Title of Invention

"A PROCESS FOR ISOLATION OF A NUCLEIC ACID MOLECULE ENCODING A NOS POLYPEPTIDE"

Abstract A process for isolation of a nucleic acid molecule encoding a NOS polypeptide selected from the group consisting of a constitutively active NOS polypeptide, a NOS polypeptide, a NOS polypeptide having an increased rate of NO production, and a NOS polypepetide having an increased reductase activity, comprising separating the nucleic acid molecule from contaminant nucleic acids.
Full Text Cross References to Related Applications
This application claims the benefit of U.S. Provisional Application No. 60/129,550, filed April 16,1999, which is herein incorporated by reference in its entirety.
Acknowledgment of Federal Support
The present invention arose in part from research funded by the following federal grant monies: HL 57665 and HL 61371.
Technical Field
The present invention relates to new NOS variants or mutants which contain structural alterations in the site of Akt dependent phosphorylation. The altered NOS proteins or peptides and their encoding nucleic acid molecules are useful as gene therapy agents for the treatment of diseases including post angioplasty restenosis, hypertension, atherosclerosis, heart failure, diabetes and diseases with defective angiogenesis.
Background of the Invention
Atherosclerosis and vascular thrombosis are a major cause of morbidity and mortality, leading to coronary artery disease, myocardial infarction, and stroke. Atherosclerosis begins with an alteration in the endothelium which lines the blood vessels. An endothelial alteration may eventually result in the development of an endothelial lesion caused, in part, by the uptake of oxidized low-density lipoprotein (LDL) cholesterol. Rupture of this lesion can lead to thrombosis and occlusion of the blood vessel. In the case of a coronary artery, rupture of a complex lesion may

precipitate a myocardial infarction, whereas in the case of a carotid artery, stroke may ensue.
In atherosclerotic coronary heart disease, endothelial dysfunction may diminish production of vasodilatory substances, such as nitric oxide. Myocardial ischemia results when autoregulatory vasodilation is prevented, whether by flow-limiting coronary arterial stenosis or by endothelial dysfunction. In both cases, arterial blood flow can no longer increase proportional to rising oxygen demands. In other situations, myocardial ischemia may occur when oxygen demands are constant but there is a primary decrease in coronary blood flow mediated via coronary artery spasm, rapid evolution of the underlying atherosclerotic plaque leading to a reduced coronary arterial lumen caliber, and/or intermittent microvascular plugging by platelet aggregates.
Balloon angioplasty is commonly used to reopen a blood vessel which is narrowed by plaque. Although balloon angioplasty is successful in a high percentage of the cases in opening the vessel, it often denudes the endothelium and injures the vessel in the process. This damage causes the migration and proliferation of vascular smooth muscle cells of the blood vessel into the area of injury to form a lesion, known as myointimal hyperplasia or restenosis. This new lesion leads to a recurrence of symptoms within three to six months after the angioplasty in a significant proportion of patients.
In atherosclerosis, thrombosis and restenosis there is also a loss of normal vascular function, such that vessels tend to constrict, rather than dilate. The excessive vasoconstriction of the vessel causes further narrowing of the vessel lumen, limiting blood flow. This can cause symptoms such as angina (if a heart artery is involved), or transient cerebral ischemia {i.e. a "small stroke", if a brain vessel is involved). This abnormal vascular function (excessive vasoconstriction or inadequate vasodilation) occurs in other disease states as well. Hypertension (high blood pressure) is caused by excessive vasoconstriction, as well as thickening, of the vessel wall, particularly in the smaller vessels of the circulation. This process may affect the lung vessels as well causing pulmonary (lung) hypertension. Other disorders known to be associated with excessive vasoconstriction, or inadequate vasodilation include transplant atherosclerosis,
-2-

congestive heart failure, toxemia of pregnancy, Raynaud's phenomenon, Prinzmetal's angina (coronary vasospasm), cerebral vasospasm, hemolytic-uremia and impotence.
A substance released by the endothelium, initially referred to as "endothelium derived relaxing factor" (EDRF), plays an important role in inhibiting these pathologic processes. EDRF is now known to be nitric oxide (NO). NO plays many roles in human physiology, including the relaxation of vascular smooth muscle, the inhibition of platelet aggregation, the inhibition of mitogenesis, the proliferation of vascular smooth muscle, and leukocyte adherence. Because NO is the most potent endogenous vasodilator, and because it is largely responsible for exercise-induced vasodilation in the conduit arteries, enhancement of NO synthesis could also improve exercise capacity in normal individuals and those with vascular disease.
Endothelial nitric oxide synthase (eNOS) is the nitric oxide synthase (NOS) isoform responsible for the maintenance of systemic blood pressure, vascular remodeling and angiogenesis (Shesely et al, 1996; Huang et al, 1995; Rudic et al, 1998; Murohara et al,, 1998). As deficient endothelial production of NO is an early, persistent feature of atherosclerosis and vascular injury, eNOS has proven to be an attractive target for vascular gene therapy. While the regulation of eNOS activation remains largely undefined, it is known that eNOS is phosphorylated in response to various forms of cellular stimulation (Michel et al., 1993; Garcia-Cardena et al, 1996; Corson et al, 1996), however, the role of phosphorylation in the regulation of nitric oxide (NO) production and the kinase(s) responsible has not been previously elucidated.
Summary of the Invention
The present inventions result, in part, from the new discovery that the serine/threonine protein kinase, Akt (protein kinase B), can directly phosphorylate eNOS on a serine residue corresponding to residue 1179 in bovine eNOS or residue 1177 in human eNOS and activate the enzyme leading to NO production. Mutant eNOS (S1179A or S1177A) is resistant to Akt phosphorylation and activation while mutant eNOS (S1179D and S1177D) or (S1179E and S1177E) is constitutively active. Moreover, using adenoviral mediated gene transfer activated Akt increases basal NO
-3-

release from endothelial cells and activation deficient Akt attenuates VEGF stimulated NO production. Thus, eNOS is a newly described Akt substrate linking signal transduction via Akt to the release of the gaseous second messenger, NO. The present inventions are also based in part on the findings that mutant eNOS (S1179D) exhibits an increase in the rate of NO production and an increase in reductase activity.
The present invention includes NOS, polypeptides or proteins and their encoding isolated nucleic acid molecules, wherein the NOS polypeptide or protein contains a substituted amino acid residue corresponding to residue 1179 of bovine eNOS, residue 1177 of human eNOS, residue 1412 of rat nNOS, or residue 1415 at human nNOS. Preferred substitutions include amino acids with negatively charged R groups, including aspartic acid and glutamic acid.
The present invention also includes NOS polypeptides or proteins and their encoding isolated nucleic acid molecules, wherein the NOS polypeptide or protein contains a substituted amino acid residue corresponding to residue 1179 of bovine eNOS, residue 1177 of human eNOS, residue 1412 of rat nNOS, or residue 1415 at human nNOS. Preferred substitutions include amino acids with non-negatively charged R groups, such as alanine.
The present invention provides methods for stimulating collateral vessel development in ischemic diseases with deficient endogenous angiogenesis, specifically peripheral vascular disease and/or myocardial ischemia in a patient comprising delivering a transgene coding for an NOS polypeptide of the invention or an Akt polypeptide.
The invention further includes a non-human transgenic animal which express an NOS polypeptide of the invention.
Lastly, the invention includes methods of identifying an agent which modulates the Akt regulated activity of NOS, comprising the general steps of: (a) exposing purified NOS, preferably eNOS or nNOS, or a cell that expresses NOS, preferably eNOS or nNOS, and Akt to an agent; and (b) measuring the Akt regulated activity of NOS, preferably eNOS or nNOS.
-4-

Brief Description of the Drawings
Figures 1A-1B. Wild-type Akt, but not kinase inactive Akt increases NO release from cells expressing membrane associated eNOS. In Fig. 1A, COS cells were transfected with plasmids for eNOS, in the absence or presence of Akt or kinase inactive Akt (K179M) and the production of NO (assayed as NO2) determined by chemiluminescence. In Fig. 1B, COS cells were transfected with various NOS plasmids as above. In both Fig. 1A and Fig. 1B, values for NO2 production were subtracted from levels obtained from cells transfected with the -galactosidase cDNA only. The inset shows the expression of proteins in total cell lysates. Data are mean±SEM, n=3-7 experiments; * denotes p Figures 2A-2D. Phosphorylation of eNOS by active Akt in vitro and in vivo. In Fig. 2A, COS cells were transfected with HA-Akt or HA-Akt(K179M), lysates were immunoprecipitated and placed into an in vitro kinase reaction with histone 2B (25 mg) or recombinant eNOS (3mg) as substrates. The top panel depicts the incorporation of 32P into the substrates and the bottom panel shows the amount of substrate by Coomassie staining of the gel. In Fig. 2B, 32P labeled wild-type or the double serine mutant of eNOS (eNOS S635/1179) was affinity purified from transfected COS cells and subjected to autoradiography (upper panel) or Western blotting (lower panel). The graphical data in Fig. 2B reflects the relative amount of labeled protein to the amount of immunoreactive eNOS in the gel. In Fig. 2C, labeled eNOS was digested with trypsin and peptides separated by RP-HPLC. The upper chromatogram documents a predominant labeled tryptic peptide that co-migrates with a unlabeled synthetic phosphopeptide standard (bottom chromatogram). The insets demonstrate by linear mode MS of labeled peptide (top) and phosphopeptide standard identical mass ions. In Fig. 2D, recombinant wild-type eNOS or eNOS S1179A were purified and equal amounts (2.4 mg) placed into an in vitro kinase reaction with recombinant Akt as described in Methods. The top panel in Fig. 2D depicts the incorporation of 32P into eNOS and the bottom panel shows the amount of substrate by Coomassie staining of the
-5-

gel. The graphical data (n=3) reflects the relative amount of labeled eNOS to the mass of eNOS (Coomassie) in the in vitro kinase reaction.
Figure 3. Evidence that serine 1179 is functionally important for Akt stimulated NO release. COS cells were transfected with plasmids for wild-type eNOS or eNOS mutants, in the absence or presence of Akt and the expression of the proteins and production of NO (assayed as NO2) determined. Interestingly, constructs with the S1179 mutation to A were not activated by Akt and mutation of S1179 to D resulted in a gain of function. In A, data are mean±SEM of 4-7 experiments; * represent significant differences (p Figures 4A-4C. Akt regulates the basal and stimulated production of NO in endothelial cells. In Fig. 4A, BLMVEC were infected with adenoviral constructs ( -gal as control, myr Akt and AA-Akt) and the amount of NO2 produced over 24hrs determined (n=3). The inset shows the expression of eNOS and Akt. In Fig. 4B, lysates from adenoviral infected BLMVEC were examined for NOS activity. Equal amounts of protein (50 mg) were incubated with various concentrations of free calcium and NOS activity determined (n=3 experiments). In Fig. 4C, BLMVEC were infected with adenoviruses as above followed by stimulation with VEGF (40 ng/ml) for 30 min and NO2 release quantified by chemiluminescence. Data are presented as VEGF stimulated release of NO2 after subtraction of basal levels. Data are mean±SEM, n=4; * represent significant differences (p Figures SA and 5B. Purity and dimer/monomer ratio of wild type and eNOS S1179D. In A and B, SDS-PAGE analysis was performed on 7.5% polyacrylamide gels stained with Coomassie Blue. Molecular mass standards (lane 1) and their size in kDa are indicated at the left. Wild type eNOS (lane 2) and eNOS S1179D (lane 3) (1 g of each) were resolved as indicated by arrowheads. In B, proteins (2 g of each) were resolved on SDS-PAGE run at 4 °C, Molecular mass standards were in lane 1.
-6-

Nonboiled samples of wild type and eNOS S1179D were resolved in lanes 2 and 3, respectively. In lane 4, wild type-eNOS was boiled in SDS sample buffer.
Figures 6A and 6B. eNOS S1179D has higher rates of NO production (A) and reductase activity (B) than does wild type eNOS. In A, the rate of NO generated from wild type (o) and S1179D (o) eNOS was determined, using the hemoglobin capture assay, as a function of L-arginine concentration, and data are presented in a double reciprocal plot. In B, DCIP and cytochrome c assays were performed in the presence or absence of CaM. Values are mean ± S.E., n - 4-6 determinations. Similar results were obtained with at least three enzyme preparations. Significant differences (p Figures 7A and 7B. NOS activities (A) and NADPH-dependent reductase (B) are increased with eNOS S1179D compared with the wild type enzyme. Hemoglobin capture (A) and NADPH-dependent cytochrome c reduction (B) assays were performed on both wild type and S1179D eNOS. In A, the rate of NO production was determined in the presence of all NOS cofactors (wild type (filled symbols) and S1179D (open symbols) eNOS). The rate of cytochrome c reduction was performed in the absence of arginine and BH4 (A) for wild type (circles) and S1179D (triangles) in the presence (filled symbols) or absence of 120 nM calmodulin (open symbols). Values are mean ± S.E., n = 3-6 determinations from at least three enzyme preparations.
Figures 8A-8D. Calmodulin- and calcium-dependent activation of NOS and reductase activities are slightly enhanced for S1179D eNOS. Calmodulin-dependent hemoglobin capture (A) and cytochrome c reduction (B) were performed on both wild type (filled symbols) and S1179D eNOS (open symbols). The rate of NO production detected by hemoglobin capture method is in the presence of all NOS cofactors, whereas cytochrome c reduction was performed in the absence of arginine and BH4. In C and D, identical experiments were performed in the presence of increasing concentrations of free calcium. The insets in C and D depict the calcium-dependent turnover of S1179D and
-7-

wild type eNOS in both NO production and cytochrome c assays. The maximal turnover rates were as follows for wild type and S1179D eNOS, respectively: A, 22 and 43 min1; B, 620 and 1400 min1; C, 58 and 100 min1; and D, 1930 and 3810 min1. Values are mean ± S.E., n - 3-6 determinations from at least three enzyme preparations.
Figures 9A and 9B. EGTA-initiated inactivation of NOS is reduced in S1179D eNOS. Hemoglobin capture (A) and reductase assays (B) were performed as described earlier, with the following modifications. The reaction was monitored for 1 min to determine the initial rate; then, EGTA was added to the reaction mixture, and the rate was monitored for an additional 1 min. The free calcium concentration in the reaction was 200 M, and the amount of EGTA added resulted in final concentrations of 0,200, 400, and 600 M chelator. The specific activities are normalized to 100% for wild type and S1179D eNOS. Values are mean ±S.E.,n = 3-6 determinations from at least three enzyme preparations, nd, no detectable activity for wild type eNOS.
Detailed Description of the Invention A. General Description
The present inventions are based, in part, upon the discovery that the serine/threonine protein kinase, Akt (protein kinase B), can directly phosphorylate eNOS on serine 1179 (serine 1177 in human eNOS), and activate the enzyme leading to NO production while mutant eNOS (S1179A) is resistant to Akt phosphorylation and activation. Moreover, using adenoviral mediated gene transfer activated Akt increases basal NO release from endothelial cells and activation deficient Akt attenuates VEGF stimulated NO production. Thus, eNOS is a newly described Akt substrate linking signal transduction via Akt to the release of the gaseous second messenger, NO. The present inventions are also based in part on the findings that mutant eNOS, for instance, S1179D, exhibits an increase in the rate of NO production and an increase in reductase activity.
The demonstration that NO production is regulated by Akt dependent phosphorylation of eNOS provides novel constitutively active eNOS mutants for use in gene therapy aimed at improving endothelial function in cardiovascular diseases
-8-

associated with dysfunction in the synthesis or biological activity of NO. Such diseases include post angioplasty restenosis, hypertension, atherosclerosis, heart failure including myocardial infarction, diabetes, and diseases with defective angiogenesis. This discovery also provides a novel therapeutic target useful for the design of drugs useful for treating diseases associated with dysfunction in the synthesis or biological activity of NO.
The present invention also provides novel constitutively active nNOS mutants which have a substituted amino acid corresponding to residue 1412 of rat nNOS or 1415 or human nNOS for use in gene therapy aimed at the treatment of diseases.
B. Specific Embodiments
Production of NOS Mutant Proteins or Polypeptides
The present invention provides NOS proteins or polypeptides, allelic variants of NOS proteins, and conservative amino acid substitutions of NOS proteins, all of which contain a mutation of a serine residue which is the site of Akt mediated phosphorylation. For instance, the proteins or polypeptides of the invention include but are not limited to: (1) human eNOS proteins which comprise a mutation of residue 1177 (Janssens et at (1992) J. Biol Chem. 267:14519-14522 which is herein incorporated by reference in its entirety) from a serine to another amino acid, such as alanine, and are resistant to Akt mediated phosphorylation; (2) bovine eNOS proteins which comprise a mutation of residue 1179 (SEQ ID NO: 2 of U.S. Patent 5,498,539, which is herein incorporated by reference in its entirety) from a serine to another amino acid, such as alanine, and are resistant to Akt mediated phosphorylation; (3) human nNOS proteins which comprise a mutation of residue 1415 from a serine to another amino acid, such as alanine, and are resistant to Akt mediated phosphorylation; (4) rat nNOS proteins which comprise a mutation of residue 1412 from a serine to another amino acid, such as alanine, and are resistant to Akt mediated phosphorylation; (5) human eNOS proteins which comprise a mutation of residue 1177 from a serine to an amino acid containing a negatively charged R group, such as aspartic or glutamic acid, and are constitutively active and exhibit increased NO production and increased reductase activity; (6) bovine eNOS proteins
-9-

which comprise a mutation of residue 1179 from a serine to an amino acid containing a negatively charged R group, such as aspartic or glutamic acid, and are constitutively active and exhibit increased NO production and increased reductase activity; (7) human nNOS proteins which comprise a mutation of residue 1415 from a serine to an amino acid containing a negatively charged R group, such as aspartic or glutamic acid, and are constitutively active and exhibit increased NO production and increased reductase activity; (8) rat nNOS proteins which comprise a mutation of residue 1412 from a serine to an amino acid containing a negatively charged R group, such as aspartic or glutamic acid, and are constitutively active and exhibit increased NO production and increased reductase activity; and (9) NOS proteins from species other than humans, cows, or rat which are modified to contain an amino acid other than serine at a position corresponding to the serine at position 1177 in the human eNOS or position 1179 in the bovine eNOS, position 1412 in the rat nNOS, and position 1415 in the human nNOS and which are either resistant to Akt phosphorylation, are constitutively active, or exhibit increased NO production and increased reductase activity. NOS mutants may also be produced by mutating other amino acids in the phosphorylation motif RXRXXS/T.
The present invention provides constitutively active NOS polypeptides, preferably eNOS or nNOS, exhibiting increased NO production and reductase activity and comprising a mutation at the serine residue at the site of Akt mediated phosphorylation. It is also within the skill of the artisan to obtain conservative variants such as substitutions,deletions, and insertions mutants of theseNOS polypeptides exhibiting mcreased NO production and reductase activity. As used herein, a conservative variant refers to alterations in the amino acid sequence that do not adversely affect the ability of constitutively active NOS, preferably eNOS or nNOS, to produce NO or the reductase activity of constitutively active NOS, preferably eNOS or nNOS. A substitution, insertion, or deletion is said to adversely affect constitutively active NOS polypeptide, when the altered sequence affects the ability of constitutive NOS, such that it does not produce NO at an mcreased level and does not have increased reductase activity as compared to the wild-type NOS. For example, the overall charge, structure or hydrophobic/hydrophilic properties of constitutive NOS can be altered without adversely
-10-

affecting the activity of constitutive NOS. Accordingly, the amino acid sequence of NOS polypeptide can be altered, for instance to render the polypeptide more hydrophobic or hydrophilic, without adversely affecting the activity of NOS.
As used herein, a "constitutively active" mutant or variant of NOS, whether modified or isolated from a natural source, refers to a NOS protein, preferably an eNOS or a nNOS, which produces NO at a rate higher than native NOS containing a serine in its unphosphorylated form at an amino acid residue corresponding to residue 1177 in human NOS or residue 1179 in bovine NOS. Preferred constitutively active variants comprise an amino acid with a negatively charged R group, such as aspartic or glutamic acid, at the amino acid residue corresponding to the serine at position 1177 in the human eNOS or position 1179 in the bovine NOS.
The present invention provides NOS proteins or polypeptides, allelic variants of NOS proteins, and conservative amino acid substitutions of NOS proteins that contain a substituted amino acid residue corresponding to residue 1177 of bovine eNOS, to residue 1179 of human eNOS, to residue 1412 of rat nNOS, and to residue 1415 of human nNOS, wherein the substituted amino acid residue comprises a non-negatively charged R group, such as alanine.
The NOS proteins, preferably eNOS or nNOS proteins, of the present invention may be in isolated form. As used herein, a protein is said to be isolated when physical, mechanical or chemical methods are employed to remove the protein from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated protein.
Also included in the invention are NOS peptides which span the Akt phosphorylation site corresponding to residue 1179 in bovine eNOS, residue 1177 in human eNOS, residue 1412 in rat nNOS or residue 1415 in human nNOS. Peptides may contain a serine at the phosphorylation site or, preferably, may contain a substitution of the serine at position corresponding to residue 1179 in bovine eNOS, residue 1177 in human eNOS, residue 1412 in rat nNOS, or residue 1415 in human nNOS. Such substitutions include, but are not limited to, amino acids with an R group that mimics serine in its phosphorylated state, such as aspartic acid or glutamic acid. Such
-11-

substitutions also include, amino acids with a non-negative R group, such as alanine. Peptides spanning this site may be about 3, 5,7,10,12,15, 17,20,25, 30, 40, 50 or more amino acids in length.
NOS proteins, polypeptides or peptides of the invention may be prepared by any means available, including recombinant expression from an NOS cDNA which has been modified to replace or alter the nucleotide triplet encoding a serine corresponding to the serine at position 1177 in the human eNOS, position 1179 in the bovine eNOS, residue 1412 in rat nNOS, or residue 1415 in human nNOS. Any available technique may be
used to mutate the nucleotide triplet encoding the serine residue, such homologous

recombination, site-directed mutagenesis or PCR mutagenesis (see,.Sambrook et al., Molecular Cloning, Cold Spring Harbor Laboratory Press, 1989). Starting cDNAs may include the human and bovine eNOS cDNAs as well as cDNAs encoding eNOS proteins of other animal species, including but not limited to rabbit, rat, murine, porcine, ovine, equine and non-human primate species.
As used herein, a nucleic acid molecule encoding a NOS protein or polypeptide, preferably eNOS or nNOS protein or polypeptide, of the invention is said to be "isolated" when the nucleic acid molecule is substantially separated from contaminant nucleic acid encoding other polypeptides from the source of nucleic acid.
The present invention further provides fragments of the encoding nucleic acid molecule. As used herein, a fragment of an encoding nucleic acid molecule refers to a small portion of the entire protein encoding sequence. The size of the fragment will be determined by the intended use. For example, if the fragment is chosen so as to encode an active portion of the protein, the fragment will need to be large enough to encode the functional region(s) of the protein, including the Akt phosphorylation site. If the fragment is to be used as a nucleic acid probe or PCR primer, then the fragment length is chosen so as to obtain a relatively small number of false positives during probing/priming to a region which spans or flanks the NOS Akt phosphorylation site.
Fragments of the encoding nucleic acid molecules of the present invention (i.e., synthetic oligonucleotides) that are used as probes or specific primers for the polymerase chain reaction (PCR), or to synthesize gene sequences encoding proteins of the invention can
-12-

easily be synthesized by chemical techniques, for example, the phosphotriester method of Matteucci,et al. 1981 J.Am. Chem. Soc. 103:3185-3191) or using automated synthesis methods. In addition, larger DNA segments can readily be prepared by well known methods, such as synthesis of a group of oligonucleotides that define various modular segments of the gene, followed by ligation of oligonucleotides to build the complete modified gene.
The encoding nucleic acid molecules of the present invention may further be modified so as to contain a detectable label for diagnostic and probe purposes. A variety of such labels are known in the art and can readily be employed with the encoding molecules herein described. Suitable labels include, but are not limited to, biotin, radiolabeled nucleotides and the like. A skilled artisan can employ any of the art known labels to obtain a labeled encoding nucleic acid molecule.
The present invention further provides recombinant DNA molecules (rDNAs) that contain an NOS coding sequence as described above. As used herein, a rDNA molecule
is a DNA moleculethat has been subjected to molecular manipulation. Methods for
generating rDNA molecules are well known in the art, for example, see Sambrqok et al, Molecular Cloning ( 1989). In the preferred rDNA molecules, a coding DNA sequence is operably linked to expression control sequences and/or vector sequences.
The choice of vector and/or expression control sequences to which one of the protein family encoding sequences of the present invention is operably linked depends directly, as is well known in the art, on the functional properties desired, e.g., protein expression, and the host cell to be transformed. A vector contemplated by the present invention is at least capable of directing the replication or insertion into the host chromosome, and preferably also expression, of the structural gene included in the rDNA molecule.
Expression control elements that are used for regulating the expression of an operably linked protein encoding sequence are known in the art and include, but are not limited to, inducible promoters, constitutive promoters, secretion signals, and other regulatory elements. Preferably, the inducible promoter is readily controlled, such as being responsive to a nutrient in the host cell's medium.
-13-

In one embodiment, the vector containing a coding nucleic acid molecule will include a prokaryotic replicon, i.e., a DNA sequence having the ability to direct autonomous replication and maintenance of the recombinant DNA molecule extrachromosomally in a prokaryotic host cell, such as a bacterial host cell, transformed therewith. Such replicons are well known in the art. In addition, vectors that include a prokaryotic replicon may also include a gene whose expression confers a detectable marker such as a drug resistance. Typical bacterial drug resistance genes are those that confer resistance to ampicillin or tetracycline.
Vectors that include a prokaryotic replicon can further include a prokaryotic or bacteriophage promoter capable of directing the expression (transcription and translation) of the coding gene sequences in a bacterial host cell, such as E. coli. A promoter is an expression control element formed by a DNA sequence that permits binding of RNA polymerase and transcription to occur. Promoter sequences compatible with bacterial hosts are typically provided in plasmid vectors containing convenient restriction sites for insertion of a DNA segment of the present invention. Typical of such vector plasmids are pUC8, pUC9, pBR322 and pBR329 available from Biorad Laboratories, (Richmond, CA), pPL and pKK223 available from Pharmacia, Piscataway, N J.
Expression vectors compatible with eukaryotic cells, preferably those compatible with vertebrate cells, can also be used to form a rDNA molecules that contains a coding sequence. Eukaryotic cell expression vectors are well known in the art and are available from several commercial sources. Typically, such vectors are provided containing convenient restriction sites for insertion of the desired DNA segment. Typical of such vectors are pSVL and pKSV-10 (Pharmacia), pBPV-l/pML2d (International Biotechnologies, Inc.), pTDTl (ATCC, #31255), and the like eukaryotic expression vectors.
Eukaryotic cell expression vectors used to construct the rDNA molecules of the present invention may further include a selectable marker that is effective in an eukaryotic cell, preferably a drug resistance selection marker. A preferred drug resistance marker is the gene whose expression results in neomycin resistance, i.e., the neomycin phosphotransferase (neo)gene. (Southern et al. (1982) J.Mol.Anal Genet. 1:327-341) Alternatively, the selectable marker can be present on a separate plasmid, and the two vectors are introduced by
-14-

co-transfection of the host cell, and selected by culturing in the appropriate drug for the selectable marker.
The present invention further provides host cells transformed or transfected with a nucleic acid molecule that encodes an NOS protein, preferably eNOS or nNOS protein, of the present invention. The host cell can be either prokaryotic or eukaryotic. Eukaryotic cells useful for expression of a protein of the invention are not limited, so long as the cell line is compatible with cell culture methods and compatible with the propagation of the expression vector and expression of the gene product. Preferred eukaryotic host cells include, but are not limited to, yeast, insect and mammalian cells, preferably vertebrate cells such as those from a mouse, rat, monkey or human cell line. Preferred eukaryotic host cells include Chinese hamster ovary (CHO) cells available from the ATCC as CCL61, NIH Swiss mouse embryo cells NIH/3T3 available from the ATCC as CRL 1658, baby hamster kidney cells (BHK), and the like eukaryotic tissue culture cell lines. Any prokaryotic host can be used to express a rDNA molecule encoding a protein of the invention. The preferred prokaryotic host is E, coli, particularly for the constitutively active NOS mutants.
Transformation or transfection of appropriate cell hosts with a rDNA molecule of the present invention is accomplished by well known methods that typically depend on the type of vector used and host system employed. With regard to transformation of prokaryotic host cells, electroporation and salt treatment methods are typically employed, see, for example, Cohen et al, (1972) Proc. Natl Acad. Set. USA 69:2110; and Maniatis et al, Molecular Cloning. A Laboratory Mammal. Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (1982). With regard to transformation of vertebrate cells with vectors containing rDNAs, electroporation, cationic lipid or salt treatment methods are typically employed, see, for example, Graham et al. 1983) Virol 52:456; Wigler et al, (1979) Proc. Natl Acad. Sci. USA 76:1373-76. Successfully transformed or transfected cells, i.e., cells that contain a rDNA molecule of the present invention, can be identified by well known techniques including the selection for a selectable marker. For example, cells resulting from the introduction of an rDNA of the present invention can be cloned to produce single colonies. Cells from those colonies can be harvested, lysed and their DNA content examined for the presence of the rDNA using a method such as that described by Southern (1975) J. Mol Biol 98:503 or
-15-

Berent et al. (1985) Biotech. 3:208, or the proteins produced from the cell assayed via an immunological method.
The present invention further provides methods for producing a NOS protein, preferably an eNOS protein or a nNOS protein, of the invention using nucleic acid molecules herein described. In general terms, the production of a recombinant form of a protein typically involves the following steps. A nucleic acid molecule is first obtained that encodes aprotein of the invention. If the encoding sequence is uninterrupted by introns, it is directly suitable for expression in any host. The nucleic acid molecule is then preferably placed in operable linkage with suitable control sequences, as described above, to form an expression unit containing the protein open reading frame. The expression unit is used to transform or transfect a suitable host and the transformed or transfected host is cultured under conditions that allow the production of the recombinant protein. Optionally the recombinant protein is isolated from the medium or from the cells; recovery and purification of the protein may not be necessary in some instances where some impurities may be tolerated.
Each of the foregoing steps can be done in a variety of ways. For example, the desired coding sequences may be obtained from genomic fragments and used directly in appropriate hosts. The construction of expression vectors that are operable in a variety of hosts is accomplished using appropriate replicons and control sequences, as set forth above. The control sequences, expression vectors, and transformation or transfection methods are dependent on the type of host cell used to express the gene and were discussed in detail earlier. Suitable restriction sites can, if not normally available, be added to the ends of the coding sequence so as to provide an excisable gene to insert into these vectors. A skilled artisan can readily adapt any host/expression system known in the art for use with the nucleic acid molecules of the invention to produce recombinant protein.
Gene therapy
Any appropriate gene delivery system combined with a suitable gene expression system using the most appropriate route of delivery is encompassed by the present invention. For instance, NOS mutant or variant genes, preferably eNOS or nNOS mutant or variant genes, of the invention or Akt genes may be transferred to the heart (or skeletal muscle),
-16-

including cardiac myocytes (and skeletal myocytes), in vitro or in vivo to direct production of the encoded protein. Particularly useful are human Akt genes and NOS mutants, preferably human eNOS, containing an amino acid with a negatively charged R group, such as aspartic or glutamic acid, at a position corresponding to serine 1177 in human eNOS. Routes of administering NOS mutant or variant genes include, but are not limited to, intravascular, intramuscular, intraperitoneal, intradermal, and intraarterial injection.
The adenovirus gene delivery system offers several advantages: adenovirus can (i) accommodate relatively large DNA inserts; (ii) be grown to high-titer, (iii) infect a broad range of mammalian cell types; and (iv) be used with a large number of available vectors containing different promoters. Also, because adenoviruses are stable in the bloodstream, they can be administered by intravenous injection. A preferred delivery vector is a helper-independent replication deficient human adenovirus 5, although other delivery means are available and may be used, including delivery of nucleic acids directly to the cells of interest (see Sawa et al. (1998)Gene Ther. 5(ll):1472-80;Labhasetwar et al. (1998) J. Pharm. Sci 87(1l):1347-50; Lin et al. (1997) Hypertension 30:307-313; Chen et al. (1997) Circ Res. 80(3):327-335; Chatmon et al. (1996) Cardiovasc. Res. 32:962-972; Harv Heart Lett. (1999) 9(8):5-6; andNabel et al. (1999)Nat. Med. 5(2):141-2.
Using the adenovirus 5 system, transfection frequencies of greater than 60% have been demonstrated in myocardial cells in vivo by a single intracoronary injection (Giordano and Hammond (1994) Clin. Res. 42:123A). Non-replicative recombinant adenoviral vectors are particularly useful in transfecting coronary endothelium and cardiac myocytes resulting in highly efficient transfection after intracoronary injection. Non-replicative recombinant adenoviral vectors are also useful for transfecting desired cells of the peripheral vascular system (see U.S. Patent 5,792,453, which is herein incorporated by reference in its entirety).
Adenoviral vectors used in the present invention can be constructed by the rescue recombination technique described in Graham et al. (1988) Virology 163:614-617. Briefly, the eNOS transgene is cloned into a shuttle vector that contains a promoter, polylinker and partial flanking adenoviral sequences from which ElA/E1B genes have been deleted. As the shuttle vector, plasmid pAC1 (Virology 163:614-617,1988) (or an analog) which encodes portions of the left end of the human adenovirus 5 genome (Virology 163:614-617,1988)
-17-

minus the early protein encoding E1A and E1B sequences that are essential for viral replication, and plasmid ACCMVPLPA (J. Biol Chem. 267:25129-25134,1992) which contains polylinker, the CMV promoter and SV40 polyadenylation signal flanked by partial adenoviral sequences from which the EA/E1B genes have been deleted can be exemplified. The use of plasmid PAC1 or ACCMVPLA facilitates the cloning process. The shuttle vector is then co-transfected with a plasmid which contains the entire human adenoviral 5 genome with a length too large to be encapsidated, into 293 cells. Co-transfection can be conducted by calcium phosphate precipitation or lipofection (Biotechniques 15:868-872,1993). Plasmid JM17 encodes the entire human adenovirus 5 genome plus portions of the vector pBR322 including the gene for ampicillin resistance (4.3 kb). Although JM17 encodes all of the adenoviral proteins necessary to make mature viral particles, it is too large to be encapsidated (40 kb versus 36 kb for wild type). In a small subset of co-transfected cells, rescue recombination between the transgene containing the shuttle vector such as plasmid pACl and the plasmid having the entire adenoviral 5 genome such as plasmid pJM17 provides a recombinant genome that is deficient in the ElA/El B sequences, and that contains the transgene of interest but secondarily loses the additional sequence such as the pBR322 sequences during recombination, thereby being small enough to be encapsidated. The CMV driven beta-galactosidase encoding adenovirus HCMVSPl lacZ (Clm. Res. 42:123A, 1994) can be used to evaluate efficiency of gene transfer using X-gal treatment.
In another embodiment, the gene encoding NOS, preferably eNOS or nNOS, may be introduced in vivo via an attenuated or defective DNA virus, such as but not limited to herpes simplex virus (HSV), papillomavims, Epstein Barr virus (EBV), adenovirus, and adeno-associated virus (AAV). Defective viruses, which entirely or almost entirely lack viral genes, are preferred. Defective virus is not infective after introduction into a cell. Use of defective, vital vectors allows for administration to cells in a specific, localized area, without concern that the vector can infect other cells. Thus, a particular locus, e.g., in the brain or spinal chord, can be specifically targeted with the vector. In a specific embodiment, a defective herpes virus 1 (HSV1) vector may be used (Kaplitt et al. (1991)Molec. Cell Neurosci. 2:320-330). In yet another embodiment, the viral vector is an attenuated adenovirus vector, such as the vector described by Stratford-Perricaudet et al. (J. Clin. Invest.
-18-

90:626-630 (1992)). In a yet a further embodiment, the vector is a defective adeno-associated virus vector (Samulski et al. (1987)J. Virol 61:3096-3101; Samulski et al. (1989)J. Virol 63:3822-3828).
The present invention also contemplates the use of cell targeting not only by delivery of the transgene into the coronary artery, or femoral artery, for example, but also the use of tissue-specific promoters. By fusing, for example, tissue-specific transcriptional control sequences of left ventricular myosin light chain-2 (MLC[2V]) or myosin heavy chain (MHC) to a transgene such as the NOS genes of the invention within the adenoviral construct, transgene expression is limited to ventricular cardiac myocytes. The efficacy of gene expression and degree of specificity provided by MLC[2V ]and MHC promoters with lacZ have been determined, using the recombinant adenoviral system of the present invention. Cardiac-specific expression has been reported previously by Lee et al. (J. Biol. Chem. 267:15875-15885 (1992)). The MLC[2Vj promoter is comprised of 250 bp, and fits easily within the adenoviral-5 packaging constraints. The myosin heavy chain promoter, known to be a vigorous promoter of transcription, provides a reasonable alternative cardiac-specific promoter and is comprised of less than 300 bp. Smooth muscle cell promoters such as SM22 alpha promoter(Kemp et al, (1995) Biochem 7310 (Pt 3):1037-43) and SM alpha actin promoter (Shimizu et al (1995) J Biol Chem 270(13):7631-43) are also available. Other promoters, such as the troponin-C promoter, while highly efficacious and sufficiently small, lacks adequate tissue specificity. By using the MLC[2V ]or MHC promoters and delivering the transgene in vivo, it is believed that the cardiac myocyte alone (that is without concomitant expression in endothelial cells, smooth muscle cells, and fibroblasts within the heart) will provide adequate expression of the NOS protein.
Limiting expression to the cardiac myocyte also has advantages regarding the utility of gene transfer for the treatment of clinical myocardial ischemia. By limiting expression to the heart, one avoids the potentially harmful effect of angiogenesis in non-cardiac tissues such as the retina. In addition, of the cells in the heart, the myocyte would likely provide the longest transgene expression since the cells do not undergo rapid turnover, expression would not therefore be decreased by cell division and death as would occur with endothelial cells. Endothelial-specific promoters are already available for this purpose. Examples of
-19-

endothelial specific promoters include the Tie-2 promoter (Schlaeger et al (1997) Proc Natl Acad Sci l;94(7):3058-63), the endothelin promoter (Lee et al. (1990) J.Biol Chem. 265:10446-10450), and the eNOS promoter (Zhang et al. (1995) JBiol Chem 270(25): 15320-6).
The present invention includes, with regard to the treatment of heart disease, targeting the heart by intracoronary or intramuscular injection with a high titer of the vector and transfecting all cell types is presently preferred. Diseases such as erectile dysfunction and cardiovascular diseases including, mycardial infarction, myocardial ischemia, heart failure, restenosis, stent stenosis, post-angioplasty stenosis, and by-pass graft failure may be treated as described using the NOS transgenes, preferably eNOS or nNOS transgenes.
Successful recombinant vectors can be plaque purified according to standard methods. The resulting viral vectors are propagated on 293 cells which provide El A and E1B functions in trans to titers in the preferred about 1010- about 1012 viral particles/ml range. Cells can be infected at 80% confluence and harvested 48 hours later. After 3 freeze-thaw cycles the cellular debris is pelleted by centrifugation and the virus purified by CsCl gradient ultracentrifugation (double CsCl gradient ultracentrifugation is preferred). Prior to in vivo injection, the viral stocks are desalted by gel filtration through Sepharose columns such as G25 Sephadex. The product is then filtered through a 30 micron filter, thereby reducing deleterious effects of intracoronary injection of unfiltered virus (life threatening cardiac arrhythmias) and promoting efficient gene transfer. The resulting viral stock has a final viral titer in the range of 1010-1012 viral particles/ml. The recombinant adenovirus must be highly purified, with no wild-type (potentially replicative) virus. Impure constructs can cause an intense immune response in the host animal. From this point of view, propagation and purification may be conducted to exclude contaminants and wild-type virus by, for example, identifying successful recombinants with PCR using appropriate primers, conducting two rounds of plaque purification, and double CsCl gradient ultracentrifugation. Additionally, the problems associated with cardiac arrhythmias induced by adenovirus vector injection into patients can be avoided by filtration of the recombinant adenovirus through an appropriately-sized filter prior to intracoronary Injection. This strategy also appears to substantially improve gene transfer and expression.
-20-

The viral stock can be in the form of an injectable preparation containing pharmaceutically acceptable carrier such as saline, for example, as necessary. The final titer of the vector in the injectable preparation is preferably in the range of about 107- about 1013 viral particles which allows for effective gene transfer. Other pharmaceutical carriers, formulations and dosages are described below. The adenovirus transgene constructs are delivered to the myocardium by direct intracoronary (or graft vessel) injection using standard percutaneous catheter based methods under fluoroscopic guidance, at an amount sufficient for the transgene to be expressed to a degree which allows for highly effective therapy. The injection may be made deeply into the lumen (about 1 cm within the arterial lumen) of the coronary arteries (or graft vessel), and preferably be made in both coronary arteries, as the growth of collateral blood vessels is highly variable within individual patients. By injecting the material directly into the lumen of the coronary artery by coronary catheters, it is possible to target the gene rather effectively, and to minimize loss of the recombinant vectors to the proximal aorta during injection. It is known that gene expression when delivered in this manner does not occur in hepatocytes and viral RNA cannot be found in the urine at any time after intracoronary injection. Any variety of coronary catheter, or a Stack perfusion catheter, for example, can be used in the present invention. In addition, other techniques known to those having ordinary skill in the art can be used for transfer of NOS genes, preferably eNOS or nNOS, to the arterial wall.
For the treatment of peripheral vascular disease, a disease characterized by insufficient blood supply to the legs, recombinant adenovirus expressing a NOS, preferably an eNOS or a nNOS, peptide or protein of the invention may be delivered by a catheter inserted into the proximal portion of the femoral artery or arteries, thereby effecting gene transfer into the cells of the skeletal muscles receiving blood flow from the femoral arteries.
In instances wherein a transgene or nucleic acid encoding an NOS, preferably an eNOS or a nNOS, or Akt protein of the invention is first transferred to endothelial or vascular smooth muscle cells in vitro, including the patients own cells, DNA may be transfected into the cells directly (see U.S. Patent 5,658,565). Generally, to transfect target cells, a plasmid vector comprising a DNA sequence encoding an Akt or NOS of the invention or a

biologically active fragment thereof may be utilized in liposome-mediated transfection of the
-21-

target cell. The stability of liposomes, coupled with the impermeable nature of these vesicles, makes them useful vehicles for the delivery of therapeutic DNA sequences (for a review, see Mannino and Gould-Forgerite (1988) BioTechniques 6(7): 682-690). Liposomes are known to be absorbed by many cell types by fusion. In one embodiment, a cationic liposome containing cationic cholesterol derivatives, such as SF-chol or DC-chol, may be utilized. The DC-chol molecule includes a tertiary amino group, a medium length spacer arm and a carbamoyl linker bond as described by Gao and Huang (Biochem. Biophys. Res. Comm. 179: 280-285, 1991).
In another embodiment regarding the use of liposome technology, the viral or nonviral based vector comprising the DNA sequence encoding a biologically active NOS protein fragment, preferably eNOS or nNOS protein fragment, is delivered to the target cell by transfection of the target cell with lipofectamine (Bethesda Research Laboratory). Lipofectamine is a 3:1 Liposome formulation of the polycationic lipid 2,3 dioleyloxy-N-[2(spemiinecarboxymido)ethyl]- N,N-dimethyl-1 -propanaminiumtric fluroacetate (DOPSA) and the neutral lipid dioleoly-phosphatidylethanolamine (DOPE).
Other non-viral modes of gene delivery include, but are not limited to: (a) direct injection of naked DNA; (b) calcium phosphate [Ca3(PO4)2] mediated cell transfection; (c) mammalian host cell transfection by electroporation; (d) DEAE-dextran mediated cell transfection; (e) polybrene mediated delivery; (f) protoplast fusion; (g) microinjection; and (h) polylysine mediated transformation, with the genetically engineered cells transferred back to the mammalian host.
Production of Transgenic Animals
Transgenic animals containing a mutant NOS gene, preferably a mutant eNOS or nNOS gene, as described herein are also included in the invention. Transgenic animals are genetically modified animals into which recombinant, exogenous or cloned genetic material has been experimentally transferred. Such genetic material is often referred to as a "transgene". The nucleic acid sequence of the transgene, in this case a form of NOS, may be integrated either at a locus of a genome where that particular nucleic acid sequence is not
-22-

otherwise normally found or at the normal locus for the transgene. The transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal.
The term "germ cell line transgenic animal" refers to a transgenic animal in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability of the transgenic animal to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic information, then they too are transgenic animals.
The alteration or genetic information may be foreign to the species of animal to which the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient In the last case, the altered or introduced gene may be expressed differently than the native gene.
Transgenic animals can be produced by a variety of different methods including transfection, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Patent No. 4,736,866; U.S. Patent No. 5,602,307; Mullins et al. (1993) Hypertension 22(4):630-633; Brenin et al (1997) Surg. Oncol. 6(2)99-110; Tuan (ed.), Recombinant Gene Expression Protocols, Methods in Molecular Biology No. 62, Humana Press (1997)).
A number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Patent No. 4,736,866); express simian SV 40 T-antigen (U.S. Patent No. 5,728,915); lack the expression of interferon regulatory factor 1 (IRF-1) (U.S. Patent No. 5,731,490); exhibit dopaminergic dysfunction (U.S. Patent No. 5,723,719); express at least one human gene which participates in blood pressure control (U.S. Patent No. 5,731,489); display greater similarity to the conditions existing in naturally occurring Alzheimer's disease (U.S. Patent No. 5,720,936); have a reduced capacity to mediate cellular adhesion (U.S. Patent No. 5,602,307); possess a bovine growth hormone gene (Clutter et al (1996) Genetics 143(4):1753-1760); or, are capable of generating a fully human antibody response (McCarthy (1997) The Lancet 349(9049) :405).
While mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal
-23-

species. Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g., Kim et al. (1997) Mol. Reprod. Dev. 46(4):515-526; Houdebine (1995) Reprod. Nutr. Dev. 35(6):609-617; Petters (1994) Reprod. Fertil. Dev. 6(5):643-645; Schnieke et al. (1997) Science 278(5346):2130-2133; and Amoah (1997) J. Animal Science 75(2):578-585).
The method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transformation of multiple nucleic acid molecules. Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Patent No. 5,489,743 and U.S. Patent No. 5,602,307. Furthermore, the production of NOS transgenic animals is well developed. For instance, transgenic mice which inducibly express or overexpress wild type eNOS have been produced (see Ohashi et al. (1998)J. Clin. Invest. 102(12):2061-71;and Drummond et al. (1998) J. Clin. Invest. 102(12):2033-4). These methods may be used to produce transgenic mice which express the NOS mutants of the invention.
Therapeutic Screening Assays
The discovery that phosphorylation of eNOS regulates its activity allows for the development of screening assays to identify agents which modulate Akt regulated NOS, preferably eNOS or nNOS, activity or expression. Any available format may be used, including in vivo transgenic animal assays, in vitro protein based assays, cell culture assays and high-throughput formats.
In many drug screening programs which test libraries of compounds, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with purified or semi-purified proteins, are often preferred as "primary" screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound. Moreover, the effects of cellular toxicity and/or bioavailabitity of the test compound can be generally ignored in the in vitro
-24-

system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an inhibition of, for instance, binding between molecules.
Cell or tissue culture based assays may be performed, for example, by plating COS-7 cells (100 mm dish) and transfecting with NOS (7.5-30 mg) and Akt (1 mg) plasmids using calcium phosphate. To balance all transfections, an expression vector for -galactosidase cDNA may be cotransfected. Twenty-four to forty-eight hours after transfection, the expression of appropriate proteins (40-80 mg) may be confirmed by Western blot analysis using eNOS mAb (9D10, Zymed), HA mAb (12CA5, Boehringer Mannheim), iNOS pAb (Zymed Laboratories) or nNOS mAb (Zymed Laboratories).
Twenty-four to forty-eight hours after transfection, media may be processed for the measurement of nitrite (NO2), the stable breakdown product of NO in aqueous solution, by NO specific chemiluminescence as described (Sessa et al, 1995). Media is deproteinized and samples containing NO2 are refluxed in glacial acetic acid containing sodium iodide. Under these conditions, NO2 is quantitatively reduced to NO which is quantified by a chemiluminescence detector after reaction with ozone in a NO analyzer (Sievers, Boulders, CO). In all experiments, controls may be prepared by inhibiting NO2 release by the use of a NOS inhibitor. In addition, NO2- release from cells transfected with the -galactosidase cDNA may subtracted to control for background levels of NO2 found in serum or media. cGMP accumulation in COS may also be used as a bioassay for the production of NO as described, In an alternative format, the conversion of 3H-L-arginine to 3H-L-citrulline may be used to determined NOS activity in COS cell or endothelial cell lysates as previously described (Garcia-Cardena et al, 1998).
For in vivo phosphorylation studies, COS cells may be transfected with the cDNAs for wild-type or S635 (control), bovine 1179A, D, or E eNOS, human 1177 D or E eNOS, rat 1412D or E nNOS, human 1415D or E nNOS and HA-Akt overnight. 36 hrs after transfection, cells are placed into dialyzed serum replete, phosphate- free Dulbecco's minimum essential medium supplemented with 80 Ci/ml of 32P orthophosphoric acid for 3 hr. A cell aliquot may be pretreated with wortmannin (500nM) in the phosphate-free media for 1 hr and during the labeling. Lysates are then harvested, NOS solubilized and partially purified by ADP sepharose affinity chromatography as previously described and the 32P
-25-

incorporation into NOS visualized after SDS-PAGE (7.5%) by autoradiography and the amount of NOS protein verified by Western blotting for NOS.
For in vitro phosphorylation studies, recombinant NOS purified from E. coli, eNOS purified from another source, or NOS peptides spanning the Akt phosphorylation site are incubated with wild-type or kinase inactive Akt immunoprecipitated from transfected COS cells. Briefly, the NOS proteins or peptides are incubated with 32P g-ATP (2 ml, specific activity 3000Ci/mmol), ATP (50 mM), DTT (1 mM), in a buffer containing HEPES (20 mM, pH= 7.4), MnCl2 (10 mM), MgCl2 (10 mM) and immunoprecipitated Akt for 20 min at room temperature.
In experiments examining the in vitro phosphorylation of wild-type and mutant NOS, recombinant Akt (lmg) purified from baculovirus infected SF9 cells, is incubated with wild-type, S1179A bovine eNOS, S1177A human eNOS, S1179 D or E bovine eNOS, S1177D or E human eNOS, S1412D or E rat nNOS, or S1415D or E human nNOS using essentially the same conditions as above. Proteins may be resolved by SDS-PAGE and 32P incorporation and the amount of protein determined by Coomassie staining as above.
The above screening assays which assay the Akt dependent phosphorylation or activation of NOS, preferably eNOS or nNOS, may be used to screen for a wide-variety of agents. For instance, agents which inhibit the dephosphorylation of NOS (phosphatase inhibitors) at an amino acid corresponding to serine 1179 in bovine eNOS, residue 1177 in human eNOS, residue 1412 in rat nNOS, or 1415 in human nNOS may be useful therapeutic molecules. Similarly, agents which activate Akt or which mimic the Akt phosphorylation site on eNOS may be useful therapeutic molecules.
Agents that are assayed in the above methods can be randomly selected or rationally selected or designed. As used herein, an agent is said to be randomly selected when the agent is chosen randomly without considering the specific sequences involved in the association of the a protein of the invention alone or with its associated substrates, binding partners, etc. An example of randomly selected agents is the use a chemical library or a peptide combinatorial library, or a growth broth of an organism.
As used herein, an agent is said to be rationally selected or designed when the agent is chosen on a nonrandom basis which takes into account the sequence of the target site and/or
-26-

its conformation in connection with the agent's action. For example, a rationally selected peptide agent can be a peptide whose amino acid sequence is similar to the Akt phosphorylation site in NOS, particularly, peptides or small molecules that mimic the NOS phosphorylation state.
The agents of the present invention can be, as examples, peptides, small molecules, vitamin derivatives, as well as carbohydrates. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
The peptide agents of the invention can be prepared using standard solid phase (or solution phase) peptide synthesis methods, as is known in the art. In addition, the DNA encoding these peptides may be synthesized using commercially available oligonucleotide synthesis instrumentation and produced recombinantly using standard recombinant production systems. The production using solid phase peptide synthesis is necessitated if non-gene-encoded amino acids are to be included.
Another class of agents of the present invention are antibodies immunoreactive with critical positions of proteins of the invention. Antibody agents are obtained by immunization of suitable mammalian subjects with peptides, containing as antigenic regions, those portions of the protein intended to be targeted by the antibodies.
Use of Agents Identified as Modulating eNOS Activity
The agents of the present invention, such as agents that inhibit the dephosphorylation of NOS (phosphatase inhibitors) at an amino acid corresponding to serine 1179 in bovine eNOS, residue 1177 in human eNOS, residue 1412 in rat nNOS, or residue 1415 in human nNOS, as well as agents which activate Akt or which mimic the Akt phosphorylation site on NOS, can be administered via parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, or buccal routes. Alternatively, or concurrently, administration may be by the oral route. The dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired. As described below, there are many methods that can readily be adapted to administer such agents.
-27-

The present invention further provides compositions containing one or more agents of the invention. While individual needs vary, a determination of optimal ranges of effective amounts of each component is within the skill of the art. Typical dosages comprise 0.1 to 100 mg/kg body wt. The preferred dosages comprise 0.1 to 10 mg/kg body wt The most preferred dosages comprise 0.1 to 1 mg/kg body wt.
In addition to the pharmacologically active agent, the compositions of the present invention may contain suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically for delivery to the site of action. Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, for example, water-soluble salts. In addition, suspensions of the active compounds as appropriate oily injection suspensions may be administered. Suitable lipophilic solvents or vehicles include fatty oils, for example, sesame oil, or synthetic fatty acid esters, for example, ethyl oleate or triglycerides. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, for example, sodium carboxymethyl cellulose, sorbitol, and/or dextran. Optionally, the suspension may also contain stabilizers. Liposomes can also be used to encapsulate the agent for delivery into the cell.
The pharmaceutical formulation for systemic administration according to the invention may be formulated for enteral, parenteral or topical administration. Indeed, all three types of formulations may be used simultaneously to achieve systemic administration of the active ingredient.
Suitable formulations for oral administration include hard or soft gelatin capsules, pills, tablets, including coated tablets, elixirs, suspensions, syrups or inhalations and controlled release forms thereof.
In practicing the methods of this invention, the compounds of this invention may be used alone or in combination, or in combination with other therapeutic or diagnostic agents. In certain preferred embodiments, the compounds of this invention may be coadministered along with other compounds typically prescribed for these conditions according to generally accepted medical practice, such as anticoagulant agents, thrombolytic agents, or other antithrombotics, including platelet aggregation inhibitors, tissue plasminogen activators,
-28-

urokinase, prourokinase, streptokinase, heparin, aspirin, or warfarin. The compounds of this invention can be utilized in vivo, ordinarily in mammals, such as humans, sheep, horses, cattle, pigs, dogs, cats, rats and mice, or in vitro.
The following working examples specifically point out preferred embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure. Other generic configurations will be apparent to one skilled in the art.
EXAMPLES
The following procedures were employed in Examples 1-2
Cell Transfections: The bovine eNOS, human iNOS, rat nNOS cDNAs in pcDNA3 and HA-tagged wild-type Akt, Akt (K179M) or myr-Akt in pCMV6 were generated by standard cloning methods. The myr-nNOS in pcDNA3 was generated by PCR, incorporating a new amino terminus containing the eNOS N-myristoylation consensus site (MGNLKSVG, SEQ ID NO:1) fused in frame to the second amino acid of the nNOS coding sequence. In preliminary experiments in COS cells, this construct was N-myristoylated based on incorporation of 3H-myristic acid whereas native nNOS is not and resulted in approximately 60% of the total protein targeted into the membrane fraction of cells whereas only 5-10% of nNOS was membrane associated in COS cells. Mutation of the putative Akt phosphorylation sites in eNOS were generated using the Quick Change site-directed mutagenesis kit (Stratagene) according to the manufacturers instructions. All mutants were verified by DNA sequencing. COS-7 cells were plated (100 mm dish) and transfected with the NOS (7.5-30 mg) and Akt (1 mg) plasmids using calcium phosphate. To balance all transfections, the expression vector for p-galactosidase cDNA was used. Twenty-four to forty-eight hours after transfection, the expression of appropriate proteins (40-80 mg) were confirmed by Western blot analysis using eNOS mAb (9D10, Zymed), HA mAb (12CA5, Boehringer Mannheim), iNOS pAb (Zymed Laboratories) or nNOS mAb (Zymed Laboratories).
-29-

NO release from transfected COS cells: 24-48 hrs after transfection, media was processed for the measurement of nitrite (NO2), the stable breakdown product of NO in aqueous solution, by NO specific chemiluminescence as described (Sessa et al., 1995). Media was deproteinized and samples containing NO2 were refluxed in glacial acetic acid containing sodium iodide. Under these conditions, NO2 was quantitatively reduced to NO which was quantified by a chemiluminescence detector after reaction with ozone in a NO analyzer (Sievers, Boulders, CO). In all experiments, NO2 release was inhibitable by a NOS inhibitor. In addition, NO2 release from cells transfected with the -galactosidase cDNA was subtracted to control for background levels of NO2 found in serum or media. In some experiments, cGMP accumulation in COS was used as a bioassay for the production of NO as described.
NOS activity assays: The conversion of 3H-L-arginine to 3H-L -citrulline was used to determine NOS activity in COS cell or endothelial cell lysates as previously described by Garcia-Cardena et al. (1998).
Phosphorylation studies in vivo and in vitro: For in vivo phosphorylation studies, COS cells were transfected with the cDNAs for wild-type or S635,1179A eNOS and HA-Akt overnight. 36 hrs after transfection, cells were placed into dialyzed serum replete, phosphate- free Dulbecco's minimum essential medium supplemented with 80 Ci/ml of 32P orthophosphoric acid for 3 hr. Some cells were pretreated with wortmannin (500nM) in the phosphate-free media for 1 hr and during the labeling. Lysates were harvested, eNOS solubilized and partially purified by ADP sepharose affinity chromatography as previously described and the 32P incorporation into eNOS visualized after SDS-PAGE (7.5%) by autoradiography and the amount of eNOS protein verified by Western blotting for eNOS. For in vitro phosphorylation studies, recombinant eNOS purified from E. coli was incubated with wild-type or kinase inactive Akt irnmunoprecipitated from transfected COS cells. eNOS was incubated with 3ZP g-ATP (2 ml, specific activity 3000Ci/mmol), ATP (50 mM), DTT (1 mM), in a buffer containing HEPES (20 mM, pH= 7.4), MnCl2 (10 mM), MgCl2 (10 mM) and irnmunoprecipitated Akt for 20 min at room temperature.
-30-

In experiments examining the in vitro phosphorylation of wild-type and mutant eNOS, recombinant Akt (lmg) purified from baculovirus infected SF9 cells, was incubated with wild-type or S1179A eNOS (2.4 mg, purified from E. coli) using essentially the same conditions as above. Proteins were resolved by SDS-PAGE and 32P incorporation and the amount of protein determined by Coomassie staining as above.
In studies identifying the labeled eNOS peptide, immunoprecipitated Akt was incubated with recombinant eNOS as above. The sample was run on SDS-PAGE, and the eNOS band digested in gel, and the resultant tryptic fragments purified by RP-HPLC. Peptide mass and 32P incorporation were monitored and the prominent labeled peak further analyzed by mass spectrometry. In other experiments, peptides corresponding to the potential Akt phosphorylation site were synthesized, purified by HPLC and verified by mass spectrometry (W.M. Keck Biotechnology Resource Center, Yale University School of Medicine). The wild-type peptide was 1174RIRTQSFSLQERHLRGAVPWA1194 (SEQ ID NO:2) and the mutant peptide was identical except S1179 was changed to an alanine. In vitro kinase reactions were essentially as described above incubating peptides (25mg) with recombinant Akt (1mg). Reactions were then spotted onto phosphocellulose filters and the amount of phosphate incorporated measured by Cerenkov counting.
Adenoviral infections and NO release in endothehal cells: Bovine lung microvascular endothelial cells (BLMVEC) were cultured in either in 100 mm dishes (for basal NO release and NOS activity assays) or C6 well plates (for stimulated NO) as previously described (Garcia-Cardena et al., 1996a). BLMVEC were infected with 200 MOI ofadenovirus containing the p-galactosidase 29, HA-tagged, inactive phosphorylation mutant Akt (AA-Akt; Alessi et al.,l996) or carboxyl terminal HA-tagged constitutively active Akt (myr-Akt) for 4 hrs. The virus was removed and cells left to recover for 18 hrs in complete medium. In preliminary experiments with the p-galactosidase virus, these conditions were optimal for infecting 100% of the cultures. For measurement of basal NO production, media was collected for NO release 24hrs after the initial infection with virus. For measurement of stimulated NO release, cells were then washed with serum-tree medium followed by stimulation with VEGF (40ng/ml) for 30 min. In some experiments the calcium
-31-

dependency of NOS was determined 24hrs after adenoviral infection. Infected cells were lysed in NOS assay buffer containing 1% NP40, and detergent soluble material used for activity. Lysates were incubated with EGTA buffered calcium to yield appropriate amounts of free calcium in the incubation.
Statistics: Data are expressed as mean ± SEM. Comparison's were made using a two-tailed, Student's t-test or ANOVA with a post-hoc test where appropriate. Differences were considered to be significant at p Example 1: Akt Modulates NO Production from eNOS To explore the possibility that a downstream effector of PI-3 kinase, Akt, could directly influence the production of NO, COS-7 cells (which do not express NOS) were co-transfected with eNOS and wild type Akt (HA-Akt), or kinase inactive Akt (HA-Akt K179M) and the accumulation of nitrite (NO2) measured by NO specific chemiluminescence. Transfection of eNOS results in an increase in NO2 accumulation, an effect that is markedly enhanced by co-transfection of wild-type Akt, but not the kinase inactive variant (Fig. 1A). Identical results were obtained using cGMP as a bioassay for biologically active NO. Under these experimental conditions, Akt was catalytically active as determined by Western blotting with a phospho-Akt specific Ab (which recognizes serine 473; not shown) and Akt activity assays (see Fig. 2A). Transfection of a constitutively active form of Akt (myr-Akt) increases cGMP accumulation (assayed in COS cells) from 5.5± 0.8 to 11.6±0.9 pmol cGMP/mg protein (in cells transfected with eNOS alone or eNOS with myr-Akt, respectively) whereas the kinase inactive Akt did not influence cGMP accumulation (5.8±0.8 pmol cGMP, n=4 experiments). As seen in the inset, equal levels of eNOS and Akt were expressed in COS cell lysates suggesting that Akt modulates eNOS thereby increasing NO production under basal conditions.
eNOS is a dually acylated peripheral membrane protein that targets into the Golgi region and plasma membrane of endothelial cells (Liu et al, 1997; Garcia-Cardena et al, 1996a; Shaal et al, 1996) and compartmentalization is required for efficient production of NO in response to agonist challenge (Sessa et al, 1995; Liu et al.,1996; Kantor et al., 1996
-32-

). To examine if eNOS activation by Akt requires membrane compartmentalization, COS-7 cells were co-transfected with cDNAs for Akt and a myristoylation, palmitoylation defective mutant of eNOS (G2A eNOS) and the release of NO quantified. As seen in Fig. 1B, Akt did not activate the non-acylated form of eNOS suggesting that compartmentalization of both proteins to the membrane is required for their functional interaction (Downward et al, 1998). Next, it was determined if Akt could activate structurally similar but distinct soluble NOS isoforms, neuronal and inducible NOS (nNOS and iNOS, respectively). Co-transfection of Akt with nNOS and iNOS did not result in a further increase in NO release demonstrating the specificity of Akt for eNOS. However, the addition of an N-myristoylation site to nNOS, in order to enhance its interactions with biological membranes, results in Akt stimulation of nNOS in a manner analogous to that seen with eNOS, suggesting that both isoforms may be susceptible to activation by Akt kinase when membrane anchored.
Example 2: Production of eNOS Multations
The above experiments imply that Akt, perhaps via phosphorylation of eNOS, can modulate NO release from intact cells. Indeed, two putative Akt phosphorylation motifs (RXRXXS/T) are present in eNOS (serines 635 and 1179 in bovine eNOS or serines 633 and 1177 in human eNOS) and one motif present in nNOS (serines 1412 in rat and 1415 human nNOS), with no obvious motifs found in iNOS. To examine if eNOS is a potential substrate for Akt phosphoiylation in vitro, COS cells were transfected with HA-Akt or HA-Akt (K179M) and kinase activity assessed using recombinant eNOS as a substrate. As seen in Fig. 2A, the active kinase phosphorylates histone 2B and eNOS (69.3±2.9 and 115.4±3.8 pmol of ATP/nmol substrate, respectively, n=3), whereas the inactive Akt did not significantly increase histone or eNOS phosphorylation. To elucidate if the putative Akt phosphorylation sites in eNOS were responsible for the incorporation of 32P, the two serines were mutated to alanine residues and the ability of Akt to stimulate wild-type and mutant eNOS phosphorylation examined in intact COS cells. Transfected cells were labeled with 32P- orthophosphate, eNOS partially purified by ADP-sepharose affinity chromatography, and the phosphorylation state and protein levels quantified. As seen in Fig 2B, co-expression of Akt results in a 2 fold enhancement in the phosphorylation of eNOS relative to non-
-33-

stimulated cells. Pretreatment of eNOS/Akt transfected cells with wortmannin abolished the Akt induced increase in phosphorylation. Moreover, mutation of serines 635 and 1179 to alanine residues abolished Akt dependent phosphorylation of eNOS suggesting that these residues could serve as potential phosphorylation sites in intact cells.
To directly identify the residues phosphorylated by Akt, wild-type eNOS was incubated with immunopurified Akt and the sites of phosphorylation determined by HPLC followed by MALDi-mass spectrometry (MALDi-MS). As seen in Fig, 2C, the primary 32P-labeled tryptic phosphopeptide co-elutes with a synthetic phosphopeptide (amino acids 1177-1185 with phosphoserine at position 1179) and has the identical mass ion as determined by linear mode MS. Using reflectron mode MALDi-MS monitoring, both the labeled tryptic peptide and the standard phosphopeptide demonstrated a loss of HP3O4 indicating that the tryptic peptide was phosphorylated. In addition, mutation of S1179 to A markedly reduces Akt-dependent phosphorylation of eNOS compared to the wild-type protein (Fig. 2D). Identical results were obtained utilizing peptides (amino acids 1174-1194) derived from wild-type or eNOS S1179A as substrates for recombinant Akt (wild-type peptide incorporated 24.6±3.7 nmol phosphate/mg compared to the alanine mutant peptide which incorporated 0.22±0.02 nmol phosphate/mg; n=5). Collectively, these data demonstrate that eNOS is a substrate for Akt and that the primary site of phosphorylation is serine 1179 (serine 1177 in human eNOS).
Next we examined the functional significance of the putative Akt phosphorylation site at serine 635 and the identified site at serine 1179. Transfection of COS cells with the double mutant eNOS S635/1179A abolishes Akt dependent NO release. Mutation of serine 635 to alanine did not attenuate NO release whereas eNOS S1179A abolishes Akt dependent activation of eNOS (Fig. 3). These results suggest that serine 1179 is functionally important for NO release. Mutation of serine 1179 into aspartic acid (eNOS S1179D) to substitute for the negative charge afforded by the addition of phosphate, partially mimics the activation state induced by Akt (S1177D in human eNOS). All site directed mutants were amply expressed (see inset Western blots) and retained NOS catalytic activity in cell lysates (in COS cells transfected with eNOS only, NOS activity was 85.3± 27.0,71.9±2.9,80.8±23.2 and
-34-

131.8±36.7 pmol L-citrulline generated/mg protein from lysates of COS cells expressing wild type, S1179A, S635,1179A and eNOS S1179D, respectively, n=3 experiments).
To examine if Akt mediates NO release from endothelial cells, bovine lung microvascular endothelial cells (BLMVEC) were infected with adenoviruses expressing activated Akt (myr-Akt), activation deficient Akt (AA-Akt) or -galactosidase as a control and the accumulation of NO measured. As seen in Fig. 4A, myr-Akt stimulates the basal production of NO from BLMVEC, whereas cells infected with -galactosidase or activation deficient Akt released low levels of NO that were close to the limits of detection. These data in conjunction with similar results in COS cells suggests that Akt phosphorylation of eNOS is sufficient to regulate NO production at resting levels of calcium. Indeed, NOS activity measured in lysates from myr-Akt infected BLMVEC demonstrates that the sensitivity of the enzyme to activation by calcium, assayed at a fixed calmodulin concentration, is enhanced relative to NOS activity seen in BLMVEC infected with the p-galactosidase virus (Fig. 4B). Interestingly, the calcium sensitivity of NOS activity in cells infected with activation deficient Akt was greatly suppressed relative to both myr -Akt and -galactosidase infected cells.
Treatment of endothelial cells with VEGF is known to activate Akt 23 and the release of NO through a mechanism partially blocked by inhibitors of PI-3 kinase (Papapetropoulos et al, 1997). To examine the functional link between VEGF as an agonist for NO release and Akt activation, BLMVEC were infected with adenoviruses for myr-Akt, AA-Akt or p-galactosidase and VEGF stimulated NO release quantified. As seen in Fig. AC, infection of endothelial cells with myr-Akt enhances VEGF driven NO production while the AA-Akt attenuates NO release. These results imply that Akt participates in the signal transduction events required for both basal and stimulated NO production in endothelial cells.
Collectively these data demonstrate that Akt can phosphorylate eNOS on serine 1179 (serine 1177 in human eNOS) and that phosphorylation enhances the ability of the enzyme to generate NO.
Example 3
-35-

Materials and Methods
eNOS Constructs and Protein Purification- Wild type bovine eNOS in the plasmid pCW was expressed as described previously with groELS in E. coli BL21 cells (Martasek et al, 1996). The S1179D mutant eNOS for expression in E. coli was generated as follows. eNOS S1179D in pcDNA 3 (Fulton et al, 1999) was digested with Xhol/Xbal, subcloned into the identical sites of eNOS in pCW, and co-expressed with groELS. Isolation of recombinant eNOS was preformed as reported previously (Roman et al, 1995; Martasek et al, 1999), with the following modifications. eNOS was eluted from 2'5'-ADP Sepharose with either 10 mM NADPH or 10 mM 2'-AMP. The amount of eNOS was quantitated using the peak absorbance at 409-412 nm, with an extinction coefficient for heme content of 0.1 M-1 cm-1 . The purity of eNOS was determined by 7.5% SDS-PAGE followed by Coomassie staining. Low temperature SDS-PAGE was performed identically, except that samples were not boiled and the electrophoresis was carried out at 4 °C in a slurry of ice/water (Klatt et al, 1995). In experiments in which NOS cofactors (L-arginine, calmodulin, and NADPH) were titrated, they were omitted from the purification and storage of the enzymes and were incubated as described below.
Assay for NOS Activity-NO production was measured by the hemoglobin capture assay as described (Kelm et al, 1988). Briefly, the reaction mixture contained eNOS (0.5-2.5 g), oxyhemoglobin (8 M), L-arginine (100 - M), BH4 (5 M), CaC12 (120 M), calmodulin (120-200 nM), and NADPH (100 M) in HEPES buffer (50 mM), pH 7.4. In the determination of calcium EC50 value for eNOS, the above reaction mixture was modified as Mows: MOPS buffer (10 mM, pH 7.6), KC1 (100 mM) and CaM (250 nM) were substituted. Under these conditions, free calcium was calculated using the WinMAXC program, version
1.8 (Stanford University), with a Kd of 2.2 X 10-8 M. The precise free calcium concentration was achieved by mixing an appropriate proportion of 10 mM K2EGTA and 10 mM CaEGTA stock solutions (Molecular Probes). NOS activity was monitored for linearity over 2 min at 401 nm, and NO production was calculated based on the change of absorbance using the
-36-

extinction coefficient of 60 mM"1 cm'. All reactions were carried out at 23°C, and each data point represents 3-8 observations. The extinction coefficient of 0.0033 M-1 cm-1 at 276 nm was used for determination of calmodulin concentration. The production of NO using this method was completely blocked by the addition of nitro-L-arginine (1 mM). When the inactivation of eNOS was determined by the addition of EGTA (200-800 M) to the reaction mixture, chelator was added 1 min after initiation of the reaction by NADPH. Identical conditions were used when NADPH-cytochrome c reductase activity was examined. These reactions contained CaM (120 nM) and CaCl2 (200 M) in a 0.5-ml volume with eNOS (0.5
g).
The conversion of L-arginine to L-citrulline was assayed as described previously by Bredt et al. (1990). Briefly, eNOS (0.25-2 g) was incubated for 3-10 min at 23 °C in the following reaction mixture: 3 pmol of L-[3H]arginine (55 Ci/mmol), 10-300 M arginine, 1 mM NADPH, 120-200 nM calmodulin, 2 mM CaC12, and 30 M BH4 in a final reaction volume of 50-100 l. The reaction was terminated by the addition of 0.5 ml of 20 mM HEPES, pH 5.5, containing 2 mM EGTA and EDTA. The reaction mixture was placed over Dowex AG50WX8, and the flow-through was counted on a Packard 1500 liquid scintillation analyzer.
Assays for Reductase Activity-- NADPH-cytochrome c reductase activity and 2,6-dichlorophenolradophenol (DCIP) reduction were measured as a change in absorbance at 550 nm as described previously by Martasek et al. (1999) and Masters et al. (1967) using an extinction coefficient of 0.021 M-1 cm-1 for both cytochrome c and DCIP. Briefly, a reaction mixture (1 ml) contained either cytochrome c (90 M); DCIP (36 M), HEPES buffer (50 mM) at pH 7.6, NaCl (250 mM), NADPH (100 M), calmodulin (120 nM), and CaC12 (200 M); or other substances as indicated. The reaction was monitored for 60 s (at 23 °C) after the addition of eNOS. When inactivation of reductase activity was determined by the addition of EGTA (200-800 M), chelator was added 1 min after initiation of the reaction and monitored for an additional 1 min. The reaction contained HEPES buffer (50 M) at pH 7.6, CaM (120 nM), and CaC12 (200 M) and was initiated with NADPH (100 M). No NaCl was added in experiments that examined EGTA inactivation of eNOS to
-37-

mimic the conditions used in the hemoglobin capture experiments. The addition of NOS inhibitors did not influence the rate of cytochrome c reduction (not shown). Determination of the calcium EC50 for eNOS was performed as described above for the hemoglobin capture assay.
Data Analysis and Statistics- All data were expressed as mean ± S.E. At least triplicate determinations were performed with a minimum of three different batches of enzymes for each data set. Wild type and mutant enzymes were purified simultaneously to control for activity variations between preparations. Statistical significance was determined using Student's t test, and p RESULTS
Expression and Purification of eNOS- Both wild type and S1179D eNOS were expressed and purified from E. coli. In a culture of 1.6 liters, approximately 2.5-4.0 mg of eNOS was typically recovered using 2'5-ADP Sepharose 4B chromatography. As seen in Fig. 5A, both enzymes were >90% pure based on Coomassie staining. These results are typical, as seen from seven independent preparations of both wild type and S1179D eNOS prepared side-by-side. Both enzymes were primarily in their dimeric form, as shown by low temperature SDS-PAGE (Fig. 5B).
eNOS S1179D Has Greater NO Synthase and Reductase Activities Than Does Wild Type eNOS- Next, the activities of wild type and S1179D eNOS were compared by measuring the rate of NO production. S1179D eNOS exhibited a higher turnover number (under optimal conditions) as compared with wild type enzyme (84 ± 6 versus 27 ± 1 min-1, n = 6 separate and paired preparations of enzymes). The Km values with L-arginine were similar for wild type and S1179D eNOS (Fig. 6A, 1.8 versus 2.5 M, respectively; see Table I).
-38-

Table I Kinetic parameters for wild type and S1179D eNOS
Substrate/cofactors Assay Catalytic Wild type S1179D determination Arginine Hemoglobin Kcat 27 ± 1 min-1 84 ± 6 min-1 capture Km 1.8 M 2.5 Hemoglobin Kcat 17±l min-1 53±3min-1 capture Km 8 M 36 M Cytochrome c Kcat +CaM 460 ± 18 min-1 840 ± 59 min-1 Kcat -CaM 70 ± 5 min-1 290 ± 9 min-1 Km+CaM 0.75 M 1.9 M NADPH -CaM 0.40 M 2.0 M L-Citrulline Kcat 22 ± 2 min1 43 ± 2 min-1 EC50 8 nM 7 nM Cytochrome c Kcat 620 ± 78 min-1 1140 ± 75 min-1 CaM EC50 13nM 21 nM Hemoglobin Kcat 58 ± 1 min-1 100 ± 3 min-1 capture (100 mM EC50 310 nM 250 nM KC1) Cytochrome c Kcat 1909 ± 33 min-1 3798 ± 54 min-1 Ca2+ (100 mM EC50 290 nM 220 nM KC1)
-39-

Because the rate of electron flux from the reductase domain to the oxygenase domain is critical for NOS catalysis, the increase in S1179D eNOS activity was examined to determine whether it could be attributed to enhanced reductase activity. When both DCIP and cytochrome c reduction were examined, a significant increase in activity for S1179D compared with wild type eNOS was observed (Fig. 6B). Furthermore, this increase was accentuated by the presence of CaM, which increased the overall activity for both enzymes. Basal cytochrome c reduction, in the absence of CaM, was 4-fold higher for S1179D compared with wild type eNOS. The magnitude of CaM-stimulated cytochrome c reduction was higher for S1179D eNOS (749 ± 35 versus 1272 ± 55 min-1 for wild type and S1179D eNOS, respectively, n =3-5); however, the level of stimulation by CaM was 8-fold for wild type eNOS compared with only 3-fold for S1179D eNOS.
Next, it was determined whether S1179D eNOS produces more superoxide than wild type eNOS, which could reduce cytochrome c. As expected, no superoxide dismutase inhibitable cytochrome c reduction was observed (as an index of superoxide anion generation) in the absence of CaM, as reported earlier for wild type eNOS (86 ± 6 versus 95 ± 8 mur1) and for S1179D eNOS (278 ± 9 versus 288 ± 7 min-1, n = 3-5). However, in the presence of CaM, superoxide dismutase reduced the rate of cytochrome c reduction for both wild type (610 ±51 versus 866±8min-l)and S1179D(1179±43 versus 1518± 19min-1) eNOS. The relative decrease in cytochrome c activity in the presence of superoxide dismutase was similar with both enzymes (30% for wild type and 22% for S1179D eNOS), suggesting that the enhanced reductase activity of S1179D compared with wild type eNOS (assayed by cytochrome c reduction) was not due to uncoupling of the enzyme.
NADPH-dependent NO Formation and Reductase Activities Are Not Different for Wild Type and S1179D eNOS- The NADPH dependence of NO production and cytochrome c reduction were examined because the NADPH binding site lies close in proximity to the Ser-1179 in eNOS. S1179D eNOS had a higher maximum turnover number (kcar) than did wild type enzyme based on NO production, assayed in the presence of CaM (Fig. 7A). The increased kcat was associated with a 4-fold increase in the Km for NADPH for S1179D eNOS compared with wild type eNOS (36 versus 8 M, respectively). The kcat for
-40-

NADPH-dependent cytochrome c reduction in the absence of CaM was greater for eNOS S1179D than wild type eNOS (290 versus 70 min-1, respectively; Fig. 7B). In the presence of CaM, the kcat for cytochrome c reduction was considerably higher for S1179D compared with wild type eNOS (840 versus 460 min-1, respectively). The Km values for NADPH were unchanged in the absence or presence of CaM (0.40 and 0.75 M for wild type eNOS and 2.0 and 1.9 M for S1179D eNOS in the absence and presence of CaM, respectively).
EC50 Values for CaM Are Unchanged between Wild Type and S1179D eNOS- To assess whether the increased activity of S1179D eNOS was attributable to changes in the affinity of the enzyme for CaM, the kinetics of NOS activity and cytochrome c reduction assayed in the presence of all NOS cofactors in excess as a function of CaM concentration were examined. The kcat for CaM activation of NOS activity was 22 min-1 for wild type and 43 min1 for S1179D eNOS. Transformation of the data, normalizing for the differences in kcat, revealed a slight shift in the curve to the left for S1179D eNOS but little difference in the EC50 values for CaM, consistent with reported data on phospho-eNOS (Mitchell et al, 1999). The EC50 values were 8 nM for wild type and 7 nM S1179D eNOS (Fig. 8A). NADPH-mediated cytochrome c reduction was measured. The kcat for CaM activation of cytochrome c reduction was about 2-fold higher for S1179D eNOS compared with wild type enzyme (1140 and 620 min1 for S1179D and wild type eNOS, respectively). Transformation of the data normalized for the differences in kcat, revealed small differences in the EC50 values for CaM between wild type and S1179D eNOS (21 versus 13 nM; Fig. 8B).
Comparison of Calcium Activation and Inactivation of eNOS- In previous experiments, it was demonstrated that the "apparent calcium sensitivity" of eNOS was enhanced in cells expressing either a majority of phospho-eNOS or S1179D eNOS, suggesting that phosphorylation changed the affinity of calcium/CaM activation (Dimmeler et al, 1999; Fulton et al. 1999). As seen m Fig. 8C, after normalization for the differences in maximal activity, the calcium dependence was slightly increased for S1179D eNOS (p -41-

determined by NO production (in the presence of 250 nM CaM). As seen in the inset to Fig. 8C, increasing concentrations of free calcium did indeed enhance S1179D eNOS turnover to a greater extent then that seen with wild type enzyme. Furthermore, the EC50 value for calcium assaying cytochrome c reduction were similar to those obtained measuring NO production (Fig. 8D; 290 and 220 nM for wild type and S1179D eNOS, respectively). Again, the Vmax for calcium activation of S1179D eNOS turnover was greater than wild type (Fig. 4D, inset).
To examine whether the inactivation of S1179D eNOS was different than that of wild type enzyme, the decay in eNOS activity after chelation of calcium with EGTA was measured. In these experiments, all NOS cofactors were added in the presence of calcium (200 uM), and NO production was monitored for 1 nun, followed by the addition of different concentrations of EGTA and monitoring for an additional 1 min. As seen in Fig. 9A, the addition of EGTA to wild type and S1179D eNOS reduced NO production in a concentration-dependent manner. However, NO production from S1179D eNOS was less sensitive to the addition of EGTA; i.e. wild type eNOS activity declined more rapidly at lower concentrations of EGTA than did S1179D eNOS activity. The greatest difference in activity between the enzymes was seen at 400 M EGTA. Furthermore, at 600 M EGTA, no activity was detected for wild type eNOS, whereas residual activity was still detected for S1179D eNOS. Similar results were obtained using cytochrome c reduction (Fig. 9B), with significant differences in activity seen with 400 and 600 uM chelator added to the reaction. However, at the highest concentration of EGTA, residual reductase activity was found for both wild type and S1179D eNOS.
In summary, bovine endothelial nitric oxide synthase (eNOS) is phosphorylated directly by the protein kinase Akt at serine 1179 (Fulton et al, 1999) and human endothelial nitric oxide synthease is phosphoryiated directly by the protein kinase Akt at serine 1177 (Dimmeler et al, 1999). Mutation of residuel 179 in bovine eNOS to the negatively charged aspartate increases nitric oxide (NO) production constitutively, in the absence of agonist challenge.
-42-

It should be understood that the foregoing discussion and examples merely present a detailed description of certain preferred embodiments. It therefore should be apparent to those of ordinary skill in the art that various modifications and equivalents can be made without departing from the spirit and scope of the invention. All references, articles and patents identified above or below are herein incorporated by reference in their entirety.
References
Additional references for which the entire citation is not given in the text are herein incorporated in their entirety.
Alessi et al., Embo J 15,6541-6551 (1996).
Bredt et al, Proc. Natl Acad. Sci U. S. A. 87,682-685 (1990).
Corson et al, Circulation Research 79,984-991 (1996).
Dimmeler et al., Nature 399,601-605 (1999).
Downward, Curr Opin Cell Biol 10,262-267 (1998).
Fulton et al., Nature 399,597-601 (1999).
Garcia-Cardena et al., Nature 392,821-824 (1998).
Garcia-Cardena et al.,J Biol Chem 271,27237-27240 (1996).
Garcia-Cardefla et at, Proc. Natl Acad. Set USA 93,6448-6453 (1996a).
Huang et al.. Nature 377,239-242 (1995).
Kantor et al., Science 274,1744-1748 (1996).
Ketm et al., Biochem. Biophys. Res. Commun. 154,236-244 (1988).
Klatt et al., EMBO J. 14,3687-3695(1995).
Liu etal., Biochemistry 35,13277-13281 (1996).
Liu et al.,J. CellBiol. 137,1525-1535 (1997).
Martasek et al., Methods Enzymol. 301,70-78 (1999).
Martasek et al, Biochem Biophys Res Commun 219,359-365 (1996).
Masters et al., Methods Enzymol. 10,565-573 (1967)
Michel et al., Proc. Natl Acad. Sci. 90,6252-6256 (1993).
-43-

Michell et al, Cwrr. Biol 9,845-848 (1999).
Murohera et al., J clin Invest 101,2567-2578 (1998).
Papapetropoulos et al.,J din Invest 100,3131-3139 (1997).
Roman et al., Proa Natl. Acad. Sci U. S. A. 92,8428-8432 (1995).
Rudic et al., J Clin Invest 101,731-736 (1998).
Sessa et al, J Biol Chem 270,17641-17644 (1995).
Shaul et al, JBiol Chem 271,6518-6522 (1996).
Shesely et al., Proc Natl Acad Sci USA 93,13176-13181 (1996).
Yano et al, Nature 396,584-587 (1998).
-44-

WE CLAIM:
1. A process for isolation of a nucleic acid molecule encoding a NOS
polypeplide selected from the group consisting of a constiutively active
NOS polypeplide, a NOS polypetide having an increased rate of NO
production, and a NOS polypeptide having an increased reduclase
activity,comprising separating the nucleic acid molecule from contaminant
nucleic acids.
2. The process as claimed In claim 1, wherein the NOS polypeplide contains a
substituted amino acid residue corresponding to residue 1179 of bovine eNOS,
residue 1177 of human eNOS, residue 1412 of rat nNOS, or residue 1415 of
human nNOS.
3. The process as claimed In claim 2, wherein the substituted amino acid residue
corresponding to residue 1179 of bovine eNOS, residue 1177 of human eNOS,
residue 1412 of rat nNOS, or residue 1415 of human nNOS, mimics a
phosphoserine.
4. The process as claimed in claim 2, wherein a serine corresponding to residue
1179 of bovine eNOS. residue 1177 of human eNOS, residue 1412 of rat nNOS,
or residue 1415 of human nNOS, has been substituted with an aspartic acid or
glutamic acid residue.
5. The process as claimed In claim 2, wherein the substituted amino acid contains
an R group that minics a phosphate group.
6. A process for the preparation of an isolated NOS polypeptide selected from the
group consisting of a constitutively active NOS polypeptide, a NOS polypeptide
with increased rate of NO production, and a NOS polypeptide with increased
reductase activity, comprising the steps of selecting a NOS cDNA,
45

modifying the nucleotlde triplet encoding a serine, followed by recombinant expression of the altered NOS cDNA.
7. The process as claimed in claim 6 which contains a substituted amino acid
residue corresponding to residue 1179 of bovine eNOS. residue 1177 of human
eNOS, residue 1412 of rat nNOS, or residue 1415 of human nNOS,
8. The process as claimed in claim 7, wherein the substituted amino acid has been
substituted with an aspartlc acid or glutamic acid residue.
9. A method of expreroing a transgene coding for a polypeptide of claim 6 or an Akt
polypeptide to promote coronary collateral vessel development, comprising
expressing the transgene from a ventricular myocyte-specific promoter, smooth
muscle cell specific promoter, or endothelial cell specific promoter.
10. The method according to claim 9, wherein the ventricular myocyte-specific
promoter has the sequences of ventricular myosin light chain-2 or myosin heavy
chain promoter.
11. A method of expressing a trensgene coding for e polypeptide of claim 6 or an AM
polypeptide to promote peripheral vascular development, comprising expressing
the transgene from a ventricular myocyte-specific promoter, smooth muscle cell specific promoter, or endothelist cell specific promoter.
12 The method as claimed in claims 9-11, wherein the trensgene is encoded by a replication-defective vector
13. The method claim 9 to 12, wherein the replication-defective vector is an
adenovlrus.
14. The method of any one of claims 9-12, wherein the transgene is transfected Into
a cell In vitro.
46

15. A method of Identifying an agent which modulates the Akt regulated activity of
NOS. comprising the steps of:
a)exposing a cell which expresses NOS to an agent; and
b) measuring the Akt regulated activity of NOS, thereby identifying an agent which modulates the Akt regulated activity of NOS.
16. The method of claim 15, wherein step (b) comprises the determination of the
phosphorylation state of NOS at an amino acid residue corresponding to residue
1179 of bovine eNOS, residue 1177 of human eNOS, residue 1412 of rat nNOS,
or residue 1415 at human nNOS.
17. The method of claim 16, wherein NOS is activated by the agent.
18. The method of claim 17, wherein the agent mimics AM mediated phosphorylation
of eNOS.
19. The method of claim 18, wherein the agent inhibits the dephosphorylation of an
amino acid residue corresponding to residue 1179 of bovine eNOS, residue 1177
of human eNOS, residue 1412 of rat nNOS, or residue 1415 at human nNOS.
20. The method of claim 15 wherein step (b) comprises the measurement of NO
production.
21. The method of claim 20, wherein No is detected by measuring nitrite
concentrations or the conversion of 3H-L-arginine to 3H-L-citrulline.
22. An in vitro method of Identifying an agent which modulates Akt regulated activity
of eNOS, comprising the steps of:

(a) exposing Akt and NOS protein or peptide to the agent; and
(b) measuring the activity of NOS.
47

23. The method of claim 22. wherein NOS activity is measured by determining the
Akt dependent phosphorylation state of NOS.
24. The method of claim 23, wherein the NOS peptlde comprises SEQ ID NO:2.
25. The method of claim 23, wherein in step (b) the activity of NOS is reductase
activity.
26. The method of claim 23, wherein in step (b), the activity of NOS is production of
NO by NOS.
27. The process as claimed in claim 1 for the isolation of an isolated nucleic acid
molecule encoding a NOS polypeplide selected from the group consisting of a
bovine eNOS that contains a substituted amino acid residue corresponding, to
residue 1179, a human eNOS that contains a substituted residue corresponding
to residue 1177, a rat nNOS that contains a substituted residue corresponding to
residue 1412 and a human nNOS that contains a substituted residue
corresponding to residue 1415, wherein the substituted amino acid residue
comprises a non-negatively charged R group.
28. The process as claimed in claim 27, wherein the substituted amino acid residue
is alanine
29. The process as claimed in claim 7, for the preparation of an isolated NOS
polypeptide selected from the group consisting of s bovine eNOS that contains a
substituted amino acid residue corresponding to residue 1179, a human eNOS
that contains a substituted residue corresponding to residue 1177, a rat nNOS
that contains s substituted residue corresponding to residue 1412, and a human
nNOS that contains a substituted residue corresponding to residue 1415,
wherein the substituted amino acid residue comprises a non-negatively charged
R group.
30. The NOS polypeptide of claim 29, wherein the substituted amino acid residue is
alanine.
-48-

49
31. The method of claim 10, wherein the endothellal cell specific promoter is Tie-2
promoter, endothelln promoter, or eNOS promoter.
32. The method of claim 10, wherein the smooth muscle cell promoter Is SM22
promoter or SM alpha actin promoter.
A process for isolation of a nucleic acid molecule encoding a NOS polypeptide selected from the group consisting of a constitutively active NOS polypeptide, a NOS polypeptide, a NOS polypeptide having an increased rate of NO production, and a NOS polypepetide having an increased reductase activity, comprising separating the nucleic acid molecule from contaminant nucleic acids.

Documents:

in-pct-2001-00998-kol abstract.pdf

in-pct-2001-00998-kol assignment.pdf

in-pct-2001-00998-kol claims.pdf

in-pct-2001-00998-kol correspondence.pdf

in-pct-2001-00998-kol description(complete).pdf

in-pct-2001-00998-kol drawings.pdf

in-pct-2001-00998-kol form-1.pdf

in-pct-2001-00998-kol form-18.pdf

in-pct-2001-00998-kol form-2.pdf

in-pct-2001-00998-kol form-26.pdf

in-pct-2001-00998-kol form-3.pdf

in-pct-2001-00998-kol form-5.pdf

in-pct-2001-00998-kol letters patent.pdf

in-pct-2001-00998-kol priority document.pdf


Patent Number 202556
Indian Patent Application Number IN/PCT/2001/998/KOL
PG Journal Number 09/2007
Publication Date 02-Mar-2007
Grant Date 02-Mar-2007
Date of Filing 25-Sep-2001
Name of Patentee YALE UNIVERSITY
Applicant Address 451,COLLEGE STREET, NEW HAVEN, CT 06550,
Inventors:
# Inventor's Name Inventor's Address
1 SESSA,WILLIAM, C 4,RICHBOROUGH ROAD,MADISON, CT 06443,
PCT International Classification Number C12N15/00
PCT International Application Number PCT/US00/09913
PCT International Filing date 2000-04-14
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/129,550 1999-04-16 U.S.A.