EP1238072A2 - Compositions et procedes d'inhibition de l'infection au virus de l'immunodeficience humaine, par regulation restrictive de genes cellulaires humains - Google Patents

Compositions et procedes d'inhibition de l'infection au virus de l'immunodeficience humaine, par regulation restrictive de genes cellulaires humains

Info

Publication number
EP1238072A2
EP1238072A2 EP00961525A EP00961525A EP1238072A2 EP 1238072 A2 EP1238072 A2 EP 1238072A2 EP 00961525 A EP00961525 A EP 00961525A EP 00961525 A EP00961525 A EP 00961525A EP 1238072 A2 EP1238072 A2 EP 1238072A2
Authority
EP
European Patent Office
Prior art keywords
seq
protein
nucleic acid
cell
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP00961525A
Other languages
German (de)
English (en)
Inventor
Tanya A. Holzmayer
Stephen J. Dunn
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
PPD Development LP
Original Assignee
SUBSIDIARY N0 3 Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by SUBSIDIARY N0 3 Inc filed Critical SUBSIDIARY N0 3 Inc
Publication of EP1238072A2 publication Critical patent/EP1238072A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to the identification of certain human genes as cellular targets for the design of therapeutic agents for suppressing human immunodeficiency virus (HIV) infection.
  • HIV human immunodeficiency virus
  • These genes encode products which are necessary for HIV infection, because HIV infection is inhibited when expression of these genes is down-regulated. Therefore, compounds that inhibit expression of these genes or function of the encoded gene products can be used as therapeutic agents for the treatment and/or prevention of HIV infection.
  • the invention relates to methods for identifying additional cellular genes as therapeutic targets for suppressing
  • HIV infection and methods of using such cellular genes and their encoded products in screening assays for selecting protective compounds that inhibit HIV infection.
  • HIV acquired immunodeficiency syndrome
  • Retroviruses are small enveloped viruses that contain a diploid, single-stranded RNA genome, and replicate via a DNA intermediate produced by a virally-encoded reverse transcriptase, an RNA-dependent DNA polymerase (Varmus, 1988, Science 240: 1427-1439).
  • HIV-1 Barre-Sinoussi etal, ibid.; Gallo etal, ibid.
  • HIV-2 Clavel et ⁇ /., 1986, Science 233:343-346; Guyaderet ⁇ /., 1987, Nature 326:662- 669. Genetic heterogeneity exists within each of these HIV subtypes.
  • CD4 + T cells are the major targets of HIV infection because the CD4 cell surface protein acts as a cellular receptor for HIY attachment (Dalgleish et al, 1984, Nature
  • nucleotide analogs are not curative, probably due to the rapid appearance of drug resistant HIV mutants (Lander et al, 1989, Science 243:1731-1734). In addition, these drugs often exhibit toxic side effects, such as bone marrow suppression, vomiting, and liver abnormalities.
  • Late-stage processing is dependent on the activity of a virally- encoded protease, and drugs including saquinavir, ritonavir, and indinavir have been developed to inhibit this protease (Pettit et al, 1993, Persp. Drug Discov. Design 1 :69-
  • RNA-based antisense and ribozymes RNA-based antisense and ribozymes
  • protein-based antiviral agents can inhibit certain stages ofthe viral life cycle.
  • Antisense polynucleo tides have been designed to complex with and sequester the HIV-1 transcripts (Holmes etal, WO 93/11230; Lipps etal, WO 94/10302; Kretschmer et al, EP 594,881; and Chatterjee et al, 1992, Science 258:1485). Furthermore, an enzymatically active R ⁇ A, termed ribozyme, has been used to cleave viral transcripts.
  • the cellular components determine host cell susceptibility to infection, and can be used as potential targets for the development of new therapeutic interventions.
  • HIV one cellular component which has been used towards this end is the cell surface molecule for HIV attachment, CD4.
  • chemokine receptors CCR2, CCR3, CCR5 or CXCR4
  • CD4 a second type of receptor
  • a chemokine receptor normally binds RANTES, MlP-l ⁇ andMIP-l ⁇ as its natural ligand.
  • CD4 first binds to the HIV gpl20 protein on the cell surface followed by binding of this complex to a chemokine receptor, resulting in viral entry into the cells (Cohen, 1997, Science 275:1261). Therefore, chemokine receptors can present an additional cellular target for the design of HIV therapeutic agents.
  • Inhibitors of HlV/chemokine receptor interactions are being tested as anti-HIV agents.
  • additional cellular targets for the design of anti-HIV therapeutics particularly intracellular targets for disrupting viral replication after viral entry into a cell.
  • the present invention relates to compositions and methods for inhibiting HIV infection by down-regulating expression of certain human cellular genes and/or inhibiting the activity of products encoded by such genes.
  • it relates to a number of human cell-derived nucleic acid molecules which inhibit HIV infection in susceptible cells.
  • the isolated nucleic acid molecules correspond to portions of cellular genes or complements thereof, and are referred to herein as genetic suppressor elements (GSEs).
  • GSEs genetic suppressor elements
  • the cellular genes encode intracellular products necessary for productive HIV infection. Additionally, small molecule inhibitors ofthe same cellular genes and their encoded products are also within the scope ofthe present invention.
  • the invention also relates to methods for identifying additional cellular genes as therapeutic targets for suppressing HIV infection, and methods for using such cellular genes and their encoded products for selecting additional inhibitors of HIV.
  • nucleic acid molecules isolated from human cells can prevent both the activation of latent HIV- 1 in a CD4 + cell line and productive HIV infection in such cells, and that such nucleic acid molecules correspond to fragments of certain human cellular genes.
  • any cellular or viral marker associated with HIV infection can be used to select for such nucleic acid molecules.
  • An example of such a marker is CD4, which is conveniently monitored by using a specific antibody.
  • GSEs Based on substantial sequence identity (90%- 100%), a number ofthe isolated GSEs correspond to portions of human cellular genes which encode different subunits of a mitochondrial enzyme complex, NADH dehydrogenase. In addition, inhibitors of this enzyme also inhibit HIV infection in susceptible host cells, including freshly isolated human CD4 + T cells.
  • GSEs have been selected which have substantial sequence identity (90% - 100%) with the following human cellular genes: 2-oxoglutarate dehydrogenase, M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein, calnexin, ADP-ribosylation factor 3, eukaryotic initiation factor 3 , protein tyrosine phosphatase, herpesvirus-associated ubiquitin-specific protease, eukaryotic initiation factor 4B, CD44, phosphatidyl-inositol 3 kinase, elongation factor 1 alpha, bone morphogenic protein- 1 , double-strand break DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG-1), lymphocyte-specific protein 1, protein phosphatase 2A, squalene synthetase, eukaryotic release factor 1 , GTP binding protein, importin beta subunit
  • TCTP1 TCTP1
  • BBC1 Nef interacting protein
  • Na + -D-glucose cotransport regulator gene protein hsp90 chaperone protein
  • FK506-binding protein Al hsp90 chaperone protein
  • Rox beta signal sequence receptor
  • tumorous imaginal disc protein TCTP1
  • cell surface heparin binding protein TCTP1
  • the GSEs selected to inhibit HIV infection several function in the sense orientation, while others function in the antisense orientation.
  • the GSEs of the invention are believed to down-regulate a cellular gene by different mechanisms.
  • the GSEs are expressed in a host cell by encoding RNA molecules that do or do not encode protein products.
  • GSEs in the sense orientation can exert their effects as transdominant mutants or RNA decoys.
  • Transdominant mutants are expressed proteins or peptides that competitively inhibit the normal function of a wild-type protein in a dominant fashion.
  • RNA decoys are protein binding sites that titrate out these proteins.
  • GSEs in the antisense orientation can exert their effects as antisense RNA; i.e. nucleic acid molecules complementary to the mRNA of the target gene. These nucleic acid molecules bind to mRNA and block the translation ofthe mRNA. Some antisense nucleic acid molecules can act directly at the DNA level to inhibit transcription. The down-regulation of a cellular gene by a GSE, in turn, removes a cellular component necessary for, for example HIV replication, resulting in an inhibition of HIV infection.
  • GSEs can be transferred into T cells, particularly CD4 + T cells which are the major cell population targeted by HIV.
  • GSEs can be transferred into hematopoietic stem cells in vitro followed by their engraftment in an autologous or histocompatible or even histoincompatible recipient.
  • any cells susceptible to HIV infection can be directly transduced or transfected with GSEs in vivo.
  • Figure 1 illustrates the percentage of CD4 + OM10.1 cells that diminish after TNF- ⁇ induction; TNF-induced cells, - ⁇ -; uninduced cells,-4-.
  • Figure 2 illustrates the percentage of intracellular p24 + CEM-ss cells containing the CF-315 sequence (SEQ ID NO: 1) after infection with HIV-1 SF2 at a TCID 50 of 1000.
  • CEM-ss cells (10 6 ) containing the C-315 construct or control vector DNA were harvested on the indicated days post infection, stained with FITC- conjugated anti-p24 monoclonal antibody and analyzed by flow cytometry. Mock infected cells, -0-; LNGFRM vector-infected cells, -D-; and C-315 infected cells, - ⁇ -.
  • Figure 3 illustrates the percentage of intracellular p24 + CEM-ss cells containing various GSEs: CF-004 (SEQ ID NO:7), CF-025 (SEQ ID NO:5), CF-113 (SEQ ID NO:8) and CF-204 (SEQ ID NO:9) after infection with HTV-1 SF2 at a TCJD 50 of 1000.
  • Controls include mock-infected, vector (LNGFRM)-infected and HlV-infected CEM-ss cells.
  • Figure 4 illustrates the percentage of intracellular p24 + CEM-ss cells containing
  • CF-001 (SEQ ID NO: 10) after infection with HIV-l Sf2 at a TCID 50 of 1000.
  • Controls include mock-infected and vector (LNGFRM)-infected cells.
  • Figure 5 illustrates the percentage of CD4 + OM10.1 cells after treatment with amytal following TNF- ⁇ induction. TNF induction, -A-; no TNF induction, - ⁇ -.
  • Figure 6 illustrates the percentage of CD4 + OM10.1 cells after treatment with mofarotene following TNF- ⁇ induction. TNF induction, - ⁇ -; no TNF induction, - ⁇ -.
  • Figure 7 illustrates the percentage of intracellular p24 + CEM-ss cells containing various GSEs: CF-527 (SEQ ID NO:41), CF-529 (SEQ ID NO:45) and CF-531 (SEQ ID NO:47) after infection with HIV-1 S ⁇ at a TCU3 50 of 1000.
  • Controls include mock- infected, vector (LNGFRM)-infected and CEM-ss cells transfected with RevM 10.
  • Figure 8 illustrates the percentage of intracellular p24 + CEM-ss cells containing the GSE CF-579 (SEQ ID NO:61) after infection with HIV-1 SF2 at a TCID 50 of 1000.
  • Controls include mock-infected, vector (LNGFRM)-infected and CEM-ss cells transfected with RevM 10.
  • Figure 9 illustrates the percentage of intracellular p24 + CEM-ss cells containing various GSEs: CF-619 (SEQ ID NO:53), CF-620 (SEQ ID NO:55) and CF-624 (SEQ ID NO:57) after infection with fflV-l SF2 at a TCID 50 of 1000.
  • Controls include mock- infected, vector (LNGFRM)-infected and CEM-ss cells transfected with RevMlO.
  • the present invention includes novel methods to identify genetic suppressor elements capable of inhibiting HIV infection, genetic suppressor elements identified by such methods, nucleic acid molecules representing host cellular genes involved in HIV infection and inhibitory compositions that inhibit HIV infection by down-regulating the expression of such cellular genes or inhibit the activity ofthe products of such cellular genes.
  • HIV infection refers to the ability of HIV to enter a host cell and/or replicate in the host cell.
  • One embodiment of the present invention is a method for isolating genetic suppressor elements, referred to herein as GSEs, comprising the steps of: 1) randomly fragmenting cell-derived cDNA into fragments; 2) inserting the fragments into expression vectors to form a random fragment expression (RFE) library; 3) transferring the expression library into a population of cells containing an inducible latent HIY-1 provirus or susceptible to HIV infection; 4) selecting a subpopulation of cells which contain a subset of the expression library enriched for GSEs by monitoring the expression of a cellular or viral marker associated with HIV infection; and 5) recovering the GSEs from the selected cell population.
  • GSEs genetic suppressor elements
  • the cell-derived cDNA is randomly- fragmented into 100-700 base pair (bp) fragments.
  • the method further includes repetition ofthe aforementioned steps so that many rounds of successive selection can be performed.
  • the method can further comprise the step of selecting GSEs by determining the continued expression of a cellular marker such as CD4 or the decreased expression of a viral marker such as p24 or gpl20 using, for example, an antibody.
  • a cell-derived RFE library can be constructed from nucleic acid molecules of any mammalian cells preferably from cDNA of HTV-susceptible cells.
  • Example 1 demonstrates that GSEs can be selected from HL-60 cells that are naturally susceptible to HIV infection and from HeLa cells which are not naturally susceptible to HIV infection due to the lack of CD4 expression.
  • CD4 expression of CD4 on the surface of HeLa cells by means of a retroviral vector renders the cells susceptible to HIV infection. Therefore, cell types not normally susceptible to HIV infection can still be useful as a source of genetic material for the construction of
  • RFE libraries It is also preferred that a normalized cDNA library is prepared (Gudkov and Roninson, 1996, Methods in Molecular Biology 69:229-231 ). DNA is first treated with enzymes to produce randomly cleaved fragments. This can be conveniently performed by DNase I cleavage in the presence of Mn "1 ⁇ (Roninson et al, U.S. Patent No. 5,217,889, column 5, lines 5-20). Thereafter, the randomly-cleaved DNA is size fractionated by gel electrophoresis. Fragments of between 100 and 700 bp are the preferred lengths for constructing RFE libraries. Single strand breaks of the size- selected fragments are repaired by methods well known in the art.
  • the fragments are ligated with 5' and 3' adaptors, which are selected to have non- cohesive restriction sites so that each fragment can be inserted into an expression vector in an oriented fashion. Further, the 5' adaptor contains a start (ATG) codon to allow the translation ofthe fragments which contain an open reading frame in the correct phase.
  • the fragments are then inserted into appropriate expression vectors. Any expression vector that results in efficient expression ofthe fragments in host cells can be used.
  • viral-based vectors such as the retroviral vectors LNCX (Miller and Rosman, 1989, BioTechniques 7:980) and LNGFRM are exemplified.
  • adenovirus, adeno-associated virus and herpes virus vectors can also be used for this purpose.
  • the ligated vectors are first transfected into a packaging cell line to produce viral particles.
  • a packaging cell line for retroviral vectors, any amphotropic packaging line such as PA317 (Miller and Buttimore, 1986, Mol. Cell. Biol. 6:2895- 2902; ATCC CRL #9078) can be used to efficiently produce virus.
  • the viral vector also contains a selectable gene, such as the neo r gene or a truncated nerve growth factor receptor (NGFR) gene, which allows isolation ofthe cells that contain the vector.
  • NGFR nerve growth factor receptor
  • the number of independent clones present in each RFE expression library can vary. In a preferred embodiment, libraries of cell-derived cDNA of about 10 6 to 10 8 independent clones can be used.
  • OM10.1 cells are used to select for GSEs, and are maintained in conventional tissue culture as described in Butera, U.S. Patent No. 5,256,534.
  • the purpose of using OM10.1 cells for the selection of GSEs is that they contain a latent HIV- 1 provirus which is inducible by
  • TNF- ⁇ TNF- ⁇ .
  • Other cell lines can be similarly engineered with an inducible HIV provirus. Examples of cell lines that are infected with latent HIV include, but are not limited to, Ul, U33, 8E5, ACH-2, LL58, THP/HIV and UHC4 (Bednarik and Folks, 1992, AIDS 6:3-16).
  • agents have been shown to be capable of inducing latent HTV- infected cells, and these include TNF- ⁇ , TNF- ⁇ , interleukins-1, -2, -3, -4 and -6, granulocyte-macrophage colony stimulating factors, macrophage-colony stimulating factors, interferon- ⁇ , transforming growth factor- ⁇ , PMA, retinoic acid and vitamin D3 (Poli and Fauci, 1992, AIDS Res. Human Retroviruses 9:191-197).
  • GSEs can be selected on the basis of their ability to directly protect HIV-susceptible cells from HIV infection using methods described herein.
  • the cell-derived RFE library can be introduced into latently HlV-infected cells or HIV-susceptible cells by any technique well known in the art that is appropriate to the vector system employed.
  • the viral vector also contains a selectable marker in addition to a random fragment of cellular DNA.
  • a suitable marker is the neo r gene, which permits selection of cells containing RFE library members using the drug G-418.
  • the viral vector contains a truncated low affinity nerve growth factor receptor (NGFR) which permits selection of the cells using an anti-NGFR monoclonal antibody.
  • NGFR nerve growth factor receptor
  • the multiplicity of infection ofthe virions ofthe library is adjusted so that pre-selection for cells that are transduced by the vector is not needed.
  • the transduced cell population is treated with 10 U/mL TNF- ⁇ for a period of 24-72 hours and preferably about 24 hours according to the method of Butera.
  • the activation of the latent HIV-1 provirus in OMlO.l can be detected by the suppression of the cell surface CD4. (It is believed that viral protein gpl20 binds to CD4 in the cytoplasm, which prevents subsequent expression of CD4 on the cell surface.)
  • Clones that are resistant to HIV replication continue to express cell surface CD4. Such clones can be selected, for example, by cell sorting using any antibody staining technique for CD4 and a fluorescence activated cell sorter (FACS).
  • FACS fluorescence activated cell sorter
  • the fraction of CD4 + cells that have been transduced with the RFE library can be compared with cells transduced with an expression library consisting ofthe vector only.
  • An increased relative difference between the cell-derived RFE library and the control library can be found with each additional round of TNF- ⁇ induction.
  • nucleic acid molecules corresponding to the GSEs can be recovered from cells that continue to express CD4 following induction ofthe latent
  • the specific GSEs are recovered from genomic DNA isolated from CD4 + cells sorted by FACS after TNF- ⁇ induction.
  • the GSEs in this population are preferably recovered by PCR amplification using primers designed from the sequences ofthe vector.
  • the recovered GSEs can be introduced into an expression vector as discussed in the Examples section herein.
  • the resultant GSEs expression library is known as a secondary library.
  • the secondary library can utilize the same or a different vector from that used for the construction of the primary library.
  • the secondary library can be transduced into another cell population and the resultant population selected, recloned and processed as described herein.
  • each individually recovered GSE can be inserted into cloning vectors for determining its specific nucleotide sequence and its orientation.
  • the sequence ofthe GSE is then compared with sequences of known genes to determine the portion ofthe cellular gene with which it corresponds.
  • the PCR products themselves can be directly sequenced to determine their nucleotide sequences.
  • the isolated GSEs can be analyzed to determine their minimal core sequences.
  • a "core sequence” is a common sequence found by comparison of GSEs with overlapping sequences. The GSEs are further tested for their ability to protect previously uninfected cells from HIV infection.
  • Another embodiment ofthe present invention includes a method for determining the core sequence of a GSE. This can be done by comparing overlapping sequences of independently derived GSEs. Alternatively, GSEs can be altered by additions, substitutions or deletions and assayed for retention of HlV-suppressive function. Alterations in the GSEs sequences can be generated using a variety of chemical and enzymatic methods which are well known to those skilled in the art. For example, oligonucleotide-directed mutagenesis can be employed to alter the GSE sequence in a defined way and/or to introduce restriction sites in specific regions within the sequence.
  • deletion mutants can be generated using DNA nucleases such as Bal 31 or Exo HI and SI nuclease. Progressively larger deletions in the GSE sequences can be generated by incubating the DNA with nucleases for increased periods of time (see Ausubel, et al, ibid., for a review of mutagenesis techniques). The altered sequences can be evaluated for their ability to suppress expression of HIV proteins such as p24 in appropriate host cells. It is within the scope of the present invention that any altered or shortened GSE nucleic acid molecules that retain their ability to suppress HIV infection can be inco ⁇ orated into recombinant expression vectors for further use.
  • the GSEs can be transferred into latently HIV infected or into HIV-susceptible host cells followed by HIV infection.
  • GSEs also can be directly selected from a RFE library for their ability to prevent productive infection by HIV, as shown in a specific embodiment exemplified in the Examples section herein. Protection experiments can be performed in any cell type that takes up the potential GSEs and which is otherwise susceptible to HIV infection. In a preferred embodiment by way of example, the CEM-ss cell line is used (Foley et al. 1965, Cancer 18:522-529).
  • CEM-ss cells as targets for quantitative infectivity of HlY-l has been described by Nara & Fischinger (1988, Nature 322:469-470).
  • Other cell lines that are susceptible to HIV infection include, but are not limited to, HUT-78, H9, Jurkat E6-1, A3.01, U-937,
  • AA-2 HeLa CD4 + and C8166.
  • freshly isolated peripheral blood leukocytes can be used.
  • the test of the potential GSEs can be performed using the same expression vector system as that employed in the RFE library transduction of cells during initial selection steps.
  • the vector system can be modified to achieve higher levels of expression, e.g. , the linkers can be employed to introduce a leader sequence that increases the translational efficiency ofthe message.
  • One such sequence is disclosed by Kozak, 1994, Biochemie 76:815-821.
  • Another way of testing the effectiveness of a potential GSE against HIV infection is to determine how rapidly HIV-1 variants develop that can negate the effects of that element.
  • Such a test includes infection of a culture of susceptible cells such as CEM-ss cells at a low multiplicity of infection and repeatedly assaying the culture to determine whether and how quickly HIV-1 infection becomes widespread.
  • the range of useful multiplicities of infection is between about 100 to 1000 tissue culture infectious units (TCID 50 ) per 10 6 CEM-ss cells.
  • the TCID 5() is determined by an endpoint method and is important for determining the input multiplicity of infection (moi).
  • a parameter that correlates with the development in the test culture of HIV-1 strains that are resistant to the effects ofthe potential GSEs is the fraction of cells that are infected in the culture.
  • This fraction can be determined by immunofluorescent staining with an antibody specific for the HIV-1 p24 antigen of fixed permeabilized cells.
  • Commercially available reagents are suitable for performing such tests (Lee et al, 1994, J. Virol. 68:8254-8264).
  • One embodiment of the present invention is an isolated nucleic acid molecule comprising a human cellular gene, or at least a portion thereof, that is necessary for HIV infection.
  • isolated nucleic acid molecules are referred to herein as "cell-derived nucleic acid molecules.”
  • a or “an” entity refers to one or more of that entity; for example, a protein refers to one or more proteins or at least one protein.
  • the terms “a” (or “an”), “one or more” and “at least one” can be used interchangeably herein.
  • the terms “comprising”, “including”, and “having” can be used interchangeably.
  • an isolated nucleic acid molecule is a nucleic acid molecule that has been removed from its natural milieu (i.e., that has been subject to human manipulation). As such, “isolated” does not reflect the extent to which the nucleic acid molecule has been purified.
  • An isolated nucleic acid molecule can include DNA, RNA, or derivatives or hybrids of either DNA or RNA.
  • An isolated nucleic acid molecule ofthe present invention can be obtained from its natural source either as an entire (i.e., complete) gene or a portion thereof corresponding to at least a portion of the gene that encodes a product necessary for productive HIV infection or necessary to inhibit HTV infection.
  • the phrase "at least a portion of an entity refers to an amount ofthe entity that is at least sufficient to have the functional aspects of that entity.
  • at least a portion of a nucleic acid sequence is an amount of a nucleic acid sequence necessary for HIV infection.
  • isolated nucleic acid molecule ofthe present invention can also be produced using recombinant DNA technology (e.g., polymerase chain reaction (PCR) amplification, cloning) or chemical synthesis.
  • Isolated nucleic acid molecules of the present invention include natural nucleic acid molecules and homologs thereof, including, but not limited to, natural allelic variants and modified nucleic acid molecules in which nucleotides have been inserted, deleted, substituted, and/or inverted in such a manner that such modifications do not substantially interfere with the nucleic acid molecule's ability to promote HIV infection or inhibit HIV infection.
  • a nucleic acid sequence complement of any nucleic acid sequence of the present invention refers to the nucleic acid sequence of the nucleic acid strand that is complementary to (i. e. , can form a complete double helix with) the strand for which the sequence is cited. It is to be noted that a double-stranded nucleic acid molecule ofthe present invention for which a nucleic acid sequence has been determined for one strand that represented by a SEQ ID NO also comprises a complementary strand having a sequence that is a complement of that SEQ ID NO.
  • nucleic acid molecules of the present invention which can be either double-stranded or single-stranded, include those nucleic acid molecules that form stable hybrids under stringent hybridization conditions with either a given SEQ ID NO denoted herein and/or with the complement of that SEQ ID NO, which may or may not be denoted herein.
  • Methods to deduce a complementary sequence are known to those skilled in the art.
  • a cell-derived nucleic acid molecule is a GSE nucleic acid molecule that is capable of inhibiting HTV infection in a susceptible cell.
  • a preferred GSE nucleic acid molecule ofthe present invention comprises a nucleic acid sequence selected from the group consisting of SEQ ID NO: 1 , SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO: 1 , SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, S
  • Highly stringent hybridization conditions can be defined as hybridization to filter-bound DNA in 0.5 M NaHPO 4 , 7% sodium dodecyl sulfate (SDS), lmM EDTA at 65°C, followed by washing in 0.1 x SSC/0.1% SDS at 68°C (Ausubel F.M. et al, eds, 1989, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Vol. I, Green
  • Moderately stringent conditions can be defined as hybridizations carried out as described above, followed by washing in 0.2 x SSC/0.1% SDS at 42°C (Ausubel et al, 1989, CURRENT PROTOCOLS FOR MOLECULAR BIOLOGY).
  • a cell-derived nucleic acid molecule of the present invention comprises a cellular gene that encodes an intracellular product necessary for productive HIV infection, referred to herein as a "target gene.”
  • a target gene ofthe present invention comprises a nucleic acid molecule that corresponds to a GSE having a nucleic acid sequence selected from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:
  • GSE nucleic acid molecules ofthe present invention include plasmids CF-315, CF-319, CF-101, CF-117, CF-025, CF-128, CF-004, CF- 113, CF-204, CF-001, CF-273, CF-311, CF-313, CF-210, CF-266, CF-302, CF-317, CF-286, CF-061, CF-280, CF-537, CF-320, CF-321, CF-322, CF-332, CF-335, CF- 42, CF-50, CF-527, CF-528, CF-529, CF-531, CF-545, CF-547, CF-619, CF-620, CF-624, CF-630, CF-579, CF-676, CF-675, CF-653, CF-674, CF-675, CF-673, CF-
  • the term "corresponds to” refers a nucleic acid sequence that is at least about 75%, more preferably about 80%, more preferably about 85%, more preferably about 90%, more preferably about 95% and more preferably about 100% identical to nucleic acid sequence SEQ ID NO: 1 , SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:ll, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO:20, SEQ ID NO:25, SEQ ID NO:27, SEQ ID NO:29, SEQ ID NO:31, SEQ ID NO:
  • GSEs SEQ ID NO:83, SEQ ID NO:85; SEQ ID NO:87; SEQ ID NO:89; SEQ ID NO:91; SEQ ID NO:93 or SEQ ID NO:95 and complements thereof.
  • the lack of complete identity between the GSEs and the target gene sequences can result from genetic polymorphism between different individuals or mutations introduced during cloning or PCR amplification.
  • a preferred embodiment of the present invention includes a target gene that encodes at least a portion a protein selected from the group consisting of NADH dehydrogenase, 2-oxoglutarate dehydrogenase, M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein, calnexin, ADP-ribosylation factor 3, eukaryotic initiation factor 3, protein tyrosine phosphatase, herpesvirus-associated ubiquitin- specific protease, eukaryotic initiation factor 4B, CD44, phosphatidyl-inositol 3 kinase, elongation factor 1 alpha, bone morphogenic protein- 1 , double-strand break DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG- 1 ), lymphocyte-specific protein 1 , protein phosphatase 2 A, squalene synthetase, eukaryotic release
  • adenosine triphosphate provides the major source of energy.
  • energy rich molecules such as NADH and FADH 2 are first formed in glycolysis, fatty acid oxidation and the citric acid cycle. When these molecules donate their electrons to molecular oxygen, free energy is released to generate ATP.
  • Oxidative phosphorylation is the process by which ATP is formed as electrons are transferred from NADH or FADH 2 to O 2 by a series of electron carriers (Stryer,
  • the 2-oxoglutarate dehydrogenase complex catalyzes oxidative decarboxylation of 2-oxoglutarate to succinyl-CoA and CO 2 , and is the rate-limiting enzyme which controls the flux of substrates through the Krebs cycle (Delvin, T.M., ed, 1992,
  • This enzyme complex is located in the inner membrane/matrix compartment of the mitochondria.
  • the complex consists of multiple copies of 2-oxoglutarate dehydrogenase (lipoamide) (OGDH or ELO; 2- oxoglutarate: lipoamide 2-oxidoreductase (decarboxylating and acceptor succinylating), EC 1.2.4.2), dihydro lipoamide succinyltransferase (designated E20; EC 2.3.1.61) and dihydrolipoamide dehydrogenase (E3; EC 1.8.1.4).
  • the coding sequence of 2- oxoglutarate dehydrogenase has been described (GenBank Accession Nos. D 10523 and
  • Pyruvate kinase/thyroid hormone binding protein p58 is a monomer of pyruvate kinase (ATP pyruvate O 2 -phosphotransferase, EC 2.7.1.40) subtype M2. Its conversion to the tetrameric pyruvate kinase is regulated by fructose 1 ,6,-bisphosphate
  • Fru-1,6-P 2 pyruvate kinase
  • mammalian cells contain low Fru-1,6-P 2 and pyruvate kinase is inactive.
  • high glucose concentration regular medium contains 5-10 mM glucose
  • high levels of Fru-1,6-P 2 are found in proliferating and tumor cells, which require high pyruvate kinase activity for growth. It has been demonstrated that low Fru-1,6-P 2 favors formation of p58 and high concentrations convert it to the tetrameric enzyme. An increase in glucose concentration could lead to multimerization of p58 which in turn activates pyruvate kinase and glycolysis.
  • thyroid hormone is released from the complex with TBP and might bind to nuclear and mitochondrial receptors and activate oxidative phosphorylation.
  • the coding sequence of pyruvate kinase/thyroid hormone binding protein has been described (GenBank Accession No. M26252; Kato et al, 1989, Proc. Natl. Acad. Sci. USA 86:7861-7865).
  • Calnexin is a type I membrane protein which functions as a molecular chaperon for secretory glycoproteins in the endoplasmic reticulum (ER) with ATP and Ca ⁇ as two cofactors involved in the substrate binding (Ou et al. , 1995, J. Biol. Chem. 270: 18051 ).
  • ADP-ribosylation factors are guanine nucleotide binding proteins of about 20kDa molecular weight that stimulate ADP-ribosyltransferase activity of cholera toxin in vitro (Tsai et al, 1991 , J Biol. Chem. 266:23053-23059). Five different ARFs from human cDNA have been cloned. ARF3 is represented by two mRNAs of 3.7 and
  • Ubiquitin-specific protease plays an important role in several cellular processes, including the regulation of gene expression, control ofthe cell cycle, DNA repair and differentiation (Hochtrasser, 1995, Curr. Opin. Cell. Biol. 7:215-223;
  • ubiquitin-dependent pathways involved in the control of gene expression is activation of NF-KB. Cleavage of pl05, a precursor of the p50 subunit of NF-KB requires ubiquitin conjugation (Palombella et al, 1994, Cell 78:773-785) and secondly the destruction of 1KB (a process which allows NF- ⁇ B to migrate to the nucleus in an active form) requires ubiquitination ofthe inhibitor in a phosphorylati on-dependent manner (Scherer et al, 1995, Proc. Natl. Acad. Sci. USA 92:11259-11263).
  • USP is characterized by the presence of two conserved active site domains and has been shown to cleave ubiquitin from model substrates (Everett et al, 1997, EMBO J. 16: 556-577).
  • the second comprises an increasing number of de-ubiquitinating proteins which can recognize and stabilize specific substrates by removing ubiquitin adducts.
  • Examples of this class include the Drosophila fat facets protein, whose de-ubiquitination is required for proper eye development (Huang et al, 1995, Science 270:1828-1831).
  • Another example is the DUB- 1 gene which is an immediate early gene that regulates cell growth (Zhu et al, 1996, Proc. Natl. Acad. Sci. USA 93:3275-3279).
  • CD44 is a broadly distributed surface receptor glycoprotein implicated in multiple physiologic cellular functions, such as extracellular matrix-cell adhesion, lymphocyte homing, lympho-hematopoiesis, T cell activation, and tumor metastasis (Shimuzu et al, 1989, J Immunol. 143:2457-2463; Huet et al, 1989, J. Immunol. 143:798-801).
  • CD44 is expressed as several different isoforms, varying between 85 to 200 kDa, depending on differential usage of 10 exons encoding a portion ofthe extracellular domain and cell type specific glycosylation. Each isoform can display some degree of functional uniqueness (Stamenkovich et al, 1989, Cell 56:1057-1062). The most widely expressed molecule is the 85-90 kDa glycoprotein, commonly referred to as CD44H, which has been demonstrated to be the major receptor for hyaluronic acid (Bartolazzi et ⁇ /., 1996,J Cell Biol. 132:1199-1208).
  • CD44H represents the principle isoform found on hematopoietic cells. It has been shown that CD44, along with other HIV receptors like CD4, can play a role in viral tropism and affects infectivity of the virus. HIV causes CD44 downmodulation in monocytes, but on a post-translational level (Guo and Hildreth, 1993, J. Immunol.
  • Tyrosine phosphorylation is important in the control of normal cellular processes such as cell proliferation and differentiation, as well as pathological events such as malignant transformation (Cantley et al, 1991, Cell 64:281-301).
  • the overall levels of tyrosine phosphorylation are modulated by the complementary and antagonistic actions of protein tyrosine kinases and protein tyrosine phosphatases. In the steady state, less that 1% of the total cellular phosphorylation is due to tyrosine phosphorylation.
  • BDP1 Phosphatase 1 mRNA and CL 100 mRNA have been described (GenBank accession No. X79568; Kim et al, 1996, Oncogene 13:2275-2279 and GenBank accession No. X68277; Keyse and Emslie, 1992, Nature 359:644-647).
  • Phosphatidylinositol 3 -kinases have been characterized as enzymes involved in receptor signal transduction. Multiple forms with different substrate specificities exist. They have been associated with a diverse range of cell surface receptors including those for growth factors, thrombin, chemotactic peptides and cytokines. It has been proposed that PI3Ks act as second messengers, possibly via the activation of certain protein kinase C isotypes (Liscovitch and Cantley, 1994, Cell 77:329-334; Toke ⁇ etaL, 1994, J Biol. Chem. 269:323598-32367). PI3K can also play a role in the regulation of protein trafficking from the Golgi to the lysosome (Volinia et al, 1995, EMBO J. 14:3339-3348).
  • HIV-1 nef expression severely impairs PI3K association with a receptor suggesting that Nef selectively affects the PI3K signaling pathway resulting in adverse effects on host cell function (Graziani et ⁇ /. , 1996,J Biol. Chem. 271 :6590-6593).
  • Nef expression is also accompanied by a decrease in basal intracellular PIK, suggesting a role for PI3K in HTV replication (Garcia, 1997, C.R. Acad. Sci. 111320:505-50 ).
  • PI3K can be activated by gpl60 and also has been implicated in Tat-mediated apoptosis (Mazerolles et al, 1997, Eur. J. Immunol.
  • inhibitors of PI3K include wortmannin and theophylline.
  • the coding sequence of phosphatidylinositol 3-kinase has been described
  • Elongation of the polypeptide chain occurs following initiation of translation. Elongation factors utilized the energy released by GTP hydrolysis to ensure selection of the proper aminoacyl-tRNA and to move the message and associated tRNAs through the decoding region ofthe ribosome (Devlin, ed, 1992, Textbook of Biochemistry, Wiley-
  • EF-l- ⁇ is an evolutionarily conserved universal cofactor of protein synthesis in all living cells. It carries aminoacyl-tRNAs to the A-site of the ribosome in GTP- dependent manner.
  • the expression levels of EF-1 ⁇ are regulated in various stages of cell life such as growth arrest, transformation and aging. Levels of EF-1 ⁇ can be a key regulator in modulating the rate of apoptosis. Reduction of EF- 1 - ⁇ expression decelerates apoptosis while overexpression accelerates the process (Duttaroy et al, 1998, Exp. Cell Res.
  • Initiation of translation occurs by the binding ofthe 40S ribosomal subunit at or near the cap structure of mRNA followed by ribosome scanning ofthe 5' untranslated region until an initiator AUG is encountered. This process is promoted by a complex group of proteins known as initiation factors. These factors participate only in initiation of translation (Devlin, ed, 1992, Textbook of Biochemistry, Wiley-Liss, Inc.).
  • Eukaryotic initiation factor 3 (eIF3) is the largest multisubunit complex involved in initiation of protein synthesis. It has a mass of 600 kDa and 10 subunits. The factor prevents association of ribosomal subunits, stabilizes methionyl-tRNA binding to the 40S subunits and promotes mRNA binding (Merrick and Hershey, 1996, in Translational Control. Hershey, Mathews and Sonenberg eds, pp31-69, Cold Spring Harbor, Cold
  • Eukaryotic initiation factor 4B is an 80 kDa polypeptide that is essential for the binding of mRNA to ribosomes. eIF4B has RNA binding activity, stimulates ATPase and RNA helicase (Mehot et al, 1996, RNA 2:38-50; Abramson et al, 1987, J. Biol. Chem. 262:3826-3832; Ray et al, 1985, J. Biol. Chem. 260:7651-7658; Rozen et al, 1990, Mol. Cell. Biol. 10:1134-1144).
  • eIF4B has been reported to have RNA annealing activity, promoting base pairing between complementary sequences in RNA strands (Altmann et al, 1995, EMBO J. 14:3820-3827). It also interacts with eIF4A and eU3 (Methot et al, 1996, Mol. Cell. Biol. 16:5328-5334). Overproduction of eIF4B results in a general inhibition of translation (Milburn et al, 1990, EMBO J. 9:2783-2790).
  • Protein extracts derived from bone can initiate the process that begins with cartilage formation and ends in de novo bone formation.
  • the protein extract is referred to as bone morphogenic protein (BMP).
  • BMP bone morphogenic protein
  • Amino acid sequence has been derived from a highly purified preparation of BMP from bovine bone. Human complementary DNA clones corresponding to three polypeptides present in a BMP preparation have been isolated, and expression of the recombinant human proteins have been obtained.
  • BMP-1 Each of the three expressed human proteins, BMP-1, BMP-2A, and BMP-3, appears to be independently capable of inducing the formation of cartilage in vivo.
  • BMP-2A and BMP-3 are new members ofthe TGF-beta supergene family, while the third, BMP-1 , is a regulatory molecule.
  • a 2.2 kb transcript was present at a high level in postmeiotic spermatids, while expression of the 3.1 kb mRNA in testis was confined to the meiotic compartment.
  • hHR21 sp mRNA was cell cycle regulated in human cells, increasing in late S phase and peaking in G2 phase. In situ hybridization showed that mHR21sp resided on chromosome 15D3, whereas hHR21sp localized to the syntenic 8q24 region.
  • Elevated expression of mHR21sp in testis and thymus indicates a role for the rad21 mammalian homologs in V(D) J immunoglobulin gene and meiotic recombination, respectively.
  • Cell cycle regulation of rad21, conserved in S. pombe and humans, is consistent with a conservation of function between S. pombe and human rad21 genes.
  • GTP -binding proteins ofthe ras superfamily are important for exocytosis from eukaryotic cells. These GTP -binding proteins can exist in two different conformations, depending on whether they are bound to GDP or GTP, and function as molecular switches that regulate a variety of cellular processes. The GTP-GDP cycle is controlled by accessory proteins that promote the exchange of bound GDP or the hydrolysis of GTP. cDNA encoding a mammalian GDP releasing protein, mss4, has been cloned (Burton et al, 1993, Nature 361:464-467).
  • Mss4 is a guanine nucleotide exchange factor that specifically binds to and promotes GDP-GTP exchange on a subset of the Rab GTPases (Burton et al.,1994, EMBO J. 13:5547-5558).
  • the Mss4 protein also stimulates GDP release from Ypt 1 and from the mammalian protein Rab3 a, but not from Ras2.
  • Mss4 shows sequence similarity to Dss4, a yeast protein with similar biochemical properties.
  • B-cell translocation gene 1 (BTG1), a member of an antiproliferative protein family including Tis-21/PC3 and Tob, regulates cell cycle progression (Rodieretal, 1999, Exp CellRes.249:337-348).
  • TheBTGl gene locushas been shown to be involved in a t(8; 12)(q24;q22) chromosomal translocation in a certain B-cell chronic lymphocytic leukemia (Rouault et al, 1992, EMBO J. 11:1663-1670).
  • the cDNA for BTGl was isolated (Rouault et al, ibid.) and contains an open reading frame of 171 amino acids.
  • BTG1 expression is maximal in the G0/G1 phases ofthe cell cycle and is down-regulated when cells progress throughout Gl. Furthermore, transfection experiments of NIH3T3 cells indicate that BTG1 negatively regulates cell proliferation.
  • the BTG1 open reading frame is 60% homologous to PC3, an immediate early gene induced by nerve growth factor in rat PC 12 cells. Sequence and Northern blot analyzes indicate that BTGl and PC3 are not cognate genes. Triiodothyronine (T3) or 8-Br-cAMP increases BTGl nuclear accumulation in confluent myoblast cultures (Rodier et al, ibid.).
  • LSP 1 lymphocyte-specific proteinl
  • Protein phosphatases can control the activity of various protein kinases.
  • Protein phosphatase 2A (PP2A) regulates cell growth and division.
  • Investigators have suggested that HIV infection activates protein phosphatase 2 A (Han etal, 1992,J. Virol. 66:4065-
  • NCp7 and Vpr form a tight complex which becomes a more potent activator of PP2 A than NCp7 alone.
  • the ability of NCp7 to activate protein PP2A is regulated by Vpr.
  • the C-terminal portion of Vpr prevents NCp7 from activating protein PP2 A while the N-terminal portion of Vpr potentiates the effect of NCp7 on the activity of PP2A.
  • Vpr acts as a targeting subunit which directs NCp7 to activate protein PP2A (Tung et al, 1997, FEBS Lett. 401:197-201).
  • SQS squalene synthetase
  • RhS phytoene synthetase
  • hSQS 2 kb cDNA
  • both mRNAs are induced 2-fold to 4-fold by the 3-hydroxy-3- methylglutaryl-coenzyme A reductase inhibitor, lovastatin.
  • Northern blot analysis of rat tissues reveals only a 2.0 kb mRNA, which is considerably up-regulated in vivo by lovastatin.
  • the termination of protein synthesis in ribosomes is governed by termination codons in messenger RNAs and by polypeptide chain release factors (RFs). Amino acid sequences of members ofthe eRFl family are highly conserved. These RF proteins are directly implicated in the termination of translation in eukaryotes ( Frolova et al. , 1994, Nature 372: 701-703).
  • Prokaryotic and eukaryotic cells incorporate the amino acid selenocysteine at a
  • UGA codon which conventionally serves as a termination signal.
  • Translation of eukaryotic selenoprotein mR ⁇ A requires a nucleotide selenocysteine insertion sequence in the 3 '-untranslated region. Erb-1 can recognize a selenocysteine insertion sequence element.
  • eRFl associates with the catalytic subunit of protein phosphatase 2A (Andjelkovic et al, 1996, EMBO J. 15:7156-67). It was postulated that eRFl also functions to recruit PP2A into polysomes, thus bringing the phosphatase into contact with putative targets among the components ofthe translational apparatus.
  • retinoic acid induction of granulocyte differentiation of HL60 cells results a transient and reversible interconversion of phosphatase 2A holoenzyme and that the C-terminus of PP2A catalytic subunit is transiently methylated in S phase of HL-60 cells (Zhu, 1997, Arch Biochem Biophys. 339:210-217).
  • G-proteins are a family of heterotrimeric guanine nucleotide-binding proteins that play important roles in signal transduction and whose expression is regulated in a tissue-specific manner.
  • G(olf) alpha is a G-protein originally believed to mediate signal transduction exclusively within the olfactory neuroepithelium and subsequently found to be a major stimulatory G-protein in the basal ganglia and several other tissues (Zigman et al, 1993, Endocrinology 133:2508-2514 ).
  • Nucleocytoplasmic transport takes place through nuclear pores. Peripheral pore structures interact with transport receptors and their cargo when these receptor complexes first encounter the pore. Protein nuclear import is mediated by basic nuclear localization signals (NLSs) that bind to the importin alpha (Imp alpha) NLS receptor.
  • NLSs basic nuclear localization signals
  • Imp beta is also necessary for nuclear import.
  • HIV-1 Rev protein binds to unspliced HIV-1 pre-mRNA and exports it from the nucleus. Rev itself can "shuttle" between the nucleus and cytoplasm. This bi-directional transport is mediated by two specific Rev sequences: a NLS, which overlaps the RNA-binding domain, and a distinct nuclear export signal (NES).
  • Imp beta supports Tat or Rev nuclear import (Truant et al. , 1999, Mol Cell Biol, 19:1210-7) through the classical NLS pathway as demonstrated by inhibition of Imp beta interaction with Tat and Rev by RanGTP but not RanGDP.
  • Importin beta also interacts with the HIV-1 protein Vpr (Jenkins et al, 1998, J
  • Vpr contains two discrete nuclear targeting signals that use two different import pathways, both of which are distinct from the classical NLS pathway. Vpr import does not appear to require Ran-mediated GTP hydrolysis and persists under conditions of low energy. Vpr bypasses many of the soluble receptors involved in import of cellular proteins. Vpr directly accesses the NPC, a property that can help to ensure the capacity of HIV to replicate in nondividing cellular hosts (Jenkins et al, ibid.). Overexpression of either Vpr or importin beta in yeast blocks nuclear transport of mRNAs.
  • Vpr F34I A mutant form of Vpr, Vpr F34I, that does not localize at the nuclear envelope, or bind to importin alpha and nucleoporins, renders HIV-1 incapable of infecting macrophages efficiently.
  • Vpr F34I still causes G2 arrest, demonstrating that the dual functions of Vpr are genetically separable (Vodicka et al, 1998, Genes Dev, 12:175-185).
  • the rodent, avian, and insect LI -like cell adhesion molecules are members ofthe immunoglobulin superfamily that have been implicated in axon growth.
  • the entire coding region of human LI CAM has been cloned and found to have a high degree of homology to mouse LI CAM, with 92% identity at the amino acid level (Hlavin et al, 1991, Genomics 11 :416-423). This similarity suggests that LI CAM is an important molecule in normal human nervous system development and nerve regeneration. This molecule has never been associated with HIV-1 life-cycle.
  • Cyclophilins have been proposed to act as chaperones in a variety of cellular processes. U4/U6 snRNP-associated cyclophilin has been cloned and sequenced (Horowitz et al, 1997, RNA 3:1374-1387).
  • the nucleotide sequence for the recepin gene, a novel human liver cDNA encoding a serpin-like molecule has been directly submitted to GenBank.
  • Arg and c-Abl represent the mammalian members ofthe Abelson family of protein-tyrosine kinases.
  • the Arg/Abl-binding protein, ArgBP2 was isolated using a segment of the Arg COOH-terminal domain as bait in the yeast two-hybrid system
  • ArgBP2 contains three COOH- terminal Src homology 3 domains, a serine/threonine-rich domain, and several potential Abl phosphorylation sites. ArgBP2 associates with and is a substrate of Arg and v-Abl, and is phosphorylated on tyrosine in v-Abl-transformed cells. ArgBP2 is widely expressed in human tissues and extremely abundant in heart. In epithelial cells, ArgBP2 is located in stress fibers and the nucleus, similar to the reported localization of c-Abl.
  • ArgBP2 is located in the Z-disks of sarcomeres. These observations indicate that ArgBP2 functions as an adapter protein to assemble signaling complexes in stress fibers, and that ArgBP2 is a link between Abl family kinases and the actin cytoskeleton.
  • Interferon (IFN)-gamma has been implicated in the pathogenesis of several autoimmune disorders and inflammatory skin diseases.
  • a cDNA detecting a 1.6 kb mRNA that accumulated in response to IFN-gamma but not in response to UN-alpha or IFN-beta has been cloned and sequenced (Flohr et al, 1992, Eur J Immunol. 22:975- 979).
  • the gene is regulated by IFN-gamma in human cell lines of epithelial origin.
  • the mRNA encodes a predicted protein of 432 amino acids and the primary structure ofthe protein demonstrates that it is a member of developmentally regulated keratin class I genes.
  • the oncoprotein 18 (Op 18) gene encodes a proliferation-related cytosolic phosphoprotein which is induced in normal lymphocytes following mitogenic stimulation.
  • the cDNA for this gene has been cloned (Zhu et al, 1989, J Biol Chem. 264:14556-14560). It is encoded by two different-sized full-length cDNAs. The two cDNAs differ in their 3'-noncoding regions as a result of alternative polyadenylation.
  • the Opl8 gene which is 6.3 kilobases in length, is comprised of five exons and four introns and exhibits features that are common to other genes involved in cellular growth and proliferation.
  • Op 18 This gene is highly conserved in several animal species and low stringency hybridization studies suggest that the p 18 gene can be a member of a family of partially homologous genes in the human genome.
  • the increase in Op 18 polypeptide in leukemia is associated with increased RNA transcription without gene amplification or rearrangement (Mel em etal, 1991, J. Biol Chem.266:17747-17753).
  • Treatment of K562 leukemia cell line with hemin that induces terminal differentiation resulted in decreased expression of Op 18.
  • Putative G-protein coupled receptor has been cloned recently (Mayer et al. , 1998, Biochim. Biophys. Acta 1395:301-308).
  • the cDNA sequence encodes a protein of 399 amino acids.
  • Northern and RNA dot blot analyzes demonstrated that the major 4.8 kb transcript is predominantly expressed in brain.
  • In situ hybridization studies of tissue sections revealed high expression in neurons of the brain and spinal cord, thymocytes, megakaryocytes, and macrophages.
  • Glucosidase alpha II is a glycoprotein involved in the processing of N-linked glycans. It resides in the endoplasmic reticulum (ER) and controls the formation of glycoproteins in the ER.
  • the glucosidase alpha II gene was cloned and the encoded protein has been shown not to contain known ER retention signals or hydrophobic regions that could represent a transmembrane domain.
  • Glucosidase alpha II has been shown to contain a single N-glycosylation site close to the amino terminus. HIV-1 contains two heavily glycosylated envelope proteins, gpl20 and gp41, which mediate attachment of virions to the glycosylated cell surface receptor molecule, CD4.
  • gp 120 and gp41 can be involved in syncytium formation and associated cytopathic effects of HIV.
  • the alpha-glucosidase inhibitor N- butyldeoxynojirimycin (NB-DNJ) is an inhibitor of HIV replication and HlY-induced syncytium formation in vitro.
  • the NB-DNJ-mediated retention of glycosylated -glycans inhibits HIV entry by a combined effect of a reduction in virion gpl20 content and a qualitative defect within the remaining gpl20, preventing it from undergoing conformational changes after CD4 binding (Fischer etal, 996, J. Virol. 70:7153-7160).
  • alpha, beta and gamma In mammals there are at least three isoforms ofthe glycolytic enzyme enolase encoded by three similar genes: alpha, beta and gamma. Structure of the human gene for alpha-enolase locus has been described (Giallongo et al, 1990, Ewr J Biochem, 190:567-573). The gene is composed of 12 exons distributed over more than 18,000 bases. The structure of this gene has a high degree of similarity to that ofthe human and rat gamma-enolase genes, with identical positions for all the intron regions.
  • the putative promoter region like that of other house-keeping genes, lacks canonical TATA and CAAT boxes, is extremely G + C-rich and contains several potential SP1 binding sites.
  • MIPl alpha The murine gene, macrophage inflammatory protein 1 alpha (MIPl alpha) is a cytokine that inhibits proliferation of bone marrow stem cells (Russell et al. , 1993, DNA Cell Biol. 12:157-175). MIPl alpha has been shown to suppress HIV-1 replication in human peripheral blood mononuclear cells. MIP 1 alpha can also suppress transcription from the HTV-1 LTR in transient transfection assays in cells of the Jurkat acute T leukemia cell line (Handen et al, 1997, FEBS Lett. 410:301-302).
  • MIPl alpha is a ligand of CCR5 and can prevent M-tropic HIV infection in vitro (Cochi, et al, 1995, Science, 270:1811-1815; Gong et al, 1998, J. Biol. Chem, 271:2599-2603). Certain individuals with elevated levels of MIPl alpha expression appear to be resistant to HIV infection (Cho et al, 1997, Biomed. Pharmacother. 51:221-229). It is not known, however, how downmodulation of intracellular expression of MIP 1 alpha can affect HIV replication in T cells.
  • the translationally-controlled tumor protein (TCTP) is a growth-related protein which is regulated at the translational level.
  • TCTP is localized on chromosome 13ql2-ql4 (MacDonald et al, 1999, Cytogenet Cell Genet, 84:128-129). Both vitamin D and PHA can induce latent HIV-1. This indicates that the TCTPl pathway overlaps with signal transduction pathways involving critical for the HIV-1 life-cycle.
  • the nef gene of human and simian immunodeficiency virus is a key factor in acquired immunodeficiency syndrome pathogenesis and virus replication.
  • Nafl Nef-associated factor 1
  • the Nafl gene generates two isoforms, Naflalpha and beta, containing four coiled-coil structures.
  • Nafl mRNA is ubiquitously expressed in human tissues with strong expression in peripheral blood lymphocytes and spleen.
  • Nafl overexpression in T cells increases surface CD4 expression. Expression of Nef suppressed this Nafl -induced augmentation of CD4 expression, providing a novel mode of Nef action in CD4 down-regulation.
  • hRSl Na + -D- Glucose cotransport regulator gene
  • pRSl (6,743 bp), which encodes a 617 amino acid protein with 74% identity to pRSl.
  • hRSl was localized to chromosome lp36.1. It is homologous to the nucleic acid sequence of pRSl, which was cloned from pig kidney cortex and encodes a membrane-associated protein involved in Na + -coupled sugar transport.
  • pRS 1 alters sugar transport by SGLT 1 from rabbit intestine or by SMIT from dog kidney which is homologous to SGLT1. In contrast, pRSl does not influence transporters from other genetic families.
  • hRSl The function of hRSl has been demonstrated by co-expression experiments of hRSl and SGLT1, from human intestine, in oocytes fromXenopus laevis. It was demonstrated that hRS 1 -protein inhibits Na + -D-glucose co- transport expressed by human SGLT 1 by decreasing both the V max and the apparent K,,, value of the transporter.
  • the analysis ofthe 5'-noncoding sequence of hRSl revealed different enhancer consensus sequences that are absent in the SGLT1 gene, e.g., several consensus sequences for steroid-binding proteins.
  • Hsp90 is an abundant molecular chaperone that is involved in the folding of a defined set of signaling molecules including steroid-hormone receptors and kinases. In vitro experiments suggest that Hsp90 contains two different binding sites for non-native proteins, which allow it to combine the properties of a promiscuous chaperone with those of a dedicated folding-helper protein.
  • the heat shock protein Hsp90 is known as an essential component of several signal transduction pathways and has now been identified as an essential host factor for hepatitis B virus replication. Hsp90 interacts with the viral reverse transcriptase to facilitate the formation of a ribonucleoprotein (RNP) complex between the polymerase and an RNA ligand (Hu et al. , 1996, Proc Natl
  • Hsp90 has not been associated with the HTV life-cycle.
  • Specific inhibitors of hsp90 e.g., anti-fungal macrolydes, geldanamycin and radocicol can be used against HIV-1 infection.
  • FK506-binding protein Al is a potent immunosuppressive agent which is 100- fold more active than cyclosporin A, a cyclic decapeptide that is used to prevent rejection after transplantation of bone marrow and organs, such as kidney, heart, and liver. It was shown that FK506 binds to a cellular protein distinct from cyclophilin which is known to bind cyclosporin A.
  • a cDNA has been isolated from human peripheral blood T-cells that encodes FK506-binding protein (FKBP) (Maki et al , 1990, Proc Natl Acad Sci USA 87: 5440-5443). The isolated cDNA contained an open reading frame encoding 108 amino acid residues.
  • the first 40 residues of the deduced amino acid sequence were identical to those ofthe reported amino-terminal sequence of bovine FKBP, indicating that the DNA sequence isolated represents the gene coding for FKBP. It is expressed in brain, lung, liver, placental cells and leukocytes. Induction of Jurkat leukemic T cells with phorbol 12-myristate 13-acetate and ionomycin did not affect the level of FKBP mRNA.
  • Proteins ofthe Myc and Mad family are involved in transcriptional regulation and mediate cell differentiation and proliferation. These molecules share a basic-helix- loop-helix leucine zipper domain (bHLHZip) and bind DNA at the E box (CANNTG) consensus by forming heterodimers with Max (Meroni et al, 1997, EMBO J. 15-
  • bandshift assays demonstrate that the Rox-Max heterodimer shows a novel DNA binding specificity, having a higher affinity for the CACGCG site compared with the canonical E box CACGTG site.
  • Transcriptional studies indicate that Rox represses transcription in both human HEK293 cells and yeast.
  • ROX expression appears to be induced in U937 myeloid leukemia cells stimulated to differentiate with 12-O-tetradecanoylphorbol-l 3-acetate.
  • SSR2 Human signal sequence receptor gene, SSR2, encodes an endoplasmic reticulum (ER) membrane protein associated with protein translocation across the ER membrane. This gene has been cloned (Chinen et al, 1995, Cytogenet Cell Genet. 70:215-217).
  • hTid-1 A human tumorous imaginal disc protein, hTid-1, which is a homo log of the Drosophila tumor suppressor protein Tid56, has been cloned (Schilling, 1998, Virology 247:74-85).
  • the hTid-1 protein is able to form complexes with the human papillomavirus E7 oncoprotein.
  • the carboxyl terminal cysteine-rich metal binding domain of E7 is the major determinant for interaction with hTid-1.
  • the hTid-1 protein is a member of the DnaJ-family of chaperones.
  • the mRNA of hTid-1 is widely expressed in human tissues, including the HPV- 18-positive cervical carcinoma cell line, HeLa, and human genital keratinocytes, the normal host cells ofthe HPVs.
  • the hTid-1 gene has been mapped to the short arm of chromosome 16.
  • the large tumor antigens of polyomaviruses encode functional J-domains that are important for viral replication and cellular transformation.
  • the ability of HPV E7 to interact with a cellular DnaJ protein suggests that these two viral oncoproteins may target common regulatory pathways through J-domains (Schilling, ibid.).
  • Heparin sulfate proteoglycans are expressed on the cell surface and their corresponding binding sites have been suggested to play an important role during the initial attachment of murine blastocysts to uterine epithelium, and human trophoblastic cell lines to uterine epithelial cell lines.
  • Three major peptide fragments from heparin- binding protein have been isolated and partial amino-terminal amino acid sequence for each peptide fragment was obtained (Raboudi, et al, 1992, J. Biol. Chem. 267, 11930-
  • HIP HP/HS interacting protein
  • Northern blot analysis detected a single transcript of 1.3 kilobases in both total RNA andpoly(A + ) RNA. Examination of human cell lines and normal tissues using both Northern blot and Western blot analyzes revealed that HIP is expressed at different levels in a variety of human cell lines and normal tissues.
  • the minimal size of a nucleic acid molecule ofthe present invention is the size required for the use ofthe nucleic acid molecule to inhibit HIV replication.
  • One of skill in the art will recognize that the length can differ if the nucleic acid molecule is in the form of RNA or DNA.
  • a nucleic acid molecule ofthe present invention can be used in a variety of applications including, but not limited to, as probes to identify additional nucleic acid molecules, as primers to amplify or extend nucleic acid molecules or in therapeutic applications to inhibit, for example, expression of a target gene ofthe present invention.
  • Such therapeutic applications include the use of such nucleic acid molecules in, for example, anti-sense-RNA and -DNA molecules, triplex formation-, ribozyme- and/or RNA drug-based technologies, that function to inhibit HIV infection, and are referred to herein as "therapeutic nucleic acid molecules.”
  • anti-sense RNA and DNA molecules can be used to directly block the translation of mRNA encoded by these cellular genes by binding to targeted mRNA and preventing protein translation.
  • Polydeoxyribonucleotides can form sequence-specific triple helices by hydrogen bonding to specific complementary sequences in duplexed DNA. Formation of specific triple helices can selectively inhibit the replication and/or expression of targeted genes by prohibiting the specific binding of functional trans-acting factors.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
  • engineered hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of cellular RNA sequences.
  • Antisense RNA showing high affinity binding to target sequences can also be used as ribozymes by addition of enzymatically active sequences known to those skilled in the art.
  • Nucleic acid molecules to be used in triplex helix formation should be single stranded and composed of deoxynucleo tides.
  • the base composition of these nucleic acid molecules must be designed to promote triple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of either purines or pyrimidines to be present on one strand of a duplex.
  • Nucleic acid molecule sequences can be pyrimidine-based, which will result in TAT and CGC triplets across the three associated strands ofthe resulting triple helix.
  • the pyrimidine-rich polynucleotides provide base complementarity to a purine-rich region of a single strand of the duplex in a parallel orientation to that strand.
  • nucleic acid molecules can be chosen that are purine-rich, for example, containing a stretch of G residues. These nucleic acid molecules will form a triple helix with a DNA duplex that is rich in GC pairs, in which the majority ofthe purine residues are located on a single strand ofthe targeted duplex, resulting in GGC triplets across the three strands in the triplex.
  • Switchback polynucleotides are synthesized in an alternating 5'-3', 3'-5' manner, such that they base pair with first one strand of a duplex and then the other, eliminating the necessity for a sizeable stretch of either purines or pyrimidines to be present on one strand of a duplex.
  • RNA molecules can be generated by in vitro and in vivo transcription of DNA sequences encoding the antisense RNA molecule. Such DNA sequences can be inco ⁇ orated into a wide variety of vectors which inco ⁇ orate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters.
  • antisense cDNA constructs that synthesize antisense RNA constitutively or inducibly, depending on the promoter used, can be introduced stably into host cells.
  • the present invention includes such therapeutic nucleic acid molecules and methods to interfere with the production of proteins encoded by a target gene necessary for HIV infection ofthe present invention by use of one or more of such technologies.
  • a preferred therapeutic nucleic acid molecule comprises at least a portion of a target gene encoding a protein selected from the group consisting of NADH dehydrogenase, 2-oxoglutarate dehydrogenase, M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein, calnexin, ADP-ribosylation factor 3, eukaryotic initiation factor 3, protein tyrosine phosphatase, he ⁇ esvirus-associated ubiquitin- specific protease, eukaryotic initiation factor 4B, CD44, phosphatidyl-inositol 3 kinase, elongation factor 1 alpha, bone mo ⁇ hogenic protein- 1 , double-strand break DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG- 1 ), lymphocyte-specific protein 1, protein phosphatase 2 A, squalene synthetase, e
  • a more preferred therapeutic nucleic acid molecule is a GSE nucleic acid molecule ofthe present invention, particularly comprising SEQ ID NO: 1, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO:l 1, SEQ ID NO:12,
  • the present invention also includes a recombinant vector, which includes a nucleic acid molecule of the present invention inserted into any vector capable of delivering the nucleic acid molecule into a host cell.
  • a vector contains heterologous nucleic acid sequences, that is nucleic acid sequences that are not naturally found adjacent to a cell-derived nucleic acid molecule of the present invention.
  • the vector can be either RNA or DNA, either prokaryotic or eukaryotic, and typically is a virus or a plasmid.
  • Recombinant vectors can be used in the cloning, sequencing, and/or otherwise manipulating of nucleic acid molecules ofthe present invention.
  • recombinant vector herein referred to as a recombinant expression molecule and described in more detail below, can be used in the expression of nucleic acid molecules ofthe present invention.
  • Preferred recombinant vectors are capable of replicating in the transformed cell.
  • a preferred nucleic acid molecule to include in a recombinant vector of the present invention is a cell-derived nucleic acid molecule of the present invention.
  • a particularly preferred nucleic acid molecule to include in a recombinant vector is at least a portion of a target gene that encodes a protein selected from the group consisting of
  • NADH dehydrogenase 2-oxoglutarate dehydrogenase
  • M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein calnexin
  • ADP-ribosylation factor 3 eukaryotic initiation factor 3
  • protein tyrosine phosphatase he ⁇ esvirus-associated ubiquitin-specific protease
  • eukaryotic initiation factor 4B CD44
  • phosphatidyl-inositol 3 kinase elongation factor 1 alpha, bone mo ⁇ hogenic protein- 1, double-strand break
  • DNA repair gene protein DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG-1), lymphocyte-specific protein 1, protein phosphatase 2A, squalene synthetase, eukaryotic release factor 1 , GTP binding protein, importin beta subunit, cell adhesion molecule LI, U-snRNP associated cyclophilin, recepin, Arg/Abl interacting protein (ArgBP2A), keratin related protein, pi 8 protein, p40 protein, glucosidase ⁇ , alpha enolase, macrophage inflammatory protein 1 alpha, tumor protein translationally-controlled 1 (TCTPl), BBC1, Nef interacting protein, Na + -D-glucose cotransport regulator gene protein, hsp90 chaperone protein, FK506-binding protein Al , Rox, beta signal sequence receptor, tumorous imaginal disc protein or cell surface heparin binding protein, or complements or homologs thereof.
  • a recombinant expression molecule ofthe present invention comprises one or more nucleic acid molecules ofthe present invention operably linked to an expression vector containing one or more regulatory sequences.
  • the phrase "operably linked” refers to insertion of a nucleic acid molecule into an expression vector in a manner such that the molecule is able to be expressed when transformed into a host cell.
  • an expression vector is a DNA or RNA vector that is capable of transforming a host cell and of effecting expression of a specified nucleic acid molecule.
  • the expression vector is also capable of replicating within the host cell.
  • Expression vectors can be either prokaryotic or eukaryotic, and are typically viruses or plasmids.
  • Expression vectors ofthe present invention include any vectors that function (/. e., direct gene expression) in recombinant cells of the present invention, including in bacterial, fungal, insect, animal, and/or plant cells.
  • nucleic acid molecules ofthe present invention can be operably linked to expression vectors containing regulatory sequences such as promoters, operators, repressors, enhancers, termination sequences, origins of replication, and other regulatory sequences that are compatible with the recombinant cell and that control the expression of nucleic acid molecules ofthe present invention.
  • a regulatory sequence includes a sequence which is capable of controlling the initiation, elongation, and termination of transcription.
  • Suitable regulatory sequences include any transcription control sequence that can function in a cell susceptible to infection by HIV. A variety of such transcription control sequences are known to those skilled in the art. Additional suitable regulatory sequences include tissue-specific promoters and enhancers as well as lymphokine-inducible promoters (e.g., promoters inducible by interferons or interleukins). Regulatory sequences ofthe present invention can also include naturally- occurring transcription control sequences naturally associated with a DNA sequence encoding a nucleic acid molecule ofthe present invention.
  • a recombinant molecule ofthe present invention is expressed as a protein
  • specific initiation signals can be required for efficient translation of inserted nucleic acid molecules.
  • initiation codon must be in phase with the reading frame ofthe inserted nucleic acid molecule to ensure translation of the entire insert.
  • exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic.
  • Preferred expression vectors ofthe present invention are derived from viruses such as retroviruses, adenovirus, adeno-associated virus, he ⁇ es viruses, or papilloma viruses. Methods which are well known to those skilled in the art can be used to construct a recombinant molecule of the present invention (Sambrook et al, 1989, MOLECULAR CLONING: ALABORATORY MANUAL, Cold Spring Harbor Laboratory, N.Y. and Ausubel et al, 1989, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Greene
  • a nucleic acid molecule of the present invention can be ligated to an adenovirus transcription-translation control complex, e.g. , the late promoter and tripartite leader sequence.
  • This chimeric gene can then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region ofthe viral genome, e.g., region El or E3, will result in a recombinant virus that is viable and capable of expressing GSEs in infected hosts (Logan & Shenk, 1984, Proc. Natl. Acad. Sci. 18.4 81:3655-3659).
  • Recombinant techniques useful for increasing the expression of nucleic acid molecules ofthe present invention include, but are not limited to, operably linking nucleic acid molecules to high-copy number plasmids, integration ofthe nucleic acid molecules into one or more host cell chromosomes, addition of vector stability sequences to plasmids, substitutions or modifications of transcription control signals (e.g., promoters, operators, enhancers), substitutions or modifications of translational control signals (e.g., ribosome binding sites, Shine-Dalgarno sequences), modification of nucleic acid molecules of the present invention to correspond to the codon usage ofthe host cell, deletion of sequences that destabilize transcripts, and use of control signals that temporally separate recombinant cell growth from recombinant protein production during fermentation.
  • the activity of an expressed recombinant protein of the present invention can be improved by fragmenting, modifying, or derivatizing the resultant protein.
  • nucleic acid molecules can be introduced as a means of increasing intracellular stability and half-life. Possible modifications include, but are not limited to, the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule or the use of phosphorothioate or 2' O-methyl rather than phosphodiesterase linkages within the polydeoxyribonucleotide backbone.
  • One embodiment ofthe present invention is an isolated protein that is necessary for HIV infection.
  • a protein is referred to herein as a "cell-derived protein.”
  • the term "protein” is intended to include both polypeptides and peptides.
  • the invention provides peptides or less than full-length fragments of such cell-derived proteins ofthe invention, wherein expression of said proteins in an HIV- susceptible cell inhibits HIV infection, replication or production of HIV progeny virus.
  • full-length proteins having at least one change in a constituent amino acid residue whereby the protein no longer supports HIV infection in said cell, and most preferably wherein the altered protein inhibits HIV infection, replication or production of HIV progeny virus.
  • an isolated, or biologically pure, protein is a protein that has been removed from its natural milieu.
  • isolated and biologically pure do not necessarily reflect the extent to which the protein has been purified.
  • An isolated cell-derived protein ofthe present invention can be obtained from its natural source, can be produced using recombinant DNA technology or can be produced by chemical synthesis.
  • an isolated cell- derived protein ofthe present invention comprises at least a portion of a protein selected from the group consisting of NADH dehydrogenase, 2-oxoglutarate dehydrogenase, M2- type pyruvate kinase/ cytosolic thyroid hormone binding protein, calnexin, ADP- ribosylation factor 3, eukaryotic initiation factor 3, protein tyrosine phosphatase, he ⁇ esvirus-associated ubiquitin-specific protease, eukaryotic initiation factor 4B, CD44, phosphatidyl-inositol 3 kinase, elongation factor 1 alpha, bone mo ⁇ hogenic protein- 1 , double-strand break DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG-1), lymphocyte-specific protein 1, protein phosphatase 2 A, squalene synthetase, eukaryotic
  • An isolated protein ofthe present invention can be identified in a straight-forward manner by the protein' s ability to inhibit HIV infection.
  • homologs include proteins in which amino acids have been deleted (e.g., a truncated version ofthe protein, such as a peptide), inserted, inverted, substituted and/or derivatized (e.g., by glycosylation, phosphorylation, acetylation, myristoylation, prenylation, palmitoylation, amidation and/or addition of glycerophosphatidyl inositol) such that the homolog has at least some ability to inhibit HIV infection, wherein expression ofthe wildtype protein is necessary for HIV infection.
  • Homologs also include at least one epitope capable of eliciting an immune response against a cell-derived protein. Techniques to measure HIV infection or the inhibition thereof, are disclosed herein.
  • Cell-derived protein homologs of the present invention can be the result of natural allelic variation or natural mutation.
  • Cell-derived protein homologs of the present invention can also be produced using techniques known in the art including, but not limited to, direct modifications to the protein or modifications to the gene encoding the protein using, for example, classic or recombinant nucleic acid techniques to effect random or targeted mutagenesis.
  • the minimal size of a cell-derived protein homolog ofthe present invention is a size sufficient to function as an inhibitor of HIV infection. The minimal size of such homolog is typically at least about 6 to about 10 residues in length.
  • the protein homolog can include a peptide, a less than full length fragment of a full-length protein, a full-length protein, multiple proteins, or portions thereof, wherein said full-length proteins most preferably comprise an altered amino acid sequence whereby the altered protein inhibits HIV infection, replication or production of infectious virus.
  • the present invention also includes mimetopes of cell-derived proteins ofthe present invention.
  • a mimetope of a cell-derived protein ofthe present invention refers to any compound that is able to mimic the activity of such a cell-derived protein (e.g., ability to inhibit HIV infection), often because the mimetope has a structure that mimics the cell-derived protein. It is to be noted, however, that the mimetope need not have a chemical structure similar to a cell-derived protein as long as the mimetope functionally mimics the protein.
  • Mimetopes can be, but are not limited to: peptides that have been modified to decrease their susceptibility to degradation; anti-idiotypic and/or catalytic antibodies, or fragments thereof; non-proteinaceous immunogenic portions of an isolated protein (e.g., carbohydrate structures); synthetic or natural organic or inorganic molecules, including nucleic acids; and/or any other peptidomimetic compounds.
  • Mimetopes of the present invention can be designed using computer- generated structures of cell-derived proteins ofthe present invention. Mimetopes can also be obtained by generating random samples of molecules, such as polynucleotides, peptides or other organic molecules, and screening such samples by affinity chromatography techniques using the corresponding binding partner.
  • a preferred mimetope is apeptidomimetic compound that is structurally and/or functionally similar to a cell-derived protein ofthe present invention.
  • a cell-derived protein ofthe present invention is a fusion protein that includes a cell-derived protein-containing domain attached to one or more fusion segments.
  • Suitable fusion segments are known to those of skill in the art depending upon the use ofthe segment, such as for protein stability or protein delivery into a cell.
  • Fusion proteins are preferably produced by culturing a recombinant cell transformed with a fusion nucleic acid molecule that encodes a cell-derived protein including the fusion segment.
  • Isolated cell-derived proteins of the present invention have the further characteristic of being encoded by a cell-derived nucleic acid molecule of the present invention.
  • a preferred cell-derived protein of the present invention is encoded by a nucleic acid molecule having a nucleic acid sequence selected from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:
  • An isolated GSE nucleic acid molecule of the present invention can suppress HIV activity by either encoding a protein or RNA product.
  • the present invention encompasses any such protein product, including a fusion protein, a leader peptide and a localization signal.
  • One embodiment of the present invention is an inhibitory composition that, when administered to an animal in an effective manner, is capable of protecting that animal from HIV infection, prophylactically or therapeutically.
  • An inhibitory composition of the present invention can include a protective compound that down- regulates the expression of a gene that encodes NADH dehydrogenase, 2-oxoglutarate dehydrogenase, M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein, calnexin, ADP-ribosylation factor 3, eukaryotic initiation factor 3, protein tyrosine phosphatase, he ⁇ esvirus-associated ubiquitin-specific protease, eukaryotic initiation factor 4B, CD44, phosphatidyl-inositol 3 kinase, elongation factor 1 alpha, bone mo ⁇ hogenic protein- 1, double-strand break DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG-1), lymphocyte-specific protein 1 , protein phosphatase 2 A, squalene synthetase, eukaryotic release factor 1
  • Such a protective compound comprises an isolated cell-derived nucleic acid molecule ofthe present invention operably linked to a regulatory sequence that controls its expression.
  • the protective compound can be an RNA- or DNA-based molecule as described herein.
  • Particularly preferred are GSE nucleic acid molecules of the present invention.
  • a functionally-active fragment of a GSE, and a GSE containing conservative nucleotide substitutions as functional equivalents of a GSE, are within the scope ofthe present invention.
  • a GSE nucleic acid molecule, or a functional equivalent thereof is operably linked to a regulatory sequence that controls its expression.
  • An inhibitory composition of the present invention can include a protective compound that inhibits the activity of a product of a gene that encodes NADH dehydrogenase, 2-oxoglutarate dehydrogenase, M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein, calnexin, ADP-ribosylation factor 3, eukaryotic initiation factor 3, protein tyrosine phosphatase, he ⁇ esvirus-associated ubiquitin- specific protease, eukaryotic initiation factor 4B, CD44, phosphatidyl-inositol 3 kinase, elongation factor 1 alpha, bone mo ⁇ hogenic protein- 1 , double-strand break DNA repair gene protein, rat guanine nucleotide releasing protein, anti-proliferative factor (BTG- 1 ), lymphocyte-specific protein 1 , protein phosphatase 2 A, squalene synthetase, euk
  • ArgBP2A Arg/Abl interacting protein
  • keratin related protein pi 8 protein
  • p40 protein glucosidase H
  • alpha enolase alpha enolase
  • macrophage inflammatory protein 1 alpha tumor protein translationally-controlled 1 (TCTPl)
  • TCTPl tumor protein translationally-controlled 1
  • BBC1 Nef interacting protein
  • Such a protective compound can include an isolated cell-derived protein of the present invention, in particular a peptide of a cell-derived protein, a mimetope of a cell-derived protein, and small molecule inhibitors ofthe activity of target gene products.
  • protective compounds e.g., proteins, mimetopes, nucleic acid molecules, and inhibitors, are disclosed herein. It is within the scope ofthe present invention that an inhibitory composition can contain one or more protective compounds.
  • Suitable inhibitors of the activity of target gene products include compounds that interact directly with the active sites of such products, usually by binding to or otherwise interacting with or otherwise modifying the products's active site.
  • Product inhibitors can also interact with other regions of the product to inhibit its activity, for example, by allosteric interaction.
  • the inhibitor of a product is identified by its ability to bind to, or otherwise interact with the product, thereby inhibiting the activity ofthe product and/or HIV infection.
  • NADH dehydrogenase a large number of small molecule inhibitors are available in the art. Such inhibitors can be used in the methods of the present invention.
  • An in vitro assay can be established to screen for additional NADH dehydrogenase complex I inhibitors by measuring membrane potential of live cells with DiC6, a dye which accumulates in the mitochondrial and cytoplasmic membrane depending on mitochondrial functional activities and ATP concentrations (Methods in
  • NADH dehydrogenase inhibitors include, but are not limited to, Amytal, Annonin VI, Aurachin A, Aurachin B, Aureothin, Benzimidazole, Bullactin, Capsaicin, Ethoxyformic anhydride, Ethoxyquin, Fenpyroximate, Mofarotene (Ro 40-8757; arotinoids), Molvizarin, Myxalamide PI, Otivarin (annonaceous acetogenins), Pethidine, Phenalamid A 2 , Phenoxan, Piericidin A, p-chloromercuribenzoate, Ranolazine (RS-
  • inhibitors of protein tyrosine phosphatase include, but are not limited to, ortho-vanadate, pervanadate, sodium vanadate, and phenylarsine oxide.
  • Inhibitors of protein tyrosine phosphatase have been described by Yousefi et al, 1994, Proc. Natl.
  • inhibitors of EF-1 ⁇ include, but are not limited to, include quercetin (3,3',4'5,7-pentahydrozyflavone) and didemnin B.
  • assay systems can be established using such gene for screening and selection of additional compounds as anti-HIV therapeutics based on their ability to down-regulate the expression ofthe gene or inhibit the activities of its gene product.
  • a cell line which naturally expresses the gene of interest or has been transfected with it can be incubated with various compounds.
  • a reduction ofthe expression ofthe gene of interest or an inhibition ofthe activities of its encoded product can be used as an indication that the compound is effective in inhibiting expression and/or the function of said gene.
  • the compounds are retested in other assays such as in OMlO.l cells or in productive HIV infection to confirm their activities against HIV infection.
  • These compounds can be screened from known organic compounds, products of peptide libraries and products of chemical combinatorial libraries.
  • One embodiment ofthe present invention is a method to identify a compound capable of inhibiting HTV infection.
  • Such a method includes the steps of (a) contacting, e.g., combining or mixing, an isolated cell-derived protein with a putative inhibitory compound under conditions in which, in the absence ofthe compound, the protein has activity, and (b) determining if the putative inhibitory compound inhibits the activity of the cell-derived protein.
  • Putative inhibitory compounds to screen include small organic molecules, antibodies (including mimetopes thereof) and substrate analogs. Methods to determine the activity if a cell-derived protein are known to those skilled in the art.
  • cell-derived nucleic acid molecules in particular GSE nucleic acid molecules, can be used to design polypeptides or peptides capable of inhibiting HIV infection.
  • a method to test inhibitory polypeptides or peptides for use as a therapeutic compound can include the steps of (a) delivering a putative inhibitory peptide to a cell susceptible to HIV infection; and (b) determining the ability of such peptide to inhibit HIV infection. Methods to deliver and determine HIV infection are disclosed herein.
  • One preferred embodiment of the present invention is the use of protective compounds ofthe present invention to protect an animal from HIV infection.
  • Preferred protective compounds ofthe present invention have been disclosed herein. Additional protection can be obtained by administering additional protective compounds, including other reagents known to inhibit HIV infection.
  • a protective compound comprising a cell-derived nucleic acid molecule can be transferred into any HIV-susceptible host cells such as CD4 + T cells or hematopoietic progenitor cells such as CD34 + cells obtained from bone marrow or mobilized peripheral blood, by any DNA transfer techniques well known in the art such as electroporation, transfection or transduction, followed by transplantation of the cells into a recipient.
  • the progeny cells express the nucleic acid molecule product and become resistant to HIV.
  • such cell-derived nucleic acid molecule is a GSE nucleic acid molecule ofthe present invention.
  • a cell-derived nucleic acid molecule can be directly administered in vivo using a gene therapy expression vector.
  • the molecules can be delivered or transferred into CD4 + T cells in both HlV-infected or uninfected individuals to protect against development of HIV infection.
  • the recombinant molecules can also be transferred into stromal cells, including macrophages.
  • a cell-derived nucleic acid molecule can be delivered into a target cell by a non- viral delivery system.
  • a GSE nucleic acid molecule can be reconstituted into liposomes for delivery to susceptible cells. Liposomes are spherical lipid bilayers with aqueous interiors.
  • oligonucleotides All molecules that are present in an aqueous solution at the time of liposome formation (in this case, oligonucleotides) are inco ⁇ orated into this aqueous interior.
  • the liposomal contents are both protected from the external microenvironment and, because liposomes fuse with cell membranes, are efficiently delivered into the cell cytoplasm, obviating the need to neutralize the oligonucleotides' negative charge.
  • Methods for introducing cell-derived nucleic acid molecules into cells or tissues include the insertion of naked nucleic acid molecule, i.e.
  • Protective compounds of the present invention can be formulated and administered through a variety of means, including systemic, localized, or topical administration. Techniques for formulation and administration can be found in "Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA. The mode of administration can be selected to maximize delivery to a desired target site in the body.
  • route of injection include, but are not limited to, intramuscular, intravenous, intraperitoneal, and subcutaneous.
  • the cell-derived nucleic acid molecules of the invention are formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • the cell-derived nucleic acid molecules can be formulated in solid or lyophilized form, then redissolved or suspended immediately prior to use.
  • a therapeutically effective dose refers to that amount of the compound sufficient to result in an inhibition of HIV infection as compared to the pre-treatment condition.
  • Suitable routes of administration can, for example, include oral, rectal, transmucosal, transcutaneous, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • parenteral delivery including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • parenteral delivery including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • parenteral delivery including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections.
  • compositions ofthe present invention can be manufactured in a manner that is itself known, e.g., by means of a conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • Pharmaceutical compositions for use in accordance with the present invention thus can be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the compounds ofthe invention can be formulated in appropriate aqueous solutions, such as physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds ofthe invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained with solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents can be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions can be used, which can optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments can be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push- fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds can be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers can be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the compositions can take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or
  • the compounds can be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection can be presented in unit dosage form, e.g. , in ampoules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions ofthe active compounds in water-soluble form. Additionally, suspensions of the active compounds can be prepared as appropriate oily injection suspensions.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions can contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension can also contain suitable stabilizers or agents which increase the solubility ofthe compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • the compounds can also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds can also be formulated as a depot preparation. Such long acting formulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds can be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical carrier for the hydrophobic compounds ofthe invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • the cosolvent system can be the VPD co- solvent system.
  • VPD is a solution of 3% w/v benzyl alcohol, 8% w/v ofthe nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the VPD co-solvent system (VPD:5W) consists of VPD diluted 1 : 1 with a 5% dextrose in water solution. This co-solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
  • co-solvent system can be varied considerably without destroying its solubility and toxicity characteristics.
  • identity ofthe co- solvent components can be varied: for example, other low-toxicity nonpolar surfactants can be used instead of polysorbate 80; the fraction size of polyethylene glycol can be varied; other biocompatible polymers can replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides can substitute for dextrose.
  • hydrophobic pharmaceutical compounds can be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as dimethylsulfoxide also can be employed, although usually at the cost of greater toxicity.
  • the compounds can be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art.
  • Sustained-release capsules can, depending on their chemical nature, release the compounds for a few weeks up to over 100 days.
  • additional strategies for protein and nucleic acid stabilization can be employed.
  • the pharmaceutical compositions also can comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • the compounds ofthe invention can be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts can be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended pu ⁇ ose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms ofthe subject being treated. Determination ofthe effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the EC50 (effective dose for 50%o increase) as determined in cell culture, i.e., the concentration ofthe test compound which achieves a half-maximal inhibition of HIV replication as assayed by the infected cells to retain CD4 expression, to reduce viral p24 or gpl20, and to prevent syncytia formation.
  • EC50 effective dose for 50%o increase
  • Such information can be used to more accurately determine useful doses in humans.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% ofthe population) and the ED50 (the dose therapeutically effective in 50% ofthe population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50.
  • Compounds which exhibit high therapeutic indices are preferred.
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See, e.g. Fingl et al, 1975, in "The Pharmacological Basis of
  • Dosage amount and interval can be adjusted individually to provide plasma levels ofthe active moiety which are sufficient to maintain the inhibitory effects.
  • Usual patient dosages for systemic administration range from 100 - 2000 mg/day. Stated in terms of patient body surface areas, usual dosages range from 50 - 910 mg/m 2 /day.
  • Usual average plasma levels should be maintained within 0.1-1000 ⁇ M. In cases of local administration or selective uptake, the effective local concentration of the compound can not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subj ect being treated, on the subj ect's body surface area, the severity ofthe affliction, the manner of administration and the judgment ofthe prescribing physician.
  • Example 1 This example describes isolation and identification of human cell-derived GSEs exhibiting HlV-suppressive activities.
  • the cDNA prepared from HL-60 cells and HeLa cells was partially digested with DNase I in the presence of MvT (Sambrook et al, 1989, MOLECULAR CLONING: A LABORATORY MANUAL, Cold Spring Harbor Laboratory, N.Y.). Under these conditions, DNase I is known to produce mostly double-stranded breaks. The resulting fragments were repaired with the Klenow fragment of DNA polymerase I and T4 polymerase and ligated to synthetic double-stranded adaptors.
  • the 5' adaptors (SEQ ID NOS: 21 and 22) were:
  • mRNA from uninduced cells was first subtracted from mRNA from cells induced with TNF- ⁇ .
  • the subtracted HL-60 library represents a modification of the procedure described in Coche et al, 1994,
  • the tracer mRNA was purified from HL-60 cells containing the LNCX plasmid at different time points after induction with TNF- ⁇ .
  • LNCX gene was used as an internal standard to monitor the enrichment ofthe sequences present in the tracer after subtraction.
  • the mRNAs isolated from induced and uninduced cells were annealed separately to oligo dT magnetic beads (available from Dyna) and the first cDNA strand was synthesized using reverse transcriptase and oligo dT as the primer.
  • the RNA strand was hydrolyzed and the second strand was synthesized on the induced population using a primer containing three ATG codons and 10 random nucleotides on the 3' end.
  • Single stranded cDNA fragments were annealed to an excess of driver cDNA attached to the magnetic beads. This procedure was repeated several times until substantial enrichment in the spiked LNCX sequence was seen.
  • ssDNA single-stranded DNA
  • GSEs from rare cDNAs since total polyA + RNA was a mixture of unequally-represented sequences.
  • the method first denatured 20 ⁇ g of cDNA by boiling for 5 min. in 25 ⁇ L of TE buffer, followed by immediate cooling on ice. Then, 25 ⁇ L of 2X hybridization solution was added, and the mixture was divided into four aliquots in Eppendorf tubes, 12.5 ⁇ L each. One to two drops of mineral oil were added to each sample to avoid evaporation, and the tubes were placed into a 68°C water bath for annealing. One tube was frozen every 12 hours.
  • each ofthe annealing mixtures was diluted with water to a final volume of 500 ⁇ L and subjected to hydroxylapatite (HAP) chromatography.
  • HAP suspension equilibrated with 0.01 M phosphate-buffered saline (PBS) was placed into Eppendorf tubes so that the volume of
  • HAP pellet was approximately 100 ⁇ L.
  • the tubes with HAP and all the solutions used below were preheated and kept at 65°C.
  • the excess of PBS was removed, and diluted annealing solution was added.
  • HAP crystals The supernatant was carefully replaced with 1 mL of preheated 0.01 M PBS, and the process was repeated.
  • the HAP pellet was suspended in 500 ⁇ L of PBS at the single-strand elution concentration determined, e.g. , 0.16M, the supernatant was collected, and the process was repeated.
  • the supernatants were combined and traces of HAP were removed by centrifugation.
  • the ssDNA was concentrated by centrifugation, and washed three times using 1 mL of water on Centricon-100.
  • the isolated ssDNA sequences were amplified by PCR with the sense primer from the adapter, using a minimal number of cycles to obtain 10 ⁇ g of the product.
  • the size ofthe PCR product that remained within the desired range (200-500 bp) was ascertained.
  • the normalization quality was tested by Southern or slot-blot hybridization with 2 P-labeled probes for high, moderate- and low-expressing genes using 0.3-1.0 ⁇ g of normalized cDNA/lane.
  • ⁇ -actin and ⁇ -tubulin cDNAs were used as probes for high-expressing genes, c-r ⁇ c and topo H cDNAs for moderately- expressing genes, and c-fos cDNA for low-expressing genes.
  • the cDNAs isolated after different annealing times were compared with the original unnormalized cDNA. The probes were ensured to have a similar size and specific activity.
  • the best-normalized ssDNA fraction was used, which produced the most uniform signal intensity with different probes, for large-scale PCR amplification to synthesize at least 20 ⁇ g of the product for cloning. More ssDNA template was used to obtain the desired amount by scaling up the number of PCR or the reaction volume.
  • the pLNGFRM vector is the same as the pLNCX vector except that the neo r gene has been replaced with a truncated low-affinity NGFR gene.
  • the cells transduced with pLNGFRM express the truncated receptor on their surface which can be easily selected by an anti -NGFR antibody and FACS.
  • the ligation mixture was transformed into E. coli.
  • the total plasmid was purified from - 100,000 recombinant clones. The size distribution ofthe cloned fragments was tested by PCR amplification using primers derived from the vector sequences adjacent to the adapters.
  • the OMlO.l cells are available from the American Type Culture Collection, Manassas, VA as CRL 10850 (Butera, U.S. Patent No. 5,256,534).
  • the C ⁇ M-ss cells are available from the NIH AIDS Research and Reference Reagent Program as Cat. No. 776.
  • HIV-1 SF2 is available from NIH AIDS Research and Reference Reagent Program as Cat. No. 275.
  • HL-60 cells are available from American Tissue Culture Collection as
  • CCL 240. HTV-lm B is available from AIDS Program as Cat. No. 398.
  • the anti-CD4 (Q4120PE) and anti-p24 (KC-57 FITC) antibodies were purchased from Sigma and Coulter, respectively. TNF- ⁇ was obtained from Boehringer Mannheim. G418 was purchased from Gibco/BRL as Geneticin.
  • the anti-NGFR antibody was obtained from the ATCC (HB-8737) as hybridoma 20.4 (U.S. Patent Nos. 4,786,593 and 4,855,241).
  • Two anti-CD4 antibodies (L77 and L120) were obtained from Becton Dickenson.
  • OMlO.l cells harboring the entire RFE library were induced with 10 U/mL of TNF- ⁇ at 37°C, and 24 hours later, were stained with an antibody and sorted for CD4 expression.
  • Genomic DNA from the CD4 + cells was purified and used for PCR amplification of inserts with the vector-derived primers. The amplified mixture was digested with EcoRI and BamHI and cloned back into the retroviral vector. The selection was repeated for additional rounds.
  • a normalized RF ⁇ library made from HeLa cells was transferred into C ⁇ M-ss cells and the neo resistant population was infected with TCID 50 of 3000/10 6 cells of HIV- 1 mg.
  • This RF ⁇ library was represented by 5 Ox 10 6 independent recombinant clones.
  • Four and seven days after infection a purified anti-CD4 monoclonal antibody,
  • L77 (available from Becton Dickinson), was added at 5 ⁇ g/mL to prevent syncytia formation. Syncytia formation is thought to be prevented by blocking the interaction between gpl20 expressed on the surface of an infected cell and CD4 on the surface of an uninfected cell. Antibody L77 does not prevent HIV infection of a cell.
  • CD4 + , p24 " cell population representing uninfected cells were sorted. Genomic DNA from the CD4 + , p24 " cells was purified and used for PCR amplification of inserts with the vector-derived primers. The amplified mixture was digested with EcoRI and BamHI and cloned back into the retroviral vector. The selection was repeated for additional rounds.
  • NGFR + cells 10 7 cells were washed twice with Assay buffer and resuspended in 200 ⁇ L Assay buffer plus 5% normal mouse serum, and 2 mL of anti-NGFR-P ⁇ antibody was added. After incubation at 4°C for 30 min., 5 mL of Assay buffer was added and the cells were centrifuged at 1200 ⁇ m for 4 min. The cells were washed twice with Assay buffer before sorting by FACS. E. Recovery of GSEs and Sequence Analysis
  • Genomic DNA was isolated from the selected population of OM10.1 or CEM-ss cells harboring putative GSEs by resuspending the cell pellet in 0.1% Triton X-100, 20 ⁇ g/mL proteinase K in lx PCR buffer, incubating at 55°C for 1 hour, and boiling for 10 minutes. Genomic DNA was used for PCR amplification using vector-derived primers, cloned into the retroviral vector, and transformed into E. coli using techniques well known in the art. Individual plasmids were purified from E. coli clones using QIAGEN plasmid kits. Inserts were sequenced by the dideoxy procedure (available from AutoRead Sequencing Kit, Pharmacia Biotech) and run on a Pharmacia LKB A.L.F. DNA sequencer. Sequences were analyzed using the DNASTAR program.
  • OMlO.l cells activation ofthe latent virus in OMlO.l cells by TNF- ⁇ leads to the production of viral protein gpl20, which binds to cytoplasmic CD4, thereby preventing its translocation to the cell surface.
  • a diminution of CD4 + OMlO.l cells also conelates with an increased production of viral protein p24 in the cells following TNF- ⁇ induction.
  • GSEs derived from cDNAs representing expressed human cellular genes were identified and isolated from a RFE library made from HL-60 cells using HIV provirus activation in OM 10.1 cells as a read-out.
  • retrovirus carrying the library was used to infect OM10.1 cells by co-cultivation or spinoculation, and NGFR selection was performed to ensure the retention ofthe viral vector.
  • NGFR selection was performed to ensure the retention ofthe viral vector.
  • the infected cells were treated with TNF- ⁇ , a small number of residual CD4 + cells were detected by an anti-CD4 antibody and sorted by FACS.
  • the GSEs contained in these cells were recovered by PCR amplification and their nucleotide sequences determined.
  • GSEs were isolated by the two aforementioned selection strategies using OMlO.l and CEM-ss cells. Six of these GSEs were shown to have substantial sequence identity with cDNAs of genes encoding different subunits ofthe
  • CF-315 (SEQ ID NO:l) is a GSE which suppresses HIV replication as an antisense molecule, which in its sense orientation has sequence identity with a gene encoding a subunit, ND6, of a mitochondrial enzyme, NADH dehydrogenase (Chomyn et al. , 1988, Science 234:614).
  • CF-315 was further shown to protect uninfected human T cells from a productive HIV- 1 infection ( Figure 2).
  • the retroviral vector, pLNGFRM containing the CF-315 nucleic acid molecule was transfened into CEM-ss cells followed by NGFR selection.
  • Vector containing plasmid DNA (denoted LNGFRM) was used as negative control.
  • the NGFR + cells were 99% CD4 + , and they were then infected with TCID 50 of 1000 of HIV-1 SF2 .
  • the infected cells were removed at 11, 14, 18, 21, 25, 28, 32, 35 and
  • Figure 2 shows that CF-315 inhibited infection of human T cells by HIV-1 SF2 , as compared with negative control of vector plasmid DNA.
  • CF-319 (SEQ ID NO:2) is a GSE which suppresses HIV replication in the sense orientation and has substantial sequence identity with another portion of the gene encoding the ND6 subunit of NADH dehydrogenase.
  • CF-101 (SEQ ID NO:3) also exhibits its HIV-suppressive activities in the sense orientation and has substantial sequence identity with a gene encoding the ND2 subunit of NADH dehydrogenase (Chomyn et al, 1985, Nature 314:592).
  • CF-117 (SEQ ID NO:4) suppresses HTV activities as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding the ND6 subunit of NADH dehydrogenase.
  • CF- 025 (SEQ ID NO: 5) suppresses HIV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding the ND2 subunit of NADH dehydrogenase.
  • CF-128 (SEQ ID NO:6) suppresses HIV infection in the sense orientation and it has substantial sequence identity with a gene encoding the ND4 subunit of NADH dehydrogenase. Since both selection strategies produced GSEs having substantial sequence identity with different subunits of NADH dehydrogenase, this enzyme complex plays an important role during HIV infection, and thus, methods to down-regulate the expression of this complex or any of its subunits can be used to inhibit HIV replication in infected cells.
  • CF-004 SEQ ID NO:7 suppresses HIV infection as a sense molecule and has substantial sequence identity with a gene encoding human 2-oxoglutarate dehydrogenase (Koike, 1995, Gene 158:261-266; Koike et al,
  • CF-113 suppresses HIV infection as a sense molecule and has substantial sequence identity with a gene encoding human M2-type pyruvate kinase/ cytosolic thyroid hormone binding protein (Kato et al, 1989, Proc. Natl. Acad. Sci. USA 86:7861-7865).
  • CF-204 suppresses HIV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding human calnexin (David et al. , 1993, J. Biol. Chem. 268:9585-9592).
  • CF-001 (SEQ ro NO: 10) suppresses HIV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding human ADP-ribosylation factor 3 (Tsai et al. , 1991 , J. Biol. Chem . 266:23053-23059).
  • CF-273 (SEQ ro NO: 11) suppresses HIV infection as a sense molecule and has substantial sequence identity with a gene encoding human eukaryotic translation initiation factor 3 (Merrick and Hershey, 1996, Translational Control, Cold Spring Harbor, NY, pp. 31-69).
  • CF-311 suppresses HIV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding a human protein tyrosine phosphatase (Kim et al. , 1996, Oncogene
  • CF-313 suppresses HIV infection as a sense molecule and has substantial sequence identity with a gene encoding a human protein tyrosine phosphatase (Keyse and Emslie, 1992, Nature 359:644-647).
  • CF-210 suppresses HIV infection as a sense molecule and has substantial sequence identity with a gene encoding he ⁇ esvirus-associated ubiquitin-specific protease (Everett et al. , 1997,
  • CF-266 (SEQ ro NO: 15) suppresses HTV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding human eIF4B (Milburn et al, 1990, EMBOJ., 9:2783-2790).
  • CF-302 (SEQ ID NO: 16) suppresses HIV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding human CD44 (Stamenkovic et ⁇ /., 991, EMBOJ. 10:243-248).
  • CF-317 suppresses HIV infection as an antisense molecule, and in its sense orientation, it has substantial sequence identity with a gene encoding human phosphatidylinositol 3 -kinase (Volinia et al, 1995, EMBO J. 14:3339-3348).
  • CF-286 suppresses HIV infection as a sense molecule and has substantial sequence identity with a gene encoding human elongation factor-l ⁇ (Uetsuki et al, 1989, J. Biol. Chem. 264:5791-5798).
  • two isolated GSEs did not share sequence homology with any known genes by sequence comparison.
  • CF-061 (SEQ ID NO: 19) was selected in OMlO.l cells.
  • CF-280 (SEQ ID NO: 20) was selected in CEM-ss cells.
  • Figure 3 shows the ability of four exemplary GSEs mentioned above in preventing productive infection of CEM-ss cells by HIV. While the GSEs were isolated as fragments of distinct cellular genes, they all inhibited HIV infection as shown by a reduction of p24 levels in the infected cells when compared with controls.
  • Figure 4 shows the HlV-suppressive activities of CF-001 which functions as an antisense molecule.
  • CF-537 is A GSE that interferes with the HIV life cycles in the sense orientation and has substantial sequence identity with a gene encoding human bone mo ⁇ hogenic protein- 1 (BMP 1-6; Wozney etal, ⁇ 9&&, Science 242:1528-1534).
  • BMP 1-6 human bone mo ⁇ hogenic protein- 1
  • SEQ ID NO:26 The complement of SEQ ID NO:25 is represented by SEQ ID NO:26.
  • CF-320 (SEQ ro NO:27) is A GSE that interferes with HIV infection in the sense orientation and has substantial sequence identity with a gene encoding double-strand break DNA repair gene protein (McKay et al. , 1996, Genomics 36:305-315).
  • the complement of SEQ ID NO:27 is represented by SEQ ID NO:28.
  • SEQ ID NO:29 is A GSE that interferes with HIV replication in the sense orientation and has substantial sequence identity with a gene encoding rat guanine nucleotide releasing protein (Burton et al, 1993, Nature 361 : 464-467).
  • the complement of SEQ ID NO:29 is represented by SEQ ID NO:30.
  • CF-322 (SEQ ID NO:31) is A GSE that interferes with HIV replication in the sense orientation and has substantial sequence identity with a gene encoding anti-proliferative factor protein (BTG-1; Rouault et al, 1992, EMBO J, 11:1663-1670).
  • BCG-1 anti-proliferative factor protein
  • SEQ ID NO:31 is represented by SEQ ro NO:32.
  • CF-332 (SEQ ID NO:33) is A GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding lymphocyte-specific protein 1 (Jongstra-Bilen et al. , 1990, J. Immunol. 144: 1104-1110).
  • the complement of SEQ ID NO:33 is represented by SEQ ID NO:34.
  • CF-335 SEQ ID NO:35
  • SEQ ID NO:35 The complement of SEQ ID NO:35 is represented by SEQ ID NO:36.
  • CF-42 (SEQ ID NO: 37) is A GSE that interferes with HIV replication in the sense orientation and has substantial sequence identity with a gene encoding squalene synthetase protein
  • SEQ ID NO:37 The complement of SEQ ID NO:37 is represented by SEQ ID NO:38.
  • CF-50 (SEQ ID NO: 39) also exhibits its HlV-suppressive activities in the sense orientation and has substantial sequence identity with a gene encoding squalene synthetase protein (Summers et al, ibid.).
  • the complement of SEQ ID NO:39 is represented by SEQ ro NO:40.
  • CF-527 (SEQ ID NO:41) is a GSE, a peptide from which interferes with HIV replication, and has substantial sequence identity with a gene encoding Eukaryotic release factor 1 protein (ERF-1; Andjelkovic et al, 1996, EMBO J.
  • SEQ IDNO:41 The complement of SEQ IDNO:42.
  • CF-528 (SEQ ID NO:43) is A GSE that interferes with HTV infection in the sense orientation and has substantial sequence identity with a gene encoding GTP binding protein (Zigman t ⁇ /., 1993, Endocrinology 133:2508-2514).
  • the complement of SEQ ro NO:43 is represented by SEQ ro NO:44.
  • CF-529 SEQ ID NO:45
  • SEQ ID NO:46 The complement of SEQ ID NO:46.
  • CF-531 (SEQ ID NO:47) is A GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding cell adhesion molecule LI protein (LCAMl; Hlavin et al, 1991, Genomics 11:416-23).
  • the complement of SEQ ID NO:47 is represented by SEQ ID NO:48.
  • CF-545 (SEQ ro NO:49) is A GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding U-snRNP associated cyclophilin protein (Horowitz et al, ibid.).
  • the complement of SEQ ID NO:49 is represented by
  • CF-547 is A GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding recepin endoprotease protein (GenBank Accession No. U03644).
  • the complement of SEQ ro NO:51 is represented by SEQ ID NO:52.
  • CF-619 is A GSE that interferes with HIV replication in the sense orientation and has substantial sequence identity with a gene encoding Arg/Abl interacting protein (ArgBP2A; GenBank Accession No. AF049884).
  • the complement of SEQ ID NO:53 is represented by SEQ ID NO:54.
  • CF-620 is A GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding keratin related protein (IFN- ⁇ regulated; Flohr et al. , ibid.).
  • the complement of SEQ ID NO:55 is represented by SEQ ID NO:56.
  • CF-624 is A GSE that interferes with HIV replication in a sense orientation and has substantial sequence identity with a gene encoding pi 8 protein (Zhu et al, ibid.).
  • the complement of SEQ ID NO:57 is represented by SEQ ID NO:58.
  • CF-630 (SEQ ID NO: 59) is A GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding p40 protein (Mayer et al, 1988, Biochim Biophys Acta 1395:301-308).
  • the complement of SEQ ID NO:59 is represented by SEQ ID NO:60.
  • CF-579 (SEQ ID NO:61) is A GSE that interferes with HIV replication in a sense orientation and has substantial sequence identity with a gene encoding glucosidase alpha H protein (GenBank AccessionNo. AJ000332).
  • the complement of SEQ ID NO:61 is represented by SEQ ID NO:62.
  • CF-287 (SEQ ro NO:77) is A GSE that interferes with HTV replication in a sense orientation and has substantial sequence identity with a gene encoding Na + -D-Glucose cotransport regulator protein (Lambotte et al. , 1996, DNA Cell Biol. 15:769-77).
  • the complement of SEQ ID NO:77 is represented by SEQ ID NO:78.
  • CF-622 (SEQ ID NO: 87) is A GSE that interferes with HTV replication as an antisense molecule and has substantial sequence identity with a gene encoding Rox protein (Meroni et al, 1997, EMBO J. 16:2892-2906).
  • the complement of SEQ ID NO:87 is represented by SEQ ID NO:88.
  • FIGS 7, 8 and 9 show the ability of seven exemplary GSEs mentioned immediately above in preventing productive infection of CEM-ss cells by HIV. While the GSEs were isolated as fragments of distinct cellular genes, they all inhibited HIV infection as shown by a reduction of p24 levels in the infected cells when compared with controls.
  • Example 2 This Example demonstrates the suppression of HIV infection by NADH dehydrogenase inhibitors
  • NADH dehydrogenase inhibitors amytal (available from Sigma, St. Louis, MO) and mofarotene (Uchida et al, 1994, Int. J. Cancer 58:891-897) were diluted in sterile culture medium and used according to the indicated concentrations .
  • OMlO.l cells were cultured in RPMI 1640 glucose-free media prior to and during incubation with NADH dehydrogenase inhibitors and TNF- ⁇ induction. The inhibitors were added to the cells followed by TNF- ⁇ induction 1-2 hours later. The expression of CD4 by the cells was assessed by anti-CD4 antibody staining and flow cytometry after 24 hour incubation at 37°C.
  • PBLs Human peripheral blood leukocytes
  • PBLs were then activated with phytohemagglutinin at 0.5 ⁇ g/mL and placed in ahumidified incubator at 37°C/5% CO 2 .
  • FIG. 5 shows that amytal inhibited the induction of latent HIV provirus in OM10.1 cells in a dose-dependent manner, as shown by its ability to retain CD4 expression by TNF- ⁇ -induced OMlO.l cells.
  • mofarotene which down-regulates mitochondrial gene expression, also inhibited HIV-1 induction at even lower concentrations (Figure 6).
  • CD4 + T cells and percentage of p24 + CD4 + T cells are CD4 + T cells and percentage of p24 + CD4 + T cells.
  • 33*% of CD4 + T cells were p24 + and only 15% of T cells were CD4 + .
  • ⁇ 32% of T cells were CD4 + , suggesting a dramatic depletion of CD4 + T cells by HIV infection.
  • the HIV-infected sample with 1 ⁇ M of mofarotene only 10% of CD4 + T cells were p24 + .
  • ⁇ 25% of T cells were
  • CD4 + indicating that mofarotene inhibited the depletion of CD4 + T cells by HIV infection.
  • the level of protection by mofarotene as measured by the percentage of p24 + CD4 + T cells and percentage of CD4 + cells in the CD3 + T cell population, diminished with decreased concentrations of mofarotene. While mofarotene alone increased the percentage of CD4 + T cells in uninfected samples, the effect was minimal (up to 4%> higher). At day 6, mofarotene did not alter the expression level of CD3 on the cell surface.
  • a NADH dehydrogenase inhibitor prevented productive infection by HIV of primary cultures of human T cells.
  • This example describes the production of a PBMC library and isolation of GSEs therefrom.
  • a cell-derived random fragment library was constructed using cDNA from peripheral blood mononuclear cells (PBMC).
  • PBMC peripheral blood mononuclear cells
  • PHA PHA
  • Cells were removed at 5, 10, and 24 hours after the addition of PHA and total RNA was isolated by Trizol extraction. All time points from all donors were then pooled.
  • Polyadenylated mRNA was purified from the total RNA by passages through an oligo-dT column.
  • cDNA was synthesized from the polyadenylated RNA with random primers using the Gibco Superscript Choice system for cDNA Synthesis (Gibco BRL, Rockville, MD).
  • the cDNA was normalized using the PCR-Select cDNA Subtraction kit (Clontech, Palo Alto, CA) based on the suppression subtractive hybridization methods of Diatchenko et al. (1996, Proc. Natl. Acad. Sci. USA 93, 6025-6030, 1996).
  • the primers used for the normalization were:
  • PBMC library was screened using the methods described above in Section IF. A total of 14 GSEs were isolated using CEM-ss cells.
  • CF-674 (SEQ ID NO:69) is
  • GSE that interferes with HIV replication in a sense orientation and has substantial sequence identity with a gene encoding human translationally controlled tumor protein 1 (GenBank Accession No. HUMCH13C4A).
  • the complement of SEQ ID NO:69 is represented by SEQ ID NO:70.
  • CF-675 (SEQ ID NO:71) is another GSE that has substantial sequence identity with a gene encoding human translationally controlled tumor protein 1 (GenBank Accession No. HUMCH13C4A).
  • the complement of SEQ ID NO:71 is represented by SEQ ID NO:72.
  • CF-679 (SEQ ro NO:83) is another GSE that has substantial sequence identity with a gene encoding human translationally controlled tumor protein 1 (GenBank Accession No. HUMCH13C4A).
  • SEQ ID NO:83 The complement of SEQ ID NO:84.
  • CF-693 (SEQ ID NO:75) is GSE that interferes with HIV replication in a sense orientation and has substantial sequence identity with a gene encoding NEF interacting protein (GenBank Accession No. HSU83843).
  • the complement of SEQ ID NO:75 is represented by SEQ ID NO:76.
  • CF- 660 (SEQ ID NO: 79) is a GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding human hsp90 chaperone protein (Rebbe et al, 1989, J Biol Chem. 264:15006-15011).
  • the complement of SEQ ID NO:79 is represented by SEQ ID NO:80.
  • CF-653 SEQ ID NO:75
  • SEQ ID NO:67 is a GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding MEP-l alpha protein (Obata et al, 1988, J Virol. 62:4381-4386).
  • the complement of SEQ ID NO:67 is represented by SEQ ID NO:68.
  • CF-676 (SEQ ID NO:63) is a GSE that interferes with HTV replication as an antisense molecule and has substantial sequence identity with a gene encoding alpha enolase protein (Yu et al, 1997, Genome Res. 7:353-358).
  • the complement of SEQ ID NO:63 is represented by SEQ ID NO:64.
  • CF-675 (SEQ ID NO:65) is another GSE that has substantial sequence identity with a gene encoding alpha enolase protein.
  • the complement of SEQ ID NO:65 is represented by SEQ ID NO:66.
  • CF-673 (SEQ ID NO:73) is a GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding BBC-1 satellite DNA protein (GenBank AccessionNo. AJ223209).
  • the complement of SEQ ID NO:73 is represented by SEQ ID NO:74.
  • CF-672 (SEQ ID NO:81) is another GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding BBC-1 satellite DNA protein.
  • SEQ ID NO:81 The complement of SEQ ID NO:82 is represented by SEQ ID NO:82.
  • CF-681 (SEQ ID NO:85) is a GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding FK506-binding protein Al (Maki et al. , 1990, Proc. Natl. Acad. Sci. USA 87:5440-5443).
  • the complement of SEQ ID NO:85 is represented by SEQ ID NO:86.
  • CF-683 (SEQ ID NO:89) is a GSE that interferes with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding beta-signal sequence receptor protein (Chinen et al, ibid.).
  • SEQ ID NO:90 The complement of SEQ ID NO:90.
  • CF-684 (SEQ ID NO:91) is a GSE that interferes with HIV replication in a sense orientation and has substantial sequence identity with a gene encoding human tumorous imaginal disc protein (Schilling et al, ibid.).
  • the complement of SEQ ID NO:91 is represented by SEQ ID NO:92.
  • CF-685 (SEQ ID NO:93) is representative of a cluster of GSEs that interfere with HIV replication as an antisense molecule and has substantial sequence identity with a gene encoding cell surface heparin binding protein (Liu et al, ibid.).
  • the complement of SEQ ID NO:93 is represented by SEQ ID NO:94.
  • SEQ ID NO:95 Another member ofthe cluster is CF-686 (SEQ ID NO:95), also having substantial sequence identity with a gene encoding cell surface heparin binding protein (Liu et al, ibid.).
  • SEQ ID NO:96 The complement of SEQ ID NO:95 is represented by SEQ ID NO:96.
  • Example 4 This example describes the identification of GSEs that inhibit translocation of the HIV protein Rev.
  • Plasmids CF-24 and CF-367 (vector without insert), and CF-203, CF-261, CF-527, CF- 529, CF-537, CF-545, CF-619, CF-653, CF-659, CF-660, CF-662 and CF-674 are transfected into cells that express a Rev protein fused to a green fluorescent protein (Rev-GFP).
  • Rev-GFP green fluorescent protein
  • Confocal microscopy is used to determine whether transport of Rev-GFP fusion protein between the nucleus and the cytoplasm of a cell transfected with a plasmid containing a GSE nucleic acid sequence is inhibited by expression ofthe GSE.
  • the observations using cells transfected with GSE are compared with cells transfected with vector alone.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Zoology (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Enzymes And Modification Thereof (AREA)

Abstract

L'invention concerne des molécules d'acides nucléiques impliquées dans l'infection au HIV, des protéines codées par de telles molécules, ainsi que des composés protecteurs comprenant ces molécules d'acides nucléiques, ces protéines, et des inhibiteurs de produits codés par lesdites molécules. En outre, l'invention concerne également des procédés d'identification d'éléments suppresseurs génétiques supplémentaires, des gènes cellulaires correspondant à de tels éléments suppresseurs génétiques, ainsi que des procédés d'utilisation de ces gènes cellulaires et des produits de ceux-ci, codés, dans des méthodes de criblage destinées à la sélection d'inhibiteurs supplémentaires du HIV.
EP00961525A 1999-09-01 2000-09-01 Compositions et procedes d'inhibition de l'infection au virus de l'immunodeficience humaine, par regulation restrictive de genes cellulaires humains Withdrawn EP1238072A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US388182 1989-07-31
US38818299A 1999-09-01 1999-09-01
PCT/US2000/024262 WO2001016322A2 (fr) 1999-09-01 2000-09-01 Compositions et procedes d'inhibition de l'infection au virus de l'immunodeficience humaine, par regulation restrictive de genes cellulaires humains

Publications (1)

Publication Number Publication Date
EP1238072A2 true EP1238072A2 (fr) 2002-09-11

Family

ID=23533027

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00961525A Withdrawn EP1238072A2 (fr) 1999-09-01 2000-09-01 Compositions et procedes d'inhibition de l'infection au virus de l'immunodeficience humaine, par regulation restrictive de genes cellulaires humains

Country Status (6)

Country Link
EP (1) EP1238072A2 (fr)
JP (1) JP2003510033A (fr)
AU (1) AU771546B2 (fr)
CA (1) CA2382030A1 (fr)
IL (1) IL148225A0 (fr)
WO (1) WO2001016322A2 (fr)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003002528A2 (fr) * 2001-06-29 2003-01-09 Subsidiary No. 3 Compositions et procedes servant a inhiber l'infection par le virus de l'immunodeficience humaine par regulation negative de genes cellulaires humains
CU23896B1 (es) * 2010-04-01 2013-05-31 Ct De Ingeniería Genética Y Biotecnología Método para inhibir la replicación del vih en células de mamíferos
TW201620887A (zh) 2014-01-10 2016-06-16 葛蘭素史克智慧財產(第二)有限公司 化合物及方法
BR112016016153A2 (pt) 2014-01-13 2017-12-12 Berg Llc composições de enolase 1 (eno1) e usos das mesmas

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL83003A (en) * 1986-07-01 1995-07-31 Genetics Inst Factors that soak bone formation
US6071743A (en) * 1997-06-02 2000-06-06 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0116322A2 *

Also Published As

Publication number Publication date
JP2003510033A (ja) 2003-03-18
WO2001016322A3 (fr) 2002-07-11
AU771546B2 (en) 2004-03-25
WO2001016322A2 (fr) 2001-03-08
IL148225A0 (en) 2002-09-12
AU7346600A (en) 2001-03-26
CA2382030A1 (fr) 2001-03-08

Similar Documents

Publication Publication Date Title
EP1984393A2 (fr) Procédés, gènes et protéines de l'apoptose
US6071743A (en) Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
CA2496891A1 (fr) Nouveau lacs de proteine cytosquelette associee a actine
US20050142538A1 (en) Viral receptor protein
US6537972B1 (en) Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
EP0951545A2 (fr) Nouvelles proteines humaines se liant a la guanosine triphosphate
AU771546B2 (en) Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
WO2001016323A2 (fr) Compositions et procedes d'inhibition de l'infection due au virus de l'immunodeficience humaine a l'aide de genes cellulaires humains a regulation negative
US6613506B1 (en) Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US20020127648A1 (en) Novel human C5a-like receptor
WO2002042457A1 (fr) Clonage et expression de la slap-2 humaine : un nouvel homologue humain de la slap contenant des domaines sh2/sh3 qui presente une expression specifique aux cellules immunitaires
WO2004061088A2 (fr) Compositions et methodes d'inhibition d'une infection par le virus de l'immunodeficience humaine par regulation vers l'aval de genes cellulaires humains
WO1998033921A1 (fr) Nouvelle proteine humaine de type cystatine
CA2264544A1 (fr) Proteines sm snrnp
WO1998026056A1 (fr) Sous-unite regulatrice de la proteine phosphatase
WO2004053094A2 (fr) Compositions et procedes permettant d'inhiber l'infection par le virus de l'immunodeficience acquise par freination des genes cellulaires humains
WO1998050545A1 (fr) Proteine humaine de type smt-3
US7018822B1 (en) Guanosine monophosphate reductase
WO1998029553A1 (fr) Nouvelle reductase de l'acide guanylique
WO1998036077A1 (fr) Nouvelle proteine humaine de type parvuline
WO1999000500A1 (fr) Phosphoproteine fixatrice de calcium
WO2002017949A9 (fr) Nouvelles fonctions de dp214

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20020327

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: PPD DEVELOPMENT, LP

17Q First examination report despatched

Effective date: 20031211

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20040622