METHODS AND COMPOUNDS TO INHIBIT ENVELOPED VIRUS RELEASE

A compound having an antiviral activity for inhibiting release of an enveloped virus from a cell is disclosed, including methods of inhibiting release of an enveloped virus from a cell. The antiviral activity of the compound includes inhibiting formation of an associative complex or disrupting formation of an associative complex. The associative complex comprises an L-domain motif of the enveloped virus and at least one cellular polypeptide, or fragment thereof, capable of binding the L-domain motif of the enveloped virus.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims benefit of priority to U.S. provisional application No. 61/745,336 filed on Dec. 21, 2012, which is incorporated by reference in its entirety.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT

Not applicable.

SEQUENCE LISTING

The instant application contains a Sequence Listing that has been submitted in ASCII format via EFS-Web and is hereby incorporated by reference in its entirety. Said ASCII copy, created in final form on Dec. 20, 2013, is named NWN01-012-US_ST25.txt, and is 68,266 bytes in size.

FIELD OF THE INVENTION

This invention relates to methods for identifying compounds that inhibit interactions with the Nedd4 family of ubiquitin ligases and Tsg101, including inhibitors of virus budding.

BACKGROUND OF THE INVENTION

There is considerable interest developing antiviral reagents to combat viral infections. The two most prevalent antiviral strategies focus on creating immunity to viral infection by use of vaccines or by interfering with a necessary virus-specific process essential to virus maintenance, replication and propagation in the host.

Vaccines have been successfully developed for many viruses to combat viral infections. So-called live vaccines containing attenuated version(s) of the target virus provide a convenient means of conferring immunity as typically only one inoculation is required. The drawbacks to most live virus vaccines lie in their limited shelf life, the requirement for maintaining appropriate storage conditions to preserve the vaccine reagent, and the possibility of revertance to high virulence due to their active replication. These drawbacks can be avoided by using so-called inactivated virus vaccines containing a completely inert virus particle or a sub-viral component like a protein. The drawback to inactive viral vaccines is that multiple inoculations are required to confer full immunity. Furthermore, vaccines have an attendant risk that adverse reactions might arise in certain populations following immunization (for example, autoimmunity responses associated with Guillain-Barré syndrome (GBS)).

Antiviral compounds that specifically target a viral replication process have also proven effective for treating some virus infections. Examples of such reagents include small molecule inhibitors selective for a given viral protein, such as a viral replicase (for example, the nucleoside analog 3′-azidothymidine for inhibiting the HIV-1 reverse transcriptase) or a viral protease (for example, Darunavir for inhibiting HIV-1 protease). Owing to their small molecular size and chemical composition, antiviral compounds can be formulated as pharmaceutical compositions having significant shelf life and can typically retain their potency over a larger temperature range during storage than many vaccines. However, HIV-1 and other virus can mutate to escape the effectiveness of the antiviral drugs when such drugs are targeted against virus-specific proteins. In particular, HIV-specific drugs have side-effects that cause patients to interrupt therapy that can lead to drug-resistant viral strains.

Generally, antiviral compounds are typically used in combinations for maximum efficacy and durability. Though most aspects of the viral replication process are susceptible to targeting and inhibition, the primary focus of antiviral inhibitor drug development is on early stage processes of viral replication, when the copy number of viral protein or nucleic acid targets is relatively low.

Late stage replication events include those associated with virus particle assembly and release from the host cell. These viral processes are more difficult targets to develop antiviral reagents. This is due in part to the vastly larger number of virus particles that result from active viral replication.

Enveloped virus particles adopt an outer membrane structure composed of the host cell membrane in its final virus form. Examples of enveloped viruses include retroviruses (for example, human immunodeficiency virus, type 1), rhabdoviruses (for example, rabies virus), and herpes viruses (for example, herpes simplex virus, type 1). For enveloped viruses, the final stages of virus replication include envelope maturation, budding and release.

No antiviral therapeutic reagents have been developed that target the processes of enveloped virus budding and release. This is due in large part to the inability to target virus-specific proteins, owing to the large number of viral proteins present during late phase infection. But more importantly, the host cell-virus interactions responsible for enveloped virus particle maturation, budding and release are only poorly understood.

BRIEF SUMMARY OF THE INVENTION

In a first aspect, a compound having an antiviral activity for inhibiting release of an enveloped virus from a cell is disclosed. In one aspect, the compound has an antiviral activity that includes inhibiting formation of an associative complex or disrupting formation of an associative complex. The associative complex comprises an L-domain motif of the enveloped virus and at least one cellular polypeptide, or fragment thereof, capable of binding the L-domain motif of the enveloped virus.

In a second aspect, a method of inhibiting release of an enveloped virus from a cell is disclosed. The method includes the step of contacting the cell with a compound having an antiviral activity. The antiviral activity includes inhibiting formation of an associative complex or disrupting formation of an associative complex. The associative complex comprises an L-domain motif of the enveloped virus and at least one cellular polypeptide, or fragment thereof, capable of binding the L-domain motif of the enveloped virus.

In a third aspect, a pharmaceutical composition comprising a compound having an antiviral activity for inhibiting release of an enveloped virus from a cell and optionally a pharmaceutically acceptable carrier is disclosed.

BRIEF DESCRIPTION OF THE DRAWINGS

The application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.

FIG. 1 depicts parallel pathways used by ASLV and HIV-1 Gag to bud from cells.

FIG. 2 depicts a first embodiment to screen in vitro for the ability of a test compound (104) to inhibit an associative complex (103) formed between a fluorescently-labeled viral peptide probe (101) containing a viral L-domain motif and a cellular ESCRT complex protein (102). The fluorescence of 101 in 103 is denoted as F1, while the fluorescence of 101 free is denoted as F2. Note that compound 104 need not bind to the same site as probe 101 on protein 102 or that probe 101 need be completely dissociated from 102 to have F2 arise.

FIG. 3 depicts a second embodiment to screen in vitro for the ability of a test compound (204) to bind to a cellular ESCRT complex protein (202) that is immobilized to substrate (201) via a linking group (203). The assay measures changes in the index of refraction upon a binding event (205) by comparing the wavelength of the reflected light (RL1) in the absence of 204 to the wavelength of the reflected light (RL2) in the presence of 204.

FIG. 4 depicts a third embodiment to screen in vivo for the ability of a test compound (304) to inhibit a reconstituted EGFP two-hybrid complex (303) formed between a partial N-terminal fusion EGFP polypeptide (denoted by small ball) that includes a viral peptide (301) containing a viral L-domain motif and a complementary partial C-terminal EGFP fusion polypeptide that includes a cellular ESCRT complex protein or a viral L-domain binding domain derived therefrom (302). The reconstituted EGFP complex 303 produces the fluorescence (Fl) analogous to native EGFP when 301 and 302 interact in vivo. If test compound 304 interferes with formation of 403, the cells produce less fluorescence.

FIG. 5A depicts EPIC label-free binding assay data with Celastrol and WWP2 (SEQ ID NO:24).

FIG. 5B depicts EPIC label-free binding assay data with Oxytetracycline and WWP2 (SEQ ID NO:24).

FIG. 5C depicts EPIC label-free binding assay data with Benserazide HCl and WWP2 (SEQ ID NO:24).

FIG. 6A depicts an exemplary assay that shows the effect of small molecule inhibitors targeting Nedd4 interaction with PPPY-containing viral Gag motif L-domain on the release of ASLV virus like particles (VLPs) from 293 cells, as adjudged by Western blot assay using an anti-ASLV Gag antibody to detect ASLV Gag protein that remains associated with the cells (cell lysates) or released from cells (VLPs) when 293 cells that express ASLV Gag protein are contacted with no inhibitor or with inhibitors (K21 [Benserazide Hydrochloride] and N20 [Oxytetracycline]) at the indicated concentrations in the media. The ASLV Gag protein denoted as Δp2b contains a deletion of the L-domain motif. The budding defect can be seen by comparing release of VLPs from untreated cells (lane 2) to K21-contacted cells (lanes 3-5) and N20-contacted cells (lanes 6-8). Lane 1 is loading control for untreated cells that do not express ASLV Gag protein. Lane 9 is a control for untreated cells that express ASLV Gag with an L-domain deletion (Δp2b). These inhibitor concentrations are not cell toxic.

FIG. 6B depicts percentage of absorbance of control (0 μM) as a function of concentration of the inhibitor K21 [Benserazide Hydrochloride] in the media.

FIG. 6C depicts VLP release (LOG(PFU/ml) as a function of time for the indicated concentrations of the inhibitor K21 [Benserazide Hydrochloride] in the media.

FIG. 6D depicts VLP release (LOG(PFU/ml) as a function of time for the indicated concentrations of the inhibitor K21 [Benserazide Hydrochloride] in the media.

FIG. 7 depicts an fluorescence-based thermal shift (FTS) assay to detect a thermal shift in protein folding of TSG101 (SEQ ID NO:33) due to the presence of inhibitor compound N16. In this case, the inhibitor compound induced thermal instability, resulting in a lower Tm for protein unfolding.

FIG. 8A depicts results of infectious HSV-1 virion production from HSV-1 infected VERO cells alone or HSV-1 infected VERO cells contacted with PTAP-motif inhibitor compounds, F15 (Esomeprazole potassium) and N16, at the indicated concentrations. The titers of infectious HSV-1 virus particles was determined from plaque assays with naïve (uninfected) VERO cell cultures infected with virions harvested from either the cultured cell media containing the VERO cells contacted with no inhibitor compound or with one of compounds F15 or N16 (“supernatant”), or from both the cultured cell media and the VERO cells contacted no inhibitor compound or with one of compounds F15 or N16 (“total”). The indicated concentrations of the PTAP-motif inhibitor compounds, F15 and N16, are not toxic to the contacted VERO cells.

FIG. 8B depicts results of MTS-based assays for evaluating cytotoxicity of cells contacted with the PTAP-motif inhibitor compounds, F15 and N16, where Absorbance at 490 nm (indicative of cell viability) is plotted as a function of inhibitor concentration present in cell culture medium in contact with the cells.

DETAILED DESCRIPTION OF THE INVENTION

Disclosed herein are methods and materials to inhibit the interaction of the above-mentioned cellular proteins or fragments thereof with L domain-containing peptides of enveloped viruses. Applicants made the seminal discovery that enveloped viruses use cellular pathways for mediating virus budding and that inhibiting these pathways results in significantly decreased rates of enveloped virus release from cell surfaces. The methods disclosed herein provide a robust, high-throughput approach to identify lead compounds having potent inhibitory effects on enveloped virus protein interactions with the components of these pathways and, thereby, virus particle release.

Referring to FIG. 1, enveloped viruses such as avian sarcoma and leukosis virus (ASLV) and human immunodeficiency virus, type 1 (HIV-1) include late assembly domains (“L-domains”) encoded within their Gag protein sequence that interact with cellular components of the endosomal sorting complex required for transport (“ESCRT”) machinery for virus budding and release from cells. The L-domains have been identified in a variety of enveloped viruses and families of enveloped viruses. Applicants have identified a consensus subset of L-domain motifs that interact with the critical ESCRT-dependent processes that enveloped viruses use to bud from cell membranes (see Table I, underlined, bold sequences).

TABLE I L-domains found in enveloped virus proteins. Amino acid sequence Virus species Virus1 Protein containing the L-domain2 SEQ ID NO: Arenavirus LFV Z AAPTAPPTGAADSIPPPYSP 1 LCMV Z TAPSSPPPYEE 2 Filovirus EboV VP40 MRRVILPTAPPEYMEAI 3 MarV VP40 NTYMQYLNPPPYADHS 4 Hepadnavirus HBV Core PPAY 5 Herpesvirus HSV-1 E PPTY 6 HSV-2 UL56 PPPY 7 CMV UL32 PTAP 8 Paramyxovirus SV5 M QSIKAFPIVINSDG 9 MuV M RLNAFPIVMGQ 10 Retrovirus ASLV p2B ATASAPPPPYVGSGLYPSL 11 (Gag) HIV-1 p6 PEPTAPPFLQSRPEPTAPPEES 12 (Gag) HTLV-I MA DPQIPPPYVEPTAP 13 (Gag) EIAV p9 QNLYPDLSEIK 14 (Gag) Rhabdovirus VSV M LGIAPPPYEEDTSMEYAPSAP 15 RV M DDLWLPPPEYVPLKEL 16 1Virus names corresponding to the abbreviations presented are as follows: LFV, Lassa fever virus; LCMV, lymphocytic choriomeningitis virus; EboV, Ebola virus; MarV, Marberg virus; HBV, hepatitis B virus; HSV-1, Herpes simplex virus, type 1; HSV-2, Herpes simplex virus, type 2; CMV, cytomegalovirus; SV5, Simian virus, type 5; MuV, Mumps virus; ASLV, avian sarcoma leucosis virus; HIV-1, human immunodeficiency virus, type 1; HTLV-I, human T-lymphotrophic virus, type 1; EIAV, equine infectious anemia virus; VSV, vesicular stomatitis virus; and RV, rabies virus. 2Underlined, bolded sequences indicate the consensus sequences within the L-domains.

One of these L-domain motifs, termed the PTAP motif (for example, from HIV-1), interacts with the TSG101 protein that becomes recruited as part of the ESCRT complexes. Another of these L-domain motifs, termed PPPY motif (also referred to as the “PY motif” or the “PY L-domain motif;” for example, from ASLV), interacts with the Nedd4 family of proteins that is also recruited by ESCRT-associated proteins. While it is often the case that certain viruses have a viral protein might encode both types of L-domains, typically only one predominates in the viral budding process through interactions with ESCRT machinery. The Applicants have devised novel, robust screening methods to identify compounds that interfere with the interaction between viral L-domains that include the PPPY motif or PTAP motif and ESCRT component, TSG101 or ESCRT-linked component, Nedd4 family proteins. These screening methods enable one to rapidly identify compounds that inhibit the interactions of both Nedd4 and TSG101 with the viral L-domain motifs, thereby providing a high-throughput strategy to obtain candidate lead compounds having utility as novel antiviral agents for inhibiting virus budding and release from infected cells.

Some candidate lead compounds can display potency at inhibiting only Nedd4- or TSG101-mediated ESCRT pathways, thereby offering specific antiviral activity for one type of virus or virus family. Yet other candidate lead compounds can display potency at inhibiting both Nedd4- or TSG101-mediated ESCRT pathways, thereby offering broad-spectrum antiviral activity to a plurality of diverse enveloped virus families. Thus, the screening methods disclosed herein contemplate identification of compounds having either narrow- or broad-spectrum antiviral effects.

One preferred embodiment of such a screening method is depicted in FIG. 2. Briefly, the assay is based upon changes in the fluorescence polarization of a fluorescently-labeled viral peptide probe 101 that includes an L-domain motif in an associative complex 103 with selected cellular ESCRT-associated protein 102 (for example, TSG101 (SEQ ID NOs:32 or 33) or a Nedd4 family polypeptide (for example, WWP1 (SEQ ID NO:29), WWP2 (SEQ ID NO:24), Nedd4 L, among others), or a fragment thereof) as a function of the presence or absence of test compound 104. Because the associative complex 103 restricts motion of the fluorescently labeled viral peptide probe 101, the latter displays characteristic fluorescent polarization properties (denoted by F1 of FIG. 2). Upon introduction of compound 104 into solutions containing the associative complex 103, an interaction between compound 104 and the associative complex 103 that alters or releases the fluorescently-labeled viral peptide probe 101 will result in a change in fluorescence polarization of the solution (denoted F2 of FIG. 2.)

Permutations and variations of the embodiment of the screening assay (FIG. 2) are recognized to one skilled in the art based upon this disclosure and fall within the scope of the invention. For example, the selection of the fluorescent label and its attachment within probe 101 can be varied, provided that associative complex 103 can form having a discernible fluorescence polarization signal. For example labels can be introduced at the amino terminus, the carboxy terminus or at suitable locations within the peptide probe using coupling chemistries that are well known in the art. In particular, the type of fluorescent label selected will depend upon a variety of factors, such as whether the spectral properties of the label can be discerned under assay conditions such as the presence of fluorescent species derived from assays components like the viral peptide sequences that form probe 101, the selected cellular ESCRT protein 102, and the test compound 104.

Likewise, the preferred choices of the viral peptide sequences for inclusion in probe 101 are routine in nature, as are the preferred choices of whether a single copy or a plurality of copies of said viral sequences are to be included in probe 101. For example, three tandemly-linked copies of the PPPY motif from the Δp2b Gag protein of ALSV (for example, SEQ ID NO:17) provided a Kd of 0.20 μM for the WWP2 polypeptide (SEQ ID NO:24), as compared to the corresponding monomer that had a Kd of 18 μM. Though any specific viral peptide sequence can be included in probe 101, preferred embodiments include L-domain motifs, including those of Table I (SEQ ID NOS: 1-16) and ESCRT-linked protein (for example, Nedd4 family polypeptides (for example, WWP1 (SEQ ID NO:29), WWP2 (SEQ ID NO:24), Nedd4 L, among others) and TSG101 polypeptide, or a fragment thereof), binding variants thereof, as well as others known in the art.

Furthermore, additional representative viral L-domain motifs can be identified from a variety of viruses belonging to different enveloped virus families using standard biochemical and molecular techniques. These L-domain motifs can be screened using modifications of the assay presented in FIG. 2. Representative viruses for this purpose include Lassa fever virus; lymphocytic choriomeningitis virus; Ebola virus; Marberg virus; West Nile Virus; hepatitis B virus; Herpes simplex virus, type 1; Herpes simplex virus, type 2; cytomegalovirus; Simian virus, type 5; Mumps virus; avian sarcoma leucosis virus; human immunodeficiency virus, type 1; human T-lymphotrophic virus, type 1; equine infectious anemia virus; vesicular stomatitis virus; and rabies virus, among others.

Likewise, the choice of cellular ESCRT component polypeptides of Nedd4 family members (for example, WWP1 (SEQ ID NO:29), WWP2 (SEQ ID NO:24), Nedd4 L, among others) and TSG101 depends upon the solubility attributes of the selected protein. For example, while the recombinant Nedd4 polypeptides are soluble under assay conditions, the full-length TSG101 was not. In the case of conducting this screening assay with TSG101, a soluble recombinant polypeptide fragment derived from the full-length protein having the binding domain for the viral L-domain motifs was prepared and used for screening assays (See SEQ ID NO:33, Table VI). One particularly sensitive assay for detecting candidate inhibitor interactions with TSG101 (SEQ ID NO:33) is based upon the ability of the candidate to alter the thermal denaturation profile of protein folding for a soluble fragment of TSG101 peptide (SEQ ID NO:33), as assessed by fluorescence methods. These assays and other aspects are described in detail in the Examples.

A high-throughput assay using the screening method of FIG. 2 was devised to screen a library of 70,000 compounds for the ability to disrupt complexes formed between Nedd4 WWP2 (SEQ ID NO:24) and the ASLV L-domain motif containing PPPY (for example, SEQ ID NO: 17). The results of this initial screen (for example with FITC-labeled probe 101 (SEQ ID NO:18)) yielded about 700 candidate compounds having the desired molecular inhibitory properties. Owing to the possibility of false-positives arising in these assays, the initial collection candidate compounds are rescreened in a secondary assay fitted with viral probe 101 containing a different fluorescent label (for example, with TAMRA-labeled probe 101 (SEQ ID NO:19)). The results of the secondary screen with the identified collection of 700 compounds provided a further reduction of viable candidates to 130 compounds having properties for inhibiting virus release from cells.

Another preferred embodiment of an in vitro screening method is depicted in FIG. 3. In this method, the ability of the candidate compound 204 to directly interact with the cellular ESCRT-linked component protein (for example, Nedd4 family of polypeptides [for example, WWP1 (SEQ ID NO:29), WWP2 (SEQ ID NO:24), Nedd4 L, among others]) or TGS101 is evaluated. According to this embodiment, a cellular ESCRT component protein 202 is preferably immobilized to a substrate 201 (for example, a microtiter plate) via a linking group 203 (for example, an amine group). The substrate preferably includes an optional reference area that provides a control to prevent non-specific interactions with non-mobilized ESCRT complex protein 202 with substrate 201. The substrate 201 is washed and irradiated with broadband light so that a baseline refractive index is measured. Test compound 204 is added to the system to permit its binding to the immobilized ESCRT component protein 202 to form a resultant associative complex 205. The substrate 201 is irradiated with broadband light again to detect a change in the wavelength of the reflected light.

Commercial instruments are available to enable high-throughput screening of test compounds 204 for their ability to bind ESCRT component protein 202 (for example, TSG101 (SEQ ID NOs:32 or 33) or a Nedd4 family polypeptide (for example, WWP1 (SEQ ID NO:29), WWP2 (SEQ ID NO:24), Nedd4 L, among others)) for example, the Epic® technology; PerkinElmer, Inc. (Watham, Mass.)). As will be apparent to one skilled in the art, either the ESCRT component protein 202 or the test compound 204 may be immobilized to substrate 201. However, it is preferable to immobilize ESCRT component protein 202 to provide a high-throughput platform for screening a plurality of test compounds 204 in parallel for their ability to form associative complex 205. These assays and other aspects are described in detail in the Examples.

This screening method can be used in conjunction with the method outlined in FIG. 2 to further refine the collection of identified compounds to identify those that directly interact with ESCRT component polypeptides. For example, the screening method was applied to 30 candidates from the collection of 130 compounds identified previously to provide seven compounds having the ability to bind directly to Nedd4 family polypeptides in the micromolar range of inhibitor compound concentrations. As further described below, two of these compounds were found to be non-toxic to cultured cells and were capable of blocking PPPPY-dependent virus-like particle (VLP) budding and release from cells.

The advantages of performing the screening method of FIG. 3 (relative, for example, to the embodiment illustrated in FIG. 2) is that one can measure direct binding interactions between test compounds 204 and ESCRT component proteins 202 in a label-free assay. In addition to being able to detect direct biomolecular interactions between test compounds 204 and ESCRT component proteins 202, one can rapidly perform thermodynamic measurements of the binding interaction (for example, Kd determinations) resulting in formation of associative complex 205. This latter advantage can provide an estimate of whether certain compounds (204) display viable binding properties supportive of continuing on with biological testing of the compounds in viral inhibition studies.

FTS Assay for Detecting Direct Binding Interactions Between Test Compounds and ESCRT Component Proteins.

The fluorescence-based thermal shift assay is based on the observation that a protein unfolds upon heating, exposing the hydrophobic residues within its tertiary structure. The unfolding temperature (Tm) is determined by the protein's primary sequence and solution environment. The FTS assay uses a fluorescent dye sensitive to a hydrophobic environment to probe protein stability and its modulation by small molecule ligands. The dye has a low fluorescence quantum yield when in a polar environment. Once in contact with the hydrophobic core normally buried within a folded protein that has become exposed during the thermal unfolding (melting) process, the quantum yield of the dye increases, thus providing a reporting signal. Furthermore, a protein's stability can be affected by ligand binding, resulting in an increase or decrease in its melting temperature. FTS assay uses the Tm shift upon binding of a ligand to identify hit compounds for drug discovery. See Pantoliano M W, Petrella E C, Kwasnoski J D, Lobanov V S, Myslik J, Graf E, Carver T, Asel E, Springer B A, Lane P, Salemme F R. (2001) High-density miniaturized thermal shift assays as a general strategy for drug discovery. J Biomol Screen 6: 429-440; Lo M C, Aulabaugh A, Jin G, Cowling R, Bard J, et al. (2004) Evaluation of fluorescence-based thermal shift assays for hit identification in drug discovery. Anal Biochem 332: 153-159, which are incorporated by reference in their entireties.

In Vivo Screening Methods-Based Molecular Genetic Assays, Cell-Based VLP Production Assays And Whole-Virus Replication Assays

As an alternative or complementary embodiment to the described screening methods in vitro, an in vivo screening method is contemplated as depicted in FIG. 4. In this method, EGFP is genetically engineered as two-half polypeptide gene cassettes (that is, N-terminal portion of EGFP and C-terminal portion of EGFP), which when expressed simultaneously in the cells, fail to provide a functionally reconstituted EGFP having fluorescence properties. The two-half EGFP polypeptide gene cassettes are engineered to include a viral L-domain motif (for examples, a PY motif or a PTAP motif) in one of the EGPF cassettes (see FIG. 4, 301) and a cellular ESCRT component protein (for example, a Nedd4 family polypeptide or a TSG101 polypeptide or fragment thereof (SEQ ID NOs:32 or 33)) in the other EGFP cassette (see FIG. 4, 302). When cells are co-transfected with expression vectors containing half-EGFP gene cassettes, the corresponding fusion proteins are expressed to enable reconstitution of EGFP functional activity via interaction between the two heterologous fusion partners, the viral L-domain motif and the corresponding ESCRT complex polypeptide in the form of an associative complex (see FIG. 4, 303). Cells (either transiently or stably expressing the EGFP constructs) can be evaluated using test compounds (see FIG. 4, 304) to assess whether the compounds reduce cellular fluorescence. These assays and other aspects are described in detail in the Examples.

Candidate compounds having an inhibitory effect of fluorescence have also been evaluated for their ability to interfere with normal cellular physiology and growth by, for example, determining cytotoxicity profiles of the compounds as a function of dose response and incubation time with the cells. One advantage of the in vivo assay is that it provides additional opportunities to survey test compounds that otherwise might not be possible with the aforementioned biochemical assays (for example, with assays involving certain ESCRT component polypeptides having limited solubility in vitro). Other further advantages of in vivo assays of this sort is that they can provide a useful model for studying compound transport and clearance in cells as would be important for determining ADME profiles (for examples, bioactivity, bioavailability, bio-inactivation, among others) at a cellular level, as well as provide additional confirmatory evidence of the biological potency of the compounds in a more meaningful, biological context.

For candidate lead compounds identified through one or more of the aforementioned screening methods, biological assays have been established to evaluate the specific antiviral inhibitory effects the compounds have on virus budding and release. In one assay, virus like particle (“VLP”) production can be evaluated as a function of test compound dose. For example, human 293 cells can permit use of ASLV gag expression systems to study viral protein expression and VLP production as a function of test compound dose response. Likewise, human 293 cells can be used to follow VLP production with HIV-1 gag expression systems as a function of test compound dose response. Follow-up experiments well within the skilled artisan's grasp include evaluating other aspects of viral replication, as monitored by standard biochemical assays (PCR, RT-PCR, western blot methods and the like) as well as cell toxicity effects. These assays and other aspects are described in detail in the Examples or are otherwise well understood in the art.

VLP production assays have provided evidence of candidate lead compounds showing antiviral inhibitory effect on virus particle release as a function of dose, experiments then can proceed to demonstrate the antiviral effect in whole virus replication assays. Three preferred whole virus replication assay systems include the rhabdovirus VSV replication system, the herpes virus KSHV replication system, HSV-1 replication assay in Vero cells, and HIV-1 virus replication system with assays in the physiological host, i.e., CD4+ T cells. These assays and other aspects are described in detail in the Examples.

The aforementioned in vitro and in vivo screening methods can be combined either in series or in parallel (and in any order) to identify compounds having either narrow-spectrum activity against a few viruses or broad-spectrum antiviral activity against many different viruses. For example, lead compounds identified that interact with Nedd4 family members can be evaluated for their ability to interact with TSG101 or to disrupt TSG101-viral L-domain interactions. In this manner, different antiviral compounds can be discerned having discrete types of inhibitory activity. Further, one can identify gradients of antiviral potency across entire classes of viruses by evaluating the dose response profiles in a combination of biochemical and biological assays with different virus families having different viral L-domain motifs, as described herein. Moreover, combinations of compounds have TSG101-specific inhibitory activity and Nedd4 family-specific inhibitory activity can be tested against virus infection to determine whether the drug combinations block virus access to the ESCRT-complex dependent pathways are blocked for enveloped virus release.

These approaches have clear utility for two simple reasons. First, L-domains encoding the aforementioned PY motifs and PTAP motifs can be found with viral proteins for single virus families. Thus, viruses having both types of L-domains can potentially utilize both pathways mediated by Nedd 4 and TSG101. Second, the L-domains used by viruses are interchangeable. Thus, there is a need for compounds to disrupt both interactions between viral L-domains with the two different pathways mediated by Nedd 4 and TSG101, wherein virus budding and release can occur from different cellular membranes.

The identified compound inhibitors have utility as antiviral therapeutic agents. The therapy is a post infection treatment that will slow down the spread of virus by preventing particles from releasing from infected cell surfaces. The accumulation of particles will enhance detection by the immune system, which will clear the infection. The human body already has an innate immunity response that targets the release of virus particles late in infection. Thus, the above approach has viability because it will complement the natural immunity mechanism.

By using the in vivo screening assays described herein, several compounds have been identified having inhibitory effects on viral L-domain motif interaction with ESCRT component polypeptides, or in the alternative, having the capability to bind directly to the ESCRT component polypeptides. These compounds are listed in Tables II-IV.

TABLE II Candidate compounds that inhibit viral budding and release. Compound Assay Identification1 Relevant Property2 NSC306711; FP Assay (FITC/TAMRA); 1 μM 813419-93-1 Epic; VLP-ASLV NSC128437 FP Assay (FITC/TAMRA); 3 μM Epic Assay CD27-G10 FP Assay (FITC/TAMRA); 1 μM Epic Assay CD23-G07 FP Assay (FITC/TAMRA); 20-30 μM     Epic Assay CD15-B10 FP Assay (FITC/TAMRA); Epic Assay 1Assays used include the fluorescence polarization assay (“FP Assay”) with SEQ ID NO: 17 coupled to either a FITC label (SEQ ID NO: 18) or a TAMRA label (SEQ ID NO: 19) (“FITC/TMR”); label-free, refractive index assay (“Epic Assay”). 2Relevant property is [compound] to achieve 50% change in fluorescence polarization in FP Assay.

Candidate compound NSC306711; 813419-93-1 has the following structure (I):

Candidate compound NSC128437 has the following structure (II):

Candidate compound CD27-G10 has the following structure (III):

Candidate compound CD23-G07 has the following structure (IV):

Candidate compound CD15-B10 has the following structure (V):

Additional candidate compounds were identified with the fluorescence polarization assay and Epic label-free binding assay using WWP2 (SEQ ID NO:24) as the target. These candidate compounds and their properties are presented in Table III. The respective binding assays using the Epic label-free system are depicted in FIG. 5.

TABLE III Candidate compounds that inhibit PY-motif interactions with WWP2 Kd IUPAC Name (μM) (Common Name) CAS Reg. No. Structure 18.5 3-Hydroxy-9β,13α- dimethyl-2-oxo-24,25,26- trinoroleana-1(10),3,5,7- tetraen-29-oic acid (Celastrol) 34157-83-0 10.6 (4S,4aR,5S,5aR,6S,12aS)-4- (dimethylamino)- 3,5,6,10,11,12a- hexahydroxy-6-methyl- 1,12-dioxo- 1,4,4a,5,5a,6,12,12a- octahydrotetracene-2- carboxamide (Oxytetracycline) 79-57-2  7.9 (RS)-2-amino-3-hydroxy-N′- (2,3,4- trihydroxybenzyl)propane- hydrazide hydrochloride (Benserazide Hydrochloride) 322-35-0

Additional candidate compounds were identified with the fluorescence polarization assay and Epic label-free binding assay using a soluble fragment of TSG101 (SEQ ID NO:33) as the target. These candidate compounds and their properties are presented in Table IV. The fluorescence-based thermal shift assay is another exemplary binding assay for identifying lead candidates (FIG. 7).

TABLE IV Candidate compounds that inhibit PTAP-motif interactions with TSG101 (SEQ ID NO: 33) IUPAC Name (Common Name) CAS Reg. No. Structure 5-methoxy-2-[(R)-[(4- methoxy-3,5- dimethylpyridin-2- yl)methane]sulfinyl]-1H-1, 3-benzodiazole, potassium (Esomeprazole potassium) 161796-78-7 (RS)-3-Methoxy-8-[(4- methoxy-3,5-dimethyl- pyridin-2-yl)methylsulfinyl]- 2,7,9- triazabicyclo[4.3.0]nona- 2,4,8,10-tetracene (Tenatoprazole) 113712-98-4 2-Phenyl-1,2- benzoselenazol-3-one (Ebselen) 60940-34-3 (4E)-5-Methyl-2-phenyl-4- {[(2,4,6-tribromophenyl)- amino]methylene}-2,4- dihydro-3H-pyrazole-3- thione [4-{[(4-Methylphenyl)- sulfonyl]amino}-3,6- dihydro-1,3,5-triazin-1(2H)- yl]acetic acid

These compounds were further evaluated as antiviral inhibitors of virus budding and release using the methods disclosed herein. Cultured T cells contacted with either Benserazide Hydrochloride (K21) or Oxytetracycline (N20) [candidate PY-binding motif inhibitors against the Nedd 4 peptide family members] and then subsequently infected with HIV-1 displayed a significant reduction in HIV-1 particle release, as adjudged by detection of HIV-1 CA protein in the cell culture media by ELISA (see Table V).

TABLE V Activity of compounds to Inhibit HTV-1 release from T-Cells in culture HIV-1 CA protein Compound [Compound]1 in culture media2 None  0 μM 3.2 ng/mL Benserazide Hydrochloride 100 μM  0.46 ng/mL  Oxytetracycline 20 μM 1.9 ng/mL 1Indicated inhibitor concentrations tested are not toxic to the contacted cells. 2Results averaged for six experiments, as detected by ELISA.

Vero cells were inoculated with HSV-1 (strain F) at a multiplicity of infection (MOI) of 0.01 for 2 hr. The cells were washed and treated with 0.10 M sodium citrate buffer to inactivate viruses on the outside of the cells (for example, in the culture medium). The culture medium was replaced with fresh culture medium containing no inhibitor compound or different concentrations (40 μM or 80 μM) of inhibitor compounds F15 or N16 (candidate PTAP motif inhibitors against the TSG101 peptide [SEQ ID NO:33]) for 48 hr. The supernatants (culture medium) and total cells (culture medium and cells) were collected for determining infectious virus titer. The virus titers were determined by standard plaque assay on Vero cells.

As shown in FIG. 8A, inhibitor compound F15 (Esomeprazole potassium) reduced infectious HSV-1 virion release from HSV-1 infected cells by more than 90% as compared to infected cells not contacted with an inhibitor compound at the higher concentration tested (80 μM) Inhibitor compound F15 (Esomeprazole potassium) also reduced the total load of infectious virions produced in the cells, whether released or not from the cells, by more than 75% relative to that observed with infected cells not contacted with an inhibitor compound at the higher concentration tested (80 μM).

Still referring to FIG. 8A, inhibitor compound N16 (Tenatoprazole) reduced infectious HSV-1 virion release from HSV-1 infected cells by more than 95% as compared to infected cells not contacted with an inhibitor compound at the higher concentration tested (80 μM). Thus, inhibitor compound N16 was slightly more effective than F15 at reducing infectious virion particle release from HSV-1 infected cells at the concentrations tested.

Furthermore, the inhibitor compounds disrupted infectious particle assembly for virions that remained associated with cells in an unreleased state (FIG. 8A). Thus, the inhibitor compounds F15 and N16 were demonstrated to inhibit HSV-1 virion release from cells.

To rule out the possibility that the reduced infectious virion production was attributed to the inhibitor compounds exhibiting a general cytotoxic effect on the host cells, cytotoxicity assays were performed on uninfected cells, wherein the cells were contacted with culture medium containing the inhibitor compounds in a concentration range from 0 μM to 100 μM. As shown in FIG. 8B, the two evaluated inhibitor compounds, F15 and N16, did not display cytotoxic effects on the Vero cells at the concentrations tested.

Thus, the methods disclosed herein can provide compounds having antiviral activity for inhibiting envelope virus release from cells. Moreover, the methods provided herein can identify compounds having antiviral activity for inhibiting formation of or disrupting an associative complex, wherein the associative complex comprises an isolated enveloped virus L-domain motif and at least one isolated cellular polypeptide, or fragment thereof, capable of binding the isolated virus L-domain motif. Furthermore the disclosed methods can yield compounds having binding affinity for at least one isolated cellular polypeptide, or fragment thereof, capable of binding the isolated virus L-domain motif.

Pharmaceutical Compositions

Pharmaceutical compositions comprising a compound having an antiviral activity for inhibiting release of an enveloped virus from a cell are contemplated herein. Such compositions can include pharmaceutically acceptable carrier. Such carriers are amenable for enhancing one or more ADME characteristics, including solubility and bioavailability of compound inhibitors in physiologically acceptable or suitable media systems or biological fluids. For example, compound inhibitors having poor solubility can be encapsulated in nanoparticles or vesicles comprising at least one micelle-forming lipid. Examples of such delivery systems are disclosed in the literature, as exemplified by one or more of the following citations, the contents of each of which are hereby incorporated by reference in their entireties: U.S. Pat. Publication No. US2002/0099164 A1 to Watterson et al.; U.S. Pat. Publication No. US2008/0008749 A1 to Pearlman et al.; and U.S. Pat. Publication No. US2013/0164379 A1 to Gartel et al.

Exemplary pharmaceutical compositions are well known in the art and fully amenable for use with the present compound inhibitors described herein. See, for example, U.S. Pat. No. 8,202,553 to Lan et al., the contents of which are hereby incorporated by reference in its entirety.

EXAMPLES Example 1 Materials and Methods Expression and Purification of Nedd4 Related Proteins.

Construction and Purification of WWP2 Protein.

The WWP2 encoding gene was excised by EcoRl and Xhol (NEB) cleavage from WWP2_pCNA3.1 construct and was ligated to pET28b(+) His-tag plasmid (Novagen) by using T4 DNA ligase (NEB) in 50 mM Tris-HCL (pH 7.5), 10 mM MgCl2, 10 mM dithiothreitol, 1 mM ATP, 25 mg/ml bovine serum albumin at 16° C. overnight. E. coli. BL21 DE3 cells (Invitrogen) were transformed with WWP2_pET28b and were expressed protein by 0.1 mM (final) IPTG induction for 3 hrs at 25° C. His-tagged WWP2 proteins were purified with His-Bind column (Novagen). Proteins were purified by manufacturer's protocol. Cells were lysed by sonication for 2 min at 4° C. in the presence of 1× binding buffer (20 mM Tris-HCl, 0.5 M NaCl, 5 mM Imidazole, pH 7.9) with a proteinase inhibitor cocktail-EDTA free (Roche), 0.1% NP40, and 1 mM PMSF. The cell debris and inclusion bodies were pelleted by centrifugation at 9000 rpm for 8 min. The supernatant fraction was then passed through a 0.45 μm filter and loaded onto a Ni+2-NTA His-Bind column at 4° C. The column was washed with Binding buffer (10 times bed volume of resin) followed by an equivalent amount of Wash buffer (20 mM Tris-HCl, 0.5 M NaCl, 60 mM Imidazole, pH 7.9). Poly H is containing protein could be eluted with Elute buffer (20 mM Tris-HCl, 0.5 M NaCl, 1 M Imidazole, pH 7.9) and dialyzes against 1× binding buffer without imidazole.

However, to remove the poly H is sequence to yield a native protein, the column was instead washed with 10 times the bed volume with thrombin cleavage buffer (20 mM Tris-HCl, pH7.5, 150 mM NaCl, 2.5 mM CaCl2). Then 1 bed volume of biotinylated thrombin solution (lu/mg of protein, Novagen) was added and the column incubated at room temperature overnight. Untagged proteins were eluted by thrombin cleavage buffer. The biotinylated thrombins were cleared by steptaviding agarose (supplied in the Novagen Thrombin cleavage kit) using a ratio of 16-μl resin per unit of enzyme. Finally, proteins were dialyzed with 1× binding buffer without imidazole. Uncleaved protein could be recovered from the column as above. The nucleotide and amino acid sequence of the WWP2 recombinant protein are shown in Table VI.

TABLE VI Nucleotide and amino acid sequences for WWP2 recombinant protein SEQ ID NO: Sequence1 23 gaattcggcttcgggatccaccATGGATTACAAGGATGACGACGATAAGATGGCATCTG CCAGCTCTAGCCGGGCAGGAGTGGCCCTGCCTTTTGAGAAGTCTCAGCT CACTTTGAAAGTGGTGTCCGCAAAGCCCAAGGTGCATAATCGTCAACC TCGAATTAACTCCTACGTGGAGGTGGCGGTGGATGGACTCCCCAGTGA GACCAAGAAGACTGGGAAGCGCATTGGGAGCTCTGAGCTTCTCTGGAA TGAGATCATCATTTTGAATGTCACGGCACAGAGTCATTTAGATTTAAAG GTCTGGAGCTGCCATACCTTGAGAAATGAACTGCTAGGCACCGCATCT GTCAACCTCTCCAACGTCTTGAAGAACAATGGGGGCAAAATGGAGAAC ATGCAGCTGACCCTGAACCTGCAGACGGAGAACAAAGGCAGCGTTGTC TCAGGCGGAGAGCTGACAATTTTCCTGGACGGGCCAACTGTTGATCTG GGAAATGTGCCTAATGGCAGTGCCCTGACAGATGGATCACAGCTGCCT TCGAGAGACTCCAGTGGAACAGCAGTAGCTCCAGAGAACCGGCACCAG CCCCCCAGCACAAACTGCTTTGGTGGAAGATCCCGGACGCACAGACAT TCGGGTGCTTCAGCCAGAACAACCCCAGCAACCGGCGAGCAAAGCCCC GGTGCTCGGAGCCGGCACCGCCAGCCCGTCAAGAACTCAGGCCACAGT GGCTTGGCCAATGGCACAGTGAATGATGAACCCACAACAGCCACTGAT CCCGAAGAACCTTCCGTTGTTGGTGTGACGTCCCCACCTGCTGCACCCT TGAGTGTGACCCCGAATCCCAACACGACTTCTCTCCCTGCCCCAGCCAC ACCGGCTGAAGGAGAGGAACCCAGCACTTCGGGTACACAGCAGCTCCC AGCGGCTGCCCAGGCCCCCGACGCTCTGCCTGCTGGATGGGAACAGCG AGAGCTGCCCAACGGACGTGTCTATTATGTTGACCACAATACCAAGAC CACCACCTGGGAGCGGCCCCTTCCTCCAGGCTGGGAAAAACGCACAGA TCCCCGAGGCAGGTTTTACTATGTGGATCACAATACTCGGACCACCACC TGGCAGCGTCCGACCGCGGAGTACGTGCGCAACTATGAGCAGTGGCAG TCGCAGCGGAATCAGCTCCAGGGGGCCATGCAGCACTTCAGCCAAAGA TTCCTCTACCAGTCTTCGAGTGCTTCGACTGACCATGATCCCCTGGGCC CCCTCCCTCCTGGCTGGGAGAAGAGACAGGACAATGGACGGGTGTATT ACGTGAACCATAACACTCGCACGACCCAGTGGGAGGATCCCCGGACCC AGGGGATGATCCAGGAACCAGCTCTGCCCCCAGGATGGGAGATGAAAT ACACCAGCGAGGGGGTGCGATACTTTGTGGACCACAATACCCGCACCA CCACCTTTAAGGATCCTCGCCCGGGGTTTGAGTCGGGGACGAAGCAAG GTTCCCCTGGTGCTTATGACCGCAGTTTTCGGTGGAAGTATCACCAGTT CCGTTTCCTCTGCCATTCAAATGCCCTACCTAGCCACGTGAAGATCAGC GTTTCCAGGCAGACGCTTTTCGAAGATTCCTTCCAACAGATCATGAACA TGAAACCCTATGACCTGCGCCGCCGGCTCTACATCATCATGCGTGGCGA GGAGGGCCTGGACTATGGGGGCATCGCCAGAGAGTGGTTTTTCCTCCT GTCTCATGAGGTGCTCAACCCTATGTATTGTTTATTTGAATATGCCGGA AAGAACAATTACTGCCTGCAGATCAACCCCGCCTCCTCCATCAACCCG GACCACCTCACCTACTTTCGCTTTATAGGCAGATTCATCGCCATGGCGC TGTACCATGGAAAGTTCATCGACACGGGCTTCACCCTCCCTTTCTACAA GCGGATGCTCAATAAGAGACCAACCCTGAAAGACCTGGAGTCCATTGA CCCTGAGTTCTACAACTCCATTGTCTGGATCAAAGAGAACAACCTGGA AGAATGTGGCCTGGAGCTGTACTTCATCCAGGACATGGAGATACTGGG CAAGGTGACGACCCACGAGCTGAAGGAGGGCGGCGAGAGCATCCGGG TCACAGAGGAGAACAAGGAAGAGTACATCATGCTGCTGACTGACTGGC GTTTCACCCGAGGCGTGGAAGAGCAGACCAAAGCCTTCCTGGATGGCT TCAACGAGGTGGCCCCGCTGGAGTGGCTGCGCTACTTTGACGAGAAAG AGCTGGAGCTGATGCTGTGCGGCATGCAGGAGATAGACATGAGCGACT GGCAGAAGAGCACCATCTACCGGCACTACACCAAGAACAGCAAGCAG ATCCAGTGGTTCTGGCAGGTGGTGAAGGAGATGGACAACGAGAAGAG GATCCGGCTGCTGCAGTTTGTCACCGGTACCTGCCGCCTGCCCGTCGGG GGATTTGCCGAACTCATCGGTAGCAACGGACCACAGAAGTTTTGCATT GACAAAGTTGGCAAGGAAACCTGGCTGCCCAGAAGCCACACCTGCTTC AACCGTCTGGATCTTCCACCCTACAAGAGCTACGAACAGCTGAGAGAG AAGCTGCTGTATGCCATTGAGGAGACCGAGGGCTTTGGACAGGAGTAA ctcgag 24 MDYKDDDDKMASASSSRAGVALPFEKSQLTLKVVSAKPKVHNRQPRINSYVEVAVDGLPS ETKKTGKRIGSSELLWNEIIILNVTAQSHLDLKVWSCHTLRNELLGTASVNLSNVLKNNG GKMENMQLTLNLQTENKGSVVSGGELTIFLDGPTVDLGNVPNGSALTDGSQLPSRDSSGT AVAPENRHQPPSTNCFGGRSRTHRHSGASARTTPATGEQSPGARSRHRQPVKNSGHSGLA NGTVNDEPTTATDPEEPSVVGVTSPPAAPLSVTPNPNTTSLPAPATPAEGEEPSTSGTQQ LPAAAQAPDALPAGWEQRELPNGRVYYVDHNTKTTTWERPLPPGWEKRTDPRGRFYYVDH NTRTTTWQRPTAEYVRNYEQWQSQRNQLQGAMQHFSQRFLYQSSSASTDHDPLGPLPPGW EKRQDNGRVYYVNHNTRTTQWEDPRTQGMIQEPALPPGWEMKYTSEGVRYFVDHNTRTTT FKDPRPGFESGTKQGSPGAYDRSFRWKYHQFRFLCHSNALPSHVKISVSRQTLFEDSFQQ IMNMKPYDLRRRLYIIMRGEEGLDYGGIAREWFFLLSHEVLNPMYCLFEYAGKNNYCLQI NPASSINPDHLTYFRFIGRFIAMALYHGKFIDTGFTLPFYKRMLNKRPTLKDLESIDPEF YNSIVWIKENNLEECGLELYFIQDMEILGKVTTHELKEGGESIRVTEENKEEYIMLLTDW RFTRGVEEQTKAFLDGFNEVAPLEWLRYFDEKELELMLCGMQEIDMSDWQKSTIYRHYTK NSKQTQWFWQVVKEMDNEKRIRLLQFVTGTCRLPVGGFAELIGSNGPQKFCIDKVGKETW LPRSHTCFNRLDLPPYKSYEQLREKLLYAIEETEGFGQE 1The upper case nucleotide sequence denotes the recombinant polypeptide coding sequence, wherein the italicized sequence encodes the FLAG tag epitope. The underlined nucleotide sequences at the 5′- and 3′-termini of the nucleotide sequence correspond to the EcoRI and XhoI restriction site sequences for introducing the recombinant insert into the pET28b vector.

The complete sequence of the pET28b expression vector that includes the WWP2 recombinant protein is presented below.

SEQ ID NO: 25: TGGCGAATGGGACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGC GTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCG CCACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAG TGCTTTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCCATCG CCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGT TCCAAACTGGAACAACACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCC GATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTTTAACAAA ATATTAACGTTTACAATTTCAGGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTT TATTTTTCTAAATACATTCAAATATGTATCCGCTCATGAATTAATTCTTAGAAAAACTCATCGA GCATCAAATGAAACTGCAATTTATTCATATCAGGATTATCAATACCATATTTTTGAAAAAGCCG TTTCTGTAATGAAGGAGAAAACTCACCGAGGCAGTTCCATAGGATGGCAAGATCCTGGTATCGG TCTGCGATTCCGACTCGTCCAACATCAATACAACCTATTAATTTCCCCTCGTCAAAAATAAGGT TATCAAGTGAGAAATCACCATGAGTGACGACTGAATCCGGTGAGAATGGCAAAAGTTTATGCAT TTCTTTCCAGACTTGTTCAACAGGCCAGCCATTACGCTCGTCATCAAAATCACTCGCATCAACC AAACCGTTATTCATTCGTGATTGCGCCTGAGCGAGACGAAATACGCGATCGCTGTTAAAAGGAC AATTACAAACAGGAATCGAATGCAACCGGCGCAGGAACACTGCCAGCGCATCAACAATATTTTC ACCTGAATCAGGATATTCTTCTAATACCTGGAATGCTGTTTTCCCGGGGATCGCAGTGGTGAGT AACCATGCATCATCAGGAGTACGGATAAAATGCTTGATGGTCGGAAGAGGCATAAATTCCGTCA GCCAGTTTAGTCTGACCATCTCATCTGTAACATCATTGGCAACGCTACCTTTGCCATGTTTCAG AAACAACTCTGGCGCATCGGGCTTCCCATACAATCGATAGATTGTCGCACCTGATTGCCCGACA TTATCGCGAGCCCATTTATACCCATATAAATCAGCATCCATGTTGGAATTTAATCGCGGCCTAG AGCAAGACGTTTCCCGTTGAATATGGCTCATAACACCCCTTGTATTACTGTTTATGTAAGCAGA CAGTTTTATTGTTCATGACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACCC CGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTGCAA ACAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTC CGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTT AGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCA GTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGG ATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAGCGAACGAC CTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCGCCACGCTTCCCGAAGGGAGA AAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCAG GGGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATT TTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGG TTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGTGG ATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAG CGAGTCAGTGAGCGAGGAAGCGGAAGAGCGCCTGATGCGGTATTTTCTCCTTACGCATCTGTGC GGTATTTCACACCGCATATATGGTGCACTCTCAGTACAATCTGCTCTGATGCCGCATAGTTAAG CCAGTATACACTCCGCTATCGCTACGTGACTGGGTCATGGCTGCGCCCCGACACCCGCCAACAC CCGCTGACGCGCCCTGACGGGCTTGTCTGCTCCCGGCATCCGCTTACAGACAAGCTGTGACCGT CTCCGGGAGCTGCATGTGTCAGAGGTTTTCACCGTCATCACCGAAACGCGCGAGGCAGCTGCGG TAAAGCTCATCAGCGTGGTCGTGAAGCGATTCACAGATGTCTGCCTGTTCATCCGCGTCCAGCT CGTTGAGTTTCTCCAGAAGCGTTAATGTCTGGCTTCTGATAAAGCGGGCCATGTTAAGGGCGGT TTTTTCCTGTTTGGTCACTGATGCCTCCGTGTAAGGGGGATTTCTGTTCATGGGGGTAATGATA CCGATGAAACGAGAGAGGATGCTCACGATACGGGTTACTGATGATGAACATGCCCGGTTACTGG AACGTTGTGAGGGTAAACAACTGGCGGTATGGATGCGGCGGGACCAGAGAAAAATCACTCAGGG TCAATGCCAGCGCTTCGTTAATACAGATGTAGGTGTTCCACAGGGTAGCCAGCAGCATCCTGCG ATGCAGATCCGGAACATAATGGTGCAGGGCGCTGACTTCCGCGTTTCCAGACTTTACGAAACAC GGAAACCGAAGACCATTCATGTTGTTGCTCAGGTCGCAGACGTTTTGCAGCAGCAGTCGCTTCA CGTTCGCTCGCGTATCGGTGATTCATTCTGCTAACCAGTAAGGCAACCCCGCCAGCCTAGCCGG GTCCTCAACGACAGGAGCACGATCATGCGCACCCGTGGGGCCGCCATGCCGGCGATAATGGCCT GCTTCTCGCCGAAACGTTTGGTGGCGGGACCAGTGACGAAGGCTTGAGCGAGGGCGTGCAAGAT TCCGAATACCGCAAGCGACAGGCCGATCATCGTCGCGCTCCAGCGAAAGCGGTCCTCGCCGAAA ATGACCCAGAGCGCTGCCGGCACCTGTCCTACGAGTTGCATGATAAAGAAGACAGTCATAAGTG CGGCGACGATAGTCATGCCCCGCGCCCACCGGAAGGAGCTGACTGGGTTGAAGGCTCTCAAGGG CATCGGTCGAGATCCCGGTGCCTAATGAGTGAGCTAACTTACATTAATTGCGTTGCGCTCACTG CCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGA GAGGCGGTTTGCGTATTGGGCGCCAGGGTGGTTTTTCTTTTCACCAGTGAGACGGGCAACAGCT GATTGCCCTTCACCGCCTGGCCCTGAGAGAGTTGCAGCAAGCGGTCCACGCTGGTTTGCCCCAG CAGGCGAAAATCCTGTTTGATGGTGGTTAACGGCGGGATATAACATGAGCTGTCTTCGGTATCG TCGTATCCCACTACCGAGATATCCGCACCAACGCGCAGCCCGGACTCGGTAATGGCGCGCATTG CGCCCAGCGCCATCTGATCGTTGGCAACCAGCATCGCAGTGGGAACGATGCCCTCATTCAGCAT TTGCATGGTTTGTTGAAAACCGGACATGGCACTCCAGTCGCCTTCCCGTTCCGCTATCGGCTGA ATTTGATTGCGAGTGAGATATTTATGCCAGCCAGCCAGACGCAGACGCGCCGAGACAGAACTTA ATGGGCCCGCTAACAGCGCGATTTGCTGGTGACCCAATGCGACCAGATGCTCCACGCCCAGTCG CGTACCGTCTTCATGGGAGAAAATAATACTGTTGATGGGTGTCTGGTCAGAGACATCAAGAAAT AACGCCGGAACATTAGTGCAGGCAGCTTCCACAGCAATGGCATCCTGGTCATCCAGCGGATAGT TAATGATCAGCCCACTGACGCGTTGCGCGAGAAGATTGTGCACCGCCGCTTTACAGGCTTCGAC GCCGCTTCGTTCTACCATCGACACCACCACGCTGGCACCCAGTTGATCGGCGCGAGATTTAATC GCCGCGACAATTTGCGACGGCGCGTGCAGGGCCAGACTGGAGGTGGCAACGCCAATCAGCAACG ACTGTTTGCCCGCCAGTTGTTGTGCCACGCGGTTGGGAATGTAATTCAGCTCCGCCATCGCCGC TTCCACTTTTTCCCGCGTTTTCGCAGAAACGTGGCTGGCCTGGTTCACCACGCGGGAAACGGTC TGATAAGAGACACCGGCATACTCTGCGACATCGTATAACGTTACTGGTTTCACATTCACCACCC TGAATTGACTCTCTTCCGGGCGCTATCATGCCATACCGCGAAAGGTTTTGCGCCATTCGATGGT GTCCGGGATCTCGACGCTCTCCCTTATGCGACTCCTGCATTAGGAAGCAGCCCAGTAGTAGGTT GAGGCCGTTGAGCACCGCCGCCGCAAGGAATGGTGCATGCAAGGAGATGGCGCCCAACAGTCCC CCGGCCACGGGGCCTGCCACCATACCCACGCCGAAACAAGCGCTCATGAGCCCGAAGTGGCGAG CCCGATCTTCCCCATCGGTGATGTCGGCGATATAGGCGCCAGCAACCGCACCTGTGGCGCCGGT GATGCCGGCCACGATGCGTCCGGCGTAGAGGATCGAGATCTCGATCCCGCGAAATTAATACGAC TCACTATAGGGGAATTGTGAGCGGATAACAATTCCCCTCTAGAAATAATTTTGTTTAACTTTAA GAAGGAGATATACCATGGGCAGCAGCCATCATCATCATCATCACAGCAGCGGCCTGGTGCCGCG CGGCAGCCATATGGCTAGCATGACTGGTGGACAGCAAATGGGTCGCGGATCCGAATTCggcttc gggatccaccATGGATTACAAGGATGACGACGATAAGatggcatctgccagctctagccgggca ggagtggccctgccttttgagaagtctcagctcactttgaaagtggtgtccgcaaagcccaagg tgcataatcgtcaacctcgaattaactcctacgtggaggtggcggtggatggactccccagtga gaccaagaagactgggaagcgcattgggagctctgagcttctctggaatgagatcatcattttg aatgtcacggcacagagtcatttagatttaaaggtctggagctgccataccttgagaaatgaac tgctaggcaccgcatctgtcaacctctccaacgtcttgaagaacaatgggggcaaaatggagaa catgcagctgaccctgaacctgcagacggagaacaaaggcagcgttgtctcaggcggagagctg acaattttcctggacgggccaactgttgatctgggaaatgtgcctaatggcagtgccctgacag atggatcacagctgccttcgagagactccagtggaacagcagtagctccagagaaccggcacca gccccccagcacaaactgctttggtggaagatcccggacgcacagacattcgggtgcttcagcc agaacaaccccagcaaccggcgagcaaagccccggtgctcggagccggcaccgccagcccgtca agaactcaggccacagtggcttggccaatggcacagtgaatgatgaacccacaacagccactga tcccgaagaaccttccgttgttggtgtgacgtccccacctgctgcacccttgagtgtgaccccg aatcccaacacgacttctctccctgccccagccacaccggctgaaggagaggaacccagcactt cgggtacacagcagctcccagcggctgcccaggcccccgacgctctgcctgctggatgggaaca gcgagagctgcccaacggacgtgtctattatgttgaccacaataccaagaccaccacctgggag cggccccttcctccaggctgggaaaaacgcacagatccccgaggcaggttttactatgtggatc acaatactcggaccaccacctggcagcgtccgaccgcggagtacgtgcgcaactatgagcagtg gcagtcgcagcggaatcagctccagggggccatgcagcacttcagccaaagattcctctaccag tcttcgagtgcttcgactgaccatgatcccctgggccccctccctcctggctgggagaagagac aggacaatggacgggtgtattacgtgaaccataacactcgcacgacccagtgggaggatccccg gacccaggggatgatccaggaaccagctctgcccccaggatgggagatgaaatacaccagcgag ggggtgcgatactttgtggaccacaatacccgcaccaccacctttaaggatcctcgcccggggt ttgagtcggggacgaagcaaggttcccctggtgcttatgaccgcagttttcggtggaagtatca ccagttccgtttcctctgccattcaaatgccctacctagccacgtgaagatcagcgtttccagg cagacgcttttcgaagattccttccaacagatcatgaacatgaaaccctatgacctgcgccgcc ggctctacatcatcatgcgtggcgaggagggcctggactatgggggcatcgccagagagtggtt tttcctcctgtctcatgaggtgctcaaccctatgtattgtttatttgaatatgccggaaagaac aattactgcctgcagatcaaccccgcctcctccatcaacccggaccacctcacctactttcgct ttataggcagattcatcgccatggcgctgtaccatggaaagttcatcgacacgggcttcaccct ccctttctacaagcggatgctcaataagagaccaaccctgaaagacctggagtccattgaccct gagttctacaactccattgtctggatcaaagagaacaacctggaagaatgtggcctggagctgt acttcatccaggacatggagatactgggcaaggtgacgacccacgagctgaaggagggcggcga gagcatccgggtcacagaggagaacaaggaagagtacatcatgctgctgactgactggcgtttc acccgaggcgtggaagagcagaccaaagccttcctggatggcttcaacgaggtggccccgctgg agtggctgcgctactttgacgagaaagagctggagctgatgctgtgcggcatgcaggagataga catgagcgactggcagaagagcaccatctaccggcactacaccaagaacagcaagcagatccag tggttctggcaggtggtgaaggagatggacaacgagaagaggatccggctgctgcagtttgtca ccggtacctgccgcctgcccgtcgggggatttgccgaactcatcggtagcaacggaccacagaa gttttgcattgacaaagttggcaaggaaacctggctgcccagaagccacacctgcttcaaccgt ctggatcttccaccctacaagagctacgaacagctgagagagaagctgctgtatgccattgagg agaccgagggctttggacaggagtaaCTCGAGCACCACCACCACCACCACTGAGATCCGGCTGC TAACAAAGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACCC CTTGGGGCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGGAACTATATCCGGAT

wherein the bold font denote the locations of the initiator and terminator codons for the recombinant peptide pro-form; the upper case letters denote pET28b vector sequences; the lower case letter denote the WWP2 coding sequences, the single-underlined sequences are the poly-His coding sequences, the double-underlined sequences encode the Thrombin cleavage site, the italicized font includes the FLAG-tag coding sequences in frame with the WWP2 coding sequences as configured in the expression vector, pET28b (Novagen).

Construction and Preparation of WWP1 Protein.

The WWP1 encoding gene was amplified by PCR reaction from FLAG-WWP1_pCNA3.1 construct and was ligated to pET28b(+) His-tag plasmid (Novagen). His-tagged WWP1 plasmids were transformed into BL21 DE3 cells. Protein expression and purification were followed the procedure of WWP2 protein preparation. The WWP1 encoding gene was amplified by PCR reaction using Deep Vent polymerase (NEB) from the FLAG-WWP1_pCNA3.1 construct using the oligodeoxynucleotides 5′-ATAGGATTCATGGCCACTGCTTCACCAAGGTCT-3′ forward primer (SEQ ID NO:26) and 5′-ATAGCGGCCGCTCATTCTTGTCCAAATCCCTCTGT-3′ reverse primer (SEQ ID NO:27) and was ligated to pET28b(+) His-tag plasmid between the EcoRl and Not cloning sites. The nucleotide and amino acid sequences for WWP1 recombinant protein are illustrated in Table VII.

TABLE VII Nucleotide and amino acid sequences for WWP1 recombinant protein1 SEQ ID NO: Sequence2 28 gaattcATGGCCACTGCTTCACCAAGGTCTGATACTAGTAATAACCACAGT GGAAGGTTGCAGTTACAGGTAACTGTTTCTAGTGCCAAACTTAAAAGA AAAAAGAACTGGTTCGGAACAGCAATATATACAGAAGTAGTTGTAGAT GGAGAAATTACGAAAACAGCAAAATCCAGTAGTTCTTCTAATCCAAAA TGGGATGAACAGCTAACTGTAAATGTTACGCCACAGACTACATTGGAA TTTCAAGTTTGGAGCCATCGCACTTTAAAAGCAGATGCTTTATTAGGAA AAGCAACGATAGATTTGAAACAAGCTCTGTTGATACACAATAGAAAAT TGGAAAGAGTGAAAGAACAATTAAAACTTTCCTTGGAAAACAAGAATG GCATAGCACAAACTGGTGAATTGACAGTTGTGCTTGATGGATTGGTGA TTGAGCAAGAAAATATAACAAACTGCAGCTCATCTCCAACCATAGAAA TACAGGAAAATGGTGATGCCTTACATGAAAATGGAGAGCCTTCAGCAA GGACAACTGCCAGGTTGGCTGTTGAAGGCACGAATGGAATAGATAATC ATGTACCTACAAGCACTCTAGTCCAAAACTCATGCTGCTCGTATGTAGT TAATGGAGACAACACACCTTCATCTCCGTCTCAGGTTGCTGCCAGACCC AAAAATACACCAGCTCCAAAACCACTCGCATCTGAGCCTGCCGATGAC ACTGTTAATGGAGAATCATCCTCATTTGCACCAACTGATAATGCGTCTG TCACGGGTACTCCAGTAGTGTCTGAAGAAAATGCCTTGTCTCCAAATTG CACTAGTACTACTGTTGAAGATCCTCCAGTTCAAGAAATACTGACTTCC TCAGAAAACAATGAATGTATTCCTTCTACCAGTGCAGAATTGGAATCTG AAGCTAGAAGTATATTAGAGCCTGACACCTCTAATTCTAGAAGTAGTTC TGCTTTTGAAGCAGCCAAATCAAGACAGCCAGATGGGTGTATGGATCC TGTACGGCAGCAGTCTGGGAATGCCAACACAGAAACCTTGCCATCAGG GTGGGAACAAAGAAAAGATCCTCATGGTAGAACCTATTATGTGGATCA TAATACTCGAACTACCACATGGGAGAGACCACAACCTTTACCTCCAGG TTGGGAAAGAAGAGTTGATGATCGTAGAAGAGTTTATTATGTGGATCA TAACACCAGAACAACAACGTGGCAGCGGCCTACCATGGAATCTGTCCG AAATTTTGAACAGTGGCAATCTCAGCGGAACCAATTGCAGGGAGCTAT GCAACAGTTTAACCAACGATACCTCTATTCGGCTTCAATGTTAGCTGCA GAAAATGACCCTTATGGACCTTTGCCACCAGGCTGGGAAAAAAGAGTG GATTCAACAGACAGGGTTTACTTTGTGAATCATAACACAAAAACAACC CAGTGGGAAGATCCAAGAACTCAAGGCTTACAGAATGAAGAACCCCTG CCAGAAGGCTGGGAAATTAGATATACTCGTGAAGGTGTAAGGTACTTT GTTGATCATAACACAAGAACAACAACATTCAAAGATCCTCGCAATGGG AAGTCATCTGTAACTAAAGGTGGTCCACAAATTGCTTATGAACGCGGC TTTAGGTGGAAGCTTGCTCACTTCCGTTATTTGTGCCAGTCTAATGCAC TACCTAGTCATGTAAAGATCAATGTGTCCCGGCAGACATTGTTTGAAGA TTCCTTCCAACAGATTATGGCATTAAAACCCTATGACTTGAGGAGGCGC TTATATGTAATATTTAGAGGAGAAGAAGGACTTGATTATGGTGGCCTA GCGAGAGAATGGTTTTTCTTGCTTTCACATGAAGTTTTGAACCCAATGT ATTGCTTATTTGAGTATGCGGGCAAGAACAACTATTGTCTGCAGATAAA TCCAGCATCAACCATTAATCCAGACCATCTTTCATACTTCTGTTTCATTG GTCGTTTTATTGCCATGGCACTATTTCATGGAAAGTTTATCGATACTGG TTTCTCTTTACCATTCTACAAGCGTATGTTAAGTAAAAAACTTACTATT AAGGATTTGGAATCTATTGATACTGAATTTTATAACTCCCTTATCTGGA TAAGAGATAACAACATTGAAGAATGTGGCTTAGAAATGTACTTTTCTGT TGACATGGAGATTTTGGGAAAAGTTACTTCACATGACCTGAAGTTGGG AGGTTCCAATATTCTGGTGACTGAGGAGAACAAAGATGAATATATTGG TTTAATGACAGAATGGCGTTTTTCTCGAGGAGTACAAGAACAGACCAA AGCTTTCCTTGATGGTTTTAATGAAGTTGTTCCTCTTCAGTGGCTACAGT ACTTCGATGAAAAAGAATTAGAGGTTATGTTGTGTGGCATGCAGGAGG TTGACTTGGCAGATTGGCAGAGAAATACTGTTTATCGACATTATACAAG AAACAGCAAGCAAATCATTTGGTTTTGGCAGTTTGTGAAAGAGACAGA CAATGAAGTAAGAATGCGACTATTGCAGTTCGTCACTGGAACCTGCCG TTTACCTCTAGGAGGATTTGCTGAGCTCATGGGAAGTAATGGGCCTCAA AAGTTTTGCATTGAAAAAGTTGGCAAAGACACTTGGTTACCAAGAAGC CATACATGTTTTAATCGCTTGGATCTACCACCATATAAGAGTTATGAAC AACTAAAGGAAAAACTTCTTTTTGCAATAGAAGAGACAGAGGGATTTG GACAAGAATGAgcggccgc 29 MATASPRSDTSNNHSGRLQLQVTVSSAKLKRKKNWFGTAIYTEVVVDGEITKTAKSSSSS NPKWDEQLTVNVTPQTTLEFQVWSHRTLKADALLGKATIDLKQALLIHNRKLERVKEQLK LSLENKNGIAQTGELTVVLDGLVIEQENITNCSSSPTIEIQENGDALHENGEPSARTTAR LAVEGTNGIDNHVPTSTLVQNSCCSYVVNGDNTPSSPSQVAARPKNTPAPKPLASEPADD TVNGESSSFAPTDNASVTGTPVVSEENALSPNCTSTTVEDPPVQEILTSSENNECIPSTS AELESEARSILEPDTSNSRSSSAFEAAKSRQPDGCMDPVRQQSGNANTETLPSGWEQRKD PHGRTYYVDHNTRTTTWERPQPLPPGWERRVDDRRRVYYVDHNTRTTTWQRPTMESVRNF EQWQSQRNQLQGAMQQFNQRYLYSASMLAAENDPYGPLPPGWEKRVDSTDRVYFVNHNTK TTQWEDPRTQGLQNEEPLPEGWEIRYTREGVRYFVDHNTRTTTFKDPRNGKSSVTKGGPQ IAYERGFRWKLAHFRYLCQSNALPSHVKINVSRQTLFEDSFQQIMALKPYDLRRRLYVIF RGEEGLDYGGLAREWFFLLSHEVLNPMYCLFEYAGKNNYCLQINPASTINPDHLSYFCFI GRFIAMALFHGKFIDTGFSLPFYKRMLSKKLTIKDLESIDTEFYNSLIWIRDNNIEECGL EMYFSVDMEILGKVTSHDLKLGGSNILVTEENKDEYIGLMTEWRFSRGVQEQTKAFLDGF NEVVPLQWLQYFDEKELEVMLCGMQEVDLADWQRNTVYRHYTRNSKQIIWFWQFVKETDN EVRMRLLQFVTGTCRLPLGGFAELMGSNGPQKFCIEKVGKDTWLPRSHTCFNRLDLPPYK SYEQLKEKLLFAIEETEGFGQE 1Recombinant protein illustrated without the His-tag sequence present (cleaved off). 2The upper case nucleotide sequence denotes the recombinant polypeptide coding sequence. The underlined nucleotide sequences at the 5′- and 3′-termini of the nucleotide sequence correspond to the EcoRI and NotI restriction site sequences for introducing the recombinant insert into the pET28b vector.

The complete sequence of the pET28b expression vector that includes the WWP1 recombinant protein is presented below.

SEQ ID NO: 30: TGGCGAATGGGACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGC GTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCG CCACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAG TGCTTTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCCATCG CCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGT TCCAAACTGGAACAACACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCC GATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTTTAACAAA ATATTAACGTTTACAATTTCAGGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTT TATTTTTCTAAATACATTCAAATATGTATCCGCTCATGAATTAATTCTTAGAAAAACTCATCGA GCATCAAATGAAACTGCAATTTATTCATATCAGGATTATCAATACCATATTTTTGAAAAAGCCG TTTCTGTAATGAAGGAGAAAACTCACCGAGGCAGTTCCATAGGATGGCAAGATCCTGGTATCGG TCTGCGATTCCGACTCGTCCAACATCAATACAACCTATTAATTTCCCCTCGTCAAAAATAAGGT TATCAAGTGAGAAATCACCATGAGTGACGACTGAATCCGGTGAGAATGGCAAAAGTTTATGCAT TTCTTTCCAGACTTGTTCAACAGGCCAGCCATTACGCTCGTCATCAAAATCACTCGCATCAACC AAACCGTTATTCATTCGTGATTGCGCCTGAGCGAGACGAAATACGCGATCGCTGTTAAAAGGAC AATTACAAACAGGAATCGAATGCAACCGGCGCAGGAACACTGCCAGCGCATCAACAATATTTTC ACCTGAATCAGGATATTCTTCTAATACCTGGAATGCTGTTTTCCCGGGGATCGCAGTGGTGAGT AACCATGCATCATCAGGAGTACGGATAAAATGCTTGATGGTCGGAAGAGGCATAAATTCCGTCA GCCAGTTTAGTCTGACCATCTCATCTGTAACATCATTGGCAACGCTACCTTTGCCATGTTTCAG AAACAACTCTGGCGCATCGGGCTTCCCATACAATCGATAGATTGTCGCACCTGATTGCCCGACA TTATCGCGAGCCCATTTATACCCATATAAATCAGCATCCATGTTGGAATTTAATCGCGGCCTAG AGCAAGACGTTTCCCGTTGAATATGGCTCATAACACCCCTTGTATTACTGTTTATGTAAGCAGA CAGTTTTATTGTTCATGACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACCC CGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTGCAA ACAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTC CGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTT AGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCA GTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGG ATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAGCGAACGAC CTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCGCCACGCTTCCCGAAGGGAGA AAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCAG GGGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATT TTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGG TTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGTGG ATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAG CGAGTCAGTGAGCGAGGAAGCGGAAGAGCGCCTGATGCGGTATTTTCTCCTTACGCATCTGTGC GGTATTTCACACCGCATATATGGTGCACTCTCAGTACAATCTGCTCTGATGCCGCATAGTTAAG CCAGTATACACTCCGCTATCGCTACGTGACTGGGTCATGGCTGCGCCCCGACACCCGCCAACAC CCGCTGACGCGCCCTGACGGGCTTGTCTGCTCCCGGCATCCGCTTACAGACAAGCTGTGACCGT CTCCGGGAGCTGCATGTGTCAGAGGTTTTCACCGTCATCACCGAAACGCGCGAGGCAGCTGCGG TAAAGCTCATCAGCGTGGTCGTGAAGCGATTCACAGATGTCTGCCTGTTCATCCGCGTCCAGCT CGTTGAGTTTCTCCAGAAGCGTTAATGTCTGGCTTCTGATAAAGCGGGCCATGTTAAGGGCGGT TTTTTCCTGTTTGGTCACTGATGCCTCCGTGTAAGGGGGATTTCTGTTCATGGGGGTAATGATA CCGATGAAACGAGAGAGGATGCTCACGATACGGGTTACTGATGATGAACATGCCCGGTTACTGG AACGTTGTGAGGGTAAACAACTGGCGGTATGGATGCGGCGGGACCAGAGAAAAATCACTCAGGG TCAATGCCAGCGCTTCGTTAATACAGATGTAGGTGTTCCACAGGGTAGCCAGCAGCATCCTGCG ATGCAGATCCGGAACATAATGGTGCAGGGCGCTGACTTCCGCGTTTCCAGACTTTACGAAACAC GGAAACCGAAGACCATTCATGTTGTTGCTCAGGTCGCAGACGTTTTGCAGCAGCAGTCGCTTCA CGTTCGCTCGCGTATCGGTGATTCATTCTGCTAACCAGTAAGGCAACCCCGCCAGCCTAGCCGG GTCCTCAACGACAGGAGCACGATCATGCGCACCCGTGGGGCCGCCATGCCGGCGATAATGGCCT GCTTCTCGCCGAAACGTTTGGTGGCGGGACCAGTGACGAAGGCTTGAGCGAGGGCGTGCAAGAT TCCGAATACCGCAAGCGACAGGCCGATCATCGTCGCGCTCCAGCGAAAGCGGTCCTCGCCGAAA ATGACCCAGAGCGCTGCCGGCACCTGTCCTACGAGTTGCATGATAAAGAAGACAGTCATAAGTG CGGCGACGATAGTCATGCCCCGCGCCCACCGGAAGGAGCTGACTGGGTTGAAGGCTCTCAAGGG CATCGGTCGAGATCCCGGTGCCTAATGAGTGAGCTAACTTACATTAATTGCGTTGCGCTCACTG CCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGA GAGGCGGTTTGCGTATTGGGCGCCAGGGTGGTTTTTCTTTTCACCAGTGAGACGGGCAACAGCT GATTGCCCTTCACCGCCTGGCCCTGAGAGAGTTGCAGCAAGCGGTCCACGCTGGTTTGCCCCAG CAGGCGAAAATCCTGTTTGATGGTGGTTAACGGCGGGATATAACATGAGCTGTCTTCGGTATCG TCGTATCCCACTACCGAGATATCCGCACCAACGCGCAGCCCGGACTCGGTAATGGCGCGCATTG CGCCCAGCGCCATCTGATCGTTGGCAACCAGCATCGCAGTGGGAACGATGCCCTCATTCAGCAT TTGCATGGTTTGTTGAAAACCGGACATGGCACTCCAGTCGCCTTCCCGTTCCGCTATCGGCTGA ATTTGATTGCGAGTGAGATATTTATGCCAGCCAGCCAGACGCAGACGCGCCGAGACAGAACTTA ATGGGCCCGCTAACAGCGCGATTTGCTGGTGACCCAATGCGACCAGATGCTCCACGCCCAGTCG CGTACCGTCTTCATGGGAGAAAATAATACTGTTGATGGGTGTCTGGTCAGAGACATCAAGAAAT AACGCCGGAACATTAGTGCAGGCAGCTTCCACAGCAATGGCATCCTGGTCATCCAGCGGATAGT TAATGATCAGCCCACTGACGCGTTGCGCGAGAAGATTGTGCACCGCCGCTTTACAGGCTTCGAC GCCGCTTCGTTCTACCATCGACACCACCACGCTGGCACCCAGTTGATCGGCGCGAGATTTAATC GCCGCGACAATTTGCGACGGCGCGTGCAGGGCCAGACTGGAGGTGGCAACGCCAATCAGCAACG ACTGTTTGCCCGCCAGTTGTTGTGCCACGCGGTTGGGAATGTAATTCAGCTCCGCCATCGCCGC TTCCACTTTTTCCCGCGTTTTCGCAGAAACGTGGCTGGCCTGGTTCACCACGCGGGAAACGGTC TGATAAGAGACACCGGCATACTCTGCGACATCGTATAACGTTACTGGTTTCACATTCACCACCC TGAATTGACTCTCTTCCGGGCGCTATCATGCCATACCGCGAAAGGTTTTGCGCCATTCGATGGT GTCCGGGATCTCGACGCTCTCCCTTATGCGACTCCTGCATTAGGAAGCAGCCCAGTAGTAGGTT GAGGCCGTTGAGCACCGCCGCCGCAAGGAATGGTGCATGCAAGGAGATGGCGCCCAACAGTCCC CCGGCCACGGGGCCTGCCACCATACCCACGCCGAAACAAGCGCTCATGAGCCCGAAGTGGCGAG CCCGATCTTCCCCATCGGTGATGTCGGCGATATAGGCGCCAGCAACCGCACCTGTGGCGCCGGT GATGCCGGCCACGATGCGTCCGGCGTAGAGGATCGAGATCTCGATCCCGCGAAATTAATACGAC TCACTATAGGGGAATTGTGAGCGGATAACAATTCCCCTCTAGAAATAATTTTGTTTAACTTTAA GAAGGAGATATACCATGGGCAGCAGCCATCATCATCATCATCACAGCAGCGGCCTGGTGCCGCG CGGCAGCCATATGGCTAGCATGACTGGTGGACAGCAAATGGGTCGCGGATCCGAATTCatggcc actgcttcaccaaggtctgatactagtaataaccacagtggaaggttgcagttacaggtaactg tttctagtgccaaacttaaaagaaaaaagaactggttcggaacagcaatatatacagaagtagt tgtagatggagaaattacgaaaacagcaaaatccagtagttcttctaatccaaaatgggatgaa cagctaactgtaaatgttacgccacagactacattggaatttcaagtttggagccatcgcactt taaaagcagatgctttattaggaaaagcaacgatagatttgaaacaagctctgttgatacacaa tagaaaattggaaagagtgaaagaacaattaaaactttccttggaaaacaagaatggcatagca caaactggtgaattgacagttgtgcttgatggattggtgattgagcaagaaaatataacaaact gcagctcatctccaaccatagaaatacaggaaaatggtgatgccttacatgaaaatggagagcc ttcagcaaggacaactgccaggttggctgttgaaggcacgaatggaatagataatcatgtacct acaagcactctagtccaaaactcatgctgctcgtatgtagttaatggagacaacacaccttcat ctccgtctcaggttgctgccagacccaaaaatacaccagctccaaaaccactcgcatctgagcc tgccgatgacactgttaatggagaatcatcctcatttgcaccaactgataatgcgtctgtcacg ggtactccagtagtgtctgaagaaaatgccttgtctccaaattgcactagtactactgttgaag atcctccagttcaagaaatactgacttcctcagaaaacaatgaatgtattccttctaccagtgc agaattggaatctgaagctagaagtatattagagcctgacacctctaattctagaagtagttct gcttttgaagcagccaaatcaagacagccagatgggtgtatggatcctgtacggcagcagtctg ggaatgccaacacagaaaccttgccatcagggtgggaacaaagaaaagatcctcatggtagaac ctattatgtggatcataatactcgaactaccacatgggagagaccacaacctttacctccaggt tgggaaagaagagttgatgatcgtagaagagtttattatgtggatcataacaccagaacaacaa cgtggcagcggcctaccatggaatctgtccgaaattttgaacagtggcaatctcagcggaacca attgcagggagctatgcaacagtttaaccaacgatacctctattcggcttcaatgttagctgca gaaaatgacccttatggacctttgccaccaggctgggaaaaaagagtggattcaacagacaggg tttactttgtgaatcataacacaaaaacaacccagtgggaagatccaagaactcaaggcttaca gaatgaagaacccctgccagaaggctgggaaattagatatactcgtgaaggtgtaaggtacttt gttgatcataacacaagaacaacaacattcaaagatcctcgcaatgggaagtcatctgtaacta aaggtggtccacaaattgcttatgaacgcggctttaggtggaagcttgctcacttccgttattt gtgccagtctaatgcactacctagtcatgtaaagatcaatgtgtcccggcagacattgtttgaa gattccttccaacagattatggcattaaaaccctatgacttgaggaggcgcttatatgtaatat ttagaggagaagaaggacttgattatggtggcctagcgagagaatggtttttcttgctttcaca tgaagttttgaacccaatgtattgcttatttgagtatgcgggcaagaacaactattgtctgcag ataaatccagcatcaaccattaatccagaccatctttcatacttctgtttcattggtcgtttta ttgccatggcactatttcatggaaagtttatcgatactggtttctctttaccattctacaagcg tatgttaagtaaaaaacttactattaaggatttggaatctattgatactgaattttataactcc cttatctggataagagataacaacattgaagaatgtggcttagaaatgtacttttctgttgaca tggagattttgggaaaagttacttcacatgacctgaagttgggaggttccaatattctggtgac tgaggagaacaaagatgaatatattggtttaatgacagaatggcgtttttctcgaggagtacaa gaacagaccaaagctttccttgatggttttaatgaagttgttcctcttcagtggctacagtact tcgatgaaaaagaattagaggttatgttgtgtggcatgcaggaggttgacttggcagattggca gagaaatactgtttatcgacattatacaagaaacagcaagcaaatcatttggttttggcagttt gtgaaagagacagacaatgaagtaagaatgcgactattgcagttcgtcactggaacctgccgtt tacctctaggaggatttgctgagctcatgggaagtaatgggcctcaaaagttttgcattgaaaa agttggcaaagacacttggttaccaagaagccatacatgttttaatcgcttggatctaccacca tataagagttatgaacaactaaaggaaaaacttctttttgcaatagaagagacagagggatttg gacaagaatgaGCGGCCGCACTCGAGCACCACCACCACCACCACTGAGATCCGGCTGCTAACAA AGCCCGAAAGGAAGCTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGG GCCTCTAAACGGGTCTTGAGGGGTTTTTTGCTGAAAGGAGGAACTATATCCGGAT

wherein the bold font denote the locations of the initiator and terminator codons for the recombinant peptide pro-form; the upper case letters denote pET28b vector sequences; the lower case letter denote the WWP1 coding sequences, the italicized font includes the leader peptide coding sequence that includes a polyhistidine motif and thrombin cleavage site derived from the expression vector, pET28b (Novagen).

Example 2 Screening for Small Molecule Compounds that Disrupt the Binding of Nedd4 Proteins to the PY Motif

Fluorescent Polarization Assay (FPA) to Detect Binding of the Nedd4 Proteins WWP1/2 to Peptides Containing the PY L-Domain Motif Sequences:

The FPA uses a fluorescein labeled peptide (FTIC-peptide) probe with three tandem PY motif binding units (FITC-ATASAPPPYVGSGGGATASAPPPYVGSGGGATASAPPPYVGSGGGRRR-OH, Biosynthesis, TX (SEQ ID NO: 18); (SEQ ID NO: 17 corresponds to SEQ ID NO: 18 without the N-terminal FITC moiety) derived from the p2 region of avian sarcoma/leucosis virus (ASLV) gag gene. With the tandem probe, a Kd of 0.2 μM was obtained from WWP2 protein dose-response study with highest protein concentration at 19.6 μM and FITC-peptide probe concentration of 5 nM. In contrast, Kd value for the probe with a single binding unit is >15 mM. Screens for compounds that disrupt the binding of the FTIC-peptide and WWP2 (SEQ ID NO:24) were carried out in 384-well plate format with 25 ml assay volume at ˜0.5 mM WWP2. Non-binding solid black plates (Corning Costar 3575) were used. The assay buffer contained 20 mM Tris and 150 mM NaCl at pH 7.9. FP was measured on either a Biotek Synergy4 or an Analyst GT plate reader. Data analysis was performed using the in-house software excelHTS. The typical Z value of a screen plate was greater than 0.7. Each assay plate included 16 negative control wells containing protein-probe mix but no compounds and 16 wells containing only probe. The difference in FP values of these wells was used as 100% readout signal to calculate the percentage of inhibition of a compound well.

The FTIC-labeled probe was premixed at a concentration of 20 nM with WWP1/2 protein at 0.5 mM. Then 25 ml of the protein-probe mix was added to assay plates by using a ViaFill dispenser. A Labcyte Echo550 acoustic transfer robot was used to transfer compounds (30 to 100 nl, equal to 10 to 50 mM) to the assay plates. The plates were shaken to ensure proper assay mix. Then FP was measured on an Analyst GT plate reader equipped with a plate stacker so that data of multiple plates were recorded in a single file to facilitate data reduction and analysis. The first read was performed one hour after compound addition then the plates were sealed and stored at 4° C. overnight before reading again after 18 hours.

Hit Confirmation by TAMRA Probe in Dose-Response Format.

The primary hits include false positives due to experimental error and fluorescent compounds that fluoresce or absorb in the FTIC emission range. After primary screen, a hit confirmation screen was performed using TAMRA-probe in dose-response format to rid false positives. TAMRA excites and emits at a longer wavelength range than FITC. The false positives caused by overlap of excitation/emission wavelengths of FITC and some fluorescent compounds can be delineated.

Example 3 Screening for Small Molecule Compounds that Bind to Nedd4 Proteins

Epic Label-Free Assay Procedure to Measure Binding of Compounds to WWP2/1:

High sensitivity biochemical plates (PerkinElmer Cat. No. 6057468) are used in WWP2 (SEQ ID NO:24) binding assay to maximize protein immobilization. The plate was activated before use with 10 μl of 200 mM EDC(N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride) (Sigma Cat. No. 03449)/50 mM sulfo-NHS(N-hydroxysulfosuccinimide) (Pierce Cat. No. 24510) diluted in H2O. After 30 min incubation at room temperature, the plates are washed 3 times with 25 μl H2O and then centrifuged inverted at 800 RPM for 1 min. WWP2 (SEQ ID NO:24) immobilization was accomplished by adding 10 μl of 80 μg/ml protein in HEPES buffer (100 mM HEPES, 150 mM NaCl, pH 7.5) followed by centrifugation at 800 RPM for 1 minute, and then incubated for 3 hours at room temperature or overnight at 4° C. Plates were then washed three times with assay buffer (HEPES buffer with 0.5% DMSO) and then followed with a final volume of 15 μl assay buffer. The binding assay was performed in 4 or 6-point dose-response format. Testing compounds were diluted with assay buffer in 384-well compound plates. Because compound stocks are in DMSO that affects the Epic label-free readout, the compound plates were prepared by Echo550 acoustic dispenser to keep DMSO content balanced for all wells. Compounds were then transferred to assay plates by a multichannel robotic liquid handler. An alternative approach was to add compounds in dose-curve format directly to assay plates using the nanoliter dispenser Echo550.

The binding of compounds to WWP2 (SEQ ID NO:24) was measured on the EnSpire label-free plate reader. After 25 minutes of thermal equilibration, a baseline reading was taken on the label-free plates with protein immobilized in assay buffer. In the next step, 25 μl of reconstituted compounds were added to the plates and mixed. The final reading was taken after 25 min of thermal equilibration and over a period of 60 minutes. The label-free responses were measured as shifts in reflected wavelength and were expressed in picometers (pm). Results were analyzed using the EnSpire label-free user interface software. The difference between the last baseline measurements and the signal max was used to determine the binding of compounds to WWP2.

Example 4 Expression of Soluble TSG101 Fragment Containing PTAP-Motif Binding Domain

The full-length nucleotide and amino acid sequence of TSG101 is presented in Table VIII. The full-length TSG101 polypeptide (SEQ ID NO:32) is not sufficiently soluble for performing the in vitro screening assays disclosed herein. Accordingly, a truncated TSG101 peptide that contains amino acids corresponding to positions 2-145 of the full-length TSG101 peptide and includes a PTAP-binding domain was prepared having sufficient solubility for these in vitro screening assays. The resultant TSG101 peptide (SEQ ID NO:33) used for these studies is illustrated in Table VIII.

TABLE VIII Nucleotide and amino acid sequences of full-length TSG101 peptide and expression construct for UEV domain of truncated-length TSG101 recombinant peptide SEQ ID NO: Sequence1 31 gaagcggaag tggtgtagtg gtgccgactt cctgttgttt gaggccgggt tgggggtgtg cgattgtgtg ggacggtctg gggcagccca gcagcggctg accctctgcc tgcggggaag ggagtcgcca ggcggccgtc ATGgcggtgt cggagagcca gctcaagaaa atggtgtcca agtacaaata cagagaccta actgtacgtg aaactgtcaa tgttattact ctatacaaag atctcaaacc tgttttggat tcatatgttt ttaacgatgg cagttccagg gaactaatga acctcactgg aacaatccct gtgccttata gaggtaatac atacaatatt ccaatatgcc tatggctact ggacacatac ccatataatc cccctatctg ttttgttaag cctactagtt caatgactat taaaacagga aagcatgttg atgcaaatgg gaagatatat cttccttatc tacatgaatg gaaacaccca cagtcagact tgttggggct tattcaggtc atgattgtgg tatttggaga tgaacctcca gtcttctctc gtcctatttc ggcatcctat ccgccatacc aggcaacggg gccaccaaat acttcctaca tgccaggcat gccaggtgga atctctccat acccatccgg ataccctccc aatcccagtg gttacccagg ctgtccttac ccacctggtg gtccatatcc tgccacaaca agttctcagt acccttctca gcctcctgtg accactgttg gtcccagtag ggatggcaca atcagcgagg acaccatccg agcctctctc atctctgcgg tcagtgacaa actgagatgg cggatgaagg aggaaatgga tcgtgcccag gcagagctca atgccttgaa acgaacagaa gaagacctga aaaagggtca ccagaaactg gaagagatgg ttacccgttt agatcaagaa gtagccgagg ttgataaaaa catagaactt ttgaaaaaga aggatgaaga actcagttct gctctggaaa aaatggaaaa tcagtctgaa aacaatgata tcgatgaagt tatcattccc acagctccct tatacaaaca gatcctgaat ctgtatgcag aagaaaacgc tattgaagac actatctttt acttgggaga agccttgaga aggggcgtga tagacctgga tgtcttcctg aagcatgtac gtcttctgtc ccgtaaacag ttccagctga gggcactaat gcaaaaagca agaaagactg ccggtctcag tgacctctac TGActtctct gataccagct ggaggttgag ctcttcttaa agtattcttc tcttcctttt atcagtaggt gcccagaata agttattgca gtttatcatt caagtgtaaa atattttgaa tcaataatat attttctgtt ttcttttggt aaagactggc ttttattaat gcactttcta tcctctgtaa actttttgtg ctgaatgttg ggactgctaa ataaaatttg ttgcataaaa aaaaaaaaaa aa 32 MAVSESQLKKMVSKYKYRDLTVRETVNVITLYKDLKPVLDSYVFNDGSSRELMNLTGTIP VPYRGNTYNIPICLWLLDTYPYNPPICFVKPTSSMTIKTGKHVDANGKIYLPYLHEWKHP QSDLLGLIQVMIVVFGDEPPVFSRPISASYPPYQATGPPNTSYMPGMPGGISPYPSGYPP NPSGYPGCPYPPGGPYPATTSSQYPSQPPVTTVGPSRDGTISEDTIRASLISAVSDKLRW RMKEEMDRAQAELNALKRTEEDLKKGHQKLEEMVTRLDQEVAEVDKNIELLKKKDEELSS ALEKMENQSENNDIDEVIIPTAPLYKQILNLYAEENAIEDTIFYLGEALRRGVIDLDVFL KHVRLLSRKQFQLRALMQKARKTAGLSDLY 33 MGSSHHHHHHSSGLVPRGSHMASENLYFQGAVSESQLKKMVSKYKYRDLTVRETVNVITLYKDLKPVL DSYVFNDGSSRELMNLTGTIPVPYRGNTYNIPICLWLLDTYPYNPPICFVKPTSSMTIKTGKHVDANG KIYLPYLHEWKHPQSDLLGLIQVMIVVFGDEPPVFSRP 1The initiator and terminator codons in the full-length TSG101 nucleotide sequence are bolded. The UEV domain of TSG101 peptide sequence for amino acids 2-145 are presented in bolded fonts in the full-length TSG101 peptide and the truncated-length TSG101 recombinant peptide. With respect to the truncated-length TSG101 recombinant peptide, the italicized font is leader peptide sequence that includes a polyhistidine motif and thrombin cleavage site derived from the expression vector, pET28b (Novagen). The underlined sequence is a TEV cleavage site introduced at the amino terminus of the TGS101 peptide sequence.

The complete sequence of the pET28b expression vector that includes the truncated TSG101 is presented below.

SEQ ID NO: 34: TGGCGAATGGGACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGGTGTGGTGGTTACGCGCAGC GTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTCCTTTCGCTTTCTTCCCTTCCTTTCTCG CCACGTTCGCCGGCTTTCCCCGTCAAGCTCTAAATCGGGGGCTCCCTTTAGGGTTCCGATTTAG TGCTTTACGGCACCTCGACCCCAAAAAACTTGATTAGGGTGATGGTTCACGTAGTGGGCCATCG CCCTGATAGACGGTTTTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGT TCCAAACTGGAACAACACTCAACCCTATCTCGGTCTATTCTTTTGATTTATAAGGGATTTTGCC GATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTTTAACAAA ATATTAACGTTTACAATTTCAGGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTT TATTTTTCTAAATACATTCAAATATGTATCCGCTCATGAATTAATTCTTAGAAAAACTCATCGA GCATCAAATGAAACTGCAATTTATTCATATCAGGATTATCAATACCATATTTTTGAAAAAGCCG TTTCTGTAATGAAGGAGAAAACTCACCGAGGCAGTTCCATAGGATGGCAAGATCCTGGTATCGG TCTGCGATTCCGACTCGTCCAACATCAATACAACCTATTAATTTCCCCTCGTCAAAAATAAGGT TATCAAGTGAGAAATCACCATGAGTGACGACTGAATCCGGTGAGAATGGCAAAAGTTTATGCAT TTCTTTCCAGACTTGTTCAACAGGCCAGCCATTACGCTCGTCATCAAAATCACTCGCATCAACC AAACCGTTATTCATTCGTGATTGCGCCTGAGCGAGACGAAATACGCGATCGCTGTTAAAAGGAC AATTACAAACAGGAATCGAATGCAACCGGCGCAGGAACACTGCCAGCGCATCAACAATATTTTC ACCTGAATCAGGATATTCTTCTAATACCTGGAATGCTGTTTTCCCGGGGATCGCAGTGGTGAGT AACCATGCATCATCAGGAGTACGGATAAAATGCTTGATGGTCGGAAGAGGCATAAATTCCGTCA GCCAGTTTAGTCTGACCATCTCATCTGTAACATCATTGGCAACGCTACCTTTGCCATGTTTCAG AAACAACTCTGGCGCATCGGGCTTCCCATACAATCGATAGATTGTCGCACCTGATTGCCCGACA TTATCGCGAGCCCATTTATACCCATATAAATCAGCATCCATGTTGGAATTTAATCGCGGCCTAG AGCAAGACGTTTCCCGTTGAATATGGCTCATAACACCCCTTGTATTACTGTTTATGTAAGCAGA CAGTTTTATTGTTCATGACCAAAATCCCTTAACGTGAGTTTTCGTTCCACTGAGCGTCAGACCC CGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTTTTCTGCGCGTAATCTGCTGCTTGCAA ACAAAAAAACCACCGCTACCAGCGGTGGTTTGTTTGCCGGATCAAGAGCTACCAACTCTTTTTC CGAAGGTAACTGGCTTCAGCAGAGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTT AGGCCACCACTTCAAGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCA GTGGCTGCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACCGG ATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTGGAGCGAACGAC CTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCGCCACGCTTCCCGAAGGGAGA AAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGGAACAGGAGAGCGCACGAGGGAGCTTCCAG GGGGAAACGCCTGGTATCTTTATAGTCCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATT TTTGTGATGCTCGTCAGGGGGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGG TTCCTGGCCTTTTGCTGGCCTTTTGCTCACATGTTCTTTCCTGCGTTATCCCCTGATTCTGTGG ATAACCGTATTACCGCCTTTGAGTGAGCTGATACCGCTCGCCGCAGCCGAACGACCGAGCGCAG CGAGTCAGTGAGCGAGGAAGCGGAAGAGCGCCTGATGCGGTATTTTCTCCTTACGCATCTGTGC GGTATTTCACACCGCATATATGGTGCACTCTCAGTACAATCTGCTCTGATGCCGCATAGTTAAG CCAGTATACACTCCGCTATCGCTACGTGACTGGGTCATGGCTGCGCCCCGACACCCGCCAACAC CCGCTGACGCGCCCTGACGGGCTTGTCTGCTCCCGGCATCCGCTTACAGACAAGCTGTGACCGT CTCCGGGAGCTGCATGTGTCAGAGGTTTTCACCGTCATCACCGAAACGCGCGAGGCAGCTGCGG TAAAGCTCATCAGCGTGGTCGTGAAGCGATTCACAGATGTCTGCCTGTTCATCCGCGTCCAGCT CGTTGAGTTTCTCCAGAAGCGTTAATGTCTGGCTTCTGATAAAGCGGGCCATGTTAAGGGCGGT TTTTTCCTGTTTGGTCACTGATGCCTCCGTGTAAGGGGGATTTCTGTTCATGGGGGTAATGATA CCGATGAAACGAGAGAGGATGCTCACGATACGGGTTACTGATGATGAACATGCCCGGTTACTGG AACGTTGTGAGGGTAAACAACTGGCGGTATGGATGCGGCGGGACCAGAGAAAAATCACTCAGGG TCAATGCCAGCGCTTCGTTAATACAGATGTAGGTGTTCCACAGGGTAGCCAGCAGCATCCTGCG ATGCAGATCCGGAACATAATGGTGCAGGGCGCTGACTTCCGCGTTTCCAGACTTTACGAAACAC GGAAACCGAAGACCATTCATGTTGTTGCTCAGGTCGCAGACGTTTTGCAGCAGCAGTCGCTTCA CGTTCGCTCGCGTATCGGTGATTCATTCTGCTAACCAGTAAGGCAACCCCGCCAGCCTAGCCGG GTCCTCAACGACAGGAGCACGATCATGCGCACCCGTGGGGCCGCCATGCCGGCGATAATGGCCT GCTTCTCGCCGAAACGTTTGGTGGCGGGACCAGTGACGAAGGCTTGAGCGAGGGCGTGCAAGAT TCCGAATACCGCAAGCGACAGGCCGATCATCGTCGCGCTCCAGCGAAAGCGGTCCTCGCCGAAA ATGACCCAGAGCGCTGCCGGCACCTGTCCTACGAGTTGCATGATAAAGAAGACAGTCATAAGTG CGGCGACGATAGTCATGCCCCGCGCCCACCGGAAGGAGCTGACTGGGTTGAAGGCTCTCAAGGG CATCGGTCGAGATCCCGGTGCCTAATGAGTGAGCTAACTTACATTAATTGCGTTGCGCTCACTG CCCGCTTTCCAGTCGGGAAACCTGTCGTGCCAGCTGCATTAATGAATCGGCCAACGCGCGGGGA GAGGCGGTTTGCGTATTGGGCGCCAGGGTGGTTTTTCTTTTCACCAGTGAGACGGGCAACAGCT GATTGCCCTTCACCGCCTGGCCCTGAGAGAGTTGCAGCAAGCGGTCCACGCTGGTTTGCCCCAG CAGGCGAAAATCCTGTTTGATGGTGGTTAACGGCGGGATATAACATGAGCTGTCTTCGGTATCG TCGTATCCCACTACCGAGATATCCGCACCAACGCGCAGCCCGGACTCGGTAATGGCGCGCATTG CGCCCAGCGCCATCTGATCGTTGGCAACCAGCATCGCAGTGGGAACGATGCCCTCATTCAGCAT TTGCATGGTTTGTTGAAAACCGGACATGGCACTCCAGTCGCCTTCCCGTTCCGCTATCGGCTGA ATTTGATTGCGAGTGAGATATTTATGCCAGCCAGCCAGACGCAGACGCGCCGAGACAGAACTTA ATGGGCCCGCTAACAGCGCGATTTGCTGGTGACCCAATGCGACCAGATGCTCCACGCCCAGTCG CGTACCGTCTTCATGGGAGAAAATAATACTGTTGATGGGTGTCTGGTCAGAGACATCAAGAAAT AACGCCGGAACATTAGTGCAGGCAGCTTCCACAGCAATGGCATCCTGGTCATCCAGCGGATAGT TAATGATCAGCCCACTGACGCGTTGCGCGAGAAGATTGTGCACCGCCGCTTTACAGGCTTCGAC GCCGCTTCGTTCTACCATCGACACCACCACGCTGGCACCCAGTTGATCGGCGCGAGATTTAATC GCCGCGACAATTTGCGACGGCGCGTGCAGGGCCAGACTGGAGGTGGCAACGCCAATCAGCAACG ACTGTTTGCCCGCCAGTTGTTGTGCCACGCGGTTGGGAATGTAATTCAGCTCCGCCATCGCCGC TTCCACTTTTTCCCGCGTTTTCGCAGAAACGTGGCTGGCCTGGTTCACCACGCGGGAAACGGTC TGATAAGAGACACCGGCATACTCTGCGACATCGTATAACGTTACTGGTTTCACATTCACCACCC TGAATTGACTCTCTTCCGGGCGCTATCATGCCATACCGCGAAAGGTTTTGCGCCATTCGATGGT GTCCGGGATCTCGACGCTCTCCCTTATGCGACTCCTGCATTAGGAAGCAGCCCAGTAGTAGGTT GAGGCCGTTGAGCACCGCCGCCGCAAGGAATGGTGCATGCAAGGAGATGGCGCCCAACAGTCCC CCGGCCACGGGGCCTGCCACCATACCCACGCCGAAACAAGCGCTCATGAGCCCGAAGTGGCGAG CCCGATCTTCCCCATCGGTGATGTCGGCGATATAGGCGCCAGCAACCGCACCTGTGGCGCCGGT GATGCCGGCCACGATGCGTCCGGCGTAGAGGATCGAGATCTCGATCCCGCGAAATTAATACGAC TCACTATAGGGGAATTGTGAGCGGATAACAATTCCCCTCTAGAAATAATTTTGTTTAACTTTAA GAAGGAGATATACCATGGGCAGCAGCCATCATCATCATCATCACAGCAGCGGCCTGGTGCCGCG CGGCAGCCATATGGCTAGCgaaaacctgtacttccagggcgcggtgtcggagagccagctcaag aaaatggtgtccaagtacaaatacagagacctaactgtacgtgaaactgtcaatgttattactc tatacaaagatctcaaacctgttttggattcatatgtttttaacgatggcagttccagggaact aatgaacctcactggaacaatccctgtgccttatagaggtaatacatacaatattccaatatgc ctatggctactggacacatacccatataatccccctatctgttttgttaagcctactagttcaa tgactattaaaacaggaaagcatgttgatgcaaatgggaagatatatcttccttatctacatga atggaaacacccacagtcagacttgttggggcttattcaggtcatgattgtggtatttggagat gaacctccagtcttctctcgtccttgataaGGATCCGAATTCGAGCTCCGTCGACAAGCTTGCG GCCGCACTCGAGCACCACCACCACCACCACTGAGATCCGGCTGCTAACAAAGCCCGAAAGGAAG CTGAGTTGGCTGCTGCCACCGCTGAGCAATAACTAGCATAACCCCTTGGGGCCTCTAAACGGGT CTTGAGGGGTTTTTTGCTGAAAGGAGGAACTATATCCGGAT

wherein the bold font denote the locations of the initiator and terminator codons for the recombinant peptide pro-form; the upper case letters denote pET28b vector sequences; the lower case letter denote the truncated TSG101 (2-145) coding sequences, the italicized font includes the leader peptide coding sequence that includes a polyhistidine motif and thrombin cleavage site derived from the expression vector, pET28b (Novagen). The underlined sequence is a TEV cleavage site introduced at the amino terminus of the TGS101 peptide sequence.

Example 5 High Throughput Fluorescence-Based Thermal Shift (FTS) Assay for TSG101

The NU-HTA has developed a robotic pipeline for small molecule protein ligand screening by FTS. See Luan C H, Light S H, Dunne S F, Anderson W F. Ligand screening using Fluorescence Thermal Shift Analysis (FTS). In Structural Genomics and Drug Discovery: Methods and Protocols. Methods Mol. Biol. 2013, Humana Press (In press), which is incorporated by reference in its entirety. The pipeline uses an Echo550 acoustic transfer robot (Labcyte, CA) for compound addition and the Mosquito robot (TTP LabTechnologies, UK) for protein dispensing followed by thermal scanning coupled with fluorescence detection which is performed on a real-time PCR machine CFX384 (Bio-Rad Laboratories). The method does not require any labeling on either protein or compound. A fluorescent dye Sypro-Orange (Invitrogen) is used for assay detection. TSG101 (SEQ ID NO:33) has a thermal unfolding profile ideal for using FTS as primary screen assay.

Compound Library Screening.

For primary screening, the TSG101 protein (SEQ ID NO:33) was premixed at a concentration of 2 uM with a 5× concentration of Sypro-Orange in Hepes buffer (100 mM HEPES, 150 mM NaCl, pH 7.5). Then 10 uL of the protein-dye mix was added to an assay plate. And 10 to 50 nanoliters of compound, equal to 10 to 50 uM, were added. The plate was shaken to ensure proper mixing and then sealed with optical seal and centrifuged. The thermal scan was performed from 10 to 95° C. with a temperature ramp rate of 1.5° C./min. The fluorescence was recorded every 10 sec. Data analysis and report generation were performed by using the in-house software excelFTS. Hit compounds identified were tested in dose-response format.

Validation of the Confirmed Hits with TSG101 by FP and EpicLF.

The confirmed hits from the primary FTS screening were validated by fluorescent polarization (FP) and Epic label-free (EpicLF) assays. The EpicLF assay was performed on an EnSpire multifunction plate reader (Perkin Elmer) to determine the Kd of hit compound binding to TSG101 (SEQ ID NO:33) as described generally in Example 3 for candidate compound binding to Nedd 4 peptide family members. The hit compounds were also tested by FP assay to determine the IC50 of disrupting TSG101 (SEQ ID NO: 33) and PTAP-probe interaction as described generally in Example 2 for candidate compound screening to determine IC50 of disrupting Nedd 4 peptide family members and PY-probe interaction.

For this purpose, tandem-linked versions of the PTAP motif from HIV-1 gag was designed and synthesized for use in these experiments. The structures of the resulting peptides are presented below.

SEQ ID NO: 20: RPGNFLQSRPEPTAPPFLQSRPEPTAPPEESFRRRR SEQ ID NO: 21: FITC-RPGNFLQSRPEPTAPPFLQSRPEPTAPPEESFRRRR SEQ ID NO: 22: TAMRA-RPGNFLQSRPEPTAPPFLQSRPEPTAPPEESFRRRR

Example 6 In Vivo Screening for Compounds that Disrupt the Interaction of TSG101 and the PTAP L-Domain Sequence in the p6 Region of HIV-1 Gag (Prophetic Example)

Cells will be transfected with suitable vector constructs to express (either transiently or stably) EGFP reconstituted from a two-hybrid system comprising a fusion protein containing PTAPP motifs and the N-terminal portion of EGFP and a fusion protein containing TSG101 polypeptides and the C-terminal portion of EGFP. Following establishment of stable EGFP expression, the ability of test compound to enter the cells and inhibit EGFP expression will be evaluated. A reduction of EGFP fluorescence as a function of test compound administration to cells will indicate that the compound inhibits formation of functional GFP complexes from the two component system. Controls will be performed that include evaluation the cytotoxicity of the test compounds having positive effect in this assay.

Plasmids for CEGFP-N1.

The plasmid pCEGFP-N1, which places CEGFP under control of the T7 promoter, was created by PCR amplification (Deep Vent polymerase) of the gene for C-terminal EGFP (from 159 to 265 amino acids) from pEGFP-N1 using the oligonucleotides 5′-ataggatccaccgg tcgccaccggtggctctggc aagaacgg catcaaggtg aacttcaa-3′ forward primer (SEQ ID NO:35) and 5′-gtcgcggccgctttacttgtacagctcgtccatg-3′ reverse primer (SEQ ID NO:36). The 4-residue linker (GGSG (SEQ ID NO:37)) is located at the beginning of the CEGFP gene. This was followed by digestion of PCR products and the plasmid pEGFP-N1 with NheI and XhoI, and then ligation with T4 DNA ligase. The C-terminal EGFP ligation was confirmed by DNA sequencing by using sequencing primer (5′-gcagagctggtttagtg-3′ forward (SEQ ID NO:38), from 561 to 577 bp of pEGFP-N1 sequences).

Plasmids for NEGFP-C3.

The plasmid pNEGFP-N1, which places NEGFP under control of the T7 promoter, was created by PCR amplification (Deep Vent polymerase) of the gene for N-terminal EGFP (from 1 to 158 amino acids) from pEGFP-C3 using the oligonucleotides 5′-cagatcc gctagcgctaccggtcgcca ccatggtgag forward primer (SEQ ID NO:39) and 5′-atactcgagatctgagtacccagagccagagccaccctgatgtcggccatgatatag-3′ reverse primer (SEQ ID NO:40). The 6-residue peptide linker (GGSGSG [(SEQ ID NO:41)]) is located at the end of the NEGFP gene. This was followed by digestion of PCR products and the plasmid pEGFP-C3 with BamHI and NotI, and then ligation with T4 DNA ligase. The N-terminal EGFP ligation was confirmed by DNA sequencing by using sequencing primer 5′ gtgggaggttttttaaa-3′ reverse (from 1451 to 1467 by of pEGFP-C3 sequences) (SEQ ID NO:42).

Construct NEGFP-2x PTAPP.

Two copies of PTAPP sequences of HIV Gag were amplified from pGBT9_HIV Gag with 2PTAP template (from Dr. Carol Carter's lab) by PCR reaction by using the oligonucleotides. This was followed by digestion of PCR products and the plasmid pNEGFP-C3 with EcoRI and BamHI, and then ligation with T4 DNA ligase.

Construct CEGFP-TSG101.

TSG101 was amplified by PCR reaction by using the oligonucleotides. This was followed by digestion of PCR products and the plasmid pNEGFP-C3 with EcoRI and BamHI, and then ligation with T4 DNA ligase.

Example 7 Biological Testing of Compounds to Inhibit Virus Budding Detected by Release Of VLPs from Cells

Transfection of 293/E Cells and Chemical Inhibitor Treatment.

293/E cells were cultured in DMEM supplemented with 10% fetal bovine serum, penicillin (1,000 units/ml), and streptomycin (1,000 μg/ml) to 50% confluence at 37° C. Expression of plasmids from p2036 was high in 293/E cells because these cells stably express the EBNA1 protein of EBV and the p2036 constructs contain the EBV FR plasmid maintenance element that EBNA 1 binds. Therefore 293/E cells were used when proteins expressed from p2036 were to be detected by western analysis. In all experiments, 24-well plates of 293/E cells were transfected with 0.5 μg of p2036-ASLV Gag with the X-treme Gene9 transfection reagent (Roche Diagnostics, Alameda, CA, USA) according to the manufacturer's instructions. After 24 h after DNA transfection, the cells were washed with 1×DPS that contained CaCl2 (0.1 mM) and MgCl2 (1 mM) [Gibco #14040-133], and 1 ml of cell culture medium (10% FBS, 1% Penicillin/Streptomycin) containing CaCl2 (0.5 mM) and MgCl2 (5 mM) was added. Thereafter, the inhibitor compounds were added to the culture medium of parallel culture wells at one of the following final concentrations: 5 μM, 10 μM, 20 μM, or 40 μM and the cells remained in contact with the culture medium containing the inhibitor compounds for 5 hr. Cells and virus-like particles (VLPs) released into the cell media were collected 5 h after chemical treatment.

Detection of Gag Proteins by Western Blotting.

For the budding assay, both media and cell lysate fractions were collected. The cell lysate fractions were prepared by suspension in radioimmune precipitation assay (RIPA) buffer [PBS containing 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, and protease inhibitor mixture tablets] 48 hours post-transfection. VLPs were purified from the cell culture medium by centrifugation through a 20% sucrose cushion at 100,000×g for 1 h at 4° C. (Beckman SW50.1 rotor). The pelleted VLPs were suspended in 100 μl of RIPA buffer containing protease inhibitor mixture tablets. For lysate fractions, Gag proteins were immunoprecipitated overnight at 4° C. with an anti-ASLV monoclonal serum (1:500-1:1000 dilution) and 20 μl of protein A-agarose beads. The precipitated proteins were separated by SDS-PAGE and transferred to a polyvinylidene difluoride membrane. After blocking of the membrane with wash buffer (10 mM Tris-HCl, pH 8.0, 150 mM NaCl, and 0.1% Tween 20) containing 5% nonfat dry milk, ASLV Gag proteins were detected with an AMV MA (p19) directed monoclonal antibody (mAb) and an anti-mouse IgG-HRP secondary antibody for ECL (Denville Scientific; Metuchen, N.J.).

Viral Replication Assays.

293/E cells in 6-well plates were transfected with HIV-1 luciferase reporter vector consisting of the following plasmids: 1.5 μg of pHIV env-Luc and 0.75 μg of VSV-G (Provided by Tom Hope, Northwestern University). Supernatants containing pseudo-typed virions were harvested 24 or 48 h after the chemical treatments, and the cells were harvested at the same time for western blot analyses. To measure titers, virus particles were filtered through a 0.45 μm filter, and infectivity was assayed by incubating in duplicate 1×105 293/E cells/well with 1 ml of pseudo typed virus in 6-well plates for 5 h. Virus was then removed and cell growth media was added. The cells were lysed in 400 μA of cell culture lysis reagent (Promega, Madison, Wis.) at 72 h post-transfection. The luciferase activity was measured with a luciferase assay kit (Promega, Madison, Wis.) and a FB12 luminometer according to manufacturer's instructions. The data are presented as the average of the duplicates.

Example 8 Biological Testing of Nedd4 Inhibitor Candidate Compounds K21 (Benserazide Hydrochloride) and N20 (Oxytetracycline) by Cell Culture Assays In Vitro Cell-Based Inhibition Assay

COS-1 cells were seeded on twelve-well plates and grown in 1 ml of DMEM with 10% fbs and 1% Penicillin/Streptomycin. The following day, well were confluency was ˜60% were selected for co-transfection with an NL4-3 derived construct (pdeltaEnv) and pIIIEnv for expression of HIV-1 virus-encoded proteins. Inhibitor treatment was done on two sets of transfected cells: one, at 5 hr post-transfection and the other, at 24 hours post-transfection. In each case, the tissue culture media was removed and replaced with treatment Nedd4 inhibitor (100 μM final concentration for K21 and 20 μM final concentration for N20) or DMSO control (1% final concentration) in DMEM with 10% FBS, 1% Penicillin/Streptomycin, 5 mM MgCl2, and 0.5 mM CaCl2. After a 24-hr treatment period, the tissue culture media was collected, cleared of debris by running through a 0.45 um syringe filter and analyzed for amount of virus particles by Elisa p24 capture assay.

Cell Death Evaluation.

The standard trypan blue assay, where dead cells are blue when visualized under a light microscope, was used to determine the percent of dead cells in the culture. Briefly, cells from a well of DMSO-treated, K21-treated and N20-treated samples were dislodged from the well with a stream of 1 ml PBS, collected, pelleted at 200 rpm for 2 min, and suspended in 300 μl trypsin-EDTA, incubated at 37° C. for 30 min. A diluted cell suspension was prepared by mixing 200 μl of the trypsin-treated cell suspension and 800 μl of PBS was used for the assay. For the assay itself: from the 1 ml of diluted cell suspension, 50 μl was taken an placed on paraffin and to this was mixed in 50 ml of 0.4% Trypan Blue dye in PBS, after exactly 2 minutes later, the mixture was loaded on a hematocytometer. The hematocytometer was placed on the light microscope stage and a blue cell count and an all cells count (blue and not blue cells) obtained. Two 50 μl aliquots were counted per sample. The results are presented in Table VII.

TABLE VII Cytotoxicity results for contacting cells with inhibitor compounds. Compound Blue cells Total cells Cytotoxicity1 DMSO 7 29 24% 9 49 18% (21%) K21 13 38 34% 17 60 28% (31%) N20 11 37 29% 7 20 35% (32%) 1Cytotoxicity (%) is determined by the fraction (blue cells)/(total cells) multiplied by 100 (%). The average of the independent experiments is shown in parentheses.

Cell Growth Based on Cell Lysate Actin Levels

The levels of actin of cell lysates prepared from DMSO-treated, K21-treated and N20-treated cells were determined by SDS-PAGE followed by Western analysis where the immunoblot were probed with mouse anti-actin antibody. The intensity of the actin bands were essentially comparable for all samples (not shown).

Effects on HIV-1 Production Accumulation of Gag-Related Proteins in the Cell

The levels of Gag-related proteins in cell lysates were immune-precipitated with polyclonal anti CA antibody. Proteins in the immune-precipitate were separated by SDS-PAGE and followed by Western analysis where the blot was probed with mouse anti-CA antibody. The commercial antibody used (NEN NEA-9306) is known to recognize the Gag precursor better than the mature p24 proteins (Dietrich et al 2001). Both GagPr55 and mature p24, as adjudged by their respective band intensities, were essentially comparable for all samples (not shown).

ELISA p24 Capture Assay of Tissue Culture Media

At 100 μM, K21 was inhibitory to virus particle release. There was a reduction in the amount of virus particle detected in the tissue culture media. This was true whether the inhibitor was added at 5 hrs post-transfection or at 24 hrs post transfection. At 20 μM, N20 was also exerted an inhibitory effect on virus particle release but it was different from that of K21. There was a reduction in the amount of virus particle detected in the tissue culture media when inhibitor was added at 5 hr post-transfection. This reduction was not seen when inhibitor was added 24 hrs post-transfection.

Specific Infectivity

The amount of infectious virus normalized to ng of p24 obtained from the ELISA assay in the tissue cultures was determined by multinuclear activation of a galactosidase indicator (MAGI) assay. In this assay, infectious unit is scored by the blue color that is assumed by infected MAGI cells (Hela cells that have been engineered to express the indicator when the infecting particle is able to simulated natural infection up until expression of HIV-1 Tat). Counts of Blue cells per ng p24 for the DMSO controls, K21-treated and N20-treated samples were comparable.

Example 9 HSV-1 Inhibition Assay by PTAP-Inhibitor Compounds F15 (Esomeprazole Potassium) and N16 (Tenatoprazole) MTS Assay

The cytotoxicity of the compounds was determined by a commercially available assay (Celltiter 96® AQueous One Solution cell proliferation assay reagent; Promega, Madison, Wis.), as described previously (Akkarawongsa et al., 2006). The CellTiter 96® AQueous One Solution Cell Proliferation Assay is a colorimetric method for determining the number of viable cells in proliferation, cytotoxicity or chemosensitivity assays. The CellTiter 96® AQueous One Solution Reagent contains a tetrazolium compound [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt; MTS] and an electron coupling reagent (phenazine ethosulfate; PES). PES has enhanced chemical stability, which allows it to be combined with MTS to form a stable solution. The CellTiter 96® AQueous Assay uses phenazine methosulfate (PMS) as the electron coupling reagent, and PMS Solution and MTS Solution are supplied separately. PES has enhanced chemical stability, which allows it to be combined with MTS to form a stable solution. Assays are performed by adding a small amount of the CellTiter 96® AQueous One Solution Reagent directly to culture wells, incubating for 1-4 hours and then recording absorbance at 490 nm with a 96-well plate reader. The quantity of formazan product as measured by the amount of 490 nm absorbance is directly proportional to the number of living cells in culture.

Briefly, Vero cells (1.5×104 cells/well) were seeded in a 96-well plate, and the plate was incubated for 24 h at 37° C. A total of 20 μl of medium containing the desired concentration of inhibitors was added to the cells. Control cells received medium only. After incubation of the cells in the presence of peptide overnight at 37° C., 20 μl of the 96 AQqueous One Solution cell proliferation assay reagent was added to each well. The plate was then incubated for 2 h at 37° C., and the absorbance at 490 nm was determined with a 96-well plate reader (Perkin Elmer).

Assay for Inhibitor Compound Effects on Infected Cells and HSV-1 Virus Production and Release.

Vero cells were grown in DMEM supplemented with 10% FBS. Viral infections were performed in DMEM supplemented with 1% heat-inactivated FBS. Vero cells were inoculated HSV-1 (strain F) at indicated concentrations, after 2 h, the cells were washed and treated with 0.1 M sodium citrate buffer (pH 3.0) for 1 min to inactivate unpenetrated viruses, washed again and incubated with fresh medium containing 1% heat-inactivated FBS. At different times after inoculation, one-half of the medium was harvested as a supernatant sample and the cells were scraped into the rest of the half medium containing released viruses as a total virus sample and lysed by sonication. Then virus titers were determined by standard plaque assay on Vero cells.

As shown in FIG. 8, inhibitor compound F15 (Esomepazole) resulted in infectious HSV-1 virion release from HSV-1 infected cells at about 10% of the level observed for virus release from infected cells not contacted with an inhibitor compound at the higher concentration tested (that is, more than a 90% reduction in virus release when F15 is present at 80 μM in the culture medium). Inhibitor compound F15 also reduced the total load of infectious virions produced in the cells, whether released or not from the cells, to about 25% of the level observed for virus release from infected cells not contacted with an inhibitor compound at the higher concentration tested (that is, more than a 75% reduction in total infectious virus when F 15 is present at 80 μM in the culture medium). As shown in FIG. 8A, inhibitor compound N16 (Tenatoprazole) reduced infectious HSV-1 virion release from HSV-1 infected cells at about 5% of the level observed for virus release from infected cells not contacted with an inhibitor compound at the higher concentration tested (that is, more than a 95% reduction in virus release when N16 is present at 80 μM in the culture medium). Thus, inhibitor compound N16 was slightly more effective than F15 at reducing infectious HSV-1 virion particle release from HSV-1 infected cells at the concentrations tested. Neither inhibitor compound was cytotoxic to the Vero cells at the concentrations tested (FIG. 8B).

Example 10 Testing of Compounds to Inhibit the Budding of KSHV (Prophetic Example)

KSHV, a member of the herpes virus family, buds from cells with a Vps4 dependence, probably using the PY motif-dependent pathway. To test the effect of inhibitors on KSHV release, of a recently reported cell line (iSLK.219) will be used that allows the doxycycline (Dox) inducible expression of the KSHV lytic transactivator protein (RTA) and is infected with recombinant KSHV.219. See Myoung J, Ganem D. Generation of a doxycycline-inducible KSHV producer cell line of endothelial origin: maintenance of tight latency with efficient reactivation upon induction. J Virol Methods. 2011, 174(1-2):12-21. PMCID: 3095772 and Vieira J, O'Hearn P M. Use of the red fluorescent protein as a marker of Kaposi's sarcoma-associated herpesvirus lytic gene expression. Virology. 2004; 325(2):225-40. The contents of these printed publication are incorporated by reference in their entirety. Upon Dox treatment of these cells, lytic reactivation results in the production of infectious KSHV carrying a GFP reporter cassette. The ability of individual dominant negative ESCRT proteins interfere with the release of infectious KSHV will be evaluated initially. For this purpose, 105 iSLK.219 cells will be plated per well in 6 well plates and transfected the next day with increasing amounts of control vectors or vectors expressing dominant negative ESCRT proteins (range 0.2 μg-2 mg/well) using Lipofectamine 2000 (Invitrogen) as instructed. Four hours after transfection, growth medium will be exchanged for medium containing 1 mg/ml Dox and cells will be incubated for 48 hours. Resulting virus containing supernatant will be cleared by centrifugation (5 min, 2000 rpm), filtered through 450 nm pore size filters and will titered on the KSHV-negative endothelial cell line SLK by serial dilutions in a 24 well format. For this, 20,000 cells will be infected with filtered virus in serial dilutions into normal growth medium (i.e. 1:1, 1:5, 1:25). Forty-eight hours after infection, SLK cells will be trypsinized, recovered by centrifugation (5 min, 1400 rpm), fixed with 4% paraformaldehyde for 20 min at room temperature, washed once with PBS, suspended in 300 ml PBS and subjected to flow cytometry on a FACS Canto II in order to establish the percentage of green cells. If ESCRT proteins are indeed required for KSHV production, a reduction in virus titer will be expected.

In a parallel positive control experiment, the effect of wild type or dominant negative ESCRT proteins on the release of a GFP-positive lentiviral vector pLCE from SLK cells will be monitored by titration and flow cytometry. The lentiviral vector pLCE is described in Zhang J, Jima D D, Jacobs C, Fischer R, Gottwein E, Huang G, et al. Patterns of microRNA expression characterize stages of human B-cell differentiation. Blood. 2009; 113(19):4586-94, the contents of which are incorporated by reference in its entirety. This experiment will be conducted as outlined above, except that ESCRT vectors will be co-transfected with 0.5 mg pLCE and 0.125 mg each of three packaging plasmids (pMDLgpRRE, pRSV-Rev and pVSV-G). Because lentiviral release depends on the ESCRT machinery, we expect that dominant negative ESCRT vectors will result in a reduction of lentiviral titers. If a dependence of KSHV budding on the ESCRT machinery can be established in the experiments outlined above, iSLK.219 cells will be used to test the effectiveness of novel small molecule inhibitors using the experimental procedure described above.

DEFINITIONS

When introducing elements of aspects of the embodiments, the articles “a,” “an,” “the,” and “said” are intended to mean that there are one or more of the elements. The terms “comprising,” “including,” and “having” are intended to be inclusive and mean that there may be additional elements other than the listed elements. The word “or” means any one member of a particular list and also includes any combination of members of that list, unless otherwise specified.

The modal verb “may” refers to the preferred use or selection of one or more options or choices among several described embodiments or features contained within the same. Where no options or choices are disclosed regarding a particular embodiment or feature contained in the same, the modal verb “may” refers to an affirmative act regarding how to make or use an aspect of a described embodiment or feature contained in the same, or a definitive decision to use a specific skill regarding a described embodiment or feature contained in the same. In this latter context, the model verb “may” has the same meaning and connotation as the auxiliary verb “can.”

The term “about” is used herein to mean approximately, roughly, around, or in the region of. When the term “about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. Preferably, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 20 percent up or down (higher or lower).

The term “associative complex” refers to two or more molecular entities (for example, two separate polypeptides; an isolated compound and an isolated polypeptide; two separate fusion proteins that interact in a two-hybrid system, two separate proteins, among others) that are present in a complex or that are capable of forming a complex.

The chemical structures described herein are named according to IUPAC nomenclature rules and include art-accepted common names and abbreviations where appropriate. The IUPAC nomenclature can be derived with chemical structure drawing software programs, such as ChemDraw® (PerkinElmer, Inc.), ChemDoodle® (iChemLabs, LLC) and Marvin (ChemAxon Ltd.). The chemical structure controls in the disclosure to the extent that an IUPAC name is misnamed or otherwise conflicts with the chemical structure disclosed herein.

The chemical structures described herein are also cataloged according to CAS Registry Nos. where appropriate. The chemical structure controls in the disclosure to the extent that an CAS Registry No. is misidentified or otherwise conflicts with the chemical structure disclosed herein.

In view of the above, it will be seen that several advantages of the invention are achieved and other advantageous results attained.

Not all of the depicted components illustrated or described may be required. In addition, some implementations and embodiments may include additional components. Variations in the arrangement and type of the components may be made without departing from the spirit or scope of the claims as set forth herein. Additional, different or fewer components may be provided and components may be combined. Alternatively or in addition, a component may be implemented by several components.

The above description illustrates the invention by way of example and not by way of limitation. This description clearly enables one skilled in the art to make and use the invention, and describes several embodiments, adaptations, variations, alternatives and uses of the invention, including what is presently believed to be the best mode of carrying out the invention. Additionally, it is to be understood that the invention is not limited in its application to the details of construction and the arrangement of components set forth in the following description or illustrated in the drawings. The invention is capable of other embodiments and of being practiced or carried out in various ways. Also, it will be understood that the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting.

Having described aspects of the invention in detail, it will be apparent that modifications and variations are possible without departing from the scope of aspects of the invention as defined in the appended claims. As various changes could be made in the above constructions, products, and methods without departing from the scope of aspects of the invention, it is intended that all matter contained in the above description and shown in the accompanying drawings shall be interpreted as illustrative and not in a limiting sense.

All references, citations, patent applications, patent publications specifically mentioned in this disclosure are hereby incorporated by reference in their entireties.

Claims

1. A compound having an antiviral activity for inhibiting release of an enveloped virus from a cell.

2. The compound of claim 1, wherein the antiviral activity comprises:

(i) inhibiting formation of an associative complex; or
(ii) disrupting formation of an associative complex,
wherein the associative complex comprises an L-domain motif of the enveloped virus and at least one cellular polypeptide, or fragment thereof, capable of binding the L-domain motif of the enveloped virus.

3. The compound of claim 2, wherein L-domain motif comprises at least one of a PY-motif or a PTAP-motif.

4. The compound of claim 2, wherein L-domain motif comprises at least one member selected from the group consisting of SEQ ID NOS:1-22.

5. The compound of claim 2, wherein the at least one cellular polypeptide comprises an ESCRT component protein.

6. The compound of claim 5, wherein the ESCRT component protein comprises at least one member selected from a Nedd 4-related family peptide and TSG101, fragments thereof, and combinations thereof.

7. The compound of claim 6, wherein ESCRT component protein comprises one of SEQ ID NOS: 24, 29, 32 and 33.

8. The compound of claim 1, wherein the enveloped virus comprises at least one member selected from the group consisting of Lassa fever virus, lymphocytic choriomeningitis virus, Ebola virus, Marberg virus, hepatitis B virus, Herpes simplex virus, type 1, Herpes simplex virus, type 2, cytomegalovirus, Simian virus, type 5, Mumps virus, avian sarcoma leucosis virus, human immunodeficiency virus, type 1, human T-lymphotrophic virus, type 1, equine infectious anemia virus, vesicular stomatitis virus, rabies virus and combinations thereof.

9. The compound of claim 1, wherein the cell comprises a host cell for supporting active replication of the enveloped virus.

10. The compound of claim 1, wherein the compound comprises at least one member selected from the group consisting of compounds having the structure of formulas (VII-(XIII): and combinations thereof.

11. A method of inhibiting release of an enveloped virus from a cell, comprising contacting the cell with a compound having an antiviral activity, said antiviral activity comprises:

(i) inhibiting formation of an associative complex; or
(ii) disrupting formation of an associative complex,
wherein the associative complex comprises an L-domain motif of the enveloped virus and at least one cellular polypeptide, or fragment thereof, capable of binding the L-domain motif of the enveloped virus.

12. The method of claim 11, wherein L-domain motif comprises at least one of a PY-motif or a PTAP-motif.

13. The method of claim 11, wherein L-domain motif comprises at least one member selected from the group consisting of SEQ ID NOS:1-22.

14. The method of claim 11, wherein the at least one cellular polypeptide comprises an ESCRT complex protein.

15. The method of claim 14, wherein the ESCRT component protein comprises at least one member selected from a Nedd 4-related family peptide and TSG101, fragments thereof, and combinations thereof.

16. The method of claim 14, wherein ESCRT component protein comprises one of SEQ ID NOS: 24, 29, 32 and 33.

17. The method of claim 11, wherein the enveloped virus comprises at least one member selected from the group consisting of Lassa fever virus, lymphocytic choriomeningitis virus, Ebola virus, Marberg virus, hepatitis B virus, Herpes simplex virus, type 1, Herpes simplex virus, type 2, cytomegalovirus, Simian virus, type 5, Mumps virus, avian sarcoma leucosis virus, human immunodeficiency virus, type 1, human T-lymphotrophic virus, type 1, equine infectious anemia virus, vesicular stomatitis virus, rabies virus and combinations thereof.

18. The method of claim 11, wherein the cell comprises a host cell for supporting active replication of the enveloped virus.

19. The method of claim 11, wherein the compound comprises at least one member selected from the group consisting of compounds having the structure of formulas (VII)-(XIII): and combinations thereof.

20. A pharmaceutical composition comprising a compound having an antiviral activity for inhibiting release of an enveloped virus from a cell and optionally a pharmaceutically acceptable carrier.

Patent History
Publication number: 20140179637
Type: Application
Filed: Dec 21, 2013
Publication Date: Jun 26, 2014
Applicants: The Research Foundation for The State University of New York (Albany, NY), Northwestern University (Evanston, IL)
Inventors: Jonathan Leis (Chicago, IL), Carol Carter (Huntington, NY)
Application Number: 14/138,053