concentration corresponding to the release of roughly one counterion when the complex is formed. (D) Antibody-binding titrations do not seem to alter the E of cp2a. (E) The τD of cp2a increases upon adding antibody to solution, consistent with the formation of a cp2a-ab2740 complex.
Motivated by the pronounced NaCl dependence observed with the antiviral aptamers, we again conducted additional experiments under conditions with elevated NaCl concentrations. Like our antibody-binding titration under standard experimental conditions, we found that even in the presence of elevated NaCl concentrations, the E of cp1a was effectively unaltered across the entire range of ab2740 concentrations we surveyed (Fig. 6A). However, we were again able to detect binding between cp1a and ab2740 at higher NaCl concentrations using our FCS analyses to calculate the τD of cp1a (Fig. 6B). Moreover, we observed the binding affinity between cp1a and ab2740 slightly, but systematically, increased upon increasing the NaCl concentration (Fig. 6C), resulting in an appKd = 0.090.05+0.09 nM at 800 mM. These findings indicate that this intermolecular interaction is hindered by repulsive intermolecular electrostatic interactions between the epitope of ab2740 and the highly conserved stretch of amino acids in NTDHCVcp it is reported to recognize. Similarly, a quantitative linkage analysis of the NaCl-dependent binding affinity data (84) shows that approximately one additional counterion is taken up from the bulk solution during binding (Fig. 6E, cyan), which presumably helps to screen this slightly repulsive binding interaction.
Given that the epitope of ab2740 consists of the highly conserved amino acids at positions 21–40 of NTDHCVcp (Fig. S1), we expected that this antibody would also efficiently bind cp2a. Indeed, a qualitative analysis of analogous sets of experiments conducted using cp2a supported this expectation. First, the E of cp2a was largely insensitive to the addition of ab2740 (Fig. 6D), whereas the τD was dependent on the concentration of ab2740 in a manner that was consistent with a single-digit nanomolar-binding affinity (Fig. 6E). Curiously, the appKd was substantially less dependent on NaCl concentration (Fig. 6C), suggesting that neither attractive nor repulsive interactions dominate, and the formation of this antibody-antigen complex does not change the number of counterions associated with the two binding partners.

Antiviral peptides promote aggregation of NTDHCVcp

Past research efforts have shown that short peptides derived from regions of NTDHCVcp believed to be involved in homotypic interactions can inhibit oligomerization of NTDHCVcp in vitro and impair viral assembly in cell culture (25, 44, 45, 85). We selected a set of four such antiviral peptides whose amino acid sequences were derived from the NTDHCVcp of a genotype 1a (H77) HCV isolate (Fig. S1) and studied their ability to interact with NTDHCVcp using single-molecule fluorescence spectroscopy.
We chose to study the peptide SL173 first due to its previously reported high binding affinity for NTDHCVcp and its antiviral potency in cell culture (45). Single-molecule measurements of cp1a were conducted under standard experimental conditions in the presence of increasing concentrations of SL173. During these antiviral peptide-binding titrations, we noted that our ability to generate transfer efficiency histograms dramatically decreased at SL173 concentrations above 1 µM due to a reduction in the relative number of high-quality bursts ( BN ). Nevertheless, for those histograms that could be generated, the corresponding E values were experimentally indistinguishable from measurements conducted in the complete absence of SL173 (Fig. 7A), suggesting that this antiviral peptide doesn’t stably interact with single molecules of NTDHCVcp. Instead, the reduction of BN , which coincided with the emergence of large, slowly diffusing, fluorescent particles, suggests that interactions between cp1a and SL173 lead to rapid formation of higher-order multimeric assembles or aggregates. To quantify the concentration of SL173 needed to produce a half-maximal effect ( EC50 ), we fit our BN data (Fig. 7B) to a cooperative ligand-binding model, which resulted in an EC50 = 3.2 ± 0.3 µM (Fig. 7B).
Although cp1a contains many positive charges, the SL173 antiviral peptide, whose sequence is derived from the amino acid residues between position 85 and 110 in the NTDHCVcp of an H77 HCV isolate, is surprisingly uncharged. As such, we expected that the mechanism of oligomerization would be largely insensitive to NaCl concentration. Indeed, the results from analogous peptide-binding titrations conducted at elevated NaCl concentrations support this notion and show only a modest increase in across the entire range of experimental conditions (Fig. 7C).
Again, the rationale for the use of these peptides stems from their perceived potential to competitively form homotypic interactions with NTDHCVcp. Because SL173 was derived from an amino acid sequence in a more variable region of NTDHCVcp, we sought to determine if SL173 would also cause cp2a to oligomerize or aggregate. Given the minimal sequence similarity between cp1a and cp2a over the region of amino acids that correspond to SL173 (Fig. S1), we were surprised to see that SL173 was nearly as effective at inducing oligomerization of cp2a as it was with cp1a and that this effect was also quite insensitive to NaCl concentration (Fig. 7C). Together, these findings suggest that the formation of higher-order oligomeric structures containing NTDHCVcp might be driven by the physicochemical properties of SL173 (e.g., hydrophobicity) rather than its ability to form sequence-specific homotypic interactions with NTDHCVcp.
Finally, we set out to determine if three other antiviral peptides had similar interactions with our two NTDHCVcp variants. We found that the of cp1a and cp2a remained largely unaltered across the entire binding titration for the SL174 antiviral peptide and values of only decreased at the highest concentrations, resulting in an > 40 µM for both high (800 mM) and low (100 mM) NaCl concentrations (Fig. 7C). Like SL173, titrations of the antiviral peptide SL175, did give rise to a decrease in corresponding to the formation of oligomeric species for both cp1a ( = 6.6 ± 0.4 µM) and cp2a ( = 17 ± 1 µM), but only at 100 mM NaCl. However, unlike SL173, which has no net charge, SL175 is charged (+2), which likely makes this binding interaction more susceptible to electrostatic contributions that can be screened at high NaCl concentrations, thus explaining why no detectable binding of SL175 was observed at 800 mM NaCl. The amino acid sequence of the antiviral peptide SL571 is the reverse of SL175 (Fig. S1), and yet, both are characterized by low-micromolar values at 100 mM NaCl and little-to-no binding at 800 mM NaCl. This finding further supports the notion that the molecular interactions between NTDHCVcp and these antiviral peptides are not necessarily governed by primary sequence and instead depend simply on the general physicochemical properties of the two binding partners.
Fig 7
Fig 7 Binding of antiviral peptides to NTD HCVcp. (A) Values of E for cp1a at varying SL173 concentrations. Sphere color corresponds to NaCl concentrations. Sphere size corresponds to the relative number of high-quality bursts ( BN ). Higher concentrations of SL173 decrease BN impairing our ability to determine E (B) For all NaCl concentrations, the value of BN decreases with increasing SL173 concentrations due to the formation of oligomeric particles containing fluorescently labeled NTDHCVcp and antiviral peptides. These data were fit to a hill-type cooperative ligand-binding equation to quantify the concentration of SL173 needed to produce a half-maximal effect ( EC50 ). (C) Values of EC50 for different antiviral peptides and their interaction with either cp1a (left) or cp2a (right). Note: Discontinuous bars represent EC50 that were > 40 µM and thus could not be accurately quantified due to the limited solubility of the antiviral peptides at higher concentrations.

Indoline alkaloids do not alter the conformation of NTDHCVcp

Finally, we wanted to characterize the interactions between NTDHCVcp and a small-molecule compound to which it had been reported to bind (43). Previously, during an initial small-molecule screen of a indoline alkaloid library, four candidate compounds were shown to greatly disrupt oligomerization of NTDHCVcp (56). Further efforts to improve these compounds ultimately resulted in a more potent and less cytotoxic compound referred to as C20 (Fig. S1), which was derived from the same indoline alkaloid scaffold associated with the initial four candidates. Whole-cell studies of C20 and its derivatives demonstrated that these compounds were able to reduce viral replication in Huh-7.5 cells infected with a genotype 2a (J6/JFH) HCV isolate (43, 56). However, the biochemical details of the molecular interaction between NTDHCVcp and these indoline alkaloid compounds have yet to be resolved. Therefore, we used single-molecule FRET spectroscopy to monitor the conformational dimensions of our two fluorescently labeled variants of NTDHCVcp in the presence of increasing concentrations of C20.
Initially, we measured samples containing cp1a in the presence of increasing concentrations of C20 under standard experimental conditions (Fig. S8). We observed that E remained unchanged (Fig. S7A) over the entire range of concentrations (0–25 µM) where C20 was soluble in solution. The same was true for analogous measurements under non-standard experimental conditions with elevated concentrations of NaCl (800 mM). These results suggest that the addition C20 does not alter the average end-to-end distance of cp1a under our experimental conditions. Next, to determine if the presence of C20 alters the diffusive behavior of cp1a, we calculated the τD of cp1a under these conditions, and it also remains unchanged at both 100 and 800 mM NaCl, regardless of the C20 concentration (Fig. S8). Then, to determine if C20 causes oligomerization of cp1a in a manner that was similar to what was observed for the antiviral peptides, we looked at BN and found that it too remains unchanged under all experimental conditions (Fig. S8). Finally, to determine if these negative findings were genotype-specific, we conducted an analogous set of measurements using fluorescently labeled cp2a. The results associated with cp2a were nearly identical to those associated with cp1a—the presence of C20 does not alter the end-to-end distance, diffusivity, or oligomerization tendency of cp2a.

DISCUSSION

Novel antiviral therapeutics for HCV may still be beneficial

Global eradication of hepatitis C virus by 2030 is a key target set by the World Health Organization (86). To achieve this goal, cost-effective pangenotypic antiviral therapeutics for HCV remain crucial. Although current medications are highly effective (87, 88), resistance-associated mutations remain a concern (89, 90), and therefore, combination therapies are often required, further increasing the financial burden associated with HCV treatment plans (91). One approach for the development of novel pangenotypic antivirals is to inhibit critical processes within the viral replication cycle that are highly conserved across all genotypes. Viral assembly is one such process and is mediated by HCVcp (19, 21, 92), which is the most conserved protein across the different HCV genotypes (15). Although HCVcp has the potential to be a promising new target for therapeutic intervention, more work is needed to assess the merits of this approach. Here, we used single-molecule spectroscopy to study fluorescently labeled variants of the N-terminal domain of HCVcp (NTDHCVcp) from either genotype 1a or 2a HCV isolates and determine the biophysical principles that govern their ability to bind to and interact with four different classes of prospective antiviral therapeutics, namely, aptamers, antibodies, peptides, and small molecules.

Free monomeric NTDHCVcp is intrinsically disordered

All of our antiviral-binding titrations contained measurements of fluorescently labeled NTDHCVcp conducted in the complete absence of antivirals. In these reference measurements, we observed that both NTDHCVcp variants (cp1a and cp2a) became increasingly compact at higher NaCl concentrations, which is a well-known phenomenon in polyelectrolytic IDPs (18, 76, 78 83) and implies that the nearly polycationic NTDHCVcp adopts a heterogenous ensemble of conformations that is dictated by repulsive intramolecular electrostatic interactions capable of being screened at high ionic strength. However, given the nature of our single-molecule experiment, one cannot completely rule out the possibility of slight structural (or functional) alterations arising from the fluorogenic probes. Nevertheless, our findings are consistent with previous studies that show the monomeric form of NTDHCVcp is intrinsically disordered under a wide range of experimental conditions (16 18, 93); some of these studies used alternative fluorophores conjugated to different positions within NTDHCVcp. Furthermore, AlphaFold (10) predictions yield structures of NTDHCVcp with low to very low confidence, which others have noted can be an effective predictor of protein disorder (13, 14). Finally, previous in vivo studies of tetra-cysteine-modified core proteins labeled with biarsenical fluorophores showed only minor changes to virus titers (94), suggesting that the protein remains functional and seems to tolerate minor molecular perturbations quite well.

Nucleic acid aptamers as antiviral therapeutics

A critical component of viral assembly involves formation of the nucleocapsid, which resides at the core of the virus (19, 92). In HCV, the nucleocapsid is formed when many positively charged core proteins condense around the negatively charged viral genome (19, 21, 92). As such, it is not at all surprising that NTDHCVcp has a high affinity for both RNA (18) and DNA (95), including the previously identified antiviral DNA aptamer, Cnew (22). In fact, previous studies have reported that other antiviral aptamers bind to HCV core proteins from different genotypes with nanomolar affinities under physiologically relevant monovalent ion concentrations (22, 23, 76, 77). Some of these studies showed that antiviral aptamers also decrease virus titers in naïve Huh-7.5 cells (22, 23). In one of these studies, four DNA and four RNA aptamers were generated using SELEX with core proteins from HCV isolates with different genotypes as the target (23). Both the RNA aptamers and the DNA aptamers had nanomolar affinities for core proteins from six different genotypes (genotypes 1 through genotype 6) (23). Our results with the DNA aptamer Cnew yield binding affinities that are comparable to those in the studies discussed above. However, our studies shed new light on the molecular mechanisms governing the interaction between antiviral aptamers and HCVcp. First, we show that NTDHCVcp undergoes a conformational change upon binding to Cnew (Fig. 4A). The compaction of NTDHCVcp that occurs after binding to nucleic acids has been observed previously in other biochemical studies of nucleocapsid protein-nucleic acid interactions (95), including interactions between NTDHCVcp and short RNAs excised from the HCV genome (18). Furthermore, we show that the binding affinity of NTDHCVcp for Cnew depends strongly on ionic strength, indicating that binding is dominated by attractive electrostatic interactions and results in the formation of what is likely to be an intrinsically disordered polyelectrolyte complex (96). Finally, we demonstrated that despite involving strong electrostatic interactions, which are often thought of as promiscuous, NTDHCVcp does have some structural specificity for Cnew (Fig. 4C), either at the level of nucleic acid primary or secondary structure. Again, such behavior was also observed previously in studies of NTDHCVcp and its preferential ability to form nucleocapsid-like particles in the presence of short RNA sequences excised from the HCV genome (18). Together, these findings paint a picture where antiviral aptamers function as particularly effective competitive inhibitors that sequester NTDHCVcp in RNA-protein complexes, thus preventing NTDHCVcp from properly condensing on the viral genome to form the nucleocapsid (76).

Antigen-binding proteins as antiviral therapeutics

Our results show that the monoclonal antibody for NTDHCVcp, ab2740, binds to both of our NTDHCVcp variants with low-nanomolar affinity. In both cases, antibody binding has no detectable effect on the conformation of NTDHCVcp (Fig. 6A and D), suggesting that amino acids far outside of the reported epitope likely remain conformationally heterogeneous. The lack of any pronounced differences between cp1a and cp2a is consistent with NTDHCVcp being extremely conserved across all of the viral genotypes. Indeed, the amino acid residues associated with the epitope (21–40) are identical between the two variants. Nevertheless, we did observe that the cp1a-ab2740 interaction was slightly dependent on NaCl concentration, whereas the cp2a-ab2740 interaction was not (Fig. 6C). Curiously, cp1a has a neutral amino acid (Q) at position 20, whereas cp2a has an acidic residue (E) at this position (Fig. S1). Ionization of this residue may help to counterbalance attractive and repulsive interactions between the two binding partners. With a neutral residue at this position, the electrostatic interactions between the hypervariable loops of ab2740 and the epitope of cp1a would be repulsive and thus could be effectively screened at elevated NaCl concentrations giving rise to slightly higher binding affinities. Although ab2740 is well suited to bind to NTDHCVcp, it is not known if the bound NTDHCVcp is functional. Even if antigen recognition were to disrupt nucleocapsid formation, ab2740 is simply too large to readily enter cells. However, the variable domains of the heavy and the light chains can be connected via a polypeptide linker to form a single-chain variable fragment (scFv) antibody, which is a much smaller biomolecule that can more readily enter cells (97, 98). Indeed, a scFv antibody that binds to a less conserved epitope on NTDHCVcp (residues 82–88) has previously been shown to inhibit viral replication (24). Therefore, engineering a scFv based on ab2740 may be an attractive starting point for novel antibody-based antiviral therapeutics for HCV.

Peptides as antiviral therapeutics

Peptides are an attractive choice for therapeutic applications because of their ease of synthesis and biocompatibility. As such, previous research efforts have identified a handful of peptides derived from NTDHCVcp that can compete in the formation of homotypic interactions. Micromolar concentrations of these peptides have been shown to both inhibit oligomerization of NTDHCVcp (as determined by fluorescence polarization and surface plasmon resonance) and impair viral replication in cell culture (as determined by immunohistochemistry and RT-PCR) (45). In our single-molecule-binding studies of antiviral peptides, none of the four appeared to stably bind to and alter the end-to-end distance (Fig. 7A) of fluorescently labeled NTDHCVcp. However, at mid-micromolar concentrations, three of these peptides (i.e., SL173, SL175, and SL571) formed large, slowly diffusing, oligomeric complexes with NTDHCVcp (Fig. 7B). The concentrations of SL175 and SL571 required for half-maximal oligomerization ( EC50 ) were dependent on NaCl concentration (Fig. 7C), indicating that electrostatic forces mediate the formation of these binding interactions. Curiously, the oligomerization-inducing behavior of these two antiviral peptides was quite similar, which seems to indicate that their ability to oligomerize with NTDHCVcp is more strongly dependent on their physicochemical properties rather than their primary amino acid sequence. It is certainly conceivable that the ability of these peptides to oligomerize with NTDHCVcp in a manner that is independent of viral RNA is related to their antiviral activity. Similar to the antiviral DNA aptamers, the peptide-induced oligomerization of NTDHCVcp could sequester core proteins away from the viral RNA ultimately preventing assembly of viral nucleocapsids—although this has yet to be properly verified.

Small molecules as antiviral therapeutics

Small molecules are the most popular therapeutics to date and constitute 90% of the overall market (46). They can be administered orally and penetrate cells to inhibit biomolecular functions. Previous work has shown that several indoline alkaloid-type compounds are capable of inhibiting the oligomerization of NTDHCVcp and impairing the viral replication in cell culture (43, 56). We studied one of these molecules (C20) to determine how it binds to and interacts with NTDHCVcp. Our investigations showed that C20 does not change any of the experimental observables we are able to monitor with our single-molecule fluorescence approach (Fig. S8). Previously, a study using an ALPHA screen assay reported that C20 inhibits the oligomerization of NTDHCVcp with an EC50 = 0.092 µM (43). If this small-molecule antiviral does indeed bind at micromolar concentrations, our results would suggest that binding does not substantially alter the conformational properties of NTDHCVcp.

Conclusions

Interactions between molecules are essential for life, and for this reason, they have been exploited for both diagnostic and therapeutic purposes. Here, we studied binding interactions between fluorescently labeled variants of the N-terminal domain of the hepatitis C virus core protein (NTDHCVcp) and four different classes of antiviral therapeutics utilizing single-molecule methods. Our findings highlight several distinguishing features associated with these interactions. First, we observed that the low-nanomolar equilibrium dissociation constants of antiviral DNA aptamers arise from strong attractive electrostatic interactions that compact the intrinsically disordered NTDHCVcp. Conversely, the binding of high-affinity antibodies does not perturb the end-to-end distance of NTDHCVcp and is weakly impeded by electrostatic repulsions between similarly charged residues in the two binding partners. Although both of these antivirals have exceptionally high binding affinities for NTDHCVcp, they also have noteworthy limitations. For example, the antibody has a molecular weight of approximately 150 kDa which limits its ability to enter the cell. Although the aptamers are much smaller (13.7 kDa), their binding mechanism is strongly dependent on electrostatic interactions, and as such, they may tightly associate with other highly positively charged biomolecules in the cellular milieu. Next, we observed that low-affinity antiviral peptides bind to NTDHCVcp, causing it to oligomerize/aggregate in the absence of viral RNA. While these peptides do associate with NTDHCVcp, they are only effective at mid-micromolar concentrations, which ultimately limits their therapeutic utility. Finally, the previously identified small-molecule antiviral compound does not appear to affect any of our experimental observables, suggesting that binding may not alter the conformation properties of HCVcp.

ACKNOWLEDGMENTS

The authors would like to acknowledge the following individuals and institutions for their valuable contributions to this research: the other members of the Holmstrom lab for their insightful feedback during the preparation of this manuscript; D. Nettels, B. Schuler, and co-workers for developing Fretica (https://schuler.bioc.uzh.ch/programs/), a user-extendable toolbox for analyzing single-molecule fluorescence data within Mathematica (Wolfram Alpha); and the University of Kansas and the National Institutes of Health (P20GM103638 to EDH) for financial support.

SUPPLEMENTAL MATERIAL

Fig. S1 - jvi.00892-23-s0001.pdf
Sequences and structures of reagents.
Fig. S2 - jvi.00892-23-s0002.pdf
Mass spectrum analysis of the genotype 2a NTDHCVcp variant (cp2a).
Fig. S3 - jvi.00892-23-s0003.pdf
Fluorophore stoichiometry histogram.
Fig. S4 - jvi.00892-23-s0004.pdf
NTDHCVcp in the absence of antivirals.
Fig. S5 - jvi.00892-23-s0005.pdf
NaCl-induced compaction of cp1a.
Fig. S6 - jvi.00892-23-s0006.pdf
Antiviral aptamer-induced compaction of cp1a.
Fig. S7 - jvi.00892-23-s0007.pdf
Binding of antiviral aptamer to cp2a.
Fig. S8 - jvi.00892-23-s0008.pdf
Binding of small molecules to NTDHCVcp.
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Organization WH. 2022. Hepatits C fact sheet (2022). Available from: https://www.who.int/news-room/fact-sheets/detail/hepatitis-c. Retrieved 24 Jun 2022.
2.
Others W a. 1999. Global surveillance and control of hepatitis C. J Viral Hepat 6:35–47.
3.
Hijikata M, Kato N, Ootsuyama Y, Nakagawa M, Shimotohno K. 1991. Gene mapping of the putative structural region of the hepatitis C virus genome by in vitro processing analysis. Proc Natl Acad Sci U S A 88:5547–5551.
4.
Grakoui A, Wychowski C, Lin C, Feinstone SM, Rice CM. 1993. Expression and identification of hepatitis C virus polyprotein cleavage products. J Virol 67:1385–1395.
5.
Hayes CN, Imamura M, Tanaka J, Chayama K. 2022. Road to elimination of HCV: clinical challenges in HCV management. Liver Int. 42:1935–1944.
6.
Zoratti MJ, Siddiqua A, Morassut RE, Zeraatkar D, Chou R, van Holten J, Xie F, Druyts E. 2020. Pangenotypic direct acting antivirals for the treatment of chronic hepatitis C virus infection: a systematic literature review and meta-analysis. EClinicalMedicine 18:100237.
7.
Keikha M, Eslami M, Yousefi B, Ali-Hassanzadeh M, Kamali A, Yousefi M, Karbalaei M. 2020. HCV genotypes and their determinative role in hepatitis C treatment. Virusdisease 31:235–240.
8.
Cabral BCA, Ramos JA, Silveira AL de M, Nascimento ÉRDS, Ferreira SB, Coelho HSM, Moura-Neto RS, Villela-Nogueira CA, Hoffmann L, Silva R. 2022. Frequency distribution of HCV resistance-associated variants in infected patients treated with direct-acting antivirals. Int J Infect Dis 115:171–177.
9.
Li H-C, Lo S-Y. 2015. Hepatitis C virus: virology, diagnosis and treatment. World J Hepatol 7:1377–1389.
10.
Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, Tunyasuvunakool K, Bates R, Žídek A, Potapenko A, Bridgland A, Meyer C, Kohl SAA, Ballard AJ, Cowie A, Romera-Paredes B, Nikolov S, Jain R, Adler J, Back T, Petersen S, Reiman D, Clancy E, Zielinski M, Steinegger M, Pacholska M, Berghammer T, Bodenstein S, Silver D, Vinyals O, Senior AW, Kavukcuoglu K, Kohli P, Hassabis D. 2021. Highly accurate protein structure prediction with alphafold. Nature 596:583–589.
11.
Tunyasuvunakool K, Adler J, Wu Z, Green T, Zielinski M, Žídek A, Bridgland A, Cowie A, Meyer C, Laydon A, Velankar S, Kleywegt GJ, Bateman A, Evans R, Pritzel A, Figurnov M, Ronneberger O, Bates R, Kohl SAA, Potapenko A, Ballard AJ, Romera-Paredes B, Nikolov S, Jain R, Clancy E, Reiman D, Petersen S, Senior AW, Kavukcuoglu K, Birney E, Kohli P, Jumper J, Hassabis D. 2021. Highly accurate protein structure prediction for the human proteome. Nature 596:590–596.
12.
Mariani V, Biasini M, Barbato A, Schwede T. 2013. lDDT: a local superposition-free score for comparing protein structures and models using distance difference tests. Bioinformatics 29:2722–2728.
13.
Guo H-B, Perminov A, Bekele S, Kedziora G, Farajollahi S, Varaljay V, Hinkle K, Molinero V, Meister K, Hung C, Dennis P, Kelley-Loughnane N, Berry R. 2022. Alphafold2 models indicate that protein sequence determines both structure and dynamics. Sci Rep 12:10696.
14.
Ruff KM, Pappu RV. 2021. Alphafold and implications for intrinsically disordered proteins. J Mol Biol 433:167208.
15.
Bukh J, Purcell RH, Miller RH. 1994. Sequence analysis of the core gene of 14 hepatitis C virus genotypes. Proc Natl Acad Sci U S A 91:8239–8243.
16.
Duvignaud J-B, Savard C, Fromentin R, Majeau N, Leclerc D, Gagné SM. 2009. Structure and dynamics of the N-terminal half of hepatitis C virus core protein: an intrinsically unstructured protein. Biochem Biophys Res Commun 378:27–31.
17.
Dolan PT, Roth AP, Xue B, Sun R, Dunker AK, Uversky VN, LaCount DJ. 2015. Intrinsic disorder mediates hepatitis C virus core–host cell protein interactions. Protein Sci. 24:221–235.
18.
Holmstrom ED, Nettels D, Schuler B. 2018. Conformational plasticity of hepatitis C virus core protein enables RNA-induced formation of nucleocapsid-like particles. J Mol Biol 430:2453–2467.
19.
Gawlik K, Gallay PA. 2014. HCV core protein and virus assembly: what we know without structures. Immunol Res 60:1–10.
20.
Moradpour D, Penin F. 2013. Hepatitis C virus proteins: From structure to function, p 113–142. In Hepatitis C virus: From molecular Virology to antiviral therapy
21.
Moradpour D, Penin F, Rice CM. 2007. Replication of hepatitis C virus. Nat Rev Microbiol 5:453–463.
22.
Shi S, Yu X, Gao Y, Xue B, Wu X, Wang X, Yang D, Zhu H. 2014. Inhibition of hepatitis C virus production by aptamers against the core protein. J Virol 88:1990–1999.
23.
Torres-Vázquez B, de Lucas AM, García-Crespo C, García-Martín JA, Fragoso A, Fernández-Algar M, Perales C, Domingo E, Moreno M, Briones C. 2022. In vitro selection of high affinity DNA and RNA aptamers that detect hepatitis C virus core protein of genotypes 1 to 4 and inhibit virus production in cell culture. J Mol Biol 434:167501.
24.
Karthe J, Tessmann K, Li J, Machida R, Daleman M, Häussinger D, Heintges T. 2008. Specific targeting of hepatitis C virus core protein by an intracellular single‐chain antibody of human origin. Hepatology 48:702–712.
25.
Strosberg AD, Kota S, Takahashi V, Snyder JK, Mousseau G. 2010. Core as a novel viral target for hepatitis C drugs. Viruses 2:1734–1751.
26.
Schwer B, Ren S, Pietschmann T, Kartenbeck J, Kaehlcke K, Bartenschlager R, Yen TSB, Ott M. 2004. Targeting of hepatitis C virus core protein to mitochondria through a novel C-terminal localization motif. J Virol 78:7958–7968.
27.
Tillmann HL. 2014. Hepatitis C virus core antigen testing: role in diagnosis, disease monitoring and treatment. World J Gastroenterol 20:6701–6706.
28.
Wang S-R, Min Y-Q, Wang J-Q, Liu C-X, Fu B-S, Wu F, Wu L-Y, Qiao Z-X, Song Y-Y, Xu G-H, Wu Z-G, Huang G, Peng N-F, Huang R, Mao W-X, Peng S, Chen Y-Q, Zhu Y, Tian T, Zhang X-L, Zhou X. 2016. A highly conserved G-rich consensus sequence in hepatitis C virus core gene represents a new anti–hepatitis C target. Sci Adv 2:e1501535.
29.
Fine SL, Martin DF, Kirkpatrick P. 2005. Pegaptanib sodium. Nat Rev Drug Discov 4:187–188.
30.
Keefe AD, Pai S, Ellington A. 2010. Aptamers as therapeutics. Nat Rev Drug Discov 9:537–550.
31.
Fang X, Tan W. 2010. Aptamers generated from cell-SELEX for molecular medicine: a chemical biology approach. Acc Chem Res 43:48–57.
32.
Kohn DB, Bauer G, Rice CR, Rothschild JC, Carbonaro DA, Valdez P, Hao Q, Zhou C, Bahner I, Kearns K, Brody K, Fox S, Haden E, Wilson K, Salata C, Dolan C, Wetter C, Aguilar-Cordova E, Church J. 1999. A clinical trial of retroviral-mediated transfer of a rev-responsive element decoy gene into CD34+ cells from the bone marrow of human immunodeficiency virus-1–infected children, blood. Blood 94:368–371.
33.
DiGiusto DL, Krishnan A, Li L, Li H, Li S, Rao A, Mi S, Yam P, Stinson S, Kalos M, Alvarnas J, Lacey SF, Yee J-K, Li M, Couture L, Hsu D, Forman SJ, Rossi JJ, Zaia JA. 2010. RNA-based gene therapy for HIV with lentiviral vector–modified CD34+ cells in patients undergoing transplantation for AIDS-related lymphoma. Sci Transl Med 2:36ra43–36ra43.
34.
Group ES. 2003. Anti-vascular endothelial growth factor therapy for subfoveal choroidal neovascularization secondary to age-related macular degeneration: phase II study results. Ophthalmology 110:979–986.
35.
Drolet DW, Green LS, Gold L, Janjic N. 2016. Fit for the eye: aptamers in ocular disorders. Nucleic Acid Ther 26:127–146.
36.
Group ES. 2002. Preclinical and phase 1A clinical evaluation of an anti-VEGF pegylated aptamer (EYE001) for the treatment of exudative age-related macular degeneration. Retina 22:143–152.
37.
Kachko A, Kochneva G, Sivolobova G, Grazhdantseva A, Lupan T, Zubkova I, Wells F, Merchlinsky M, Williams O, Watanabe H, Ivanova A, Shvalov A, Loktev V, Netesov S, Major ME. 2011. New neutralizing antibody epitopes in hepatitis C virus envelope glycoproteins are revealed by dissecting peptide recognition profiles. Vaccine 30:69–77.
38.
Chiba J, Ohba H, Matsuura Y, Watanabe Y, Katayama T, Kikuchi S, Saito I, Miyamura T. 1991. Serodiagnosis of hepatitis C virus (HCV) infection with an HCV core protein molecularly expressed by a recombinant baculovirus. Proc Natl Acad Sci U S A 88:4641–4645.
39.
Yasui K, Wakita T, Tsukiyama-Kohara K, Funahashi SI, Ichikawa M, Kajita T, Moradpour D, Wands JR, Kohara M. 1998. The native form and maturation process of hepatitis C virus core protein. J Virol 72:6048–6055.
40.
Moradpour D, Wakita T, Tokushige K, Carlson RI, Krawczynski K, Wands JR. 1996. Characterization of three novel monoclonal antibodies against hepatitis C virus core protein. J Med Virol 48:234–241.
41.
Lun Y-Z, Cheng J, Zhong Y-W, Yu Z-G, Wang Q, Wang F, Feng J. 2011. Cloning, expression and identification by immunohistochemistry of humanized single-chain variable fragment antibody against hepatitis C virus core protein. Pol J Microbiol 60:13–17.
42.
Fosgerau K, Hoffmann T. 2015. Peptide therapeutics: current status and future directions. Drug Discov Today 20:122–128.
43.
Ni F, Kota S, Takahashi V, Strosberg AD, Snyder JK. 2011. Potent inhibitors of hepatitis C core dimerization as new leads for anti-hepatitis C agents. Bioorg Med Chem Lett 21:2198–2202.
44.
Kota S, Scampavia L, Spicer T, Beeler AB, Takahashi V, Snyder JK, Porco JA, Hodder P, Strosberg AD. 2010. A time-resolved fluorescence–resonance energy transfer assay for identifying inhibitors of hepatitis C virus core dimerization. Assay Drug Dev Technol 8:96–105.
45.
Kota S, Coito C, Mousseau G, Lavergne J-P, Strosberg AD. 2009. Peptide inhibitors of hepatitis C virus core oligomerization and virus production. J Gen Virol 90:1319–1328.
46.
Wang H. 2019. Small vs. big: Understanding the differences between small molecule drugs and biologic drugs. Immpress magazine.
47.
Bourne C, Lee S, Venkataiah B, Lee A, Korba B, Finn MG, Zlotnick A. 2008. Small-molecule effectors of hepatitis B virus capsid assembly give insight into virus life cycle. J Virol 82:10262–10270.
48.
Litterman N, Lipinski C, Ekins S. 2015. Small molecules with antiviral activity against the ebola virus. F1000Res 4:38.
49.
Panoutsopoulos AA. 2020. Known drugs and small molecules in the battle for COVID-19 treatment. Genes Dis 7:528–534.
50.
Cai L, Jiang S. 2010. Development of peptide and small‐molecule HIV‐1 fusion inhibitors that target gp41. ChemMedChem 5:1813–1824.
51.
Rizk JG, Lippi G, Henry BM, Forthal DN, Rizk Y. 2022. Prevention and treatment of monkeypox. Drugs 82:1–7.
52.
Jacobson IM, Pawlotsky J-M, Afdhal NH, Dusheiko GM, Forns X, Jensen DM, Poordad F, Schulz J. 2012. A practical guide for the use of boceprevir and telaprevir for the treatment of hepatitis C. J. Viral Hepat. 19:1–26.
53.
Lenz O, Vijgen L, Berke JM, Cummings MD, Fevery B, Peeters M, De Smedt G, Moreno C, Picchio G. 2013. Virologic response and characterisation of HCV genotype 2–6 in patients receiving TMC435 monotherapy (study TMC435-C202). J Hepatol 58:445–451.
54.
Kong J, Chen C, Balsells-Padros J, Cao Y, Dunn RF, Dolman SJ, Janey J, Li H, Zacuto MJ. 2012. Synthesis of the HCV protease inhibitor vaniprevir (MK-7009) using ring-closing metathesis strategy. J Org Chem 77:3820–3828.
55.
Taylor JG, Zipfel S, Ramey K, Vivian R, Schrier A, Karki KK, Katana A, Kato D, Kobayashi T, Martinez R, Sangi M, Siegel D, Tran CV, Yang Z-Y, Zablocki J, Yang CY, Wang Y, Wang K, Chan K, Barauskas O, Cheng G, Jin D, Schultz BE, Appleby T, Villaseñor AG, Link JO. 2019. Discovery of the pan-genotypic hepatitis C virus NS3/4A protease inhibitor voxilaprevir (GS-9857): a component of Vosevi®. Bioorg Med Chem Lett 29:2428–2436.
56.
Wei W, Cai C, Kota S, Takahashi V, Ni F, Strosberg AD, Snyder JK. 2009. New small molecule inhibitors of hepatitis C virus. Bioorg Med Chem Lett 19:6926–6930.
57.
Nakai K, Okamoto T, Kimura-Someya T, Ishii K, Lim CK, Tani H, Matsuo E, Abe T, Mori Y, Suzuki T, Miyamura T, Nunberg JH, Moriishi K, Matsuura Y. 2006. Oligomerization of hepatitis C virus core protein is crucial for interaction with the cytoplasmic domain of E1 envelope protein. J Virol 80:11265–11273.
58.
Boulant S, Vanbelle C, Ebel C, Penin F, Lavergne J-P. 2005. Hepatitis C virus core protein is a dimeric alpha-helical protein exhibiting membrane protein features. J Virol 79:11353–11365.
59.
Stryer L, Haugland RP. 1967. Energy transfer: a spectroscopic ruler. Proc Natl Acad Sci U S A 58:719–726.
60.
Schuler B, Lipman EA, Steinbach PJ, Kumke M, Eaton WA. 2005. Polyproline and the “spectroscopic ruler” revisited with single-molecule fluorescence. Proc Natl Acad Sci U S A 102:2754–2759.
61.
LeBlanc SJ, Kulkarni P, Weninger KR. 2018. Single molecule FRET: a powerful tool to study intrinsically disordered proteins. Biomolecules 8:140.
62.
Schuler B, Soranno A, Hofmann H, Nettels D. 2016. Single-molecule FRET spectroscopy and the polymer physics of unfolded and intrinsically disordered proteins. Annu Rev Biophys 45:207–231.
63.
Metskas LA, Rhoades E. 2020. Single-molecule FRET of intrinsically disordered proteins. Annu Rev Phys Chem 71:391–414.
64.
Nasir I, Bentley EP, Deniz AA. 2020. Ratiometric single‐molecule FRET measurements to probe conformational subpopulations of intrinsically disordered proteins. Curr Protoc Chem Biol 12:e80.
65.
Information NC f. B, Camacho C. 2008. BLAST (R) command line applications user manual. National Center for Biotechnology Information (US).
66.
Colina R, Casane D, Vasquez S, García-Aguirre L, Chunga A, Romero H, Khan B, Cristina J. 2004. Evidence of Intratypic recombination in natural populations of hepatitis C virus. J Gen Virol 85:31–37.
67.
Kato T, Furusaka A, Miyamoto M, Date T, Yasui K, Hiramoto J, Nagayama K, Tanaka T, Wakita T. 2001. Sequence analysis of hepatitis C virus isolated from a fulminant hepatitis patient. J Med Virol 64:334–339.
68.
Zosel F, Holla A, Schuler B. 2020. Labeling of proteins for single-molecule fluorescence spectroscopy. Chemistry.
69.
Kapanidis AN, Laurence TA, Lee NK, Margeat E, Kong X, Weiss S. 2005. Alternating-laser excitation of single molecules. Acc Chem Res 38:523–533.
70.
Müller BK, Zaychikov E, Bräuchle C, Lamb DC. 2005. Pulsed interleaved excitation. Biophys J 89:3508–3522.
71.
Nettes D, Schuler B. 2020. Fretica. Available from: https://schuler.bioc.uzh.ch/programs
72.
Holmstrom ED, Andrea H, Wenwei Z, Daniel N, Robert BB, Benjamin S. 2018. Accurate transfer efficiencies, distance distributions, and ensembles of unfolded and intrinsically disordered proteins from single-molecule FRET. Meth Enzymol 611:287–325.
73.
Laurence T, Fore S, Huser T. 2005. A fast, flexible algorithm for calculating correlations in fluorescence correlation spectroscopy. Opt Lett.
74.
Rhoades E, Ramlall TF, Webb WW, Eliezer D. 2006. Quantification of α-synuclein binding to lipid vesicles using fluorescence correlation spectroscopy. Biophys J 90:4692–4700.
75.
Kota S, Madoux F, Chase P, Takahashi V, Liu Q, Mercer BA, Cameron M, Strosberg AD, Roush W, Hodder P. 2014. Ml322, A small molecule inhibitor of dimerization of the core protein of hepatitis C virus (HCV), . In Probe reports from the NIH molecular libraries program [Internet]
76.
Sen S, Holmstrom E.D. 2022 RNA nanotechnology and Therapeutics, p 193–209. In A single-molecule FRET approach for investigating the binding mechanisms of anti-viral Aptamers in RNA nanotechnology and Therapeutics.. CRC Press, Boca Raton.
77.
Lee S, Kim YS, Jo M, Jin M, Lee D, Kim S. 2007. Chip-based detection of hepatitis C virus using RNA aptamers that specifically bind to HCV core antigen. Biochem Biophys Res Commun 358:47–52.
78.
Maity H, Baidya L, Reddy G. 2022. Salt-induced transitions in the conformational ensembles of intrinsically disordered proteins. J Phys Chem B 126:5959–5971.
79.
Maity H, Muttathukattil AN, Reddy G. 2018. Salt effects on protein folding thermodynamics. J Phys Chem Lett 9:5063–5070.
80.
Müller-Späth S, Soranno A, Hirschfeld V, Hofmann H, Rüegger S, Reymond L, Nettels D, Schuler B. 2010. From the cover: Charge interactions can dominate the dimensions of intrinsically disordered proteins. Proceedings of the National Academy of Sciences of the United States of America 107:14609–14614.
81.
Garaizar A, Espinosa JR. 2021. Salt dependent phase behavior of intrinsically disordered proteins from a coarse-grained model with explicit water and ions. J Chem Phys 155:125103.
82.
Hazra MK, Levy Y. 2021. Biophysics of phase separation of disordered proteins is governed by balance between short- and long-range interactions. J Phys Chem B 125:2202–2211.
83.
Wohl S, Jakubowski M, Zheng W. 2021. Salt-dependent conformational changes of intrinsically disordered proteins. J Phys Chem Lett 12:6684–6691.
84.
Wyman J, Gill SJ. 1990. Binding and linkage: Functional chemistry of biological macromolecules. University Science Books.
85.
Li W, Kou X, Xu J, Zhou W, Zhao R, Zhang Z, Fang X. 2018. Characterization of hepatitis C virus core protein dimerization by atomic force microscopy. Anal Chem 90:4596–4602.
86.
Dhiman RK, Premkumar M. 2020. Hepatitis C virus elimination by 2030: conquering mount improbable. Clin Liver Dis (Hoboken) 16:254–261.
87.
Kish T, Aziz A, Sorio M. 2017. Hepatitis C in a new era: a review of current therapies. P T 42:316–329.
88.
Lin MV, Chung R. 2014. Recent FDA approval of sofosbuvir and simeprevir. Implications for current HCV treatment. Clin Liver Dis (Hoboken) 3:65–68.
89.
Courcambeck J, Bouzidi M, Perbost R, Jouirou B, Amrani N, Cacoub P, Pèpe G, Sabatier J-M, Halfon P. 2006. Resistance of hepatitis C virus to NS3–4A protease inhibitors: mechanisms of drug resistance induced by R155Q, A156t, D168A and D168V mutations. Antiviral Therapy 11:847–856.
90.
Pawlotsky J-M. 2016. Hepatitis C virus resistance to direct-acting antiviral drugs in interferon-free regimens. Gastroenterology 151:70–86.
91.
Roebuck MC, Liberman JN. 2019. Assessing the burden of illness of chronic hepatitis C and impact of direct-acting antiviral use on healthcare costs in medicaid. Am J Manag Care 25:S131–S139.
92.
Lindenbach BD, Rice CM. 2013. The INS and outs of hepatitis C virus entry and assembly. Nat Rev Microbiol 11:688–700.
93.
Ivanyi-Nagy R, Darlix J-L. 2010. Intrinsic disorder in the core proteins of flaviviruses. Protein Pept Lett 17:1019–1025.
94.
Counihan NA, Rawlinson SM, Lindenbach BD. 2011. Trafficking of hepatitis C virus core protein during virus particle assembly. PLoS Pathog 7:e1002302.
95.
Holmstrom ED, Liu Z, Nettels D, Best RB, Schuler B. 2019. Disordered RNA chaperones can enhance nucleic acid folding via local charge screening. Nat Commun 10:2453.
96.
Schuler B, Borgia A, Borgia MB, Heidarsson PO, Holmstrom ED, Nettels D, Sottini A. 2020. Binding without folding - the biomolecular function of disordered polyelectrolyte complexes. Curr Opin Struct Biol 60:66–76.
97.
Schouten A, Roosien J, Bakker J, Schots A. 2002. Formation of disulfide bridges by a single-chain Fv antibody in the reducing ectopic environment of the plant cytosol. J Biol Chem 277:19339–19345.
98.
Ahmad ZA, Yeap SK, Ali AM, Ho WY, Alitheen NBM, Hamid M. 2012. scFv antibody: principles and clinical application. Clinical & developmental immunology 2012:980250.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 97Number 1031 October 2023
eLocator: e00892-23
Editor: J.-H. James Ou, University of Southern California, Los Angeles, California, USA
PubMed: 37772835

History

Received: 14 June 2023
Accepted: 10 August 2023
Published online: 29 September 2023

Permissions

Request permissions for this article.

Keywords

  1. HCV
  2. core protein
  3. nucleocapsid
  4. single-molecule
  5. FRET
  6. antivirals

Contributors

Authors

Sudip Nepal
Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
Author Contributions: Data curation, Formal analysis, Investigation, Visualization, Writing – original draft, and Writing – review and editing.
Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, USA
Department of Chemistry, University of Kansas, Lawrence, Kansas, USA
Author Contributions: Conceptualization, Data curation, Formal analysis, Funding acquisition, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing – original draft, and Writing – review and editing.

Editor

J.-H. James Ou
Editor
University of Southern California, Los Angeles, California, USA

Notes

The authors declare no conflict of interest.

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy