CA2402877A1 - Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same - Google Patents

Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same Download PDF

Info

Publication number
CA2402877A1
CA2402877A1 CA002402877A CA2402877A CA2402877A1 CA 2402877 A1 CA2402877 A1 CA 2402877A1 CA 002402877 A CA002402877 A CA 002402877A CA 2402877 A CA2402877 A CA 2402877A CA 2402877 A1 CA2402877 A1 CA 2402877A1
Authority
CA
Canada
Prior art keywords
seq
ifi206
sequence
polypeptide
polynucleotide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002402877A
Other languages
French (fr)
Inventor
David A. Lewin
Sean Adams
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genentech Inc
CuraGen Corp
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority claimed from PCT/US2001/008333 external-priority patent/WO2001068830A1/en
Publication of CA2402877A1 publication Critical patent/CA2402877A1/en
Abandoned legal-status Critical Current

Links

Abstract

The present invention provides novel mouse interferon-inducible proteins (IFI206 and naturally occuring variants) modulated under conditions that affect metabolic status and the polynucleotides that identify and encode IFI206. The invention provides for genetically engineered expression vectors and host cells comprising the nucleic acid sequence encoding IFI206 and for a method for producing the protein. The invention also provides pharmaceutical compositions containing IFI206 and the use of such compositions for the prevention or treatment of diseases associated with the expression of IFI206.
Additionally, the invention provides antisense molecules to IFI206 and their use in the treatment of diseases associated with the expression of IFI206. The invention also provides diagnostic assays that utilize polynucleotides that hybridize with naturally occurring sequences encoding IFI206 and antibodies that specifically bind to the protein.

Description

IF1206, A NOVEL INTERFERON-INDUCED POLYPEPTIDE, AND
NUCLEIC ACIDS ENCODING THE SAME
RELATED APPLICATIONS
This application claims priority to U.S. provisional application Serial No.
60/188,716 filed 03/13/2000.
BACKGROUND
Obesity and Metabolic Disorders Obesity is the most prevalent metabolic disorder in the United States affecting on the order of 35% of adults at an estimated cost of 300,000 lives and $70 billion in direct and indirect costs. As an epidemic, it is growing due to the increase in the number of children who can be considered overweight or obese. Obesity is defined as an excess of body fat, frequently resulting in a significant impairment of health. Obesity results when adipocyte size or number in a person's body increases to levels that may result in one or more of a number of physical and psychological disorders. A normal-sized person has between 30 and 35 billion fat cells. When a person gains weight, these fat cells increase in size at first and later in number. Obesity is influenced by genetic, metabolic, biochemical, psychological, and behavioral factors. As such, obesity is a complex disorder that must be addressed on several fronts to achieve a lasting positive clinical outcome (ADAReport, 1997; Perusse and Bouchard, 1999; Pi-Sunjer and Panel, 1998).
Obese individuals are prone to ailments including: type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea. Sleep apnea is episodes of not breathing during sleep that correlates with higher incidence of stroke and heart attack, two other factors contributing to obesity-linked morbidity and mortality among the clinically obese (ADAReport, 1997; Pi-Sunjer and Panel, 1998).
There are several well-established treatment modes ranging from non-pharmaceutical to pharmaceutical clinical intervention. Non-pharmaceutical intervention includes diet, exercise, psychiatric treatment, and surgical treatments to reduce food consumption or remove fat (i.e: liposuction).
Appetite suppressants and energy expenditure/nutrient-modifying agents represent the focus of pharmacological intervention. Dexfenfluramine (REDUXX~') and sibutramine (MERIDIA~) are members of the first class and beta3-adrenergic agonists and orlistat (XENICAL~) are representative of the latter (Dunlop and Rosenzweig-Lipson, 1998).
Animal models have provided strong evidence that genetic make-up is influential in the determining the nature and extent of obesity. 40-80% of variation in body mass index (BMI, a measure of obesity correlating weight and height) can be attributed to genetic factors (Bouchard, 1995; Pi-Sunjer and Panel, 1998). While human obesity does not generally follow a Mendelian inheritance pattern (Weigle and Kuijper, 1996), there are several rodent models that do (Spiegelman and Flier, 1996; Weigle and Kuijper, 1996). As human obesity is a complex trait, it is not surprising that single mutations in rodents might not explain the etiology of obesity in all humans although there are examples of humans with genetic lesions analogous to those found in rodents (Clement et a1.,_1998; Montague et al., 1997).
Interestingly, there are animal models for complex phenotypes, such as hypertension and stroke, which are obese, too. This suggests that these animals may represent a more telling model for understanding the complexities of human obesity (Pomp, 1997; Van Zwieten et al., 1996; Wexler et al., 1980).
There are several rodent models of obesity that result in the inheritance of a single genetic lesion. Monogenetic obesity syndromes in mice that are well characterized but rarely, if ever, observed in humans include: obese (ob), aberrant termination of the translation of the satiety factor leptin.
Mutations of the leptin receptor result in the obese diabetic mouse (db) phenotype. Agouti (A'') is a coat color mutant that is obese. Normally only expressed in the skin, in the mutant animals it is ubiquitously expressed and may antagonize the binding of melanocyte stimulating hormone (MSH). MSH is derived from adrenocorticotropic hormone (ACTH) a major pituitary hormone that results from the proteolytic processing of the pro-hormone proopiomelanocortin (POMC). The fat phenotype is the consequence of a mutation in the hypothalamic pro-hormone converting enzyme carboxypeptidase E. The least well-characterized obese mouse mutant is tub. tub encodes a cytosolic protein that may influence the processing of hypothalamic neuropeptide hormones such as neuropeptide Y (NPY, an appetite stimulating hormone) and POMC (Aron et al., 1997; Guan et al., 1998; Spiegelman and Flier, 1996;
Weigle and Kuijper, 1996). Recently, a POMC knockout mouse was reported that has a phenotype analogous to several mouse models for obesity, particularly that of Ay. The POMC knockout has early onset obesity and has yellow hair color as well as adrenal insufficiency due to the apparent morphological absence of their adrenal gland. As there is no detectable corticosterone in these animals and corticosterones increase food intake, it is surprising that they are obese. The obese phenotype can be treated with a-MSH, a peptide hormone derived from POMC (Yaswen et al., 1999).
Other animal models include fa/fa (fatty) rats, which bear many similarities to the ob/ob and db/db mice, discussed above. One difference is that, while fa/fa rats are very sensitive to cold, their capacity for non-shivering thermogenesis is normal. It is well established that thermogenesis and metabolism are closely coupled endocrinologically. Torpor, a condition analogous to hibernation and lethargy, seems to play a larger part in the maintenance of obesity in fa/fa rats than in the mice mutants. Further, several desert rodents, such as the spiny mouse, do not become obese in their natural habitats, but do become so when fed on standard laboratory feed (Tartaglia, 5,861,485, 1999).
Adipose Tissues Brown Adipose Tissue (BAT), also known as multilocular adipose tissue, is so called because of the its color due to the large number of capillaries and mitochondria in the cells making up this tissue. BAT is primarily found in the shoulder region and flanks of human embryo and newborn infant, it then disappears in the first months of life. In animals, particularly hibernating animals and rodents, it is more abundant. BAT has features of an endocrine organ; it is vascularized by capillaries and it receives direct sympathetic innervation. Sympathetic neurotransmission leads to the release of the catecholamines noradrenaline and adrenaline resulting in the activation of a hormone-sensitive lipase. This results in the hydrolysis of triglycerides that are converted to fatty acids and glycerol leading to an increase in oxygen consumption and heat production by uncoupling of the mitochondria) proton gradient from the formation of ATP via the activity of uncoupling proteins (UCPs; (Gura, 1998). BAT stimulation by catecholamines results in non-shivering thermogenesis (Junqueira et al., 0-8385-0590-2, 1998; Palou et al., 1998; Schrauwen et al., 1999).
Evidence of BAT as an endocrine organ comes from the work of Himms-Hagen done in the late 1960's. The conclusion that BAT is an endocrine organ comes from the observations that age and temperature acclimation affect the degree to which glucose carbon is incorporated into the lipids of BAT, an indication of metabolic activity under non-shivering thermogenic conditions (Himms-Hagen, 1969a). In addition, experiments involving the removal of BAT from rats acclimated to different temperatures and the effects upon enhanced calorigenic response to catecholamines lead to the following observations (1) removal of interscapular BAT (IBAT) from cold-acclimated rats has no immediate effect on the calorigenic response of rats to catecholamines. The significance being that BAT is not the organ directly responsive to this stimulus. (2) With time (days), there is a progressive loss of the enhanced catecholamines response by rats that have had IBAT removed, suggesting that BAT is responsible for the long-term maintenance of the catecholamines-induced thermogenic response.
Interestingly, the ability of IBAT to maintain the enhanced response correlated with the duration of exposure to cold. This suggests that BAT has short term and long-term effects on acclimation. With long-term cold acclimation there may be a proliferation of BAT into regions, other than that occupied by IBAT, thus maintaining the catecholamines response (Himms-Hagen, 1969b). Other work showing that transplantation of IBAT from cold-acclimated animals into those raised in the warm can confer a thermogenic response under condition that normally would not support the endocrine nature of BAT.
The role of this endocrine organ in the maintenance of body weight as well as thermogenesis was demonstrated by the ablation of BAT in a transgenic mouse model using a BAT-specific promoter (UCP1) controlling the expression of diphtheria toxin during the development of this tissue.
Animals were found to be unable to maintain core body temperature when exposed to the cold and obesity develops in the absence of hyperphagia. The significance of this latter observation is that in the absence of BAT the mice have increased metabolic efficiency. That is to say, in the absence of BAT
and UCP, there is a net accumulation of energy stored in the form of fat.
Finally, in the case of one strain of mice with only a transient ablation of BAT, the metabolic defect is ameliorated with the reemergence of BAT (Friedman, 1993; Lowell et al., 1993). These data taken together support the contention that BAT is an endocrine organ with an indirect but pivotal role in the metabolic status of organisms in which it is observed.
Endocrine organs regulate metabolism and in doing so, perforce, must regulate gene expression. Only a small set of genes have been shown to involved in metabolism related to brown adipose tissue as an endocrine tissue (Charon et al., 1995; Collins et al., 1999; Denjean et al., 1999; Foellmi-Adams et al., 1996; Savontaus et al., 1998). Regardless of the mechanism of BAT-mediated non-shivering thermogenesis, genes modulated in response to mouse husbandry below the thermal neutral zone of these animals represent important markers of metabolic response, or lack thereof, potential drug targets for metabolic disorders, and/or in the case of secreted/integral membrane proteins drugs themselves.
Interferons Interferons (IFNs) are a part of the group of intercellular messenger proteins known as cytokines. IFNa is the product of a multigene family of at least 16 members, whereas IFN(i is the product of a single gene. a- and (3-IFNs are also known as type I IFNs. Type I IFNs are produced in a variety of cell types. Biosynthesis of type I IFNs is stimulated by viruses and other pathogens and by various cytokines and growth factors. IFNy, also known as type II IFN, is produced in T-cells and natural killer cells. Biosynthesis of type II IFN is stimulated by antigens to which the organism has been sensitized.
Both a- and 8-IFNs are immunomodulators and anti-inflammatory agents, activating macrophages, T-cells and natural killer cells.
IFNs are part of the body's natural defense to viruses and tumors.
They exert these defenses by affecting the function of the immune system and by direct action on pathogens and tumor cells. IFNs mediate these multiple effects in part by inducing the synthesis of many cellular proteins.
Some interferon-inducible (1F1) genes are induced equally well by a-, (i-, and y-IFNs. Other IFI genes are preferentially induced by the type I or by the type.
II IFNs. _ The various proteins produced by IFI genes possess antitumor, 15. antiviral and immunomodulatory functions. The expression of tumor antigens by cancer cells is increased in the presence of IFNa, thus rendering the cancer cells more susceptible to immune rejection. The IFI proteins synthesized in response to viral infections are known to inhibit viral functions , such as cell penetration, uncoating, RNA and protein synthesis, assembly and release (Hardman et al., 1996). Type II IFN stimulates expression of major histocompatibility complex (MHC) proteins. For this reason it is thus used in immune response enhancement (De Maeyer and De Maeyer-Guignard, 1998;
Janeway and Travers, 1997).
Interferons may be grouped into three categories. IFNa (leukocyte) interferon is made by white blood cells; IFN(i (fibroblast) interteron is made by skin cells; and IFNy (immune) interferon is made by lymphocytes after stimulation by antigen. Host response to infection includes changes in metabolic state, for example the regulation of hepatic fatty acid biosynthesis.
In response to IFNa fatty acid biosynthesis is stimulated, but the mechanism appears to be different from that of other cytokines such as interleukin-1 (IL-1) and tumor necrosis factor (TNF) since only treatment with the former in conjunction with either one of the two later cytokines can stimulate lipogenesis. IL-1 and TNF cannot act synergistically with each other, but can do so with IFNa (Grunfeld and Feingold, 1992). However, there is an older observation that TNF can affect the thermogenic activity of BAT, the core temperature, the rate of food intake and body weight, and resting oxygen consumption of rats. In this work there seems to be a less robust response to IFNy (Coombes et al., 1987).
In addition to changes in fatty acid metabolism and biosynthesis that might be induced by treatment with IFNs and/or other cytokines, it has been observed that treatments can induce the expression of inducible nitric oxide synthase (iNOS) when mice or cell lines (NIH 3T3L1) are treated with multiple cytokines such as IFNy, TNFa, and bacterial endotoxin lipopolysaccharide (LPS) together. Alone, these agents do not induce the expression of iNOS
(Kaput et al., 1999).
Interestingly, IFNa and IFN~ have been shown to affect the composition of BAT in suckling mice. Morphological changes included reduction in the number and size of mitochondria as well as the inclusions in the cristae. In addition, there was a change in the total amounts of lipids in the BAT and a reduction in the thickness of white adipose tissue in treated animals. The changes described were analagous to those observed in older animals (Sbarbati et al., 1995). The response of BAT in rats treated with TNF, and a much-reduced response to INFy treatment, was limited to juveniles;
adult rats were less responsive to treatment .(Coombes et al., 1987).
These observations suggest that IFNs or more specifically, interferon-modulated genes may play a role in the composition and distribution of BAT
and WAT. If so, then these IFNresponsive genes may represent targets or agents of therapeutic intervention in metabolic disease, if not excellent markers for the assessment of such compounds.
IFNinduced genes in mice cluster on chromosome 1 in the region 95.2 cM from the centromere in close proximity to the erythroid a-spectrin locus and the serum amyloid P-component locus. This region corresponds to _$-human 1 q21-23. The genes that form the interferon-inducible gene cluster contain canonical seven acid repeat regions as well as conserved non-coding regions in the promotor regions. These genes appear to have evolved because of gene duplication then subsequently diverged. There are many known interferon-inducible genes, the founding members of the mouse 200 series genes are 201, 202abc, 203, 204, and 205/D3. The p202 and p204 gene has been localized to the cytoplasm and nucleus of cells. Constitutive over-expression of p202 in transfected cells inhibits cell growth. p202 binds the cell growth regulatory retinoblastoma protein (pRb) in vitro and in vivo.
The 202 protein is a 52kD phosphoprotein that can bind to the pRb as well as a number of other transcription factors such as c-Jun, c-Fos, NFKB, and AP-1 (Min et al., 1996). The 72kD gene product of the 204 gene is also a phosphoprotein (Choubey and Lengyel, 1992; Choubey and Lengyel, 1993;
Choubey et al., 1989; Tannenbaum et al., 1993; Wang et al., 1999).
A human IFI gene known as 6-16 encodes an mRNA that is highly induced by type I IFNs in a variety of human cells (Kelly et al., 1986). After induction, 6-16 mRNA constitutes as much as 0.1 % of the total cellular mRNA. The 6-16 mRNA is present at only very low levels in the absence of type I IFN, and is only weakly induced by type II IFN.
The 6-16 mRNA encodes a hydrophobic protein of 130 amino acids.
The first 20 to 23 amino acids comprise a putative signal peptide. Protein 6-16 has at least two predicted transmembrane regions culminating in a negatively charged C-terminus.
The p27 gene encodes a protein with 41 % amino acid sequence identity to the 6-16 protein. The p27 gene is expressed in some breast tumor cell lines and in a gastric cancer cell line. In other breast tumor cell lines, in the HeLa cervical cancer cell line, and in fetal lung fibroblasts, p27 expression occurs only upon a-IFN induction. In one breast tumor cell line, p27 is independently induced by estradiol and by IFN (Rasmussen et al., 1993).
Expression of p27 was analyzed in 21 primary invasive breast carcinomas, 1 breast cancer bone metastasis, and 3 breast fibroadenomas.
High levels of p27 were found in about one-half of the primary carcinomas _g_ and in the bone metastasis, but not in the fibroadenomas. These observations suggest that certain breast tumors may produce high levels of, or have increased sensitivity to, type I IFN as compared to other breast tumors (Rasmussen et al., 1993). In addition, the p27 gene is expressed at significant levels in normal tissues including colon, stomach and lung, but not expressed in placenta, kidney, liver or skin (Rasmussen et al., 1993).
The small IFI gene products may contribute to viral resistance. A
hepatitis-C virus (HCV)-induced gene, 130-51, was isolated from a cDNA
library prepared from chimpanzee liver during the acute phase of the infection.
The protein product of this gene has 97% identity to the human 6-16 protein (Kato et al., 1992). The investigators suggest that HCV infection actively induces IFN expression, which in turn induces expression of IFI genes including 130-51. IFI genes may be important in viral infections, such as in hepatitis, including hepatoxicity induced by inflammation.
The IFI proteins synthesized in response to viral infections are known to inhibit viral functions such as penetration, uncoating, RNA or protein synthesis, assembly or release. The 130-51 protein may inhibit one or more of these functions in HCV. A particular virus may be inhibited in multiple functions by IFI proteins. In addition, the principle inhibitory effect exerted by IFI proteins differs among the virus families (Hardman et al., 1996).
The IFI proteins of the invention may provide the basis for clinical diagnosis of diseases associated with their induction. These proteins may be useful in the diagnosis and treatment of tumors, viral infections, inflammation, or conditions associated with impaired immunity. Furthermore, these proteins may be used for investigations of the control of gene expression by IFNs and other cytokines in normal and diseased cells.
In murine models of inflammatory bowel disease, systemic administration of interleukin (IL)-12 and IL-18 to wild-type BALB/c mice induces liver injury and intestinal inflammation. The nature of the injury and the induced hepatotoxcicity includes prominent intestinal mucosal inflammation and fatty liver, leading to piloerection, bloody diarrhoea, and weight loss. IL-12 and IL-18 induce striking elevations in serum levels of IFNy that would be expected to result in the expression of interferon-induced genes. The major symptoms of IL-12- and IL-18-induced toxicity are similar to those found in endotoxin-induced septic shock. TNF-a knockout mice induce intestinal mucosal inflammation. Furthermore, they have diffuse and dense infiltration of small fat droplets in their hepatocytes associated with an increase in serum levels of liver enzymes representing the fatty liver (steatosis). Fatty liver is dependent upon IFNy that may induce the expression of interferon-induced genes in the liver and other tissues, thereby affecting the metabolism of fatty acids (Chikano et al., 2000; Nakamura et al., 2000).
Although obesity-related fatty livers are vulnerable to damage from endotoxin, the involved mechanisms remain obscure. (Guebre-Xabier et al., 2000) determined if immunologic priming might be involved in this process by determining if fatty livers resemble normal livers that have been sensitized to endotoxin damage by Propionibacterium acnes infection. The latter induces interleukin (IL)-12 and -18, causing a selective reduction of CD4+NK T cells, diminished IL-4 production, deficient production of T-helper type 2 (Th-2) cytokines (e.g., IL-10), and excessive production of Th-1 cytokines (e.g., interteron-y). Liver and spleen lymphocyte populations and hepatic cytokine production were compared in genetically obese, ob/ob mice (a model for obesity-related fatty liver) and lean mice. Obese mice have a selective reduction of hepatic CD4+NK T cells. Serum IL-18 is also increased basally, and the hepatic mRNA levels of IL-18 and -12 are greater after endotoxin challenge. Thus, up-regulation of IL-18 and IL-12 in fatty livers may reduce hepatic CD4+NK T cells. In addition, mononuclear cells from fatty livers have decreased expression of the adhesion molecule, leukocyte factor antigen-1 (LFA-1), which is necessary for the hepatic accumulation of CD4+NK T cells.
Consistent with reduced numbers of hepatic CD4+NK T cells, mononuclear cells from fatty livers produce less IL-4. Furthermore, after endotoxin treatment, hepatic induction of IL-10 is inhibited, while that of IFNy is enhanced. Thus, fatty livers have inherent immunologic alterations that may predispose them to damage from endotoxin and other insults that induce a proinflammatory cytokine response.

The role of the IFN-inducible p204 as growth regulator has been investigated by transfecting an expression vector constitutively expressing p204 into several cell lines. Like pRB and p107, p204 is a potent growth inhibitor in sensitive cells, as demonstrated by cell focus assays. Since stable transfectants of sensitive lines constitutively overexpressing p204 cannot be established in vitro, investigators have used an inducible promoter to express p204. It has been shown that proliferation of B6MEF fibroblasts lacking endogenous p204 is strongly inhibited by transient p204 expression in the nucleus. p204 delays G1 progression into the S-phase and cells accumulate with a DNA content equivalent to cells arrested in late G1. The role of p204 in the control of cell growth in vivo has been investigated by generating _ transgenic mice in which the IFI204 gene was constitutively expressed in all tissues. The over expression of the p204 transgene is compatible with embryo development up to the four-cell stage in an in vitro follow-up of 4.5 days. However, no viable animals with an intact copy of the transgene were obtained, suggesting that high and constitutive levels of p204 expression can impair normal embryo development. These findings indicate that p204 plays a negative role in growth regulation and provide new information about the molecular mechanisms exploited by IFNs to inhibit cell proliferation (Lembo et al., 1998). Mutations affecting the expression of interferon-induced proteins may play a role in controlling cellular proliferation as observed in cancer as well as in cellular differentiation. For example, the human interferon induced protein IF116 has been found to play a role in hematopoiesis. 1F116 is expressed in CD34+ and monocytoid daughter cells, but is rapidly and markedly down-regulated at the corresponding stages of polymorphonuclear anderythroid development. This differential expression of IFI 16 in myeloid precursor subpopulations and its perceived molecular properties are consistent with a possible role in regulating myelopoiesis (Dawson et al., 1998; Landolfo et al., 1998).
Cancer Cachexia Cachexia is a wasting phenomenon observed in almost half of cancer patients. Cachexia is a result of tumor-induced distant metabolic changes disproportionate to tumor burden. Weight loss by cancer patients is most prevalent in those with pancreatic and gastric cancers, but is not limited to these cancers. Cachexia-induced weight loss may lead to respiratory distress, a major contributing factor to mortality among cancer patients as metabolic changes lead to loss of adipose tissue and skeletal muscle mass, particularly as respiratory muscle is affected. Knowledge about the mechanisms of cachexia may lead to better therapeutic and clinical interventions that complement chemotherapy (DeWys et al., 1980; Tisdale, 1999).
INFy prevents cancer cachexia in a mouse model, perhaps by the down regulation of the enzyme lipoprotein lipase and/or the up regulation of triglyceride lipase. In such a case, the IFNy mediated modulation of these genes andlor other indirect regulation of their activity would require the activity of signal transduction and/or transcription factors (Mori et al., 1996x;
Tisdale, 1999). Interleukin-12's (IL-12) activity in preventing cachexia in a murine model is at least in part due to the ability of IL-12 to down regulate the expression of IL-6 and INF-y (Mori et al., 1996b).
Understanding the mechanisms involved in INF-induced gene expression increases the usefulness of animal models for cachexia.
Understanding of such models is instrumental in the development of effective therapy. Interferon-induced genes act as markers for INF activities, for example in the case of genes that are modulated in response to thermogenic conditions that are known to affect metabolic status. Genes modulated under these conditions, as well as with IFN treatment, make it possible to dissect the roles of multiple proteins in complex pathways that are specific for adipose tissues (WAT and BAT) and skeletal muscle by monitoring the modulation of .
INF-affected genes.
SUMMARY

The invention is based in part upon the discovery of novel nucleic acid sequences encoding novel polypeptides. Nucleic acids encoding the polypeptides disclosed in the invention, and derivatives and fragments thereof, will hereinafter be collectively designated as "1F1206" nucleic acid or polypeptide sequences.
In one aspect, the invention provides an isolated IFI206 nucleic acid molecule encoding an IF1206 polypeptide that includes a nucleic acid sequence that has identity to the nucleic acids disclosed in SEQ ID NOS:1 or 3. In some embodiments, the IFI206 nucleic acid molecule can hybridize under stringent conditions to a nucleic acid sequence complementary to a nucleic acid molecule that includes a protein-coding sequence of an IFI206 sequence. The invention also includes an isolated nucleic acid that encodes an IF1206 polypeptide, or a fragment, homolog, analog or derivative thereof.
For example, the nucleic acid can encode a polypeptide at least 80% identical to a polypeptide comprising the amino acid sequences of SEQ ID NOS:2, 4 or 15. The nucleic acid can be, for example, a genomic DNA fragment or a cDNA molecule that includes the nucleic acid sequence of any of SEQ ID
NOS: 2, 4 or 15.
Also included in the invention is an oligonucleotide, e.g., an oligonucleotide which includes at least 6 contiguous nucleotides of an IFI206 nucleic acid (e.g., SEQ ID NOS:1 or 3) or a complement of said oligonucleotide.
Also included in the invention are substantially purified IF1206 (SEQ ID
N0:2, 4 or 15). In some embodiments, the IF1206 include an amino acid sequence that is substantially identical to the amino acid sequence of a human IF1206.
The invention also features antibodies that immunoselectively-bind IF1206.
In another aspect, the invention includes pharmaceutical compositions that include therapeutically- or prophylactically-effective amounts of a therapeutic and a pharmaceutically-acceptable carrier. The therapeutic can be, e.g., an IF1206, an IF1206, or an antibody specific for an IF1206. In a further aspect, the invention includes a kit containing, in one or more containers, a therapeutically- or prophylactically-effective amount of this pharmaceutical composition.
In a further aspect, the invention includes a method of producing a polypeptide by culturing a cell that includes an IFI206, under conditions allowing for expression of the IF1206 encoded by the DNA. If desired, the IF1206 can then be recovered.
In another aspect, the invention includes a method of detecting the presence of an IF1206 in a sample. In the method, a sample is contacted with a compound that selectively binds to the polypeptide under conditions allowing for formation of a complex between the polypeptide and the compound. The complex is detected, if present, thereby identifying the IF1206 within the sample.
The invention also includes methods to identify specific cell or tissue types based on their expression of an IF1206.
Also included in the invention is a method of detecting the presence of an IF1206 molecule in a sample by contacting the sample with an IF1206 probe or primer, and detecting whether the nucleic acid probe or primer bound to an IFI206 molecule in the sample.
In a further aspect, the invention provides a method for modulating the activity of an IF1206 by contacting a cell sample that includes the IF1206 with a compound that binds to the IF1206 in an amount sufficient to modulate the activity of said polypeptide. The compound can be, e.g., a small molecule, such as a nucleic acid, peptide, polypeptide, peptidomimetic, carbohydrate, lipid or other organic (carbon containing) or inorganic molecule, as further described herein.
Also within the scope of the invention is the use of therapeutics in the manufacture of a medicament for treating or preventing disorders or syndromes related to obesity, including, e.g., type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea, as well as those directly related to interferons, such as metabolic disorders, tumors, viral infections, inflammation, cancer (including renal, bladder and ovarian carcinomas, leukemias, and Kaposi's sarcoma), cancer cachexia, infections by viruses or other pathogens (such as HCV and leishmania), and conditions associated with inflammation or immune impairment such as rheumatoid and osteoarthritis and Acquired Immunodeficiency Syndrome (AIDS). The Therapeutic can be, e.g., an IFI206, an IF1206, or an IF1206 -specific antibody, or biologically-active derivatives or fragments thereof.
The invention further includes a method for screening for a modulator of disorders or syndromes including, e.g.,. type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea, as well as those directly related to interferons, such as metabolic disorders, tumors, viral infections, inflammation, cancer, cancer cachexia, infections by viruses or other pathogens, and conditions associated with inflammation or immune impairment such as rheumatoid and osteoarthritis and Acquired ' Immunodeficiency Syndrome (AIDS). The method includes contacting a test compound with an IF1206 and determining if the test compound binds to the IF1206. Binding of the test compound to the IF1206 indicates the test compound is a modulator of activity, or of latency or predisposition to the aforementioned disorders or syndromes.
Also within the scope of the invention is a method for screening for a modulator of activity, or of latency or predisposition to disorders or syndromes including, e.g., type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea, as well as those directly related to interferons, such as metabolic disorders, tumors, viral infections, inflammation, cancer, cancer cachexia, infections by, viruses or other pathogens, and conditions associated with inflammation or immune impairment such as rheumatoid and osteoarthritis and Acquired Immunodeficiency Syndrome (AIDS), by administering a test compound to a test animal at increased risk for the aforementioned disorders or syndromes.

The test animal expresses a recombinant polypeptide encoded by an IFI206.
Expression or activity of IF1206 is then measured in the test animal, as is expression or activity of the protein in a control animal which recombinantly-expresses I F1206 and is not at increased risk for the disorder or syndrome.
Next, the expression of IF1206 in both the test animal and the control animal is compared. A change in the activity of IF1206 in the test animal relative to the control animal indicates the test compound is a modulator of latency of the disorder or syndrome.
In yet another aspect, the invention includes a method for determining the presence of or predisposition to a disease associated with altered levels of an IF1206, an IF1206, or both, in a subject (e.g., a human subject). The method includes measuring the amount of the IF1206 in a test sample from the subject and comparing the amount of the polypeptide in the test sample to the amount of the IF1206 present in a control sample. An alteration in the level of the IF1206 in the test sample as compared to the control sample indicates the presence of or predisposition to a disease in the subject.
Preferably, the predisposition includes, e.g., Type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea, as well as those directly related to interferons, such as metabolic disorders. Also, the expression levels of the new polypeptides of the invention can be used in a method to screen for various cancers.
In a further aspect, the invention includes a method of treating or preventing a pathological condition associated with a disorder in a mammal by administering to the subject an IF1206, an IFI206, or an IF1206 -specific antibody to a subject (e.g., a human subject), in an amount sufficient to alleviate or prevent the pathological condition. In preferred embodiments, the disorder, includes, e.g., Type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea, as well as those directly related to interferons, such as metabolic disorders.

In yet another aspect, the invention can be used in a method to identity the cellular components that interact with the IFI206 and polypeptides, including cellular receptors and downstream effectors, by any one of a number of techniques commonly employed in the art. These include but are not limited to the finro-hybrid system, affinity purification, co-precipitation with Abs or other specific-interacting molecules.
Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG 1 shows Global Sequence Similarity (GCG:GAP) (A), Multiple Alignment Analysis (BeStFlt (Genetics computer Group_(GCG), 1999)) demonstrating the relationship between SEQ_ID_NO 2 and SEQ_ID_NO 4 (B) and PHYLIP Protein Distance Analysis. Neighbor-Joining/UPGMA
method version 3.572c, tree is unrooted and negative branch lenghts are allowed. (C) Partial ClustalW analysis (Thompson, et al., Nucleic Acids Research, 22(22):4673-4680) of SEQ_ID_NO 2 and SEQ_ID_NO_3 and proteins encoded by of mouse and human genes encoding polypeptides of interferon-induced genes (AIM2, GENBANK-ID:AF024714; GENBANK-ID:HUMIF116~acc:M63838; IF116B, GENBANK-ID:AF208043; IF1202, GENBANK-ID:MUSINA202~acc:M31418; IF1202B, GENBANK-ID:AF140672;
IF1204, GENBANK-ID:MUSINA204~acc:M31419; IF1205D3, GENBANK-ID:MUSLPSINDA~acc:M74123; IF13, GENBANK-ID:AF022371;MNDA, GENBANK-ID:HUMMCNDA~acc:M81750). (B) and (C) demonstrate the relationship between SEQ_ID_NO 2 and SEQ_ID_NO 4 and protein sequences to known interferon-induced genes.
FIG 2 shows hydrophobicity plots ((GCG), 1999) for IF1206 (A; SEQ ID
N0:1) and its naturally occurring variant (B; SEQ ID N0:3); the X axis reflects amino acid position, and the positive Y axis, hydrophobicity.
FIG 3 shows the radiation hybrid map of IF1206 (SEQ ID N0:2/SEQ ID
N0:4) as generated using Auto-RHMAPPER (Stein et al., 1995).

FIG 4 shows DOTPLOT and COMPARE analysis of polypeptide sequence from IF1206 variants.
FIG 5 shows that IF1206 variants are primarily expressed in WAT, BAT, skeletal muscle, and to a much lesser extent in cardiac muscle.
FIG 6 shows the modulation of expression of the IFI206's expression during the development of NIH3T3LI cells in culture. Expression of IF1206 family members is variable during maturation of mouse NIH3T3L1 pre-ad ipocytes.

The inventors have identified a gene and polypeptide that is expressed in response to interferon stimulation, IF1206.
Definitions Unless defined otherwise, all technical and scientific terms have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. The definitions below are presented for clarity.
The recommendations of (Demerec et al., 1966) where these are relevant to genetics are adapted herein. To distinguish between genes (and related nucleic acids) and the proteins that they encode, the abbreviations for genes are indicated by italicized (or underlined) text while abbreviations for the proteins start with a capital letter and are not italicized. Thus, IFI206 or IF1206 refers to the nucleotide sequence that encodes IF1206.
"Isolated," when referred to a molecule, refers to a molecule that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that interfere with diagnostic or therapeutic use.
"Container" is used broadly to mean any receptacle for holding material or reagent. Containers may be fabricated of glass, plastic, ceramic, metal, or any other material that can hold reagents. Acceptable materials will not react adversely with the contents.

1. Nucleic acid-related definitions (a) control sequences Control sequence are DNA sequences that enable the expression of an operably-linked coding sequence in a particular host organism. Prokaryotic control sequences include promoters, operator sequences, and ribosome , binding sites. Eukaryotic cells utilize promoters, polyadenylation signals, and enhancers.
(b) operably-linked Nucleic acid is operably-linked when it is placed into a functional relationship with another nucleic acid sequence. For example, a promoter or enhancer is operably-linked to a coding sequence if it affects the transcription of the sequence, or a ribosome-binding site is operably-linked to a coding sequence if positioned to facilitate translation. Generally, "operably-linked"
means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by conventional recombinant DNA methods.
(c) isolated nucleic acids An isolated nucleic acid molecule is purified from the setting in which it.
is found in nature and is separated from at least one contaminant nucleic acid molecule. Isolated IFI206 molecules are distinguished from the specific IF1206 molecule, as it exists in cells. However, an isolated IFI206 molecule includes IFI206 molecules contained in cells that ordinarily express the where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells.
2. Protein-related definitions (a) purified polypeptide When the molecule is a purified polypeptide, the polypeptide will be purified (1 ) to obtain at least 15 residues of N-terminal or internal amino acid sequence using a sequenator, or (2) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or silver stain.
Isolated polypeptides include those expressed heterologously in genetically-engineered cells or expressed in vitro, since at least one component of the IF1206 natural environment will not be present. Ordinarily, isolated polypeptides are prepared by at least one purification step.
(b) active polypeptide An active IF1206 or IF1206 fragment retains a biological and/or an immunological activity of native or naturally-occurring IF1206. Immunological activity refers to the ability to induce the production of an antibody against an antigenic epitope possessed by a native IF1206; biological activity refers to a function, either inhibitory or stimulatory, caused by a native IF1206 that excludes immunological activity. A biological activity of IF1206 includes, for example, binding of nucleic acids, such as binding mRNA expressed in BAT.
(c) Abs Antibody may be single anti-IF1206 monoclonal Abs (including agonist, antagonist, and neutralizing Abs), anti-IF1206 antibody compositions with polyepitopic specificity, single chain anti-IF1206 Abs, and fragments of anti-IF1206 Abs. A "monoclonal antibody" refers to an antibody obtained from a population of substantially homogeneous Abs, i.e., the individual Abs comprising the population are identical except for naturally-occurring mutations that may be present in minor amounts (d) epitope tags An epitope tagged polypeptide refers to a chimeric polypeptide fused to a "tag polypeptide". Such tags provide epitopes against which Abs can be made or are available, but do not interfere with polypeptide activity. To reduce anti-tag antibody reactivity with endogenous epitopes, the tag polypeptide is preferably unique. Suitable tag polypeptides generally have at least six amino acid residues and usually between about 8 and 50 amino acid residues, preferably between 8 and 20.amino acid residues). Examples of epitope tag sequences include HA from Influenza A virus and FLAG.

The invention is based, in part, upon the discovery of novel nucleic acid sequences that encode novel polypeptides, particularly interferon-inducible proteins. The nucleic acids, and their encoded polypeptides, are collectively designated herein as "1F1206".
The novel IFI206 of the invention include the nucleic acids whose sequences are provided in Tables 1 and 3, or a fragment thereof. The invention also includes a mutant or variant IFI206, any of whose bases may be changed from the corresponding base shown in Tables 1 and 3 while still encoding a protein that maintains the activities and physiological functions of the IF1206 fragment, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including complementary nucleic acid fragments. The invention additionally includes nucleic acids or nucleic acid fragments, or .
complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized.
These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as anti-sense binding nucleic acids in therapeutic applications in a subject.
In the mutant or variant nucleic acids, and their complements, up to 20% or more of the bases may be so changed.
The novel IF1206 of the invention include the protein fragments whose sequences are provided in Tables 2, 4 and 5 inclusive. The invention also includes an IF1206 mutant or variant protein, any of whose residues may be changed from the corresponding residue shown in Tables 2, 4 and 5 while still encoding a protein that maintains its native activities and physiological functions, or a functional fragment thereof. In the mutant or variant IF1206, up to 20% or more of the residues may be so changed. The invention further encompasses Abs and antibody fragments, such as Fab or (Fab)2, that bind immunospecifically to any of the IF1206 of the invention.
The IF1206 nucleic acid (Table 1 ) comprises a start codon at nucleotides 290-292 (bold, underline); a stop codon at nucleotides 1708-1710 (bold, dash underline), and a putative polyadenylation site at nucleotides 1770-1777 (bold, double-underlined).
Table 1. 1F1206 nucleotide fragment (SEQ ID N0:1 ).
cgattcgaattcggccacactggccggatcctctagagatccctcgacctcgacccacgc~60 gtccgagcacagtgagagacacccagtgctgctcaagaagtgaaacaactctgagagtat120 cctaaccactggtgtcttcctttataccccatttttcactttctcagttactgaattatc180 tgcctacctactcaaaccaagcaggccacttctgttgttgaagatctcagcacctgtaca240 ttgctgccgaaattccagggagtataaccaacaacttgaaagatggagaa~aatataag 300 agacttgttctgctggaaggacttgaatgtatcaataagcatcaattcaatttatttaag360 tcattgatggtcaaagatttaaatctggaagaagacaaccaagagaaatataccacgttt420 cagattgctaacatgatggtaaagaaatttccagctgatgctggattggacaaactgatc480 aacttttgtgaacgtgtaccaactcttaaaaaacgtgctgaaattcttaaaaaagagaga540 tcagaagtaacaggagaaacatcactggaaataaataggcaagaagcaagtcctgcaaca600 cctacatcaactacaagccacatgttagcatctgaaagaggcaagacttccacaaccacc660 actgagacccaggaagagacttccacagcccagtcggggacttccacagctcacgcgggg720 acttctacagcaccggcggggactttcacaactcagaaaagaaaaagtaggagagaagaa780 gagactggagtgaaaaagagcaaggcgtctaaggaaccagatcagcctccctgttgtgaa840 gaacccacagccaggtgccagtcaccaatactccacagctcatcttcagcttcatctaac900 attccttcagctacgaaccaaaaaccacaaccccagaaccagaacattcccagaggtgct960 gttctccactcagagcccctgacagtgatggtgctcactgcaacagacccgtttgaatat1020 gaatcaccagaacatgaagtaaagaacatgtttcatgctacagtggctacagtgagccag1080 tatttccatgtgaaagttttcaacatcaacttgaaagagaagttcacaaaaaagaatttt1140 atcatcatatccaattactttgagagcaaaggcatcctggagatcaatgagacttcctct1200 gtgttaaaggctgatcctgaccaaatgattgaagtgcccaacaatattatcagaaatgca1260 aatgccagtcctaagatctgtgatattcaaaagggtacttctggagcagtgttctatgga1320 gtgtttacattacacaagaaaaaagtgaaaacacagaacacaagctatgaaataaaagat1380 ggttcaggaagtatagaagtggaggggagtggacaatggcacaacatcaactgtaaggaa1440 ggagataagctccacctcttctgctttcacctgaaaagagaaagaggacaaccaaagtta1500 gtgtgtggagaccacagtttcgtcaagatcaaggtcaccaaggctgggaaaaaaaaggaa1560 gcatcaactgtcctgtcaagcacaaaaaatgaagaagaaaataattacccaaaagatgga1620 attaaggtagagatgccagactattcacgtctaaatgacagctttagtagtatatccaag1680 catttaataaccttcatacctgatttc,~g~ttttgtattttcatttgaaaaaatttctta1740 ttgttctgtttttctatgaass~aaaatttgatttaatttctctactgtaaaaataataa1800 acatgtctttttaaagggacatcaaaaaaaaagaaggagggaggggagggggttggtata1860 agaaaaaccggggcggccg 1879 A polypeptide encoded by SEQ ID N0:1 is presented in Table 2. The polypeptide described in SEQ ID N0:1 is likely nuclear, PSORT predicts nuclear localization to nucleus with certainty = 0.8800, to the microbody (peroxisome) with certainty=0.3000, to the mitochondrial matrix space with certainty=0.1000 and to the lysosome (lumen) with certainty=0.1000 (Nakai and Fiorton, 1999) . ProDom (Protein Domain Database) analysis demonstrates that SEQ ID N0:1 is an IFI protein of the class described by prdm: 3409 p36 (8) if16(2) ifi4(2) ifi2(2) // protein interferon induction interferon-activatable myeloid differentiation repeat y-interferon=inducible IFI-16 interferon-inducible p=1.2e-82 (Altschul et al., 1990).
Table 2. 1F1206 polypeptide sequence (SEQ ID N0:2).
Met Glu Asn Glu Tyr Lys Arg Leu Val Leu Leu Glu Gly Leu Glu Cys Ile Asn Lys His Gln Phe Asn Leu Phe Lys Ser Leu Met Val Lys Asp Leu Asn Leu Glu Glu Asp Asn Gln Glu Lys Tyr Thr Thr Phe Gln Ile Ala Asn Met Met Val Lys Lys Phe Pro Ala Asp Ala Gly Leu Asp Lys Leu Ile Asn Phe Cys Glu Arg Val Pro Thr Leu Lys Lys Arg Ala Glu Ile Leu Lys Lys Glu Arg Ser Glu Val Thr Gly Glu Thr Ser Leu Glu Ile Asn Arg Gln Glu Ala 5er Pro Ala Thr Pro Thr Ser Thr Thr Ser His Met Leu Ala Ser Glu Arg Gly Lys Thr Ser Thr Thr Thr Thr Glu Thr Gln Glu Glu Thr Ser Thr Ala Gln Ser Gly Thr Ser Thr Ala His Ala Gly Thr Ser Thr Ala Pro Ala Gly Thr Phe Thr Thr Gln Lys Arg Lys Ser Arg Arg Glu Glu Glu Thr Gly Val Lys Lys Ser Lys Ala Ser Lys Glu Pro Asp Gln Pro Pro Cys Cys Glu Glu Pro Thr Ala Arg Cys Gln Ser Pro Ile Leu His Ser Ser Ser Ser Ala Ser Ser Asn Ile Pro Ser Ala Thr Asn Gln Lys Pro Gln Pro Gln Asn Gln Asn Ile Pro Arg Gly Ala Val Leu His Ser Glu Pro Leu Thr Val Met Val Leu Thr Ala Thr Asp Pro Phe Glu Tyr Glu Ser Pro Glu His Glu Val Lys Asn Met Phe His Ala Thr Val Ala Thr Val Ser Gln Tyr Phe His Val Lys Val Phe Asn Ile Asn Leu Lys Glu Lys Phe Thr Lys Lys Asn Phe Ile Ile Ile Ser Asn Tyr Phe Glu Ser Lys Gly Ile Leu Glu Ile Asn Glu Thr Ser Ser Val Leu Lys Ala Asp Pro Asp Gln Met Ile Glu Val Pro Asn Asn Ile Ile Arg Asn Ala Asn Ala Ser Pro Lys Ile Cys Asp Ile Gln Lys Gly Thr Ser Gly Ala Val Phe Tyr Gly Val Phe Thr Leu His Lys Lys Lys Val Lys Thr Gln Asn Thr Ser Tyr Glu Ile Lys Asp Gly Ser Gly Ser Ile Glu Val Glu Gly Ser Gly Gln Trp His Asn Ile Asn Cys Lys Glu Gly Asp Lys Leu His Leu Phe Cys Phe His Leu Lys Arg Glu Arg Gly Gln Pro Lys Leu Val Cys Gly Asp His Ser Phe Val Lys Ile Lys Val Thr Lys Ala Gly Lys Lys Lys Glu Ala Ser Thr Val Leu Ser .. Ser Thr Lys.Asn Glu Glu Glu Asn Asn T~yr Pro Lys~Asp Gly ~Ile-Lys Val Glu Met Pro Asp Tyr Ser Arg Leu Asn Asp Ser Phe Ser Ser Ile Ser Lys His Leu Ile Thr Phe Ile Pro Asp Phe Table 3 presents an analysis of the physical.characteristics of SEQ ID
N0:2 (Pace et al., 1995). The SEQ ID N0:2 polypeptide consists of 475 amino acids with a calculated molecular weight of 53095.5 Daltons and a predicted isoelectric point of 8.18 ((GCG), 1999). The conditions at which this analysis is valid are: pH 6.5, 6.0 M guanidium hydrochloride, 0.02 M
phosphate buffer.
Table 3 Amino acid composition, molecular weight, and structural analysis of SEQIDN0:2 The naturally-occuring variant of interferon-inducible polypeptide 206 (1F1206) nucleic acid (SEQ ID N0:3, Table 4) comprises a. start codon at nucleotides 290-292 (bold, underline); a stop codon at nucleotides 1708-1710 (bold, dash underline), and a putative polyadenylation site at nucleotides 1770-1777 (bold, double-underlined).

Table 4 IF1206b nucleotide fragment, a naturally-occuring variant (SEQ
ID N0:3) cgattcgaattcggccacactggccggatcctctagagatccctcgacctcgacccacgc60 gtccgagcacagtgagagacacccagtgctgctcaagaagtgaaacaactctgagagtat120 cctaaccactggtgtcttcctttataccccatttttcactttctcagttactgaattatc180 tgcctacctactcaaaccaagcaggccacttctgttgttgaagatctcagcacctgtaca240 ttgctgccgaaattccagggagtataaccaacaacttgaaagatggagaa 300 taaatataag agacttgttctgctggaaggacttgaatgtatcaataagcatcaattcaatttatttaag360 tcattgatggtcaaagatttaaatctggaagaagacaaccaagagaaatataccacgttt420 cagattgctaacatgatggtaaagaaatttccagctgatgctggattggacaaactgatc480 aacttttgtgaacgtgtaccaactcttaaaaaacgtgctgaaattcttaaaaaagagaga540 tcagaagtaacaggagaaacatcactggaaataaataggcaagaagcaagtcctgcaaca600 cctacatcaactacaagccacatgttagcatctgaaagaggcgagacttccacaacccag660 gaagagacttccacagcccagtccgggccttcgacagctcctgcgcggactttaacagcc720 cagaaaagaaaaagtaggagagaagaagagactggagtgaaaaagagcaaggcgtctaag780 gaaccagatcagcctccctgttgtgaagaacccacagccaggtgccagtcaccaatactc840 cacagctcatcttcagcttcatctaacattccttcagctacgaaccaaaaaccacaaccc900 cagaaccagaacattcccagaggtgctgttctccactcagagcccctgacagtgatggtg960 ctcactgcaacagacccgtttgaatatgaatcaccagaacatgaagtaaagaacatgttt1020 catgctacagtggctacagtgagccagtatttccatgtgaaagttttcaacatcaacttg1080 aaagagaagttcacaaaaaagaattttatcatcatatccaattactttgagagcaaaggc1140 atcctggagatcaatgagacttcctctgtgttaaaggctgatcctgaccaaatgattgaa1200 gtgcccaacaatattatcagaaatgcaaatgccagtcctaagatctgtgatattcaaaag1260 ggtacttctggagcagtgttctatggagtgtttacattacacaagaaaaaagtgaaaaca1320 cagaacacaagctatgaaataaaagatggttcaggaagtatagaagtggaggggagtgga1380 caatggcacaacatcaactgtaaggaaggagataagctccacctcttctgctttcacctg1440 aaaagagaaagaggacaaccaaagttagtgtgtggagaccacagtttcgtcaagatcaag1500 gtcaccaaggctgggaaaaaaaaggaagcatcaactgtcctgtcaagcacaaaaaatgaa1560 gaagaaaataattacccaaaagatggaattaaggtagagatgccagactatcacgtctaa1620 a~gacagctttagtagtatatccaagcatttaataaccttcatacctgatttctgatttt1680 gtattttcatttgaaaaaatttcttattgttctgtttttctatgaaaataaaatttgatt1740 taatttctctactgtaaaaa.taataaaca~ agggacatcaaaaaaaaaga1800 gtctttttaa aggagggaggggagggggttggtataagaaaaaccggggc 1840 A polypeptide encoded by SEQ ID N0:3 is presented in Table 5. The polypeptide described in SEQ ID N0:3 is likely nuclear, PSORT predicts nuclear localization to nucleus with certainty = 0.8800, to the microbody (peroxisome) with certainty=0.3000, to the mitochondrial matrix space with certainty=0.1000 and to the lysosome (lumen) with certainty=0.1000, (Nakai and Horton, 1999) . ProDom (Protein Domain Database) analysis demonstrates that SEQ ID N0:3 is an IFI protein of the class described by ~rdm: 3409 p36 (8~ if16(2) ifi4(2) ifi2(2) // protein interferon induction interferon-activatable myeloid differentiation repeat ~r-interferon-inducible IFI-16 interferon-inducible p=2.7e-83 LAltschul et al., 1990).
Table 5 IF1206b, a naturally-occuring variant polypeptide sequence (SEQ ID
N0:4) Met Glu Asn Glu Tyr Lys Arg Leu Val Leu Leu Glu Gly Leu Glu Cars Ile Asn Lys His Gln Phe Asn Leu Phe Lys Ser Leu Met Val Lys Asp Leu Asn Leu Glu Glu Asp Asn Gln Glu Lys Tyr Thr Thr Phe Gln Ile Ala Asn Met Met Val Lys Lys Phe Pro Ala Asp Ala Gly Leu Asp Lys Leu Ile Asn Phe Cps Glu Arg Val Pro T'hr Leu Lys Lys Arg Ala Glu Ile Leu Lys Lys Glu Arg Ser Glu Val Thr Gly Glu Thr Ser Leu Glu Ile Asn Arg Gln Glu Ala Ser Pro Ala Thr Pro Thr Ser Thr Thr Ser His Met Leu Ala Ser Glu Arg Gly Glu Thr Ser Thr Thr Gln Glu Glu 115 120 , 125 Thr Ser Thr Ala Gln Ser Gly Pro Ser Thr Ala Pro Ala Arg Thr Leu Thr Ala Gln Lys Arg Lys Ser Arg Arg Glu Glu Glu Thr Gly Val Lys 145 150 155 ' 160 Lys Ser Lys Ala Ser Lys Glu Pro Asp Gln Pro Pro Cars Cps Glu Glu Pro Thr Ala Arg Cars Gln Ser Pro Ile Leu His Ser Ser Ser Ser Ala Ser Ser Asn Ile Pro Ser Ala Thr Asn Gln Lys Pro Gln Pro Gln Asn Gln Asn Ile Pro Arg Gly Ala Val Leu His Ser Glu Pro Leu Thr Val Met Val Leu Thr Ala Thr Asp Pro Phe Glu Tyr Glu Ser Pro Glu His Glu Val Lys Asn Met Phe His Ala Thr Val Ala Thr Val Ser Gln Tyr Phe His Val Lys Val Phe Asn Ile Asn Leu Lys Glu Lys Phe Thr Lys Lys Asn Phe Ile Ile Ile Ser Asn Tyr Phe Glu Ser Lys Gly Ile Leu Glu Ile Asn Glu Thr Ser Ser Val Leu Lys Ala Asp Pro Asp Gln Met Ile Glu Val Pro Asn Asn Ile Ile Arg Asn Ala Asn Ala Ser Pro Lys Ile Cps Asp Ile Gln Lys Gly Thr Ser Gly Ala Val Phe Tyr Gly Val Phe Thr Leu His Lys Lys Lys Val Lys Thr Gln Asn Thr Ser Tyr Glu Ile Lys Asp Gly Ser Gly Ser Ile Glu Val Glu Gly Ser Gly Gln Trp His Asn Ile Asn Cps Lys Glu Gly Asp Lys Leu His Leu Phe Cars Phe His Leu Lys Arg Glu Arg Gly Gln Pro Lys Leu Val Cps Gly Asp His 385 390 395 400 ' Ser Phe Val Lys Ile Lys Val Thr Lys Ala Gly Lys Lys Lys Glu Ala Ser Thr Val Leu Ser Ser Thr Lys Asn Glu Glu Glu Asn Asn err Pro Lys Asp Gly Ile Lys Val Glu Met Pro Asp 'I~rr His Val Table 6 presents an analysis of the physical characteristics of SEQ ID
N0:4 (Pace et al., 1995). The SEQ ID N0:4 polypeptide consists of 445 amino acids with a calculated molecular weight of 49899.1 Daltons and a predicted isoelectric point of 8.17 ((GCG), 1999). The conditions at which this analysis is valid are: pH 6.5, 6.0 M guanidium hydrochloride, 0.02 M
phosphate buffer.
Table 6 Amino acid composition, molecular weight, and structural analysis of SEQIDN0:4 Values assuming all Cys residues appear as half cystines ,;:::::::::::::::::::::::::~:::::8::::::::::::::::;::::
................................................................~
~ 279 nm 2 ~ 282 ................................................................:::::::::::::::
::::::::::~.::::::::::::::::::::::::0 nm : ;
276 nm '278 ;
nm ;' . ................................................
'..Extinction Coefficient ::........................::................................................168 19030 18708 i 18245 . 17690 ..
.. ~ ~ :: .
. :: ' ............. .........................:
:~........................:........................:
.........................
.................................................;.........................
...............................................................................
............ .........................:.......................Ø337 Optical Densit ..........................Ø366 0..355 .
........................:
y ,.......................:~ .... .
0 .375 : 381 .. 0 Values assuming no Cys residues appear as half cystines:

::............. ..... .. .... ~ _ . ..
.. 76 nm 278 nm ~ 279 nm 280 282 nm 2 nm .. ... .. . ._ ... .... . s ......... . ...
"............:.........~.......................~...~...........,:..............
..........i;........................... ...... ...
Extinction Coefficient ... .....
,:.........................
;: ;:18200 .,~........................,:........................
18450 f 17765 ..16400 17210 , : .

.. .
,.
.................................:::::::::::::::::::::::::::::::;;:::::::::::::
::::::::::::;::::::::::::::::::::::::: :::::::::::::::::::::::::
;::::::::::::::::::::::::;
.................................. .. :::::::::::::::::::::::, Optical Density 0 , . ;:
................................................................:~...... 370 :: ;' 0.329 ........ 365 U 0 _356........v:........................
'..................:0.345_......
~ 0~..................: ...... :......

The cDNA sequence encoding mouse IF1206c (SEQ ID N0:14) derived from a mouse brown adipose tissue (BAT) cDNA library is shown in Table 7.
Start codon "ATG" (bold, underlined) and stop codon "TAG" (bold, dash underlined), and the putative polyadenylation sites are (bold, double-underlined) are indicated. The cDNA clone was obtained from the library upon PCR amplification cloning methods utilizing specific oligos, followed by further identification of positive clones via common analysis employing a 32P-labeled probe:
SEQ ID N0:16 (IF1206.snr1 PCR oligo):
CATCATGTTAGCAATCTGAAACGTGGTATATTTCT
SEQ ID N0:17 (IF1206.snf1 PCR oligo):
GTAAAGAAATTTCCAGCTGATGCTGGATTGG
SEQ ID N0:18 (IF1206.p1 probe):
CTTCCTGGGTTGCGGAAGTCTCGCCTCTTTCAGATG

Table 7 IF1206c nucleotide fragment, a naturally-occuring variant (SEQ
ID N0:14) agcacagtgagagacacccagtgctgctcaagaagtgaaacaactctgagagtatcctaa60 ccactggtgtcttcctttataccccatttttcactttctcagttactgaattatctgcct120 acctactcaaaccaagcaggccacttctgttgttgaagatctcagcacctgtacattgct180 gccgaaattccagggagtataaccaacaacttgaaaaatggagaatgaatataagagact240 tgttctgctggaaggacttgaatgtatcaataagcatcaattcaatttatttaagtcatt300 gatggtcaaagatttaaatctggaagaagacaaccaagagaaatataccacgtttcagat360 tgctaacatgatggtaaagaaatttccagctgatgctggattggacaaactgatcaactt420 ttgtgaacgtgtaccaactcttaaaaaacgtgcagaaattcttaaaaaagagagatcaga480 agtaacaggagaaacatcactggaaataaataggcaagaagcaggtcctgcaacacctac540 atcaactacaagccacatgttagcatctgaaagaggcgagacttccgcaacccaggaaga600 gacttccacaggccagaaaaggaagccaggtggagagattaggtctgtctcccagccaag660 gccagtcaggaaccagaggggagctgggctggcaaggaaaggttggggtgtgctggctga720 aggagagaaaggagagaaaggagagaaaggaaagaaggaaggagagaaagaaagaaagaa780 agaaaggaaggaaggaaggaaggaagaaagaaaaagaaagaaagaaagaaagaaagaaag890 aaagaaagaaagaaagaaagaaagaaagaaagaaagaaagacagaccacaggtttgtcat900 cttcagcctccaggtttgtcatcttcagcctccaggtttgtcatcttcagcctccaggtt960 tgtcatcttcagcctccaggtttgtcatcttcagcctccaggtttgtcatcttcagcctc1020 caggtttgtcatcttcagcctccaggtttgtcatcttcagcctccaggtttgtcatcttc1080 agcctccaggtttgtcatcttcagcctccacaggtttgtcatcttcagcctccaggtagg1140 tggggtaggctctggctctgtgtcctgcctttac~agactagcacaccagcaaaccaaatt1200 cccatctcgtcagagtagcagtaagggcaagcccaggggggtagtgtgccacccagtgac1260 ccattgatccttgggtaatggtcctctctgtccataaggctcaggagtcacagaaggtcc1320 agctatctcaaccccacactcttgggaacacctccccgcctttttagaacagtaagttct1380 ctgtggcctcatgctgttctgagagccccttggtgctgccacttctccctgtgctctctc1440 attcccttctgcttcctgcacatctgctgaacccacgtcatttccggtactgcctagtta1500 gtcctggaaaaaactctcttggccattggcaggaatcagtgtagaaaagtttgcaggaca1560 tccctggctttccagagcatgcagaatcagtgtagctcatgacactgtcagacactttag1620 acacgagagaaattcttaagagacctacgcctttgacctctcagatggcacggccgctgt1680 acacagggaagtgttcactttccttgagacgggaagctggcttcaggttcctatggaata1740 gagttttctttccttattcccttttcacctaacagttttgctcttcagacagctgcccat1800 tccctaagcctcgcctagaaaccataacacagatgtacctagatgaatgagccaagcaac1860 tgagaaacagcaaggaaactggaaggcttgaggtgggaatatgaaggtcaagacaagaat1920 tagggagctgaaaagatggctcatcagttgactgctcttccagaggtcctgagttcaatt1980 cccagcaaccacatgatggctcgcaaccatctataataggatccacacactcttctggtg2040 tgtctgaagacagctacagtgtactcataataaataaagtaaataaatttaaaaaaaaaa2100 aaaaaatggagaatgaat 2118 Table 8 shows the polypeptide sequence (SEQ ID N0:15) of the open reading frame of the polynucleotide sequence shown in Table 7.

Table 8 IF1206c, a naturally-occuring variant polypeptide sequence (SEQ ID N0:15) Met Glu Asn Glu Tyr Lys Arg Leu Val Leu Leu Glu Gly Leu Glu Cys Ile Asn Lys His Gln Phe Asn Leu Phe Lys Ser Leu Met Val Lys Asp Leu Asn Leu Glu Glu Asp Asn Gln Glu Lys Tyr Thr Thr Phe Gln Ile Ala Asn Met Met Val Lys Lys Phe Pro Ala Asp Ala Gly Leu Asp Lys Leu Ile Asn Phe Cys Glu Arg Val Pro Thr Leu Lys Lys Arg Ala Glu Ile Leu Lys Lys Glu Arg Ser Glu Val Thr Gly Glu Thr Ser Leu Glu Ile Asn Arg Gln Glu Ala Gly Pro Ala Thr Pro Thr Ser Thr Thr Ser His Met Leu Ala Ser Glu Arg Gly Glu Thr Ser Ala Thr Gln Glu Glu Thr Ser Thr Gly Gln Lys Arg Lys Pro Gly Gly Glu Ile Arg Ser Val Ser Gln Pro Arg Pro Val Arg Asn Gln Arg Gly Ala Gly Leu Ala Arg Lys Gly Trp Gly Val Leu Ala Glu Gly Glu Lys Gly Glu Lys Gly Glu Lys Gly Lys Lys Glu Gly Glu Lys Glu Arg Lys Lys Glu Arg Lys Glu Gly Arg Lys Glu Glu Arg Lys Arg Lys Lys Glu Arg Lys Lys Glu Arg Lys Lys Glu Arg Lys Lys Glu Arg Lys Lys Glu Arg Lys Thr Asp His Arg Phe Val Ile Phe Ser Leu Gln Val Cars His Leu Gln Pro Pro Gly Leu Ser Ser Ser Ala Ser Arg Phe Val Ile Phe Ser Leu Gln Val Cps His Leu Gln Pro Pro Gly Leu Ser Ser Ser Ala Ser Arg Phe Val Ile Phe Ser Leu Gln Val Cars His Leu Gln Pro Pro Gly Leu Ser Ser Ser Ala Ser Arg Phe Val Ile Phe Ser Leu His Arg Phe Val Ile Phe Ser Leu Gln Val Gly Gly Val'Gly Ser Gly Ser Val Ser Cars Leu Table 9 presents an analysis of the physical characteristics of SEQ ID
N0:15 (Pace et al., 1995). The SEQ ID N0:15 polypeptide consists of 318 amino acids with a calculated molecular weight of 35984.1 Daltons and a predicted isoelectric point of 10.67 ((GCG), 1999). The conditions at which this analysis is valid are: pH 6.5, 6.0 M guanidium hydrochloride, 0.02 M

phosphate buffer. 1F1206 function may be assigned by analyzing protein similarity.
Table 9 Amino acid composition, molecular weight, and structural analysis of SEQIDN0:15 .......................
..........:.....:..::::::................:............:::.:.......:.:...:....:.
::::. ..............::...:...::.:
::::.................::.........:::::.::...:................._._._....:........
.:.. .....:.....:.::.
""Values'assuming aIL.Cys.residues appear .as'half.cystines ...... ....... ..
:...........................................................................s.~
...........................................................;...................
....."........................:
.. .. . . , . . .. .
'27 m " 278 nm ; 279 nm...280.nm....282.nm..' :::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::::
;:::::::::::::::::::::::: ,::::::::::::::::::::::::, .........................:.:8 :..::............."...:::.............::....
. 8781 . 610 .. 8300 Extinction Coefficient 8735 ~ 8710.......
~:..............................................' Optical Densit .....................~w0.243~......~, 0.244.. .... .Ø242 ~' 0 ' 0w ..........................................y...................
~........................~~.........................:~.~....~.~...._~239 ::.....:231 Values assuming no Cys residues appear as half cystines ... ..:. ......:.. .. 276 nm.. 278..nm ~ 279 nm 280 nm . 282 nm ..................................................................,...:........
............,:......................::..............:.........::.:............:
...........".:.::....................:
Extinction Coefficient 8300 ' 8350 ' 8250 8000 ................................................................:.'............
............: '_.8400.......... ~........................: .........
..............:,.......................
............~........................~........................"................
........;,........................,,........................,..................
.....;;........................, .~ Optical Density 0.231 0.233 0.232 0.229 .222 ' ;. . . .. ~~ 0 The invention also includes polypeptides having 80-100%, including 81, 82, 83, 84, 85, 86, 87, 88, 89, 89.2, 90, 91, 92, 93, 94, 95, 96, 97, 98 and 99%, sequence identity to SEQ ID NOS: 2, 4, and 15, excluding those polypeptides that are identical to SEQ ID NOS:22 and 24, preferably excluding those polypeptides having 80-100%, including 81, 82, 83, 84, 85, 86, 87, 88, 89, 89.2, 90, 91, 92, 93, 94, 95, 96, 97, 98 and 99%, sequence identity to SEQ ID NOS:22 and 24. The invention also includes nucleotides encoding these polypeptides. SEQ ID N0:22 corresponds to IF1204, and SEQ ID N0:24 corresponds to IF1205D3. SEQ ID N0:21 and SEQ ID N0:23 are the corresponding nucleotide sequences.
One method, using the EMOTIF database (Huang and Brutlag, 2001;
Nevill-Manning et al., 1998), assesses IF1206 polypeptide sequence against a database of protein motifs (consensus sequences, consensi) that correspond to evolutionarily and/or functionally conserved regions of proteins. Such an approach mines databases of known protein motifs, generates new protein motifs, or tests new motifs against databases of known proteins such as SwissProt.
One such motif tested against SwissProt using EMOTIF-Scan identifies a number of interferon-induced genes, matches IF1206, and a number of proteins that have in common the ability to bind RNA or DNA. Of particular note, is the identification of the fly SUS gene, a mRNA binding protein (Voelker et al., 1991 ), the yeast mRNA binding protien RNA15 (Minvielle-Sebastia et al., 1994), among other nucleotide binding proteins such as the mouse ribosomal binding protein L6 also known as HTLV-I tax responsive element binding protein 107 (TAXREB107) which binds to DNA (Morita et al., 1993).
Table 10 shows the consensus sequence for IFI-induced genes from human and mouse that were generated using the software EMOTIF (Huang and Brutlag, 2001; Nevill-Manning et al., 1998) and represented in the single letter abbreviation for amino acids. Residues in []'s indicate that any of those amino acids may be used at that position; a "." Indicates that any amino acid-or no amino acid-may occupy this position in the motif.

Table 10 Consensus motifs SEQ ID Consensus sequence Probability'ide t fying2~3 NO

M[FYWLI]HATVA[TAS].(STKR][QE][FYW]1 in 102' IFI proteins [F [HRKFYW]V KRMLI]V[FYLI]

7 [F HATVA ST] ND IFI proteins M..[EKQ]YK.[ILV][ILV]LL.G[FLY][DE].[ILM

8 ..[FLY]..[FILMV]K.[FLY][ILMV]..[DE][FL1 in 10Z' IFI proteins .[ILV]

IFI proteins, poly-nucleotide-[FLY]..[FILMV]K.[FLY][ILMV]..[DE][FLY].(IND binding LV] protiens, nucleotide binding proteins [KQR]E.Y..[FILMY][KQR][ILV)[AST][DN].

M..KF...[AS].L.KLI.[FILMY].[EKQ].[ILMV]..1 in 1022 IFI proteins L[EKQR]......L[KR .E[KR

IFI proteins, 11 [KQR]E.Y..[FILMV](KQR][ILV][AST][DN]ND mYloido--genic glycoproteins IFI proteins, Poly-nucleotide-12 T.[TGVS][TASE][QKAER][KR][RK][KRVN]ND binding ..[EQKRIL]....K proteins, nucleotide-binding proteins 13 C[KREQ].G[DESTNQ][KRTS][LIV].LND IFI proteins 'Probability of matching a random sequence, using emotif maker calculation of requency of hits to SwissProt.
ND, not done.

ZUtility determined by identification of proteins in SwissProt using search engine and allowing a single mis-match.

3Mis-matches allowable and still capable of identifying IFI
proteins in SwissProt using emotif scan.

The nucleic acids and proteins of the invention are potentially useful in the treatment of Type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, reproductive organ cancers, fatty liver, viral infections, inflammation, allergies, steatosis, hepatoxicity, inflammary bowel disease, septic shock, and related conditions and sleep apnea, as well as those directly related to interferons, such as metabolic disorders.
The IFI206 and proteins of the invention are useful in potential therapeutic applications implicated in Type II diabetes mellitus (NIDDM), hypertension, coronary heart disease, hypercholesterolemia, osteoarthritis, gallstones, cancers of the reproductive organs, and sleep apnea, as well as those directly related to interferons, such as metabolic disorders. For example, a cDNA encoding IF1206 may be useful in gene therapy, and IF1206 protein may be useful when administered to a subject in need thereof. The novel nucleic acid encoding IF1206, and the IFI206 protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
These materials are further useful in the generation of Abs that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods.
IF1206 polynucleotides One aspect of the invention pertains to isolated nucleic acid molecules.
that encode IF1206 or biologically-active portions thereof. Also included in the invention are nucleic acid fragments sufficient for use as hybridization probes to identify IF1206-encoding nucleic acids (e.g., IFI206 mRNAs) and fragments for use as polymerase chain reaction (PCR) primers for the amplification and/or mutation of IF1206 molecules. A "nucleic acid molecule" includes DNA
molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs. The nucleic acid molecule may be single-stranded or double-stranded, but preferably comprises double-stranded DNA.
probes Probes are nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), 100 nt, or many (e.g., 6,000 nt) depending on the specific use. Probes are used to detect identical, similar, or complementary nucleic acid sequences. Longer length probes can be obtained from a natural or recombinant source, are highly specific, and much slower to hybridize than shorter-length oligomer probes. Probes may be single- or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA-like technologies.
Probes are substantially purified oligonucleotides that will hybridize under stringent conditions to at least optimally12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence of SEQ ID NOS:1 ' or 3; or an anti-sense strand nucleotide sequence of SEQ ID NOS:1 or 3; or of a naturally occurring mutant of SEQ ID NOS:1 or 3.
The full- or partial length native sequence IFI206 may be used to "pull out" similar (homologous) sequences (Ausubel et al., 1987; Sambrook, 1989), such as: (1 ) full-length or fragments of IF1206 cDNA from a cDNA library from any species (e.g. human, murine, feline, canine, bacterial, viral, retroviral, yeast), (2) from cells or tissues, (3) variants within a species, and (4) homologues and variants from other species. To find related sequences that may encode related genes, the probe may be designed to encode unique sequences or degenerate sequences. Sequences may also be genomic sequences including promoters, enhancer elements and introns of native sequence IFI206.
For example, IFI206 coding region in another species may be isolated using such probes. A probe of about 40 bases is designed, based on IFI206, and made. To detect hybridizations, probes are labeled using, for example, radionuclides such as 32P or 35S, or enzymatic labels such as alkaline phosphatase coupled to the probe via avidin-biotin systems. Labeled probes are used to detect nucleic acids having a complementary sequence to that of IFI206 in libraries of cDNA, genomic DNA or mRNA of a desired species.
Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis-express an IF1206, such as by measuring a level of an IFI206 in a sample of cells from a subject e.g., detecting IF1206 mRNA levels or determining whether a genomic IFI206 has been mutated or deleted.
2. isolated nucleic acid An isolated nucleic acid molecule is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid.
Preferably, an isolated nucleic acid is free of sequences that naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3'-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, isolated IFI206 molecules can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell/tissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.). Moreover, an isolated nucleic acid molecule, such as a cDNA molecule, can be substantially free of.other cellular material or culture medium when produced by recombinant techniques, or of chemical precursors or other chemicals when chemically synthesized.
A nucleic acid molecule of the invention, e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NOS: 2, 4 or 15, or a complement of this aforementioned nucleotide sequence, can be isolated using standard molecular biology techniques and the provided sequence information. Using all or a portion of the nucleic acid sequence of SEQ ID NOS: 2, 4 or 15 as a hybridization probe, IFI206 molecules can be isolated using standard hybridization and cloning techniques (Ausubel et al., 1987; Sambrook, 1989).
PCR amplification techniques can be used to amplify IFI206 using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers. Such nucleic acids can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to IFI206 sequences can be prepared by standard synthetic techniques, e.g., an automated DNA synthesizer.
3. oligonucleotide An oligonucleotide comprises a series of linked nucleotide residues, which oligonucleotide has a sufficient number of nucleotide bases to be used in a PCR reaction or other application. A short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue. Oligonucleotides comprise portions of a nucleic acid sequence having about 10 nt, 50 nt, or 100 nt in length, preferably about 15 nt to 30 nt in length. In one embodiment of the invention, an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides of SEQ ID NOS:1 or 3, or a complement thereof. Oligonucleotides may be chemically synthesized and may also be used as probes.
4. complementary nucleic acid sequences; binding In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS: 2, 4 or 15, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of an IF1206). A nucleic acid molecule that is complementary to the nucleotide sequence shown in SEQ ID NOS:1 or 3, is one that is sufficiently complementary to the nucleotide sequence shown in SEQ ID NOS:1 or 3, that it can hydrogen bond with little or no mismatches to the nucleotide sequence shown in SEQ ID NOS:1 or 3, thereby forming a stable duplex.
"Complementary" refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a nucleic acid molecule, and the term "binding"
means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like. A physical interaction can be either direct or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates.
Nucleic acid fragments are at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, respectively, and are at most some portion less than a full-length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice.
5. derivatives, and analogs Derivatives are nucleic acid sequences or amino acid sequences formed from the native compounds either directly or by modification or partial substitution. Analogs are nucleic acid sequences or amino acid sequences that have a structure similar to, but not identical to, the native compound but differ from it in respect to certain components or side chains. Analogs may be synthetic or from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type. Homologs are nucleic acid sequences or amino acid sequences of a particular gene that are derived from different species.
Derivatives and analogs may be full length or other than full length, if the derivative or analog contains a modified nucleic acid or amino acid, as described below. Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the aforementioned proteins under stringent, moderately stringent, or low stringent conditions (Ausubel et al., 1987).
6. homology A "homologous nucleic acid sequence" or "homologous amino acid sequence," or variations thereof, refer to sequences characterized by a homology at the nucleotide level or amino acid level as discussed above.
Homologous nucleotide sequences encode those sequences coding for isoforms of IF1206. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA.
Alternatively, different genes can encode isoforms. In the invention, homologous nucleotide sequences include nucleotide sequences encoding for an IF1206 of species other than humans, including, but not limited to:
vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat cow, horse, and other organisms. Homologous nucleotide sequences also include, but are not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein. A homologous nucleotide sequence does not, however, include the exact nucleotide sequence encoding human IF1206. Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NOS:2, 4 or 15, as well as a polypeptide possessing IF1206 biologicaE
activity. Various biological activities of the IF1206 are described below.
7. open reading frames The open reading frame (ORF) of an IFI206 gene encodes IF1206. An ORF is a nucleotide sequence that has a start codon (ATG) and terminates with one of the three "stop" codons (TAA, TAG, or TGA). In this invention, however, an ORF may be any part of a coding sequence that may or may not comprise a start codon and a stop codon. To achieve a unique sequence, preferable IFI206 ORFs encode at least 50 amino acids.
1F1206 polypeptides 1. mature An IF1206 can encode a mature IF1206. A "mature" form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide or precursor form or proprotein. The naturally' occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full-length gene product, encoded by the corresponding gene.
Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an open reading frame described herein. The product "mature"
form arises, again by way of nonlimiting example, as a result of one or more naturally occurring processing steps as they may take place within the cell, or host cell, in which the gene product arises. Examples of such processing steps leading to a "mature" form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an open reading frame, or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protein that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine. Alternatively, a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+1 to residue N remaining. Further as used herein, a "mature" form of a polypeptide or protein may arise from a step of post-translational modification other than a proteolytic cleavage event. Such additional processes include, by way of non-limiting example, glycosylation, myristoylation or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them.
2. active An active IF1206 polypeptide or IF1206 polypeptide fragment retains a biological and/or an immunological activity similar, but not necessarily identical, to an activity of a naturally-occuring (wild-type) IF1206 polypeptide of the invention, including mature forms. A particular biological assay, with or without dose dependency, can be used to determine IF1206 activity. A nucleic acid fragment encoding a biologically-active portion of IF1206 can be prepared by isolating a portion of SEQ ID NOS: 2, 4 or 15 that encodes a polypeptide having an IF1206 biological activity (the biological activities of the IF1206 are described below), expressing the encoded portion of IF1206 (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of IF1206. Immunological activity refers to the ability to induce the production of an antibody against an antigenic epitope possessed by a native IF1206; biological activity refers to a function, either inhibitory or stimulatory, caused by a native IF1206 that excludes immunological activity.
IF1206 nucleic acid variants and hybridization 1. variant polynucleotides, genes and recombinant genes The invention further encompasses nucleic acid molecules that differ from the nucleotide sequences shown in SEQ ID NOS:1 or 3 due to degeneracy of the genetic code and thus encode the same IF1206 as that encoded by the nucleotide sequences shown in SEQ ID NO NOS: 1 or 3. An isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NOS:2, 4or15.
In addition to the IFI206 sequences shown in SEQ ID NOS:2, 4 or 15, DNA sequence polymorphisms that change the amino acid sequences of the IF1206 may exist within a population. For example, allelic variation among individuals will exhibit genetic polymorphism in IFI206. The terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame (ORF) encoding IF1206, preferably a vertebrate IF1206. Such natural allelic variations can typically result in 1-5% variance in IFI206.
Any and all such nucleotide variations and resulting amino acid polymorphisms in the IF1206, which are the result of natural allelic variation and that do not alter the functional activity of the IF1206 are within the scope of the invention.
Moreover, IFI206 from other species that have a nucleotide sequence that differs from the human sequence of SEQ ID NOS:1 or 3, are contemplated. Nucleic acid molecules corresponding to natural allelic variants and homologues of the IFI206 cDNAs of the invention can be isolated based on their homology to the IFI206 of SEQ ID NOS:1 or 3 using cDNA-derived probes to hybridize to homologous IFI206 sequences under stringent conditions.

"1F1206 variant polynucleotide" or "1F1206 variant nucleic acid sequence" means a nucleic acid molecule which encodes an active IF1206 that (1) has at least about 80% nucleic acid sequence identity with a nucleotide acid sequence encoding a full-length native IF1206, (2) a full-length native IF1206 lacking the signal peptide, (3) an extracellular domain of an IF1206, with or without the signal peptide, or (4) any other fragment of a full-length IF1206. Ordinarily, an IF1206 variant polynucleotide will have at least about 80% nucleic acid sequence identity, more preferably at least about 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% nucleic acid sequence identity and yet more preferably at least about 99% nucleic acid sequence identity with the nucleic acid sequence encoding a full-length native IF1206. An IF1206 variant polynucleotide may encode full-length native IF1206 lacking the signal peptide, an extracellular domain of an IF1206, with or without the signal sequence, or any other fragment of a full-length IF1206. Variants do not encompass the native nucleotide sequence.
Ordinarily, IF1206 variant polynucleotides are at least about 30 nucleotides in length, often at least about 60, 90, 120, 150, 180, 210, 240, 270, 300, 450, 600 nucleotides in length, more often at least about 900 nucleotides in length, or more.
"Percent (%) nucleic acid sequence identity" with respect to IF1206-encoding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in the IFI206 sequence of interest, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining % nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.

When nucleotide sequences are aligned, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) can be calculated as follows:
%nucleic acid sequence identity - W~Z ~ 100 where W is the number of nucleotides cored as identical matches by the sequence alignment program's or algorithm's alignment of C and D
and Z is the total number of nucleotides in D.
When the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D
will not equal the % nucleic acid sequence identity of D to C.
2. Stringency Homologs (i.e., nucleic acids encoding IF1206 derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning.
The specificity of single stranded DNA to hybridize complementary fragments is determined by the "stringency" of the reaction conditions.
Hybridization stringency increases as the propensity to form DNA duplexes decreases. In nucleic acid hybridization reactions, the stringency can be chosen to either favor specific hybridizations (high stringency), which can be used to identify, for example, full-length clones from a library. Less-specific hybridizations (low stringency) can be used to identify related, but not exact, DNA molecules (homologous, but not identical) or segments.
DNA duplexes are stabilized by: (1 ) the number of complementary base pairs, (2) the type of base pairs, (3) salt concentration (ionic strength) of the reaction mixture, (4) the temperature of the reaction, and (5) the presence of certain organic solvents, such as formamide which decreases DNA duplex stability. In general, the longer the probe, the higher the temperature required for proper annealing. A common approach is to vary the temperature: higher relative temperatures result in more stringent reaction conditions. (Ausubel et al., 1987) provide an excellent explanation of stringency of hybridization reactions.
To hybridize under "stringent conditions" describes hybridization protocols in which nucleotide sequences at least 60% homologous to each other remain hybridized. Generally, stringent conditions are selected to be about 5°C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium.
(a) high stringency "Stringent hybridization conditions" conditions enable a probe, primer or oligonucleotide to hybridize only to its target sequence. Stringent conditions are sequence-dependent and will differ. Stringent conditions comprise: (1 ) low ionic strength and high temperature washes (e.g. 15 mM
sodium chloride, 1.5 mM sodium citrate, 0.1 % sodium dodecyl sulfate at 50°C); (2) a denaturing agent during hybridization (e.g. 50% (v/v) formamide, 0.1 % bovine serum albumin, 0.1 % Ficoll, 0.1 % polyvinylpyrrolidone, 50mM
sodium phosphate buffer (pH 6.5; 750 mM sodium chloride, 75 mM sodium citrate at 42°C); or (3) 50% formamide. Washes typically also comprise SSC (0.75 M NaCI, 75 mM sodium citrate), 50 mM sodium phosphate (pH
6.8), 0.1 % sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 Ng/ml), 0.1 % SDS, and 10% dextran sulfate at 42°C, with washes at 42°C in 0.2 x SSC (sodium chloride/sodium citrate) and 50%
formamide at 55°C, followed by a high-stringency wash consisting of 0.1 x SSC containing EDTA at 55°C. Preferably, the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99%

homologous to each other typically remain hybridized to each other. These conditions are presented as examples and are not meant to be limiting.
(b) moderate stringency "Moderately stringent conditions" use washing solutions and hybridization conditions that are less stringent (Sambrook, 1989), such that a polynucleotide will hybridize to the entire, fragments, derivatives or analogs of SEQ ID NOS:1 or 3. One example comprises hybridization in 6X SSC, 5X
Denhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA
at 55°C, followed by one or more washes in 1X SSC, 0.1% SDS at 37°C.
The temperature, ionic strength, etc., can be adjusted to accommodate experimental factors such as probe length. Other moderate stringency conditions are described in (Ausubel et al., 1987; Kriegler, 1990).
(c) low stringency "Low stringent conditions" use washing solutions and hybridization conditions that are less stringent than those for moderate stringency (Sambrook, 1989), such that a polynucleotide will hybridize to the entire, fragments, derivatives or analogs of SEQ ID NOS:1 or 3. A non-limiting example of low stringency hybridization conditions are hybridization in 35%
formamide, 5X SSC, 50 mM Tris-HCI (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10%
(wt/vol) dextran sulfate at 40°C, followed by one or more washes in 2X
SSC, mM Tris-HCI (pH 7.4), 5 mM EDTA, and 0.1 % SDS at 50°C. Other conditions of low stringency, such as those for cross-species hybridizations are described in (Ausubel et al., 1987; Kriegler, 1990; Shilo and Weinberg, 25 1981).
3. Conservative mutations In addition to naturally-occurring allelic variants of IFI206, changes can be introduced by mutation into SEQ ID NO NOS:1 or 3 sequences that incur alterations in the amino acid sequences of the encoded IF1206 that do not alter IF1206 function. For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NOS:2, 4 or 15. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the IF1206 without altering their biological activity, whereas an "essential" amino acid residue is required for such biological activity. For example, amino acid residues that are conserved among the IF1206 of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well-known in the art.
Useful conservative substitutions are shown in Table 6, "Preferred substitutions." Conservative substitutions whereby an amino acid of one class is replaced with another amino acid of the same type fall within the scope of the subject invention so long as the substitution does not materially alter the biological activity of the compound. If such substitutions result in a change in biological activity, then more substantial changes, indicated in Table 7 as exemplary are introduced and the products screened for IF1206 polypeptide biological activity.
Table A Preferred substitutions Original residueExemplary substitutionsPreferred substitutions Ala A Val, Leu, Ile Val Arg (R) Lys, Gln, Asn Lys Asn N) Gln, His, Lys, Arg Gln Asp (D) Glu Glu Cys (C) Ser Ser Gln Q Asn Asn Glu (E) Asp Asp GI G Pro, Ala Ala His (H) Asn, Gln, Lys, Arg Arg Ile (I) Leu, Val, Met, Ala, Leu Phe, Norleucine Leu (L) Norleucine, Ile, Val,Ile Met, Ala, Phe Lys (K) Arg, Gln, Asn Arg Met M Leu, Phe, Ile Leu Phe (F) Leu, Val, Ile, Ala, Leu Tyr Pro P Ala Ala Ser(S) Thr Thr Thr (T) Ser Ser Tr W T r, Phe T r Tyr (Y) Trp, Phe, Thr, Ser Phe Val (V) Ile, Leu, Met, Phe, Leu Ala, Norleucine Non-conservative substitutions that effect (1 ) the structure of the polypeptide backbone, such as a ~i-sheet or a-helical conformation, (2) the charge or (3) hydrophobicity, or (4) the bulk of the side chain of the target site can modify IF1206 polypeptide function or immunological identity. Residues are divided into groups based on common side-chain properties as denoted in Table B. Non-conservative substitutions entail exchanging a member of one of these classes for another class. Substitutions may be introduced into conservative substitution sites or more preferably into non-conserved sites.
Table B Amino acid classes Class Amino acids hydro hobic Norleucine, Met, Ala, Val, Leu, Ile neutral h drophilic Cys, Ser, Thr acidic As , Glu basic Asn, Gln, His, Lys, Arg disru t chain conformationGI , Pro aromatic Tr , Tyr, Phe The variant polypeptides can be made using methods known in the art such as oligonucleotide-mediated (site-directed) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter, 1986;
Zoller and Smith, 1987), cassette mutagenesis, restriction selection mutagenesis (Wells et al., 1985) or other known techniques can be performed on the cloned DNA to produce the IF1206 variant DNA (Ausubel et al., 1987;
Sambrook, 1989).
In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 45%, preferably 60%, more preferably 70%, 80%, 90%, and most preferably about 95% homologous to SEQ ID
NOS:2, 4 or 15.

A mutant IF1206 can be assayed for blocking adipocyte differentiation in vitro.
4. Anti-sense nucleic acids Using antisense and sense IF1206 oligonucleotides cari prevent IF1206 polypeptide expression. These oligonucleotides bind to target nucleic acid sequences, forming duplexes that block transcription or translation of the target sequence by enhancing degradation of the duplexes, terminating prematurely transcription or translation, or by other means.
Antisense or sense oligonucleotides are singe-stranded nucleic acids, either RNA or DNA, which can bind target IFI206 mRNA (sense) or IFI206 DNA (antisense) sequences. Anti-sense nucleic acids can be designed according to Watson and Crick or Hoogsteen base pairing rules. The anti-sense nucleic acid molecule can be complementary to the entire coding region of IFI206 mRNA, but more preferably, to only a portion of the coding or noncoding region of IFI206 mRNA. For example, the anti-sense oligonucleotide can be complementary to the region surrounding the translation start site of IFI206 mRNA. Antisense or sense oligonucleotides may comprise a fragment of the IF1206 DNA coding region of at least about 14 nucleotides, preferably from about 14 to 30 nucleotides. In general, antisense RNA or DNA molecules can comprise at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 bases in length or more.
Among others, (Stein and Cohen, 1988; van der Krol et al., 1988a) describe methods to derive antisense or a sense oligonucleotides from a given cDNA
sequence.
Examples of modified nucleotides that can be used to generate the anti-sense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the anti-sense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been sub-cloned in an anti-sense orientation such that the transcribed RNA will be complementary to a target nucleic acid of interest.
To introduce antisense or sense oligonucleotides into target cells (cells containing the target nucleic acid sequence), any gene transfer method may be used. Examples of gene transfer methods include (1 ) biological, such as gene transfer vectors like Epstein-Ban- virus or conjugating the exogenous DNA to a ligand-binding molecule, (2) physical, such as electroporation and injection, and (3) chemical, such as CaPOa precipitation and oligonucleotide-lipid complexes.
An antisense or sense oligonucleotide is inserted into a suitable gene transfer retroviral vector. A cell containing the target nucleic acid sequence is '20 contacted with the recombinant retroviral vector, either in vivo or ex vivo.
Examples of suitable retroviral vectors include those derived from the murine retrovirus M-MuLV, N2 (a retrovirus derived from M-MuLV), or the double copy vectors designated DCTSA, DCTSB and DCTSC (WO 90/13641, 1990).
To achieve sufficient nucleic acid molecule transcription, vector constructs in which the transcription of the anti-sense nucleic acid molecule is controlled by a strong pol II or pol III promoter are preferred.
To specify target cells in a mixed population of cells cell surface receptors that are specific to the target cells can be exploited. Antisense and sense oligonucleotides are conjugated to a ligand-binding molecule, as described in (WO 91 /04753, 1991 ). Ligands are chosen for receptors that are specific to the target cells. Examples of suitable ligand-binding molecules include cell surface receptors, growth factors, cytokines, or other ligands that bind to cell surtace receptors or molecules. Preferably, conjugation of the ligand-binding molecule does not substantially interfere with the ability of the receptors or molecule to bind the ligand-binding molecule conjugate, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell.
Liposomes efficiently transfer sense or an antisense oligonucleotide to cells (WO 90/10448, 1990). The sense or antisense oligonucleotide-lipid complex is preferably dissociated within the cell by an endogenous lipase.
The anti-sense nucleic acid molecule of the invention may be an x-anomeric nucleic acid molecule. An a-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual a-units, the strands run parallel to each other (Gautier et al., 1987). The anti-sense nucleic acid molecule can also comprise a 2'-0-methylribonucleotide (Inoue et al., 1987x) or a chimeric RNA-DNA analogue (Inoue et al., 1987b).
In one embodiment, an anti-sense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes, such as hammerhead ribozymes (Haseloff and Gerlach, 1988) can be used to catalytically cleave IFI206 mRNA transcripts and thus inhibit translation. A ' ribozyme specific for an IF1206-encoding nucleic acid can be designed based on the nucleotide sequence of an IFI206 cDNA (i.e., SEQ ID NOS:1 or 3). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an IF1206-encoding mRNA (Cech et al., U.S. Patent No. 5,116,742, 1992; Cech et al., U.S. Patent No. 4,987,071, 1991 ). 1F1206 mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules (Bartel and Szostak, 1993).
Alternatively, IFI206 expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the IFI206 (e.g., the IF1206 promoter and/or enhancers) to form triple helical structures that prevent transcription of the IFI206 in target cells (Helene, 1991; Helene et al., 1992; Maher, 1992).
Modifications of antisense and sense oligonucleotides can augment their effectiveness. Modified sugar-phosphodiester bonds or other sugar linkages (WO 91/06629, 1991), increase in vivo stability by conferring resistance to endogenous nucleases without disrupting binding specificity to target sequences. Other modifications can increase the affinities of the oligonucleotides for their targets, such as covalently linked organic moieties (WO 90/10448, 1990) or poly-(L)-lysine. Other attachments modify binding specificities of the oligonucleotides for their targets, including metal complexes 'or intercalating (e.g. ellipticine) and alkylating agents.
For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids (Hyrup and Nielsen, 1996).
"Peptide nucleic acids" or "PNAs" refer to nucleic acid mimics (e.g., DNA
mimics) in that the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained.
The neutral backbone of PNAs allows for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols (Hyrup and Nielsen, 1996; Perry-O'Keefe et al., 1996).
PNAs of IF1206 can be used in therapeutic and diagnostic applications.
For example, PNAs can be used as anti-sense or antigene agents for sequence-specific modulation of gene expression by inducing transcription or~
translation arrest or inhibiting replication. 1F1206 PNAs may also be used in the analysis of single base pair mutations (e.g., PNA directed PCR clamping;
as artificial restriction enzymes when used in combination with other enzymes, e.g., S, nucleases (Hyrup and Nielsen, 1996); or as probes or primers for DNA sequence and hybridization (Hyrup and Nielsen, 1996; Perry-O'Keefe et al., 1996).
PNAs of IF1206 can be modified to enhance their stability or cellular uptake. Lipophilic or other helper groups may be attached to PNAs, PNA-DNA dimmers formed, or the use of liposomes or other drug delivery techniques. For example, PNA-DNA chimeras can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerises) to interact with the DNA portion while the PNA portion provides high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds befinreen the nucleobases, and orientation (Hyrup and Nielsen, 1996). The synthesis of PNA-DNA chimeras can be performed (Finn et al., 1996; Hyrup and Nielsen, 1996). For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5' end of DNA (Finn et al., 1996; Hyrup and Nielsen, 1996). PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment (Finn et al., 1996). Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment (Petersen et al., 1976).
The oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (Lemaitre et al., 1987; Letsinger et al., 1989) or PCT Publication No. W088/09810) or the blood-brain barrier (e.g., PCT Publication No. WO 89/10134). In addition, oligonucleotides can be modified with hybridization-triggered cleavage agents (van der Krol et al., 1988b) or intercalating agents (Zon, 1988). The oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like.
1F1206 polypeptides One aspect of the invention pertains to isolated IF1206, and biologically-active portions derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-IF1206 Abs. In one embodiment, native IF1206 can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, IF1206 are produced by recombinant DNA techniques. Alternative to recombinant expression, an IF1206 or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
1. Polypeptides An IF1206 polypeptide includes the amino acid sequence of IF1206 whose sequences are provided in SEQ ID NOS:2, 4 or 15. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residues shown in SEQ ID NOS:2, 4 or 15, while still encoding a protein that maintains its IF1206 activities and physiological functions, or a functional fragment thereof.
2. Variant IF1206 polypeptides In general, an IF1206 variant that preserves IFI206-like function and includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further includes the possibility of inserting an additional residue or residues between finro residues of the parent protein as well as the possibility of deleting one or more residues from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above.
"1F1206 polypeptide variant" means an active IF1206 polypeptide having at least: (1 ) about 80% amino acid sequence identity with a full-length native sequence IF1206 polypeptide sequence, (2) a IF1206 polypeptide sequence lacking the signal peptide, (3) an extracellular domain of a IF1206 polypeptide, with or without the signal peptide, or (4) any other fragment of a full-length 1F1206 polypeptide sequence. For example, IF1206 polypeptide variants include IF1206 polypeptides wherein one or more amino acid residues are added or deleted at the N- or C- terminus of the full-length native amino acid sequence. A IF1206 polypeptide variant will have at least about 80% amino acid sequence identity, preferably at least about 81 % amino acid sequence identity, more preferably at least about 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% amino aCld sequence identity and most preferably at least about 99% amino acid sequence identity with a full-length native sequence IF1206 polypeptide sequence. A IF1206 polypeptide variant may have a sequence lacking the signal peptide, an extracellular domain of a IF1206 polypeptide, with or without the signal peptide, or any other fragment of a full-length IF1206 polypeptide sequence. Ordinarily, IF1206 variant polypeptides are at least about 10 amino acids in length, often at least about 20 amino acids in length, more often at least about 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, or 300 amino acids in length, or more.
"Percent (%) amino acid sequence identity" is defined as the percentage of amino acid residues that are identical with amino acid residues in the disclosed IF1206 polypeptide sequence in a candidate sequence when the two sequences are aligned. To determine % amino acid identity, sequences are aligned and if necessary, gaps are introduced to achieve the maximum % sequence identity; conservative substitutions are not considered as part of the sequence identity. Amino acid sequence alignment procedures to determine percent identity are well known to those of skill in the art.
Often publicly available computer software such as BLAST, BLAST2, ALIGN2 or Megalign (DNASTAR) software is used to align peptide sequences. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
When amino acid sequences are aligned, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) can be calculated as:
%amino acid sequence identity - ~ ~ 100 where X is the number of amino acid residues scored as identical matches by the sequence alignment program's or algorithm's alignment of A and B
and Y is the total number of amino acid residues in B.
If the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A.
3. Isolatedlpurified polypeptides An "isolated" or "purified" polypeptide, protein or biologically active fragment is separated and/or recovered from a component of its natural environment. Contaminant components include materials that would typically interfere with diagnostic or therapeutic uses for the polypeptide, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous materials. Preferably, the polypeptide is purified to a sufficient degree to obtain at least 15 residues of N-terminal or internal amino acid sequence. To be substantially isolated, preparations having less than 30% by dry weight of non-IF1206 contaminating material (contaminants), more preferably less than 20%, 10% and most preferably less than 5% contaminants. An isolated, recombinantly-produced IF1206 or biologically active portion is preferably substantially free of culture medium, i.e., culture medium represents less than 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the IF1206 preparation. Examples of contaminants include cell debris, culture media, and substances used and produced during in vitro synthesis of IF1206.
4. Biologically active Biologically active portions of IF1206 include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the IF1206 (SEQ ID NOS:2 or 4) that include fewer amino acids than the full-length IF1206, and exhibit at least one activity of an IF1206.
Biologically active portions comprise a domain or motif with at least one activity of native IF1206. A biologically active portion of an IFI206 can be a polypeptide that is, for example, 10, 25, 50, 100 or more amino acid residues in length. Other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native IF1206.
Biologically active portions of IF1206 may have an amino acid sequence shown in SEQ ID NOS:2 or4, or substantially homologous to SEQ
ID NOS:2 or 4, and retains the functional activity of the protein of SEQ ID
NOS:2 or 4, yet differs in amino acid sequence due to natural allelic variation or mutagenesis. Other biologically active IF1206 may comprise an amino acid sequence at least 45% homologous to the amino acid sequence of SEQ ID
NOS:2 or 4, and retains the functional activity of native IF1206.
5. Determining homology between two or more sequences "1F1206 variant" means an active IF1206 having at least: (1 ) about 80%
amino acid sequence identity with a full-length native sequence IF1206 sequence, (2) an IF1206 sequence lacking the signal peptide, (3) an extracellular domain of an IF1206, with or without the signal peptide, or (4) any other fragment of a full-length IF1206 sequence. For example, IF1206 variants include IF1206 wherein one or more amino acid residues are added or deleted at the N- or C- terminus of the full-length native amino acid sequence. An IF1206 variant will have at least about 80% amino acid sequence identity, preferably at least about 81 % amino acid sequence identity, more preferably at least about 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98% amino acid sequence identity and most preferably at least about 99% amino acid sequence identity with a full-length native sequence IF1206 sequence. An IF1206 variant may have a sequence lacking the signal peptide, an extracellular domain of an IF1206, with or without the signal peptide, or any other fragment of a full-length IF1206 sequence. Ordinarily, IF1206 variant polypeptides are at least about 10 amino acids in length, often at least about 20 amino acids in length, more often at least about 30, 40, 50, 60, 70, 80, 90, 1.00, 150, 200, or 300 amino acids in length, or more. ' .

"Percent (%) amino acid sequence identity" is defined as the percentage of amino acid residues that are identical with amino acid residues in the disclosed IF1206 sequence in a candidate sequence when the two sequences are aligned. To determine % amino acid identity, sequences are aligned and if necessary, gaps are introduced to achieve the maximum sequence identity; conservative substitutions are not considered as part of the sequence identity. Amino acid sequence alignment procedures to determine percent identity are well known to those of skill in the art. Often publicly available computer software such as BLAST, BLAST2, ALIGN2 or Megalign (DNASTAR) software is used to align peptide sequences. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
When amino acid sequences are aligned, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) can be calculated as:
%amino acid sequence identity - ~ ~ 1 ~0 where X is the number of amino acid residues scored as identical matches by the sequence alignment program's or algorithm's alignment of A and B
and Y is the total number of amino acid residues in B.
If the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A.
6. Chimeric and fusion proteins Fusion polypeptides are useful in expression studies, cell-localization, bioassays, and IF1206 purification. An IF1206 "chimeric protein" or "fusion protein" comprises IF1206 fused to a non-IF1206 polypeptide. A non-IF1206 polypeptide is not substantially homologous to IF1206 (SEQ ID NOS:2 or 4).
An IF1206 fusion protein may include any portion to the entire IF1206, including any number of the biologically active portions. 1F1206 may be fused to the C-terminus of the GST (glutathione S-transferase) sequences. Such fusion proteins facilitate the purification of recombinant IF1206. In certain host cells, (e.g. mammalian), heterologous signal sequences fusions may ameliorate IF1206 expression and/or secretion. Additional exemplary fusions are presented in Table C.
Other fusion partners can adapt IF1206 therapeutically. Fusions with members of the immunoglobulin (1g) protein family are useful in therapies that inhibit IF1206 ligand or substrate interactions, consequently suppressing IF1206-mediated signal transduction in vivo. Such fusions, incorporated into pharmaceutical compositions, may be used to treat proliferative and differentiation disorders, as well as modulating cell survival. IF1206-Ig fusion polypeptides can also be used as immunogens to produce anti-IF1206 Abs in a subject, to purify IF1206 ligands, and to screen for molecules that inhibit interactions of IF1206 with other molecules.
Fusion proteins can be easily created using recombinant methods. A
nucleic acid encoding IF1206 can be fused in-frame with a non-IF1206 encoding nucleic acid, to the IF1206 NHZ- or COO- -terminus, or internally.
Fusion genes may also be synthesized by conventional techniques, including automated DNA synthesizers. PCR amplification using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (Ausubel et al., 198,7) is also useful. Many vectors are commercially available that facilitate sub-cloning IFI206 in-frame to a fusion moiety.
Table C Useful non-IFI206 fusion polypeptides Reporter in vitro in vivo Notes Reference Human growthRadioimmuno- none Expensive, (Selden et al., hormone assay insensitive,1986) (hGH) narrow linear range.

(3-glucu- Colorimetric,colorimetricsensitive, (Gallagher, ronidase fluorescent, (histo- broad linear1992) , or (GUS) chemi- chemical range, non-luminescent staining iostopic.
with X- luc Green Fluorescent fluorescent can be used(Chalfie et al., fluorescent in live 1994) cells;

protein resists (GFP) photo-and related bleaching molecules (RFP, BFP, IF1206, etc.) Luciferase bioluminsecentBio- protein (de Wet is et al., (firefly) luminescent unstable, 1987) difficult to reproduce, signal is brief ChlorampheniChromato- none Expensive (Gorman , et coal graphy, radioactiveal., 1982) acetyltransferadifferential substrates, se (CAT) extraction, time-fluorescent, consuming, or immunoassay insensitive, narrow linear range (3-galacto-colorimetric,colorimetricsensitive, (Alam and sidase fluorescence,(histochemicalbroad linearCook, 1990) chemi- staining range; some with luminscence X-gal), bio-cells have luminescent high in live cells endogenous activity Secrete colorimetric,none Chem- (Berger et al., alkaline bioluminescent, iluminscence1988) phosphatase chemi- assay is (SEAP) luminescent sensitive and broad linear range; some cells have endogenouse alkaline phosphatase activity Therapeutic applications of IF1206 1. Agonists and antagonists "Antagonist" includes any molecule that partially or fully blocks, inhibits, or neutralizes a biological activity of endogenous IF1206. Similarly, "agonisY' includes any molecule that mimics a biological activity of endogenous IF1206.
Molecules that can act as agonists or antagonists include Abs or antibody fragments, fragments or variants of endogenous IF1206, peptides, antisense oligonucleotides, small organic molecules, etc.
2. Identifying antagonists and agonists To assay for antagonists, IF1206 is added to, or expressed in, a cell along with the compound to be screened for a particular activity. If the compound inhibits the activity of interest in the presence of the IF1206, that compound is an antagonist to the IF1206; if IF1206 activity is enhanced, the compound is an agonist.
(a) Specific examples of potential antagonists and agonist Any molecule that alters IF1206 cellular effects is a candidate antagonist or agonist. Screening techniques well known to those skilled in the art can identify these molecules. Examples of antagonists and agonists include: (1) small organic and inorganic compounds, (2) small peptides, (3) Abs and derivatives, (4) polypeptides closely related to IF1206, (5) antisense DNA and RNA, (6) ribozymes, (7) triple DNA helices and (8) nucleic acid aptamers.

Small molecules that bind to the IF1206 active site or other relevant part of the polypeptide and inhibit the biological activity of the IF1206 are antagonists. Examples of small molecule antagonists include small peptides, peptide-like molecules, preferably soluble, and synthetic non-peptidyl organic or inorganic compounds. These same molecules, if they enhance IF1206 activity, are examples of agonists.
Almost any antibody that affects IF1206's function is a candidate antagonist, and occasionally, agonist. Examples of antibody antagonists include polyclonal, monoclonal, single-chain, anti-idiotypic, chimeric Abs, or humanized versions of such Abs or fragments. Abs may be from any species in which an immune response can be raised. Humanized Abs are also contemplated.
Alternatively, a potential antagonist or agonist may be a closely related protein, for example, a mutated form of the IF1206 that recognizes an IF1206-interacting protein but imparts no effect, thereby competitively inhibiting action. Alternatively, a mutated IF1206 may be constitutively activated and may act as an agonist.
Antisense RNA or DNA constructs can be effective antagonists.
Antisense RNA or DNA molecules block function by inhibiting translation by hybridizing to targeted mRNA. Antisense technology can be used to control gene expression through triple-helix formation or antisense DNA or RNA, both of which depend on polynucleotide binding to DNA or RNA. For example, the 5' coding portion of the IFI206 sequence is used to design an antisense RNA
oligonucleotide of from about 10 to 40 base pairs in length. A DNA
oligonucleotide is designed to be complementary to a region of the gene involved in transcription (triple helix) (Beat and Dervan, 1991; Cooney et al., 1988; Lee et al., 1979), thereby preventing transcription and the production of the IF1206. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into the IF1206 (antisense) (Cohen, 1989; Okano et al., 1991 ). These oligonucleotides can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of the IF1206. When antisense DNA is used, oligodeoxyribonucleotides derived from the translation-initiation site, e.g., between about -10 and +10 positions of the target gene nucleotide sequence, are preferred.
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA. Ribozymes act by sequence-specific hybridization to the complementary target RNA, followed by endonucleolytic cleavage.
Specific ribozyme cleavage sites within a potential RNA target can be identified by known techniques (WO 97/33551, 1997; Rossi, 1994).
To inhibit transcription, triple-helix nucleic acids that are single-stranded and comprise deoxynucleotides are useful antagonists. These oligonucleotides are designed such that triple-helix formation via Hoogsteen base-pairing rules is promoted, generally requiring stretches of purines or pyrimidines (WO 97/33551, 1997).
Because an IF1206 activity may include nucleic acid binding, such as BAT mRNA, molecules that compete for IF1206 nucleic acid binding sites) can be effective intracellular competitors. Aptamers are short oligonucleotide sequences that can be used to recognize and specifically bind almost any molecule. The systematic evolution of ligands by exponential enrichment (SELEX) process (Ausubel et al., 1987; Ellington and Szostak, 1990; Tuerk and Gold, 1990) is powerful and can be used to find such aptamers.
Aptamers have many diagnostic and clinical uses; almost any use in which an antibody has been used clinically or diagnostically, aptamers too may be used. In addition, are cheaper to make once they have been identified, and can be easily applied in a variety of formats, including administration in pharmaceutical compositions, in bioassays, and diagnostic tests (Jayasena, 1999).
Anti-IF1206 Abs The invention encompasses Abs and antibody fragments, such as Fab or (Fab)2, that bind immunospecifically to any IF1206 epitopes.
"Antibody" (Ab) comprises single Abs directed against IF1206 (anti-IF1206 Ab; including agonist, antagonist, and neutralizing Abs), anti-IF1206 Ab compositions with poly-epitope specificity, single chain anti-IF1206 Abs, and fragments of anti-IF1206 Abs. A "monoclonal antibody" is obtained from a population of substantially homogeneous Abs, i.e., the individual Abs comprising the population are identical except for possible naturally-occurring mutations that may be present in minor amounts. Exemplary Abs include polyclonal (pAb), monoclonal (mAb), humanized, bi-specific (bsAb), and heteroconjugate Abs.
1. Polyclonal Abs (pAbs) Polyclonal Abs can be raised in a mammalian host, for example, by one or more injections of an immunogen and, if desired, an adjuvant.
Typically, the immunogen and/or adjuvant are injected in the mammal by multiple subcutaneous or intraperitoneal injections. The immunogen may include IF1206 or a fusion protein. Examples of adjuvants include Freund's complete and monophosphoryl Lipid A synthetic-trehalose dicorynomycolate (MPL-TDM). To improve the immune response, an immunogen may be conjugated to a protein that is immunogenic in the host, such as keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. Protocols for antibody production are described by (Ausubel et al., 1987; Harlow and Lane, 1988). Alternatively, pAbs may be made in chickens, producing IgY molecules (Schade et al., 1996).
2. Monoclonal Abs (mAbs) Anti-IF1206 mAbs may be prepared using hybridoma methods (Milstein and Cuello, 1983). Hybridoma methods comprise at least four steps: (1 ) immunizing a host, or lymphocytes from a host; (2) harvesting the mAb secreting (or potentially secreting) lymphocytes, (3) fusing the lymphocytes to immortalized cells, and (4) selecting those cells that secrete the desired (anti-IF1206) mAb.
A mouse, rat, guinea pig, hamster, or other appropriate host is immunized to elicit lymphocytes that produce or are capable of producing Abs that will specifically bind to the immunogen. Alternatively, the lymphocytes may be immunized in vitro. If human cells are desired, peripheral blood lymphocytes (PBLs) are generally used; however, spleen cells or lymphocytes from other mammalian sources are preferred. The immunogen typically includes IF1206 or a fusion protein.
The lymphocytes are then fused with an immortalized cell line to form hybridoma cells, facilitated by a fusing agent such as polyethylene glycol (coding, 1996). Rodent, bovine, or human myeloma cells immortalized by transformation may be used, or rat or mouse myeloma cell lines. Because pure populations of hybridoma cells and not unfused immortalized cells are preferred, the cells after fusion are grown in a suitable medium that contains one or more substances that inhibit the growth or survival of unfused, immortalized cells. A common technique uses parental cells that lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT). In this case, hypoxanthine, aminopterin and thymidine are added to ' the medium (HAT medium) to prevent the growth of HGPRT-deficient cells while permitting hybridomas to grow.
Preferred immortalized cells fuse efficiently, can be isolated from mixed populations by selecting in a medium such as HAT, and support stable and high-level expression of antibody after fusion. Preferred immortalized cell lines are murine myeloma lines, available from the American Type Culture Collection (Manassas, VA). Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human mAbs (Kozbor et al., 1984; Schook, 1987).
Because hybridoma cells secrete antibody extracellularly, the culture media can be assayed for the presence of mAbs directed against IF1206 (anti-IF1206 mAbs). Immunoprecipitation or in vitro binding assays, such as radio immunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA), measure the binding specificity of mAbs (Harlow and Lane, 1988; Harlow and Lane, 1999), including Scatchard analysis (Munson and Rodbard, 1980).
Anti-IF1206 mAb secreting hybridoma cells may be isolated as single clones by limiting dilution procedures and sub-cultured (coding, 1996).
Suitable culture media include Dulbecco's Modified Eagle's Medium, RPMI-1640, or if desired, a protein-free or -reduced or serum-free medium (e.g., Ultra DOMA PF or HL-1; Biowhittaker; Walkersville, MD). The hybridoma cells may also be grown in vivo as ascites.
The mAbs may be isolated or purified from the culture medium or ascites fluid by conventional Ig purification procedures such as protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, ammonium sulfate precipitation or affinity chromatography (Harlow and Lane, 1988; Harlow and Lane, 1999).
The mAbs may also be made by recombinant methods (U.S. Patent No. 4166452, 1979). DNA encoding anti-IF1206 mAbs can be readily isolated and sequenced using conventional procedures, e.g., using oligonucleotide probes that specifically bind to murine heavy and light antibody chain genes, to probe preferably DNA isolated from anti-IF1206-secreting mAb hybridoma cell lines. Once isolated, the isolated DNA fragments are sub-cloned into expression vectors that are then transfected into host cells such as simian COS-7 cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce Ig protein, to express mAbs. The isolated DNA
fragments can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4816567, 1989; Morrison et al., 1987), or by fusing the Ig coding sequence to all or part of the coding sequence for a non-Ig polypeptide. Such a non-Ig polypeptide can be substituted for the constant domains of an antibody, or can be substituted for the variable domains of one antigen-combining site to create a chimeric bivalent antibody.
3. Monovalent Abs The Abs may be monovalent Abs that consequently do not cross-link with each other. For example, one method involves recombinant expression of Ig light chain and modified heavy chain. Heavy chain truncations generally at any point in the F~ region will prevent heavy chain cross-linking.
Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted, preventing crosslinking. In vitro methods are also suitable for preparing monovalent Abs. Abs can be digested to produce fragments, such as Fab fragments (Harlow and Lane, 1988; Harlow and Lane, 1999).
4. Humanized and human Abs Anti-IF1206 Abs may further comprise humanized or human Abs.
Humanized forms of non-human Abs are chimeric Igs, Ig chains or fragments (such as F", Fab, Fab', F~ab~~z or other antigen-binding subsequences of Abs) that contain minimal sequence derived from non-human Ig.
Generally, a humanized antibody has one or more amino acid residues introduced from a non-human source. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. Humanization is accomplished by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody (Jones et al., 1986; Riechmann et al., 1988; Verhoeyen et al., 1988). Such "humanized" Abs are chimeric Abs (U.S. Patent No. 4816567, 1989), wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
In practice, humanized Abs are typically human Abs in which some CDR
residues and possibly some FR residues are substituted by residues from analogous sites in rodent Abs. Humanized Abs include human Igs (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit, having the desired specificity, affinity and capacity. In some instances, corresponding non-human residues replace F" framework residues of the human Ig. Humanized Abs may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody comprises substantially all of at least one, and typically two, variable domains, in which most if not all of the CDR regions correspond to those of a non-human Ig and most if not all of the FR regions are those of a human Ig consensus sequence. The humanized antibody optimally also comprises at least a portion of an Ig constant region (F~), typically that of a human Ig (Jones et al., 1986; Presta, 1992; Riechmann et al., 1988).

Human Abs can also be produced using various techniques, including phage display libraries (Hoogenboom et al., 1991; Marks et al., 1991) and the preparation of human mAbs (Boerner et al., 1991; Reisfeld and Sell, 1985).
Similarly, introducing human Ig genes into transgenic animals in which the endogenous Ig genes have been partially or completely inactivated can be exploited to synthesize human Abs. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire (U.S. Patent No. 5545807, 1996; U.S. Patent No. 5545806, 1996; U.S. Patent No. 5569825, 1996; U.S. Patent No. 5633425, 1997; U.S. Patent No.
5661016, 1997; U.S. Patent No. 5625126, 1997; Fishwild et al., 1996;
Lonberg and Huszar, 1995; Lonberg et al., 1994; Marks et al., 1992).
5. Bi-specific mAbs Bi-specific Abs are monoclonal, preferably human or humanized, that have binding specificities for at least two different antigens. For example, a binding specificity is IF1206; the other is for any antigen of choice, preferably a cell-surface protein or receptor or receptor subunit.
Traditionally, the recombinant production of bi-specific Abs is based on the co-expression of two Ig heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, 1983). Because of the random assortment of Ig heavy and light chains, the resulting hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the desired bi-specific structure. The desired antibody can be purified using affinity chromatography or other techniques (WO 93/08829, 1993; Traunecker et al., 1991 ).
To manufacture a bi-specific antibody (Suresh et al., 1986), variable domains with the desired antibody-antigen combining sites are fused to Ig constant domain sequences. The fusion is preferably with an Ig heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. Preferably, the first heavy-chain constant region (CH1) containing the site necessary for light-chain binding is in at least one of the fusions.
DNAs encoding the Ig heavy-chain fusions and, if desired, the Ig light chain, are inserted into separate expression vectors and are co-transfected into a suitable host organism.
The interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers that are recovered from ~ recombinant cell culture (WO 96/27011, 1996). The preferred interface comprises at least part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the intertace of the first antibody molecule are replaced with larger side chains (e.g.
tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chains) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This mechanism increases the yield of the heterodimer over unwanted end products such as homodimers.
Bi-specific Abs can be prepared as full length Abs or antibody fragments (e.g. F~ab~~2 bi-specific Abs). One technique to generate bi-specific Abs exploits chemical linkage. Intact Abs can be proteolytically cleaved to generate F~ab')z fragments (Brennan et al., 1985). Fragments are reduced with a dithiol complexing agent, such as sodium arsenite, to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The generated Fab fragments are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab~-TNB derivatives is then reconverted to the Fab~-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bi-specific antibody. The produced bi-specific Abs can be used as agents for the selective immobilization of enzymes.
Fab~ fragments may be directly recovered from E. coli and chemically coupled to form bi-specific Abs. For example, fully humanized bi-specific F(ab')2 Abs can be produced (Shalaby et al., 1992). Each Fab~ fragment is separately secreted from E. coli and directly coupled chemically in vitro, forming the bi-specific antibody.
Various techniques for making and isolating bi-specific antibody fragments directly from recombinant cell culture have also been described.
For example, leucine zipper motifs can be exploited (Kostelny et al., 1992).

Peptides from the Fos and Jun proteins are linked to the Fab~ portions of two different Abs by gene fusion. The antibody homodimers are reduced at the hinge region to form monomers and then re-oxidized to form antibody heterodimers. This method can also produce antibody homodimers. The "diabody" technology (Holliger et al., 1993) provides an alternative method to generate bi-specific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (V~) by a linker that is too short to allow pairing between the two domains on the same chain. The VH and V~ domains of one fragment are forced to pair with the complementary V~ and VH domains of another fragment, forming two antigen-binding sites. Another strategy for making bi-specific antibody fragments is the use of single-chain F~ (sF") dimers (Gruber et al., 1994).
Abs with more than two valencies are also contemplated, such as tri-specific Abs (Tutt et al., 1991 ).
Exemplary bi-specific Abs may bind to two different epitopes on a given IF1206. Alternatively, cellular defense mechanisms can be restricted to a particular cell expressing the particular IF1206: an anti-IF1206 arm may be .
combined with an arm that binds to a leukocyte triggering molecule, such as a T-cell receptor molecule (e.g. CD2, CD3, CD28, or B7), or to F~ receptors for IgG (F~yR), such as F~yRI (CD64), F~yRll (CD32) and F~yRlll (CD16). Bi-specific Abs may also be used to target cytotoxic agents to cells that express a particular IF1206. These Abs possess an IF1206-binding arm and an arm that binds a cytotoxic agent or a radionuclide chelator.
6. Heteroconjugate Abs Heteroconjugate Abs, consisting of two covalently joined Abs, have been proposed to target immune system cells to unwanted cells (4,676,980, 1987) and for treatment of human immunodeficiency virus (HIV) infection (WO
91 /00360, 1991; WO 92/20373, 1992). Abs prepared in vitro using synthetic protein chemistry methods, including those involving cross-linking agents, are contemplated. For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents include iminothiolate and methyl-4-mercaptobutyrimidate (4,676,980, 1987).
7. Immunoconjugates Immunoconjugates may comprise an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin or fragment of bacterial, fungal, plant, or animal origin), or a radioactive isotope (i.e., a radioconjugate).
Useful enzymatically-active toxins and fragments include Diphtheria A
chain, non-binding active fragments of Diphtheria toxin, exotoxin A chain from' Pseudomonas aeruginosa, ricin A chain, abrin A chain, modeccin A chain, a-sarcin, Aleurites fordii proteins, Dianthin proteins, Phytolaca americana proteins, Momordica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated Abs, such as 2'2 Bi, '3' I, '3' In, 9~Y, and '$6Re.
Conjugates of the antibody and cytotoxic agent are made using a variety of bi-functional protein-coupling agents, such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bi-functional derivatives of imidoesters (such as dimethyl adipimidate HCI), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6- diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared (Vitetta et al., 1987). '4C-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugating radionuclide to antibody (WO
94/11026, 1994).
In another embodiment, the antibody may be conjugated to a "receptor"
(such as streptavidin) for utilization in tumor pre-targeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a streptavidin "ligand" (e.g., biotin) that is conjugated to a cytotoxic agent (e.g., a radionuclide).
8. ~ffector function engineering The antibody can be modified to enhance its effectiveness in treating a disease, such as cancer. For example, cysteine residues) may be introduced into the F~ region, thereby allowing interchain disulfide bond formation in this region. Such homodimeric Abs may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC) (Caron et al., 1992; Shopes, 1992). Homodimeric Abs with enhanced anti-tumor activity can be prepared using hetero-bifunctional cross-linkers (Wolff et al., 1993). Alternatively, an antibody engineered with dual F~ regions may have enhanced complement lysis (Stevenson et al., 1989).
9. Immunoliposomes Liposomes containing the antibody may also be formulated (U.S.
Patent No. 4485045, 1984; U.S. Patent No. 4544545, 1985; U.S. Patent No.
5013556, 1991; Eppstein et al., 1985; Hwang et al., 1980). Useful liposomes can be generated by a reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG- PE). Such preparations are extruded through filters of defined pore size to yield liposomes with a desired diameter. Fab~ fragments of the antibody can be conjugated to the liposomes (Martin and Papahadjopoulos, 1982) via a disulfide-interchange reaction. A
chemotherapeutic agent, such as Doxorubicin, may also be contained in the liposome (Gabizon et al., 1989). Other useful liposomes with different compositions are contemplated.
10. Diagnostic applications of Abs directed against IFI206 Anti-IF1206 Abs can be used to localize and/or quantitate IF1206 (e.g., for use in measuring levels of IF1206 within tissue samples or for use in diagnostic methods, etc.). Anti-IF1206 epitope Abs can be utilized as pharmacologically-active compounds.

Anti-IF1206 Abs can be used to isolate IF1206 by standard techniques, such as immunoaffinity chromatography or immunoprecipitation. These approaches facilitate purifying endogenous IF1206 antigen-containing polypeptides from cells and tissues. These approaches, as well as others, can be used to detect IF1206 in a sample to evaluate the abundance and pattern of expression of the antigenic protein. Anti-IF1206 Abs can be used to monitor protein levels in tissues as part of a clinical testing procedure; for example, to determine the efficacy of a given treatment regimen. Coupling the antibody to a detectable substance (label) allows detection of Ab-antigen complexes. Classes of labels include fluorescent, luminescent, bioluminescent, and radioactive materials, enzymes and prosthetic groups.
Useful labels include horseradish peroxidase, alkaline phosphatase, (3-galactosidase, acetylcholinesterase, streptavidin/biotin, avidin/biotin, umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride, phycoerythrin, luminol, luciferase, luciferin, aequorin, and'25I, '3' I, ssS or 3H.
11. Antibody therapeutics .
Abs of the invention, including polyclonal, monoclonal, humanized and fully human Abs, can be used therapeutically. Such agents will generally be employed to treat or prevent a disease or pathology in a subject. An antibody preparation, preferably one having high antigen specificity and affinity generally mediates an effect by binding the target epitope(s). Generally, administration of such Abs may mediate one of two effects: (1) the antibody may prevent ligand binding, eliminating endogenous ligand binding and subsequent signal transduction, or (2) the antibody elicits a physiological result by binding an effector site on the target molecule, initiating signal transduction.
A therapeutically effective amount of an antibody relates generally to the amount needed to achieve a therapeutic objective, epitope binding affinity, administration rate, and depletion rate of the antibody from a subject.
Common ranges for therapeutically effective doses may be, as a nonlimiting .
example, from about 0.1 mg/kg body weight to about 50 mg/kg body weight.

Dosing frequencies may range, for example, from twice daily to once a week.
12. Pharmaceutical compositions of Abs Anti-IF1206 Abs, as well as other IF1206 interacting molecules (such as aptamers) identified in other assays, can be administered in pharmaceutical compositions to treat various disorders. Principles and considerations involved in preparing such compositions, as well as guidance in the choice of components can be found in (de Boer, 1994; Gennaro, 2000; Lee, 1990).
Because IF1206 is intracellular, Abs that are internalized are preferred when whole Abs are used as inhibitors. Liposomes may also be used as a delivery vehicle for intracellular introduction. Where antibody fragments are used, the smallest inhibitory fragment that specifically binds to the epitope is preferred. For example, peptide molecules can be designed that bind a preferred epitope based on the variable-region sequences of a useful antibody. Such peptides can be synthesized chemically and/or produced by recombinant DNA technology (Marasco et al., 1993). Formulations may also contain more than one active compound for a particular treatment, preferably those with activities that do not adversely affect each other. The composition may comprise an agent that enhances function, such as a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent.
The active ingredients can also be entrapped in microcapsules prepared by coacervation techniques or by interfacial polymerization; for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions.
The formulations to be used for in vivo administration are highly preferred to be sterile. This is readily accomplished by filtration through sterile filtration membranes or any of a number of techniques.
Sustained-release preparations may also be prepared, such as semi-permeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (Boswell and Scribner, U.S. Patent No.
3,773,919, 1973), copolymers of L-glutamic acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as injectable microspheres composed of lactic acid-glycolic acid copolymer, and poly-D-(-)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods and may be preferred.
IFI206 recombinant expression vectors and host cells Vectors are tools used to shuttle DNA between host cells or as a means to express a nucleotide sequence. Some vectors function only in prokaryotes, while others function in both prokaryotes and eukaryotes, enabling large-scale DNA preparation from prokaryotes for expression in eukaryotes. Inserting the DNA of interest, such as IF1206 nucleotide sequence or a fragment, is accomplished by ligation techniques and/or mating protocols well-known to the skilled artisan. Such DNA is inserted such that its integration does not disrupt any necessary components of the vector. In the case of vectors that are used to express the inserted DNA protein, the introduced DNA is operably-linked to the vector elements that govern its transcription and translation.
Vectors can be divided into two general classes: Cloning vectors are replicating plasmid or phage with regions that are non-essential for propagation in an appropriate host cell, and into which foreign DNA can be inserted; the foreign DNA is replicated and propagated as if it were a component of the vector. An expression vector (such as a plasmid, yeast, or animal virus genome) is used to introduce foreign genetic material into a host cell or tissue in order to transcribe and translate the foreign DNA. In expression vectors, the introduced DNA is operably-linked to elements, such as promoters, that signal to the host cell to transcribe the inserted DNA.

Some promoters are exceptionally useful, such as inducible promoters that control gene transcription in response to specific factors. Operably-linking IFI206 or anti-sense construct to an inducible promoter can control the expression of IFI206 or fragments, or anti-sense constructs. Examples of classic inducible promoters include those that are responsive to a-interferon, heat-shock, heavy metal ions, and steroids such as glucocorticoids (Kaufman, 1990) and tetracycline. Other desirable inducible promoters include those that are not endogenous to the cells in which the construct is being introduced, but, however, is responsive in those cells when the induction agent is exogenously supplied.
Vectors have many difference manifestations. A "plasmid" is a circular double stranded DNA molecule into which additional DNA segments can be introduced. Viral vectors can accept additional DNA segments into the viral genome. Certain vectors are capable of autonomous replication in a host cell (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. In general, useful expression vectors are often plasmids. However, other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses) are contemplated.
Recombinant expression vectors that comprise IFI206 (or fragments) regulate IFI206 transcription by exploiting one or more host cell-responsive (or that can be manipulated in vitro) regulatory sequences that is operably-linked to IFI206. "Operably-linked" indicates that a nucleotide sequence of interest is linked to regulatory sequences such that expression of the nucleotide sequence is achieved.
Vectors can be introduced in a variety of organisms and/or cells (Table D). Alternatively, the vectors can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.

Table D Examples of hosts for cloning or expression Organisms Examples Sources and References*

Prokaryotes E. coli K 12 strain MM294 ATCC 31,446 X1776 ATCC 31,537 W3110 ATCC 27,325 K5 772 ATCC 53,635 Enterobacter Erwinia Klebsiella EnterobacteriaceaeProteus Salmonella (S.

tyhpimurium) Serratia (S. marcescans) Shigella Bacilli (B. subtilis and 8.

licheniformis) Pseudomonas (P.

aeruginosa) Streptomyces Eukaryotes Yeasts Saccharomyces cerevisiae Schizosaccharomyces pombe Kluyveromyces (Fleer et al., 1991) K. lactis MW98-8C, (de Louvencourt et al., , CBS683, CBS4574 1983) K. fragilis ATCC 12,424 K. bulgaricus ATCC 16,045 K. wickeramii ATCC 24,178 K. waltii ATCC 56,500 K, drosophilarum ATCC 36,906 K. thermotolerans K. marxianus; yarrowiaEPO 402226, 1990 Pichia pastoris (Sreekrishna et al., Candida Trichoderma reesia Neurospora crassa (Case et al., 1979) Torulopsis Rhodotorula Table D Examples of hosts for cloning or expression Organisms Examples Sources and References*

Schwanniomyces (S.

occidentalis) Neurospora Penicillium F Tolypocladium (WO 91/00357, 1991) Fil t i ung ous amen Aspergillus (A. nidulansIYelton, 1984 #229;

and A. niger) Kelly, 1985 #219;

Tilburn, 1983 #227 I ~rosophila S2 ll rt b t nve Spodoptera Sf9 e e ce s ra Chinese Hamster Ovary (CHO) Vertebrate cellssimian COS

*Unreferenced cells are generally available from American Type Culture Collection (Manassas, VA).

Vector choice is dictated by the organism or cells being used and the desired fate of the vector. Vectors may replicate once in the target cells, or may be "suicide" vectors. In general, vectors comprise signal sequences, origins of replication, marker genes, enhancer elements, promoters, and transcription termination sequences. The choice of these elements depends on the organisms in which the vector will be used and are easily determined.
Some of these elements may be conditional, such as an inducible or conditional promoter that is turned "on" when conditions are appropriate.
Examples of inducible promoters include those that are tissue-specific, which relegate expression to certain cell types, steroid-responsive, or heat-shock reactive. Some bacterial repression systems, such as the lac operon, have been exploited in mammalian cells and transgenic animals (Fleck et al., 1992;
Wyborski et al., 1996; Wyborski and Short, 1991 ). Vectors often use a selectable marker to facilitate identifying those cells that have incorporated the vector. Many selectable markers are well known in the art for the use with prokaryotes, usually antibiotio-resistance genes or the use of autotrophy and auxotrophy mutants.
Using antisense and sense IF1206 oligonucleotides can prevent IF1206~
polypeptide expression. These oligonucleotides bind to target nucleic acid sequences, forming duplexes that block transcription or translation of the target sequence by enhancing degradation of the duplexes, terminating prematurely transcription or translation, or by other means.
Antisense or sense oligonucleotides are singe-stranded nucleic acids, either RNA or DNA, which can bind target IF1206 mRNA (sense) or IF1206 DNA (antisense) sequences. According to the present invention, antisense or sense oligonucleotides comprise a fragment of the IF1206 DNA coding region of at least about 14 nucleotides, preferably from about 14 to 30 nucleotides.
In general, antisense RNA or DNA molecules can comprise at least 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 bases in length or more. Among others, (Stein and Cohen, 1988; van der Krol et al., 1988a) describe methods to derive antisense or a sense oligonucleotides from a given cDNA sequence.
Modifications of antisense and sense oligonucleotides can augment their effectiveness. Modified sugar-phosphodiester bonds or other sugar linkages (WO 91/06629, 1991), increase in vivo stability by conferring resistance to endogenous nucleases without disrupting binding specificity to target sequences. Other modifications can increase the affinities of the oligonucleotides for their targets, such as covalently linked organic moieties (WO 90/10448, 1990) or poly-(L)-lysine. Other attachments modify binding specificities of the oligonucleotides for their targets, including metal complexes or intercalating (e.g. ellipticine) and alkylating agents.
To introduce antisense or sense oligonucleotides into target cells (cells containing the target nucleic acid sequence), any gene transfer method may be used and are well known to those of skill in the art. Examples of gene transfer methods include 1 ) biological, such as gene transfer vectors like Epstein-Barr virus or conjugating the exogenous DNA to a ligand-binding molecule (WO 91/04753, 1991), 2) physical, such as electroporation, and 3) _79-chemical, such as CaP04 precipitation and oligonucleotide-lipid complexes (WO 90/10448, 1990).
The terms "host cell" and "recombinant host cell" are used interchangeably. Such terms refer not only to a particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term.
Methods of eukaryotic cell transfection and prokaryotic cell transformation are well known in the art. The choice of host cell will dictate the preferred technique for introducing the nucleic acid of interest. Table ##, which is not meant to be limiting, summarizes many of the known techniques in the art. Introduction of nucleic acids into an organism may also be done with ex vivo techniques that use an in vitro method of transfection, as well as established genetic techniques, if any, for that particular organism.
Table E Methods to introduce nucleic acid into cells Cells Methods References Notes (Cohen et al., 1972;

ProkaryotesCalcium chlorideHanahan, 1983; Mandel b and Hi a, 1970 t i ( ac er a) Electroporation(Shigekawa and Dower, Eukaryotes N-(2-Hydroxyethyl)piperazine-N'-(2-ethanesulfonicCells may acid be (HEPES) buffered shocked" with saline solution (Chen and glycerol or Okayama, 1988; dimethylsulfoxid Mammalian Calcium Graham and van der a (DMSO) to Eb, cells phosphate 1973; Wigler et increase al., transfection 1978) transfection efficiency BES (N,N-bis(2-(Ausubel et hydroxyethyl)-2- al., 1987).
aminoethanesulfonic acid) buffered solution (Ishiura et al., Table E Methods to introduce nucleic acid into cells Cells Methods References Notes Most useful for transient, but not stable, Diethylaminoethyl(Fujita et al., transfections 1986;

.
(DEAE)-DextranLopata et al., 1984;
Chloroquine can transfection Selden et al., 1986)be used to increase efficiency.

(Neumann et al., Especially 1982; useful ElectroporationPotter, 1988; Potterfor hard-to-et al., 1984; Wong transfect and Neumann, 1982) lymphoc tes.

Cationic lipid(Elroy-Stein and Applicable Moss, to 1990; Felgner et both in vivo reagent al., and 1 gg7; Rose et al.,in vitro transfection 1991;

Whitt et al., 1990)transfection.

Production exemplified by (Cepko et al., 1984;

Miller and Buttimore,Lengthy process, 1986; Pear et al., many packaging 1993) Infection in vitro lines available and in at Retroviral vivo: (Austin and ATCC.

Cepko, 1990; BodineApplicable et to al., 1991; Fekete both in vivo and and Cepko, 1993; Lemischkain vitro et al., 1986; Turnertransfection.
et al., 1990; Williams et al., 1984) (Chaney et al., 1986;

Polybrene Kawai and Nishizawa,.

Can be used to establish cell Microinjection(Capecchi, 1980) lines carrying integrated copies of IF1206 DNA

se uences.

(Rassoulzadegan et al., Protoplast 1 g82; Sandri-Goldin fusion et al., 1981; Schaffner, (Luckow, 1991; Miller,Useful for Insect Baculovirus in vitro cells lggg; O'Reilly et production (in vitro)systems al., of 1 g92 proteins with _81 _ Table E Methods to introduce nucleic acid into cells Cells Methods References Notes eukaryotic modifications.

Electroporation(Becker and Guarente, 1991 ) Lithium acetate(Gietz et al., 1998;
Ito et Yeast al., 1983) Spheroplast (Beggs Laborious, 1978; Hinnen et can fusion , produce 1978) al.

, aneu loids.

(Bechtold and Pelletier, Agrobacterium 1998; Escudero and transformationHohn, 1997; Hansen and Chilton, 1999;

Touraev and al., 1997) Biolistics (Finer et al., 1999;

(microprojectiles)Hansen and Chilton, 1999; Shillito, (Fromm et al., 1985;
Ou-Lee et al., 1986;
Rhodes Plant cellsElectroporationet al., 1988; Saunders et (general (protoplasts) al., 1989) reference: May be combined with liposomes (Trick (Hansen and al., and 1997) W
h rig t, polyethylene 1999)) glycol (PEG) (Shillito, 1999) treatment May be combined with Liposomes electroporation (Trick and al., 1997 in plants (Leduc and al., 1996;

microin'ectionZhou and al., 1983 Seed imbibition(Trick and al., 1997) Laser beam (Hoffman, 1996 Silicon carbide(Thompson and al., whiskers 1995) Vectors often use a selectable marker to facilitate identifying those cells that have incorporated the vector. Many selectable markers are well known in the art for the use with prokaryotes, usually antibiotio-resistance _82_ genes or the use of autotrophy and auxotrophy mutants. Table F lists often-used selectable markers for mammalian cell transfection.
Table F Useful selectable markers for eukaryote cell transfection Selection Reference Selectable Marker Action Conversion of Xyl-Media includes A to Xyl-ATP, (Kaufman 9-Vii-Adenosine D-xylofuranosyl which incorporateset al., deaminase (ADA) adenine (Xyl-A) into nucleic 1 gg6) acids, killing cells.
ADA

detoxifies MTX competitive inhibitor of DHFR.

In absence (Simonsen of Methotrexate exogenous Dihydrofolate (MTX) and reductase (DHFR)and dialyzed purines, cellsLevinson, serum (purine-free require DHFR, 1 gg3) media) a necessary enzyme in purine biosynthesis.

6418, an aminoglycoside Aminoglycoside detoxified (Southern phosphotransferase by APH, 6418 interferes and Berg, ("APH" with "neo"

, ribosomal function1982) , "G418") and consequently, translation.

Hygromycin-B, an aminocyclitol Hygromycin-B- detoxified by HPH, (Palmer et phosphotransferasehygromycin-B disrupts protein1987) al.

(HPH) translocation , and promotes mistranslation.

Forward selectionForward:

(TK+): Media Aminopterin (HAT) forces incorporates cells to synthesze Thymidine kinaseaminopterin. dTTP from (Littlefield , (TK) Reverse selectionthymidine, 1 g64) a (TK-): Media pathway requiring incorporates TK.

bromodeoxyuridineReverse: TK

(BrdU). phosphorylates Table F Useful selectable markers for eukaryote cell transfection Selection Reference Selectable Marker Action BrdU, which incorporates into nucleic acids, killing cells.

A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce IF1206. Accordingly, the invention provides methods for producing IF1206 using the host cells of the invention.
In one embodiment, the method comprises culturing the host cell of the invention (into which a recombinant expression vector encoding IF1206 has been introduced) in a suitable medium, such that IF1206 is produced. In another embodiment, the method further comprises isolating IF1206 from the medium or the host cell.
Transgenic IF1206 animals Transgenic animals are useful for studying the function andlor activity of IFI206 and for identifying and/or evaluating modulators of IF1206 activity.
"Transgenic animals" are non-human animals, preferably mammals, more preferably a rodents such as rats or mice, in which one or more of the cells include a transgene. Other transgenic animals include primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A "transgene" is exogenous DNA
that is integrated into the genome of a cell from which a transgenic animal develops, and that remains in the genome of the mature animal. Transgenes preferably direct the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal with the purpose of preventing expression of a naturally encoded gene product in one or more cell types or tissues (a "knockout" transgenic animal), or serving as a marker or indicator of an integration, chromosomal location, or region of recombination (e.g.
crelloxP mice). A "homologous recombinant animal" is a non-human animal, such as a rodent, in which endogenous IFI206 has been altered by an exogenous DNA molecule that recombines homologously with endogenous IF1206 in a (e.g. embryonic) cell prior to development the animal. Host cells with exogenous IF1206 can be used to produce non-human transgenic animals, such as fertilized oocytes or embryonic stem cells into which IFI206-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals or homologous recombinant animals.
1. Approaches to transgenic animal production A transgenic animal can be created by introducing IFI206 into the male pronuclei of a fertilized oocyte (e.g., by microinjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal (pffa). The IFI206 cDNA sequences.(SEQ ID N0:1) can be introduced as a transgene into the genome of a non-human animal. Alternatively, a homologue of IF1206, such as the naturally-occuring variant of IF1206 (SEQ ID
N0:3), can be used as a transgene. Intronic sequences and polyadenylation signals can also be included in the transgene to increase transgene expression. Tissue-specific regulatory sequences can be operably-linked to the IFI206 transgene to direct expression of IFI206 to particular cells.
Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art, e.g. (Evans et al., U.S. Patent No. 4,870,009, 1989; Hogan, 0879693843, 1994; Leder and Stewart, U.S. Patent No. 4,736,866, 1988;
Wagner and Hoppe, US Patent No. 4,873,191, 1989). Other non-mice transgenic animals may be made by similar methods. A transgenic founder animal, which can be used to breed additional transgenic animals, can be identified based upon the presence of the transgene in its genome and/or expression of the transgene mRNA in tissues or cells of the animals.
Transgenic (e.g. IFI206) animals can be bred to other transgenic animals carrying other transgenes.
2. Vectors for transgenic animal production To create a homologous recombinant animal, a vector containing at least a portion of IFI206 into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, IFI206. 1F1206 can be a murine gene (SEQ ID N0:1 ), or other IFI206 homologue, such as the naturally occurring variant (SEQ ID N0:3). In one approach, a knockout vector functionally disrupts the endogenous IFI206 gene upon homologous recombination, and thus a non-functional IF1206 protein, if any, is expressed.
Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous IFI206 is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of endogenous IF1206). In this type of homologous recombination vector, the altered portion of the IFI206 is flanked at its 5'- and 3'-termini by additional nucleic acid of the IFI206 to allow for homologous recombination to occur between the exogenous IFI206 carried by the vector and an endogenous IFI206 in an embryonic stem cell. The additional flanking IF1206 nucleic acid is sufficient to engender homologous .
recombination with endogenous IF1206. Typically, several kilobases of flanking DNA (both at the 5'- and 3'-termini) are included in the vector (Thomas and Capecchi, 1987). The vector is then introduced into an embryonic stem cell line (e.g., by electroporation), and cells in which the introduced IF1206 has homologously-recombined with the endogenous IFI206 are selected (Li et al., 1992).
3. Introduction of IF1206 transgene cells during development Selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (Bradley, 1987). A chimeric embryo can then be implanted into a suitable pffa and the embryo brought to term.
Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously-recombined DNA by germline transmission of the transgene.
Methods for constructing homologous recombination vectors and homologous recombinant animals are described (Berns et al., WO 93/04169, 1993;
Bradley, 1991; Kucherlapati et al., WO 91 /01140, 1991; Le Mouellic and Brullet, WO 90/11354, 1990).
Alternatively, transgenic animals that contain selected systems that allow for regulated expression of the transgene can be produced. An example of such a system is the crelloxP recombinase system of bacteriophage P1 (Lakso et al., 1992). Another recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al., 1991 ). If a crelloxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be produced as "double" transgenic animals, by mating an animal containing a transgene encoding a selected protein to another containing a transgene encoding a recombinase.
Clones of transgenic animals can also be produced (Wilmut et al., 1997). In brief, a cell from a transgenic animal can be isolated and induced to exit the growth cycle and enter Go phase. The quiescent cell can then be fused to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured to develop to a morula or blastocyte and then transferred to a pffa. The offspring borne of this female foster animal will be a clone of the "parent" transgenic animal.
Pharmaceutical compositions The IFI206 nucleic acid molecules, IF1206 polypeptides, and anti-IF1206 Abs (active compounds) of the invention, and derivatives, fragments, analogs and homologs thereof, can be incorporated into pharmaceutical compositions. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. A
"pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration (Gennaro, 2000). Preferred examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5%
human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. Except when a conventional media or agent is incompatible with an active compound, use of these compositions is contemplated. Supplementary active compounds can also be incorporated , into the compositions.

_87_ 1. General considerations A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration, including intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents;
antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
2. Injectable formulations Pharmaceutical compositions suitable for injection include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, CREMOPHOR EL~° (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid so as to be administered using a syringe. Such compositions should be stable during manufacture and storage and must be preserved against contamination from microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (such as glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures.
Proper fluidity can be maintained, for example, by using a coating such as lecithin, by maintaining the required particle size in the case of dispersion and by using surfactants. Various antibacterial and antifungal agents, for _88_ example, parabens, chlorobutanol, phenol, ascorbic acid, and thimerosal, can contain microorganism contamination. Isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, and sodium chloride can be included in the composition. Compositions that can delay absorption include agents such as aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incorporating the active compound (e.g., an IF1206 or anti-IF1206 antibody) in the required amount in an appropriate solvent with one or a combination of ingredients as required, followed by sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium, and the other required ingredients as discussed. Sterile powders for the preparation of sterile injectable solutions, methods of preparation include vacuum drying and freeze-drying that yield a powder containing the active ingredient and any desired ingredient from a sterile solutions.
3. Oral compositions Oral compositions generally include an inert diluent or an edible carrier.
They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients aid used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally.
Pharmaceutically compatible binding agents, and/or adjuvant materials can be included. Tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, PRIMOGEL, or corn starch; a lubricant such as magnesium stearate or STEROTES; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
4. Compositions for inhalation _89_ For administration by inhalation, the compounds are delivered as an aerosol spray from a a nebulizer or a pressurized container that contains a suitable propellant, e.g., a gas such as carbon dioxide.
5. Systemic administration Systemic administration can also be transmucosal or transdermal. For transmucosal or transdermal administration, penetrants that can permeate the target barriers) are selected. Transmucosal penetrants include, detergents, bile salts, and fusidic acid derivatives. Nasal sprays or suppositories can be used for transmucosal administration. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams.
The compounds can also be prepared in the form of suppositories (e.g., with bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
6. Carriers In one embodiment, the active compounds are prepared with carriers that protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Such materials can be obtained commercially from ALZA Corporation (Mountain View, CA) and NOVA
Pharmaceuticals, Inc. (Lake Elsinore, CA), or prepared by one of skill in the art. Liposomal suspensions can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, such as in (Eppstein et al., US Patent No. 4,522,811, 1985).
7. Unit dosage Oral formulations or parenteral compositions in unit dosage form can be created to facilitate administration and dosage uniformity. Unit dosage form refers to physically discrete units suited as single dosages for the subject to be treated, containing a therapeutically effective quantity of active compound in association with the required pharmaceutical carrier. The specification for the unit dosage forms of the invention are dictated by, and directly dependent on, the unique characteristics of the active compound and the particular desired therapeutic effect, and the inherent limitations of compounding the active compound.
8. Gene therapy compositions The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (Nabel and Nabel, US Patent No. 5,328,470, 1994), or by stereotactic injection (Chen et al., 1994). The pharmaceutical preparation of a gene therapy vector can include an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.
9. Kits for pharmaceutical compositions The pharmaceutical compositions can be included in a kit, container, pack, or dispenser together with instructions for administration. When the invention is supplied as a kit, the different components of the composition may be packaged in separate containers and admixed immediately before use.
Such packaging of the components separately may permit long-term storage without losing the active components' functions.
Kits may also include reagents in separate containers that facilitate the execution of a specific test, such as diagnostic tests or tissue typing. For example, IFI206 DNA templates and suitable primers may be supplied for . internal controls.
(a) Containers or vessels The reagents included in the kits can be supplied in containers of any sort such that the life of the different components are preserved, and are not adsorbed or altered by the materials of the container. For example, sealed glass ampules may contain lyophilized luciferase or buffer that have been packaged under a neutral, non-reacting gas, such as nitrogen. Ampoules may consist of any suitable material, such as glass, organic polymers, such as polycarbonate, polystyrene, etc., ceramic, metal or any other material typically employed to hold reagents. Other examples of suitable containers include simple bottles that may be fabricated from similar substances as ampules, and envelopes, that may consist of foil-lined interiors, such as aluminum or an alloy. Other containers include test tubes, vials, flasks, bottles, syringes, or the like. Containers may have a sterile access port, such as a bottle having a stopper that can be pierced by a hypodermic injection needle. Other containers may have two compartments that are separated by a readily removable membrane that upon removal permits the components to mix. Removable membranes may be glass, plastic, rubber, etc.
(b) Instructional materials Kits may also be supplied with instructional materials. Instructions may be printed on paper or other substrate, and/or may be supplied as an electronic-readable medium, such as a floppy disc, CD-ROM, DVD-ROM, Zip disc, video tape, audio tape, etc. Detailed instructions may not be physically associated with the kit; instead, a user may be directed to an Internet web site specified by the manufacturer or distributor of the kit, or supplied as electronic mail.
Screening and detection methods The isolated nucleic acid molecules of the invention can be used to express IF1206 (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect IFI206 mRNA (e.g., in a biological sample) or a genetic lesion in an IFI206, and to modulate IF1206 activity, as described below. In addition, IF1206 polypeptides can be used to screen drugs or compounds that modulate the IF1206 activity or expression as well as to treat disorders characterized by insufficient or excessive production of IF1206 or production of IF1206 forms that have decreased or aberrant activity compared to IF1206 wild-type protein, or modulate biological function that involve IF1206 (e.g. obesity). In addition, the anti-IF1206 Abs of the invention can be used to detect and isolate IF1206 and modulate IF1206 activity.
1. Screening assays The invention provides a method (screening assay) for identifying modalities, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs), foods, combinations thereof, etc., that effect IF1206, a stimulatory or inhibitory effect, inlcuding translation, transcription, activity or copies of the gene in cells. The invention also includes compounds identified in screening assays.
Testing for compounds that increase or decrease IF1206 activity are desirable. A compound may modulate IF1206 activity by affecting: (1 ) the number of copies of the gene in the cell (amplifiers and deamplifiers); (2) increasing or decreasing transcription of the IFI206 (transcription up-regulators and down-regulators); (3) by increasing or decreasing the translation of IFI206 mRNA into protein (translation up-regulators and down-regulators); or (4) by increasing or decreasing the activity of IF1206 itself (agonists and antagonists).
(a) effects of compounds To identify compounds that affect IF1206 at the DNA, RNA and protein levels, cells or organisms are contacted\ with a candidate compound and the corresponding change in IF1206 DNA, RNA or protein is assessed (Ausubel et al., 1987). For DNA amplifiers and deamplifiers, the amount of IFI206 DNA is measured, for those compounds that are transcription up-regulators and down-regulators the amount of IF1206 mRNA is determined; for translational up- and down-regulators, the amount of IF1206 polypeptides is measured.
Compounds that are agonists or antagonists may be identified by contacting cells or organisms with the compound, and then measuring, for example, adipocyte differentiation in vitro.
In one embodiment, many assays for screening candidate or test compounds that bind to or modulate the activity of IFI206 or polypeptide or biologically-active portion are available. Ttest compounds can be obtained using any of the numerous approaches in combinatorial library methods, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution;
the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptides, while the other four approaches encompass peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, 1997).
(b) small molecules A "small molecule" refers to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention. Examples of methods for the synthesis of molecular libraries can be found in: (Carell et al., 1994x; Carell et al., 1994b; Cho et al., 1993;
DeWitt et al., 1993; Gallop et al., 1994; Zuckermann et al., 1994).
Libraries of compounds may be presented in solution (Houghten et al., 1992) or on beads (Lam et al., 1991 ), on chips (Fodor et al., 1993), bacteria, spores (Ladner et al., US Patent No. 5,223,409, 1993), plasmids (Cull et al., 1992) or on phage (Cwirla et al., 1990; Devlin et al., 1990; Felici et al., 1991;
Ladner et al., US Patent No. 5,223,409, 1993; Scott and Smith, 1990). A cell-free assay comprises contacting IF1206 or biologically-active fragment with a known compound that binds IF1206 to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with IF1206, where determining the ability of the test compound to interact with IF1206 comprises determining the ability of the IF1206 to preferentially bind to or modulate the activity of an IFI206 target molecule.
(c) cell-free assays The cell-free assays of the invention may be used with both soluble or a membrane-bound forms of IF1206. In the case of cell-free assays comprising the membrane-bound form, a solubilizing agent to maintain IF1206 in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, TRITON~ X-100 and others from the TRITON~ series, THESIT~, Isotridecypoly(ethylene glycol ether)n, N-dodecyl--N,N-dimethyl-3-ammonio-1-propane sulfonate, 3-(3-cholamidopropyl) dimethylamminiol-1-propane sulfonate (CHAPS), or 3-(3-cholamidopropyl)dimethylamminiol-2-hydroxy-1-propane sulfonate (CHAPSO).
(d) immobilization of target molecules to facilitate screening In more than one embodiment of the assay methods, immobilizing either IF1206 or its partner molecules can facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate high throughput assays. Binding of a test compound to IF1206, or interaction of IF1206 with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants, such as microtiter plates, test tubes, and micro-centrifuge tubes. A fusion protein can be provided that adds a domain .
that allows one or both of the proteins to be bound to a matrix. For example, GST-IF1206 fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (SIGMA Chemical, St. Louis, MO) or glutathione derivatized microtiter plates that are then combined with the test compound or the test compound and either the non-adsorbed target protein or IF1206, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described.
Alternatively, the complexes can be dissociated from the matrix, and the level of IF1206 binding or activity determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in screening assays. Either IF1206 or its target molecule can be immobilized using biotin-avidin or biotin-streptavidin systems. Biotinylation can be accomplished using many reagents, such as biotin-NHS
(N-hydroxy-succinimide; PIERCE Chemicals, Rockford, IL), and immobilized in wells of streptavidin-coated 96 well plates (PIERCE Chemical).

Alternatively, Abs reactive with I F1206 or target molecules, but which do not interfere with binding of the IF1206 to its target molecule, can be derivatized to the wells of the plate, and unbound target or IF1206 trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described for the GST-immobilized complexes, include immunodetection of complexes using Abs reactive with IF1206 or its target, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the IF1206 or target molecule.
(e) scn?ens to identify modulators Modulators of IF1206 expression can be identified in a method where a cell is contacted with a candidate compound and the expression of IF1206 mRNA or protein in the cell is determined. The expression level of IFI206 mRNA or protein in the presence of the candidate compound is compared to IF1206 mRNA or protein levels in the absence of the candidate compound.
The candidate compound can then be identified as a modulator of IF1206 mRNA or protein expression based upon this comparison. For example, when expression of IF1206 mRNA or protein is greater (i.e., statistically , significant) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of IF1206 mRNA or protein expression. Alternatively, when expression of IF1206 mRNA or protein is less (statistically significant) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of IF1206 mRNA or protein expression. The level of IF1206 mRNA or protein expression in the cells can be determined by methods described for detecting IF1206 mRNA or protein.
(i) hybrid assays In yet another aspect of the invention, IF1206 can be used as "bait" in two-hybrid or three hybrid assays [Saifer, 1994 #38; Zervos, 1993 #382;
Madura, 1993 #383; Bartel, 1993 #384; Iwabuchi, 1993 #385; Brent, 1994 #386] to identify other proteins that bind or interact with IF1206 (F1206-binding proteins (IF1206-bps)) and modulate IF1206 activity. Such IF1206-bps are also likely to be involved in the propagation of signals by the IF1206 as, for example, upstream or downstream elements of an IF1206 pathway.
The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for IF1206 is fused to a gene encoding the DNA
binding domain of a known transcription factor (e.g., GAL4). The other ' construct, a DNA sequence from a library of DNA sequences that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey"
proteins are able to interact in vivo, forming an IF1206-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably-linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected, and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the IF1206-interacting protein.
The invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein.
2. Detection assays Portions or fragments of IFI206 cDNA sequences identified herein (and the complete IFI206 gene sequences) are useful in themselves. By way of non-limiting example, these sequences can be used to: (1 ) identify an individual from a minute biological sample (tissue typing); and (2) aid in forensic identification of a biological sample.
(a) Tissue typing The IF1206 sequences of the invention can be used to identify individuals from minute biological samples. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes and probed on _97_ a Southern blot to yield unique bands. The sequences of the invention are useful as additional DNA markers for "restriction fragment length polymorphisms" (RFLP; (Smulson et al., US Patent No. 5,272,057, 1993)).
Furthermore, the IFI206 sequences can be used to determine the actual base-by-base DNA sequence of targeted portions of an individual's genome. 1F1206 sequences can be used to prepare two PCR primers from the 5'- and 3'-termini of the sequences that can then be used to amplify an the corresponding sequences from an individual's genome and then sequence the amplified fragment.
Panels of corresponding DNA sequences from individuals can provide .
unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the invention can be used to obtain such identification sequences from individuals and from tissue. The IFI206 sequences of the invention uniquely represent portions of an individual's genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. The allelic variation between individual humans occurs with a frequency of about once ever 500 bases. Much of the allelic variation is due to single nucleotide polymorphisms (SNPs), which include RFLPs.
Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification purposes. Because greater numbers of polymorphisms occur in noncoding regions, fewer sequences are necessary to differentiate individuals. Noncoding sequences can positively identify individuals with a ' panel of 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as those in SEQ ID NOS:1 or 3 are used, a more appropriate number of primers for positive individual identification would be 500-2,000.
Predictive medicine The invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring _98_ clinical trials are used for prognostic (predictive) purposes to treat an individual prophylactically. Accordingly, one aspect of the invention relates to diagnostic assays for determining IF1206 and/or nucleic acid expression as well as IF1206 activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with aberrant IF1206 expression or activity, including obesity. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with IF1206, nucleic acid expression or activity. For example, mutations in IFI206 can be assayed in a biological sample. Such assays can be used for prognostic or predictive purpose to prophylactically treat an individual prior to the onset of a disorder characterized by or associated with IF1206, nucleic acid expression, or biological activity.
Another aspect of the invention provides methods for determining IF1206 activity, or nucleic acid expression, in an individual to select appropriate therapeutic or prophylactic agents for that individual (referred to herein as "pharmacogenomics"). Pharmacogenomics allows for the selection of modalities (e.g., drugs, foods) for therapeutic or prophylactic treatment of an individual based on the individual's genotype (e.g., the individual's genotype to determine the individual's ability to respond to a particular agent).
Another aspect of the invention pertains to monitoring the influence of modalities (e.g., drugs, foods) on the expression or activity of IF1206 in clinical trials.
1. Diagnostic assays An exemplary method for detecting the presence or absence of IF1206 in a biological sample involves obtaining a biological sample from a subject and contacting the biological sample with a compound or an agent capable of detecting IF1206 or IFI206 nucleic acid (e.g., mRNA, genomic DNA) such that the presence of IF1206 is confirmed in the sample. An agent for detecting IFI206 mRNA or genomic DNA is a labeled nucleic acid probe that can hybridize to IFI206 mRNA or genomic DNA. The nucleic acid probe can be, _99_ for example, a full-length IF1206 nucleic acid, such as the nucleic acid of SEQ
ID NOS: 1 or 3, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to IFI206 mRNA or genomic DNA.
An agent for detecting IF1206 polypeptide is an antibody capable of binding to IF1206, preferably an antibody with a detectable label. Abs can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment (e.g., Fab or F(ab')2) can be used. A labeled probe or antibody is coupled (i.e., physically linking) to a detectable substance, as well as indirect detection of the probe or antibody by reactivity with another reagent that is directly labeled.
Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin.
The term "biological sample" includes tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. The detection method of the invention can be used to detect IFI206 mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of IFI206 mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of IF1206 polypeptide include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of IFI206 genomic DNA include Southern hybridizations and fluorescence in situ hybridization (FISH). Furthermore, in vivo techniques for detecting IF1206 include introducing into a subject a labeled anti-IF1206 antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample from the subject contains protein molecules, and/or mRNA molecules, and/or genomic DNA molecules.
A prefen-ed biological sample is blood.
In another embodiment, the methods further involve obtaining a biological sample from a subject to provide a control, contacting the sample with a compound or agent to detect IFI206, mRNA, or genomic DNA, and comparing the presence of IFI206, mRNA or genomic DNA in the control sample with the presence of IF1206, mRNA or genomic DNA in the test sample.
The invention also encompasses kits for detecting IF1206 in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of detecting IF1206 or IFI206 mRNA in a sample; reagent and/or equipment for determining the amount of IF1206 in the sample; and reagent and/or equipment for comparing the amount of IF1206 in the sample with a standard. The compound or agent can be packaged in a suitable container.
The kit can further comprise instructions for using the kit to detect IF1206 or nucleic acid.
2. Prognostic assays The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with aberrant IF1206 expression or activity. For example, the assays described herein, can be used to identify a subject having or at risk of developing a disorder associated with IF1206, nucleic acid expression or activity. Alternatively, the prognostic assays can be used to identify a subject having or at risk for developing a disease or disorder. Tthe invention provides a method for identifying a disease or disorder associated with aberrant IF1206~
expression or activity in which a test sample is obtained from a subject and IF1206 or nucleic acid (e.g., mRNA, genomic DNA) is detected. A test sample is a biological sample obtained from a subject. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
Pognostic assays can be used to determine whether a subject can be administered a modality (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, food, etc.) to treat a disease or disorder associated with aberrant IF1206 expression or activity. Such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder. The invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant IF1206 expression or activity in which a test sample is obtained and IF1206 or nucleic acid is detected (e.g., where the presence of IF1206 or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with aberrant IFI206 expression or activity).
The methods of the invention can also be used to detect genetic lesions in an IFI206 to determine if a subject with the genetic lesion is at risk for a disorder characterized by aberrant cell proliferation, differentiation or obesity. Methods include detecting, in a sample from the subject, the presence or absence of a genetic lesion_characterized by at an alteration affecting the integrity of a gene encoding an IF1206 polypeptide, or the mis-' expression of IFI206. Such genetic lesions can be detected by ascertaining:
(1 ) a deletion of one or more nucleotides from IFI206; (2) an addition of one or more nucleotides to IFI206; (3) a substitution of one or more nucleotides in IF1206, (4) a chromosomal rearrangement of an IF1206 gene; (5) an alteration in the level of a IFI206 mRNA transcripts, (6) aberrant modification of an IFI206, such as a change genomic DNA methylation, (7) the presence of a non-wild-type splicing pattern of an IFI206 mRNA transcript, (8) a non-wild-type level of IFI206, (9) allelic loss of IFI206, and/or (10) inappropriate post-translational modification of IF1206 polypeptide. There are a large number of known assay techniques that can be used to detect lesions in IFI206. Any biological sample containing nucleated cells may be used.
In certain embodiments, lesion detection may use a probe/primer in a polymerase chain reaction (PCR) (e.g., (Mullis, US Patent No. 4,683,202, 1987; Mullis et al., US Patent No. 4,683,195, 1987), such as anchor PCR or rapid amplification of cDNA ends (RACE) PCR, or, alternatively, in a ligation chain reaction (LCR) (e.g., (Landegren et al., 1988; Nakazawa et al., 1994), the latter is particularly useful for detecting point mutations in IFI206-genes (Abravaya et al., 1995). This method may include collecting a sample from a patient, isolating nucleic acids from the sample, contacting the nucleic acids with one or more primers that specifically hybridize to IFI206 under conditions such that hybridization and amplification of the IFI206 (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
Alternative amplification methods include: self sustained sequence replication (Guatelli et al., 1990), transcriptional amplification system (Kwoh et al., 1989); Q(i Replicase (Lizardi et al., 1988), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules present in low abundance.
Mutations in IFI206 from a sample can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA' is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
Hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high-density arrays containing hundreds or thousands of oligonucleotides probes, can identify genetic mutations in IFI206 (Cronin et al., 1996; Kozal et al., 1996). For example, genetic mutations in IFI206 can be identified in two-dimensional arrays containing light-generated DNA probes as described in Cronin, et al., supra. Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations.
This is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the IFI206 and detect mutations by comparing the sequence of the sample IF1206-with the corresponding wild-type (control) sequence. Examples of sequencing reactions include those based on classic techniques (Maxam and Gilbert, 1977; Sanger et al., 1977). Any of a variety of automated sequencing procedures can be used when performing diagnostic assays (Naeve et al., 1995) including sequencing by mass spectrometry (Cohen et al., 1996; Griffin and Griffin, 1993; Koster, W094/16101, 1994).
Other methods for detecting mutations in the IFI206 include those in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al., 1985). In general, the technique of "mismatch cleavage" starts by providing heteroduplexes formed by hybridizing (labeled) RNA or DNA containing the wild-type IF1206 sequence with potentially mutant RNA or DNA obtained from a sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as those that arise from base pair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with S, nuclease to enzymatically digest the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. The digested material is then separated by size on denaturing polyacrylamide gels to determine the mutation site (Grompe et al., 1989; Saleeba and Cotton, 1993). The control DNA or RNA can be labeled for detection.
Mismatch cleavage reactions may employ one or more proteins that recognize mismatched base pairs in double-stranded DNA (DNA mismatch .
repair) in defined systems for detecting and mapping point mutations in IFI206 cDNAs obtained from samples of cells. For example, the mutt enzyme of E.

coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G!T mismatches (Hsu et al., 1994). According to an exemplary embodiment, a probe based on a wild-type IFI206 sequence is hybridized to a cDNA or other DNA product from a test cell(s), The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like (Modrich et al., US Patent No. 5,459,039, 1995).
Electrophoretic mobility alterations can be used to identify mutations in IF1206. For example, single strand conformation polymorphism (SSCP) may be used to detect differences in electrophoretic mobility befinreen mutant and wild type nucleic acids (Cotton, 1993; Hayashi, 1992; Orita et al., 1989).
Single-stranded DNA fragments of sample and control IFI206 nucleic acids are denatured and then renatured. The secondary structure of single-stranded nucleic acids varies according to sequence; the resulting alteration in electrophoretic mobility allows detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a sequence changes. The subject method may use heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al., 1991).
The migration of mutant or wild-type fragments can be assayed using denaturing gradient gel electrophoresis (DGGE; (Myers et al., 1985). In DGGE, DNA is modified to prevent complete denaturation, for example by adding a GC clamp of approximately 40 by of high-melting GC-rich DNA by PCR. A temperature gradient may also be used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA
(Rossiter and Caskey, 1990).
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found (Saiki et al., 1986; Saiki et al., 1989). Such allele-specific oligonucleotides are hybridized to PCR-amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used. Oligonucleotide primers for specific amplifications may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization (Gibbs et al., 1989)) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerise extension (Prosser, 1993). Novel restriction site in the region of the mutation may be introduced to create cleavage-based detection (Gasparini et al., 1992). Certain amplification may also be performed using Taq ligase for amplification (Barany, 1991 ). In such cases, ligation occurs only if there is a perfect match at the 3'-terminus of the 5' sequence, allowing detection of a known mutation by scoring for amplification.
The described methods may be performed, for example, by using pre-packaged kits comprising at least one probe (nucleic acid or antibody) that may be conveniently used, for example, in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving IF1206.
Furthermore, any cell type or tissue in which IF1206 is expressed may , be utilized in the prognostic assays described herein.
3. Pharmacogenomics Agents, or modulators that have a stimulatory or inhibitory effect on IF1206 activity or expression, as identified by a screening assay can be administered to individuals to treat, prophylactically or therapeutically, disorders, including obesity. In conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between a subject's genotype and the subject's response to a foreign modality, such as a food, compound or drug) may be considered. Metabolic differences of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug.
Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens.
Accordingly, the activity of IF1206, expression of IFI206 nucleic acid, or mutations) in an individual can be determined to guide the selection of appropriate agents) for therapeutic or prophylactic treatment.
Pharmacogenomics deals with clinically significant hereditary variations in the response to modalities due to altered modality disposition and abnormal action in affected persons (Eichelbaum and Evert, 1996; Linder et al., 1997).
In general, two pharmacogenetic conditions can be differentiated: (1 ) genetic conditions transmitted as a single factor altering the interaction of a modality with the body (altered drug action) or (2) genetic conditions transmitted as single factors altering the way the body acts on a modality (altered drug metabolism). These pharmacogenetic conditions can occur either as rare defects or as nucleic acid polymorphisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.
As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymorphisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) explains the phenomena of some patients who show exaggerated drug response and/or serious toxicity after taking the standard and safe dose of a drug. These polymorphisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the CYP2D6 gene is highly polymorphic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers due to mutant CYP2D6 and CYP2C19 frequently experience exaggerated drug responses and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM shows no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite morphine. At the other extreme are the so-called ultra-rapid metabolizers who are unresponsive to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
The activity of IF1206, expression of IFI206 nucleic acid, or mutation content of IF1206 in an individual can be determined to select appropriate agents) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymorphic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with an IF1206 modulator, such as a modulator identified by one of the described exemplary screening assays.
4. Monitoring effects during clinical trials Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of IF1206 (e.g., the ability to modulate aberrant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay to increase IFI206 expression, protein levels, or up-regulate IF1206 activity can be monitored in clinical trails of subjects exhibiting decreased IFI206 expression, protein levels, or down-regulated IF1206 activity. Alternatively, the effectiveness of an agent determined to decrease IF1206 expression, protein levels, or down-regulate IF1206 activity, can be monitored in clinical trails of subjects exhibiting increased IFI206 expression, protein levels, or up-regulated IF1206 activity.
In such clinical trials, the expression or activity of IF1206 and, preferably, other genes that have been implicated in, for example, obesity can be used as a "read out" or markers for a particular cell's responsiveness.
For example, genes, including IFI206, that are modulated in cells by treatment with a modality (e.g., food, compound, drug or small molecule) can be identified. To study the effect of agents on cellular proliferation disorders, for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of IFI206 and other genes implicated in .
the disorder. The gene expression pattern can be quantified by Northern blot analysis, nuclear run-on or RT-PCR experiments, or by measuring the amount of protein, or by measuring the activity level of IF1206 or other gene products.
In this manner, the gene expression pattern itself can serve as a marker, indicative of the cellular physiological response to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent.
The invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, food or other drug candidate identified by the screening assays described herein) comprising the steps of (1 ) obtaining a pre-administration sample from a subject; (2) detecting the level of expression of an IF1206, mRNA, or genomic DNA in the.
preadministration sample; (3) obtaining one or more post-administration samples from the subject; (4) detecting the level of expression or activity of the IF1206, mRNA, or genomic DNA in the post-administration samples; (5) comparing the level of expression or activity of the IF1206, mRNA, or genomic DNA in the pre-administration sample with the IF1206, mRNA, or genomic DNA in the post administration sample or samples; and (6) altering the administration of the agent to the subject~accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of IF1206 to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of IF1206 to lower levels than detected, i.e., to decrease the effectiveness of the agent.

5. Methods of treatment The invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant IF1206 expression or activity. The disorders include obesity. Furthermore, these same methods of treatment may be used to induce weight loss, or enhance weight loss, by changing the level of expression or activity of IF1206.
6. Disease and disorders Diseases and disorders that are characterized by increased IF1206 levels or biological activity may be treated with therapeutics that antagonize .
(i.e., reduce or inhibit) activity. Antognists may be administered in a therapeutic or prophylactic manner. Therapeutics that. may be used include:
(1 ) IF1206 peptides, or analogs, derivatives, fragments or homologs thereof;
(2) Abs to an IF1206 peptide; (3) IF1206 nucleic acids; (4) administration of antisense nucleic acid and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences) that are used to eliminate endogenous function of by homologous recombination (Capecchi, 1989); or (5) modulators (i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or Abs specific to IF1206) that alter the interaction between IF1206 and its binding partner.
Diseases and disorders that are characterized by decreased IF1206 levels or biological activity may be treated with therapeutics that increase (i.e., are agonists to) activity. Therapeutics that upregulate activity may be administered therapeutically or prophylactically. Therapeutics that may be used include peptides, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability.
Similary, the same therapeutics used to treat diseases and disorders may also be used to decrease obesity or induce weight gain.
Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or IFI206 mRNAs). Methods include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like).
7. Prophylactic methods The invention provides a method for preventing, in a subject, a disease or condition associated with an aben-ant IF1206 expression or activity, by administering an agent that modulates IF1206 expression or at least one IF1206 activity. Subjects at risk for a disease that is caused or contributed to by aberrant IF1206 expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the IF1206 aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending on the type of IF1206 aberrancy, for example, an IF1206 agonist or IF1206 antagonist can be used to treat the subject. The appropriate agent can be determined based on screening assays.
8. Therapeutic methods Another aspect of the invention pertains to methods of modulating IF1206 expression or activity for therapeutic purposes. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of IF1206 activity associated with the cell. An agent that modulates IF1206 activity can be a nucleic acid or a protein, a naturally occurring cognate ligand of IFI206, a peptide, an IF1206 peptidomimetic, or other small molecule. The agent may stimulate IF1206 activity. Examples of such stimulatory agents include active IF1206 and a .
1F1206 nucleic acid molecule that has been introduced into the cell. In another embodiment, the agent inhibits IF1206 activity. Examples of inhibitory agents include antisense IFI206 nucleic acids and anti-IF1206 Abs. Modulatory methods can be performed in vitro (e.g., by.culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the invention provides methods of treating an individual afflicted with a disease or disorder characterized by aberrant expression or activity of an IF1206 or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay), or combination of agents that modulates (e.g., up-regulates or down-regulates) IF1206 expression or activity. In another embodiment, the method involves administering an IF1206 or nucleic acid molecule as therapy to compensate for reduced or aberrant IF1206 expression or activity.
Stimulation of IF1206 activity is desirable in situations in which IF1206 is abnormally down-regulated and/or in which increased IF1206 activity is likely to have a beneficial effect. One example of such a situation is where a subject has a disorder characterized by aberrant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders). Another example of such a situation is obesity.
9. Determination of the biological effect of the therapeutic Suitable in vitro or in vivo assays can be performed to determine the effect of a specific therapeutic and whether its administration is indicated for treatment of the affected tissue.
In various specific embodiments, in vitro assays may be performed with representative cells of the types) involved in the patient's disorder, to determine if a given therapeutic exerts the desired effect upon the cell type(s).
Modalities for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo testing, any of the animal model system known in the art may be used prior to administration to human subjects.
10. Prophylactic and therapeutic uses of the compositions of the in vention IF1206 nucleic acids and proteins are useful in potential prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to obesity.

As an example, a cDNA encoding IF1206 may be useful in gene therapy, and the protein may be useful when administered to a subject in need thereof. By way of non-limiting example, the compositions of the invention will have efficacy for treatment of patients suffering from infertility.
IFI206 nucleic acids, or fragments thereof, may also be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein is to be assessed. A further use could be as an anti-bacterial molecule (i.e.,-some peptides have been found to possess anti-bacterial properties). These materials are further useful in the generation of Abs that immunospecifically bind to the novel substances of the invention for use in therapeutic or diagnostic methods.
Examples 1. cDNA library construction The KIDNNOT05 cDNA library was constructed from tissue removed from a female infant kidney with anoxia (lot #RU95-04-0274; International Institute of Advanced Medicine, Exton Pa.). The frozen tissue was immediately homogenized and cells lysed with a Brinkmann Homogenizer Polytron PT-3000 (Brinkmann Instruments Inc., Westbury N.Y.) in a guanidinium isothiocyanate solution. Lysates were then loaded on a 5.7 M
CsCI cushion and ultracentrifuged in a SW28 swinging bucket rotor for 18 hours at 25,000 rpm at ambient temperature. The RNA was extracted once with acid phenol at pH 4.0 and precipitated with 0.3 M sodium acetate and 2.5 volumes of ethanol, resuspended in DEPC-treated water and DNAse treated for 25 min at 37°C. The reaction was stopped with an equal volume of pH
8.0 phenol, and the RNA was as above. The RNA was isolated using the Qiagen Oligotex kit (QIAGEN Inc, Chatsworth Calif.) and used to construct the cDNA
library.
The RNA was handled according to the recommended protocols in the Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (catalog #18248-013; Gibco/BRL). cDNAs were fractionated on a Sepharose CL4B column (catalog #275105, Pharmacia), and those cDNAs exceeding 400 by were ligated into pSport I. The plasmid pSport I was subsequently transformed into DHSa.TM. competent cells (Cat. #18258-012, Gibco/BRL).
2. Isolation and sequencing of cDNA clones Plasmid DNA was released from the cells and purified using the Miniprep Kit (Catalogue # 77468; Advanced Genetic Technologies Corporation, Gaithersburg Md.). This kit consists of a 96 well block with reagents for 960 purifications. The recommended protocol was employed except for the following changes: 1 ) the 96 wells were each filled with only ml of sterile Terrific Broth (Catalog # 22711, LIFE TECHNOLOGIES.TM., Gaithersburg Md.) with carbenicillin at 25 mg/L and glycerol at 0.4%; 2) the bacteria were cultured for 24 hours after the wells were inoculated and then lysed with 60 NI of lysis buffer; 3) a centrifugation step employing the Beckman GS-6R @2900 rpm for 5 min was performed before the contents of the block were added to the primary filter plate; and 4) the optional step of adding isopropanol to TRIS buffer was not routinely performed. After the last step in the protocol, samples were transferred to a Beckman 96-well block for storage.
The cDNAs were sequenced by the method of Sanger F and AR
Coulson (1975; J Mol Biol 94:441 f), using a Hamilton Micro Lab 2200 (Hamilton, Reno Nev.) in combination with four Pettier Thermal Cyclers (PTC200 from MJ Research, Watertown Mass.) and Applied Biosystems 377 or 373 DNA Sequencing Systems (Perkin Elmer), and reading frame was determined.
3. Homologies with cDNA clones and deduced proteins Each cDNA was compared to sequences in GenBank using a search algorithm developed by Applied Biosystems and incorporated into the INHERIT- 670 Sequence Analysis System. In this algorithm, Pattern Specification Language (TRW Inc, Los Angeles Calif.) was used to determine regions of homology. The three parameters that determine how the sequence comparisons run were window size, window offset, and error tolerance. Using a combination of these three parameters, the DNA database was searched for sequences containing regions of homology to the query sequence, and the appropriate sequences were scored with an initial value. Subsequently, these homologous regions were examined using dot matrix homology plots to distinguish regions of homology from chance matches. Smith-Waterman alignments were used to display the results of the homology search.
Peptide and protein sequence homologies were ascertained using the INHERIT.TM. 670 Sequence Analysis System in a way similar to that used in DNA sequence homologies. Pattern Specification Language and parameter windows were used to search protein databases for sequences containing regions of homology which were scored with an initial value. Dot-matrix homology plots were examined to distinguish regions of significant homology from chance matches.
BLAST, which stands for Basic Local Alignment Search Tool (Altschul S F (1993) J Mol Evol 36:290-300; Altschul, S F et al (1990) J Mol Biol 215:403-10), was used to search for local sequence alignments. BLAST
produces alignments of both nucleotide and amino acid sequences to determine sequence similarity. Because of the local nature of the alignments, BLAST is especially useful in determining exact matches or in identifying homologs. BLAST is useful for matches which do not contain gaps. The fundamental unit of BLAST algorithm output is the High-scoring Segment Pair (HSP) An HSP consists of two sequence fragments of arbitrary but equal lengths whose alignment is locally maximal and for which the alignment score meets or exceeds a threshold or cutoff score set by the user. The BLAST
approach is to look for HSPs between a query sequence and a database sequence, to evaluate the statistical significance of any matches found, and to report only those matches which satisfy the user-selected threshold of significance. The parameter E establishes the statistically significant threshold for reporting database sequence matches. E is interpreted as the upper bound of the expected frequency of chance occurrence of an HSP (or set of HSPS) within the context of the entire database search. Any database sequence whose match satisfies E is reported in the program output.

4. Northern analyses Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide.
sequence to a membrane on which RNAs from a particular cell type or tissue have been bound (Sambrook et al. supra).
Analogous computer techniques use BLAST (Altschul SF 1993 and 1990, supra) to search for identical or related molecules in nucleotide databases such as GenBank. This analysis is much faster than multiple, membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or homologous.
~' The basis of the search is the product score which is defined as:
EQU1 and it takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match will be exact within a 1-2% error; and at 70, the match will be exact. Homologous molecules are usually identified by selecting those which show product scores between 15 and 40, although lower scores may identify related molecules.
The results of the search are reported as a list of libraries in which the full length sequence, or parts thereof, is represented, the abundance of the sequence, and the percent abundance. Abundance directly reflects the number of times a particular transcript is present in a cDNA library, and percent abundance is abundance divided by the total number of sequences examined in the library.
5. Real-time quantitative PCR analysis (TaqMan system) to quantify mouse IFI206 abundance Total RNA preparations from liver or pulverized SKM of individual mice were made (Ultraspec reagent; Biotecx Laboratories, Houston TX) and assayed for mRNA abundance using quantitative real-time reverse-transcriptase PCR (RT-~PCR) following digestion of samples with DNAse per manufacturers instructions (GIBCO BRL, Grand Island NY). This system employed primers and probes specific to murine IFI206. 18S primers/probe were purchased from Perkin-Elmer Applied Biosystems (Foster City, CA).
Reactions and detection were carried out using a Model 7700 Sequence Detector and TaqMan reagents (PE Applied Biosystems; Boston, MA) in a volume of 50 ~,L and containing: 100 ng RNA, 3 mM MgCl2, reaction Buffer A
(1X), 12.5 U MuLV reverse transcriptase, 1.25 U TaqGold, forward and reverse primers (0.01 O.D. ea.), and 0.1wM probe (Note: 18S analyses employed 240 pg RNA, 5.5 mM MgCl2, and 0.05 ~.M probe/primer). Cycling conditions were: 50°C 15 min and 95°C 10 min, followed by 40 cycles of 95°C
sec and 60°C 1 min. 18S mRNA abundance was used as a loading 10 control, and all values reported herein represent 18S-corrected values.
TaaMan Oligo Seguences:
SEQ ID N0:19 15 <mulFlhlog.for1 >TGGAAATAAATAGGCAAGAAAGCA
SEQ ID N0:20 <mulFlhlog.rev1 >TCTCGCCTTCTTTCAGATGTAACA
SEQ ID N0:5 <mulFlhlog.probe1 >TCCTGCACACCTACATCAACTACAAGCCAC
Examples 6 and 7 are prophetic:
6. Extension of IFI206 to full length or to recover regulatory elements The nucleic acid sequence encoding full length IF1206 (SEQ ID N0:2) is used to design oligonucleotide primers for extending a partial nucleotide sequence to full length or for obtaining 5' sequences from genomic libraries.
One primer is synthesized to initiate extension in the antisense direction (XLR) and the other is synthesized to extend sequence in the sense direction (XLF). Primers allow the extension of the known IF1206 nucleotide sequence "outward" generating amplicons containing new, unknown nucleotide sequence for the region of interest The initial primers are designed from the cDNA using OLIGO® 4.06 Primer Analysis Software (National Biosciences), or another appropriate program, to be 22-30 nucleotides in length, to have a GC content of 50% or more, and to anneal to the target sequence at temperatures about 68°-72°C Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations is avoided.
The original, selected cDNA libraries, or a human genomic library are used to extend the sequence; the latter is most useful to obtain 5' upstream regions. If more extension is necessary or desired, additional sets of primers are designed to further extend the known region.
By following the instructions for the XL-PCR kit (Perkin Elmer) and thoroughly mixing the enzyme and reaction mix, high fidelity amplification is obtained. Beginning with 40 pmol of each primer and the recommended concentrations of all other components of the kit, PCR is performed using the Pettier Thermal Cycler (PTC200; MJ Research, Watertown Mass.) and the following parameters:
Step 1 94°C for 1 min (initial denaturation) Step 2 65°C for 1 min Step 3 68°C for 6 min Step 4 94°C for 15 sec Step 5 65°C for 1 min Step 6 68°C for 7 mih Step 7 Repeat step 4-6 for 15 additional cycles Step 8 94°C for 15 sec Step 9 65°C for 1 min Step 10 68°C for 7:15 min Step 11 Repeat step 8-10 for 12 cycles Step 12 72°C for 8 min Step 13 4°C (and holding) A 5-10 NI aliquot of the reaction mixture is analyzed by electrophoresis on a low concentration (about 0.6-0.8%) agarose mini-gel to determine which reactions were successful in extending the sequence. Bands thought to contain the largest products were selected and cut out of the gel. Further purification involves using a commercial gel extraction method such as QIAQuick.TM. (QIAGEN Inc). After recovery of the DNA, Klenow enzyme was used to trim single-stranded, nucleotide overhangs creating blunt ends which facilitate religation and cloning.
After ethanol precipitation, the products are redissolved in 13 NI of ligation buffer, 1 NI T4-DNA ligase (15 units) and 1 NI T4 polynucleotide kinase are added, and the mixture is incubated at room temperature for 2-3 hours or overnight at 16°C Competent E. coli cells (in 40 NI of appropriate media) are transformed with 3 NI of ligation mixture and cultured in 80 NI of SOC medium (Sambrook J et al, supra). After incubation for one hour at 37°C, the whole transformation mixture is plated on Luria Bertani (LB)-agar (Sambrook J et al, supra) containing 2×Carb. The following day, several colonies are randomly picked from each plate and cultured in 150 NI of liquid LB/2×Carb medium placed in an individual well of an appropriate, commercially-available, sterile 96-well microtiter plate. The following day, 5 NI
of each overnight culture is transfer-ed into a non-sterile 96-well plate and after dilution 1:10 with water, 5 NI of each sample is transferred into a PCR
array.
For PCR amplification, 18 NI of concentrated PCR reaction mix (3.3×) containing 4 units of rTth DNA polymerise, a vector primer and one or both of the gene specific primers used for the extension reaction are added to each well. Amplification is performed using the following conditions:.
Step 1 94°C for 60 sec Step 2 94°C for 20 sec Step 3 55°C for 30 sec Step 4 72°C for 90 sec Step 5 Repeat steps 2-4 for an additional 29 cycles Step 6 72°C for 180 sec Step 7 4°C (and holding) Aliquots of the PCR reactions are run on agarose gels together with molecular weight markers. The sizes of the PCR products are compared to the original partial cDNAs, and appropriate clones are selected, ligated into plasmid and sequenced.
7: Labeling and use of hybridization probes Hybridization probes derived from SEQ ID N0:2 are employed to screen cDNAs, genomic DNAs or mRNAs. Although the labeling of oligonucleotides, consisting of about 20 base-pairs, is specifically described, essentially the same procedure is used with larger cDNA fragments.
Oligonucleotides are designed using state-of-the-art software such as OLIGO
4.06 (National Biosciences), labeled by combining 50 pmol of each oligomer and 250 mCi of y adenosine triphosphate (Amersham, Chicago IIL) and T4 polynucleotide kinase (DuPont NEN; Boston Mass.). The labeled oligonucleotides are substantially purified with Sephadex G-25 super fine resin column (Pharmacia). A portion containing l07 counts per minute of each of the sense and antisense oligonucleotides is used in a typical membrane based hybridization analysis of human genomic DNA digested with one of the following endonucleases (Ase I, Bgl II, Eco RI, Pst I, Xba 1, or Pvu II; DuPont NEN).
The DNA from each digest is fractionated on a 0.7 percent agarose gel and transferred to nylon membranes (Nytran Plus, Schleicher & Schuell, Durham N.H.). Hybridization is carried out for 16 hours at 40°C To remove nonspecific signals, blots are sequentially washed at room temperature under increasingly stringent conditions up to 0.1 ×saline sodium citrate and 0.5% sodium dodecyl sulfate. After XOMAT AR.TM. film (Kodak, Rochester N.Y.) is exposed to the blots in a Phosphoimager cassette (Molecular Dynamics, Sunnyvale Calif.) for several hours, hybridization patterns are compared visually.
EQUIVALENTS
Although particular embodiments have been disclosed herein in detail, this has been done by way of example for purposes of illustration only, and is not intended to be limiting with respect to the scope of the appended claims that follow. In particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein. Other aspects, advantages, and modifications considered to be within the scope of the following claims.
References U.S. Patent No. 4166452. Apparatus for testing human responses to stimuli.
1979.
U.S. Patent No. 4485045. Synthetic phosphatidyl cholines useful in forming liposomes. 1984.
U.S. Patent No. 4544545. Liposomes containing modified cholesterol for organ targeting. 1985.
4,676,980. Target specific cross-linked heteroantibodies. 1987.
U.S. Patent No. 4816567. Recombinant immunoglobin preparations. 1989.
WO 90/10448. Covalent conjugates of lipid and oligonucleotide. 1990.

WO 90/13641. Stably transformed eucaryotic cells comprisng a foreign transcribable DNA under the control of a pol III promoter. 1990.
EPO 402226. Transformation vectors for yeast Yarrowia. 1990.
WO 91/00360. Bispecific reagents for AIDS therapy. 1991.
WO 91/04753. Conjugates of antisense oligonucleotides and therapeutic uses thereof. 1991.
U.S. Patent No. 5013556. Liposomes with enhanced circulation time. 1991.
WO 91/00357. New strain with filamentous fungi mutants, process for the production of recombinant proteins using said strain, and strains and proteins. 1991.
WO 91/06629. Oligonucleotide analogs with novel linkages. 1991.
WO 92/20373. Heteroconjugate antibodies for treatment of HIV infection.
1992.
WO 93/08829. Compositions that mediate killing of HIV-infected cells. 1993.
WO 94/11026. Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cells. 1994.
WO 96/27011. A method for making heteromultimeric polypeptides. 1996.
U.S. Patent No. 5545807. Production of antibodies from transgenic animals.
1996.

U.S. Patent No. 5545806. Ransgenic <sic> non-human animals for producing heterologous antibodies. 1996.
U.S. Patent No. 5569825. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes. 1996.
WO 97/33551. Compositions and methods for the diagnosis, prevention, and treatment of neoplastic cell growth and proliferation. 1997.
U.S. Patent No. 5633425. Transgenic non-human animals capable of producing heterologous antibodies. 1997.
U.S. Patent No. 5661016. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes. 1997.
U.S. Patent No. 5625126. Transgenic non-human animals for producing heterologous antibodies. 1997.
(GCG), G.C.G. 1999. Wisconsin Package: SeqLab, SeqWeb, Madison, Wisconsin.
Abravaya, K., J.J. Carrino, S. Muldoon, and H.H. Lee. 1995. Detection of point mutations with a modified ligase chain reaction (Gap- LCR). Nucleic Acids Res. 23:675-82.
ADAReport. 1997. Position of the American Dietetic Association: weight management. J Am Diet Assoc. 97:71-4.
Alam, J., and J.L. Cook. 1990. Reporter genes: Application to the study of mammalian gene transcription. Anal. Biochem. 188:245-254.

Altschul, S.F., W. Gish, W. Miller, E.W. Myers, et al. 1990. Basic local alignment search tool. J Mol Biol. 215:403-10.
Aron, D., J. Findling, and J. Tyrell. 1997. Hypothalamus & Pituitary. In Basic &
Clinical Endocrinology. F. Greenspan and G. Strewler, editors.
Appleton & Lange, Stamford, CT. 95-156.
Austin, C.P., and C.L. Cepko. 1990. Cellular migration patterns in the developing mouse cerebral cortex. Development. 110:713-732.
Ausubel, F.M., R. Brent, R.E. Kingston, D.D. Moore, et al. 1987. Current protocols in molecular biology. John Wiley & Sons, New York.
Barany, F. 1991. Genetic disease detection and DNA amplification using cloned thermostable ligase. Proc Natl Acad Sci U S A. 88:189-93.
Bartel, D.P., and J.W. Szostak. 1993. Isolation of new ribozymes from a large pool of random sequences [see comment]. Science. 261:1411-8.
Beal, P.A., and P.B. Dervan. 1991. Second structural motif for recognition of DNA by oligonucleotide- directed triple-helix formation. Science.
251:1360-3.
Bechtold, N., and G. Pelletier. 1998. In planta Agrobacterium-mediated transformation of adult Arabidopsis thaliana plants by vacuum infiltration. Methods Mol Biol. 82:259-66.
Becker, D.M., and L. Guarente. 1991. High-efficiency transformation of yeast by electroporation. Methods Enzymol. 194:182-187.
Beggs, J.D. 1978. Transformation of yeast by a replicating hybrid plasmid.
Nature. 275:104-109.

Berger, J., J. Hauber, R. Hauber, R. Geiger, et al. 1988. Secreted placental alkaline phosphatase: A powerful new qunatitative indicator of gene expression in eukaryotic cells. Gene. 66:1-10.
WO 93/04169. GENE TARGETING IN ANIMAL CELLS USING ISOGENIC
DNA CONSTRUCTS. 1993.
Bodine, D.M., K.T. McDonagh, N.E. Seidel, and A.W. Nienhuis. 1991. Survival and retrovirus infection of murine hematopoietic stem cells in vitro:
effects of 5-FU and method of infection. Exp. Hematol. 19:206-212.
Boerner, P., R. Lafond, W.Z. Lu, P. Grams, et al. 1991. Production of antigen-specific human monoclonal antibodies from in vitro-primed human splenocytes. J Immunol. 147:86-95.
U.S. Patent No. 3,773,91'9. Polylactide-drug mixtures. 1973.
Bouchard, C. 1995. Genetics of obesity: an update on molecular markers. Int J Obes Relat Metab Disord. 19 Suppl 3:S10-3.
Bradley. 1987. Teratocarcinomas and Embryonic Stem Cells: A Practical Approach. Oxford University Press, Inc., Oxford. 268 pp.
Bradley, A. 1991. Modifying the mammalian genome by gene targeting. Curr Opin 8iotechnol. 2:823-9.
Brennan, M., P.F. Davison, and H. Paulus. 1985. Preparation of bispecific antibodies by chemical recombination of monoclonal immunoglobulin G1 fragments. Science. 229:81-3.

Capecchi, M.R. 1980. High efficiency transformation by direct microinjection of DNA into cultured mammalian cells. Cell. 22:479.
Capecchi, M.R. 1989. Altering the genome by homologous recombination.
Science. 244:1288-92.
Carell, T., E.A. Wintner, and J. Rebek Jr. 1994a. A novel procedure for the synthesis of libraries containing small organic molecules. Angewandte Chemie International Edition. 33:2059-2061.
Carell, T., E.A. Wintner, and J. Rebek Jr. 1994b. A solution phase screening procedure for the isolation of active compounds from a molecular library. Angewandte Chemie International Edition. 33:2061-2064.
Caron, P.C., W. Laird, M.S. Co, N.M. Avdalovic, et al. 1992. Engineered humanized dimeric forms of IgG are more effective antibodies. J Exp Med. 176:1191-5.
Carter, P. 1986. Site-directed mutagenesis. Biochem J. 237:1-7.
Case, M.E., M. Schweizer, S.R. Kushner, and N.H. Giles. 1979. Efficient transformation of Neurospora crassa by utilizing hybrid plasmid DNA.
Proc Natl Acad Sci U S A. 76:5259-63.
U.S. Patent No. 5,116,742. RNA ribozyme restriction endoribonucleases and methods. 1992.
U.S. Patent No. 4,987,071. RNA ribozyme polymerises, dephosphorylases, restriction endoribonucleases and methods. 1991.

Cepko, C.L., B.E. Roberts, and R.E. Mulligan. 1984. Construction and applications of a highly transmissible murine retrovirus shuttle vector.
Cell. 37:1053-1062.
Chalfie, M., Y. tu, G. Euskirchen, W.W. Ward, et al. 1994. Green fluorescent protein as a marker for gene expression. Science. 263:802-805.
Chaney, W.G., D.R. Howard, J.W. Pollard, S. Sallustio, et al. 1986. High-frequency transfection of CHO cells using Polybrene. Somatic Cell Mol.
Genet. 12:237.
Charon, C., S. Krief, F. Diot-Dupuy, A.D. Strosberg, et al. 1995. Early alterations in the brown adipose tissue adenylate cyclase system of pre-obese Zucker rat fa/fa pups: decreased G-proteins and beta 3-adrenoceptor activities. Biochem J. 312:781-8.
Chen, C., and H. Okayama. 1988. Calcium phosphate-mediated gene transfer: A highly efficient system for stably transforming cells with plasmid DNA. BioTechniques. 6:632-638.
Chen, S.H., H. D. Shine, J.C. Goodman, R.G. Grossman, et al. 1994. Gene therapy for brain tumors: regression of experimental gliomas by adenovirus-mediated gene transfer in vivo. Proc Natl Acad Sci U S A.
91:3054-7.
Chikano, S., K. Sawada, T. Shimoyama, S.I. Kashiwamura, et al. 2000. IL-18 and IL-12 induce intestinal inflammation and fatty liver in mice in an IFN-gamma dependent manner. Gut. 47:779-86.
Cho, C.Y., E.J. Moran, S.R. Cherry, J.C. Stephans, et al. 1993. An unnatural biopolymer. Science. 261:1303-5.

Choubey, D., and P. Lengyel. 1992. Interferon action: nucleolar and nucleoplasmic localization of the interferon-inducible 72-kD protein that is encoded by the Ifi 204 gene from the gene 200 cluster. J Cell Biol.
116:1333-41.
Choubey, D., and P. Lengyel. 1993. Interferon action: cytoplasmic and nuclear localization of the interferon-inducible 52-kD protein that is encoded by the Ifi 200 gene from the gene 200 cluster. J Interferon Res. 13:43-52.
Choubey, D., J. Snoddy, V. Chaturvedi, E. Toniato, et al. 1989. Interferons as gene activators. Indications for repeated gene duplication during the evolution of a cluster of interferon-activatable genes on murine chromosome 1. J Biol Chem. 264:17182-9.
Clement, K., C. Vaisse, N. Lahlou, S. Cabrol, et al. 1998. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction [see comments]. Nature. 392:398-401.
Cohen, A.S., D.L. Smisek, and B.H. Wang. 1996. Emerging technologies for sequencing antisense oligonucleotides: capillary electrophoresis and mass spectrometry. Adv Chromatogr. 36:127-62.
Cohen, J.S. 1989. Oligodeoxynucleotides: Antisense inhibitors of gene expression. CRC Press, Boca Raton, FL. 255 pp.
Cohen, S.M.N., A.C.Y. Chang, and L. Hsu. 1972. Nonchromosomal antibiotic resistance in bacteria: Genetic transformation of Escherichia coli by R-factor DNA. Proc. Natl. Acad. Sci. USA. 69:2110.
Collins, S., K.W. Daniel, and E.M. Rohlfs. 1999. Depressed expression of adipocyte beta-adrenergic receptors is a common feature of congenital and diet-induced obesity in rodents. Int J Obes Relat Metab Disord.
23:669-77. . . .. . . . .. . . , .
Coombes, R.C., N.J. Rothwell, P. Shah, and M.J. Stock. 1987. Changes in thermogenesis and brown fat activity in response to tumour necrosis factor in the rat. Biosci Rep. 7:791-9.
Cooney, M., G. Czemuszewicz, E.H. Postel, S.J. Flint, et al. 1988. Site-specific oligonucleotide binding represses transcription of the human o-myc gene in vitro. Science. 241:456-9.
Cotton, R.G. 1993. Current methods of mutation detection. Mutat Res.
285:125-44.
Cronin, M.T., R.V. Fucini, S.M. Kim, R.S. Masino, et al. 1996. Cystic fibrosis mutation detection by hybridization to light-generated DNA probe arrays. Hum Mutat. 7:244-55.
Cull, M.G., J.F. Miller, and P.J. Schatz. 1992. Screening for receptor ligands using large libraries of peptides linked to the C terminus of the lac repressor. Proc Natl Acad Sci U S A. 89:1865-9.
Cwirla, S.E., E.A. Peters, R.W. Barrett, and W.J. Dower. 1990. Peptides on phage: a vast library of peptides for identifying ligands. Proc Natl Acad Sci U S A. 87:6378-82.
Dawson, M.J., N.J. Elwood, R.W. Johnstone, and J.A. Trapani. 1998. The IFN-inducible nucleoprotein IFI 16 is expressed in cells of the monocyte lineage, but is rapidly and markedly down-regulated in other myeloid precursor populations. J Leukoc Biol. 64:546-54.

de Boer, A.G. 1994. Drug absorption enhancement: Concepts, possibilities, limitations and trends. Harwood Academic Publishers, Langhome, PA.
de Louvencourt, L., H. Fukuhara, H. Heslot, and M. Wesolowski. 1983.
Transformation of Kluyveromyces lactis by killer plasmid DNA. J
Bacteriol. 154:737-42.
De Maeyer, E., and J. De Maeyer-Guignard. 1998. Type I interferons. Int Rev Immunol. 17:53-73.
de Wet, J.R., K.V. Wood, M. DeLuca, D.R. Helinski, et al. 1987. Sturcture and expression in mammalian cells. Mol. Cell Biol. 7:725-737.
Demerec, M., E.A. Adelberg, A.J. Clark, and P.E. Hartman. 1966. A proposal for a uniform nomenclature in bacterial genetics. Genetics. 54:61-76.
Denjean, F., J. Lachuer, A. Geloen, F. Cohen-Adad, et al. 1999. Differential regulation of uncoupling protein-1, -2 and -3 gene expression by sympathetic innervation in brown adipose tissue of thermoneutral or cold-exposed rats. FEBS Lett. 444:181-5.
Devlin, J.J., L.C. Panganiban, and P.E. Devlin. 1990. Random peptide libraries: a source of specific protein binding molecules. Science.
249:404-6.
DeWitt, S.H., J.S. Kiely, C.J. Stankovic, M.C. Schroeder, et al. 1993.
"Diversomers": an approach to nonpeptide, nonoligomeric chemical diversity. Proc Natl Acad Sci U S A. 90:6909-13.
DeWys, W.D., C. Begg, P.T. Lavin, P.R. Band, et al. 1980. Prognostic effect of weight loss prior to chemotherapy in cancer patients. Eastern Cooperative Oncology Group. Am J Med. 69:491-7.

Dunlop, J., and S. Rosenzweig-Lipson. 1998. Therapeutic approaches to obesity. Exp. Opin. Ther. Patents. 8:1683-1694.
Eichelbaum, M., and B. Evert. 1996. Influence of pharmacogenetics on drug disposition and response. Clin Exp Pharmacol Physiol. 23:983-5.
Ellington, A.D., and J.W. Szostak. 1990. In vitro selection of RNA molecules that bind specific ligands. Nature. 346:818-22.
Elroy-Stein, O., and B. Moss. 1990. Cytoplasmic expression system based on constitutive synthesis of bacteriophage T7 RNA polymerase in mammalian cells. Proc. Natl. Acad. Sci. USA. 87:6743-6747.
US Patent No. 4,522,811. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides Serial injection of muramyldipeptides and~liposomes enhances the anti-infective activity of muramyldipeptides. 1985.
Eppstein, D.A., Y.V. Marsh, M. van der Pas, P.L. Felgner, et al. 1985.
Biological activity of liposome-encapsulated murine interferon gamma is mediated by a cell membrane receptor. Proc Natl Acad Sci U S A.
82:3688-92.
Escudero, J., and B. Hohn. 1997. Transfer and integration of T-DNA without cell injury in the host plant. Plant Cell. 9:2135-2142.
U.S. Patent No. 4,870,009. Method of obtaining gene product through the generation of transgenic animals. 1989.

Fekete, D.M., and C.L. Cepko. 1993. Retroviral infection coupled with tissue transplantation limits gene transfer in the chick embryo. Proc. Natl.
Acad. Sci. USA. 90:2350-2354.
Felgner, P.L., T.R. Gadek, M. Holm, R. Roman, et al. 1987. Lipofectin: A
highly efficient, lipid-mediated DNA/transfection procedure. Proc. Natl.
Acad. Sci. USA. 84:7413-7417.
Felici, F., L. Castagnoli, A. Musacchio, R. Jappelli, et al. 1991. Selection of antibody ligands from a large library of oligopeptides expressed on a multivalent exposition vector. J Mol Biol. 222:301-10.
Fieck, A., D.L. Wyborski, and J.M. Short. 1992. Modifications of the E.coli Lac repressor for expression in eukaryotic cells: effects of nuclear signal sequences on protein activity and nuclear accumulation. Nucleic Acids Res. 20:1785-91.
Finer, J.J., K.R. Finer, and T. Ponappa. 1999. Particle bombardment-mediated transformation. Current Topics in microbiology and immunology. 240:59-80.
Finn, P.J., N.J. Gibson, R. Fallon, A. Hamilton, et al. 1996. Synthesis and properties of DNA-PNA chimeric oligomers. Nucleic Acids Res.
24:3357-63.
Fishwild, D.M., S.L. O'Donnell, T. Bengoechea, D.V. Hudson, et al. 1996.
High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice [see comments]. Nat Biotechnol.
14:845-51.

Fleer, R., P. Yeh, N. Amellal, I. Maury, et al. 1991. Stable multicopy vectors for high-level secretion of recombinant human serum albumin by Kluyveromyces yeasts. Biotechnology (N Y). 9:968-75.
Fodor, S.P., R.P. Rava, X.C. Huang, A.C. Pease, et al. 1993. Multiplexed biochemical assays with biological chips. Nature. 364:555-6.
Foellmi-Adams, L.A., B.M. Wyse, D. Hen-on, J. Nedergaard, et al. 1996.
Induction of uncoupling protein in brown adipose tissue. Synergy between norepinephrine and pioglitazone, an insulin-sensitizing agent.
Biochem Pharmacol. 52:693-701.
Friedman, J.M. 1993. Obesity. Brown fat and yellow mice [news; comment].
Nature. 366: 720-1.
Fromm, M., L.P. Taylor, and V. Walbot. 1985. Expression of genes transferred into monocot and dicot plant cells by electroporation. Proc. Natl. Acad.
Sci. USA. 82:5824-5828.
Fujita, T., H. Shubiya, T. Ohashi, K. Yamanishi, et al. 1986. Regulation of human interleukin-2 gene: Functional DNA sequences in the 5' flanking region for the gene expression in activated T lymphocytes.
Cell. 46:401-407.
Gabizon, A., R. Shiota, and D. Papahadjopoulos. 1989. Pharmacokinetics and tissue distribution of doxorubicin encapsulated in stable liposomes with long circulation times. J Natl Cancer Inst. 81:1484-8.
Gallagher, S.R. 1992. GUS protocols: Using the GUS gene as a reporter of gene expression. Academic Press, San Diego, CA.

Gallop, M.A., R.W. Barrett, W.J. Dower, S.P. Fodor, et al. 1994. Applications of.combinatorial technologies to drug discovery. 1. Background and peptide combinatorial libraries. J Med Chem. 37:1233-51.
Gasparini, P., A. Bonizzato, M. Dognini, and P.F. Pignatti. 1992. Restriction site generating-polymerise chain reaction (RG-PCR) for the probeless detection of hidden genetic variation: application to the study of some common cystic fibrosis mutations. Mol Cell Probes. 6:1-7.
Gautier, C., F. Morvan, B. Rayner, T. Huynh-Dinh, et al. 1987. Alpha-DNA. IV:
Alpha-anomeric and beta-anomeric tetrathymidylates covalently linked to intercalating oxazolopyridocarbazole. Synthesis, physicochemical properties and poly (rA) binding. Nucleic Acids Res. 15:6625-41.
Genetics Computer Group_(GCG). 1999. Wisconsin Package. Wisconsin Package, SeqLabO, SeqWeb~, Madison, Wisc.
Gennaro, A.R. 2000. Remington: The science and practice of pharmacy.
Lippincott, Williams & Wilkins, Philadelphia, PA.
Gibbs, R.A., P.N. Nguyen, and C.T. Caskey. 1989. Detection of single DNA
base differences by competitive oligonucleotide priming. Nucleic Acids Res. 17:2437-48.
Gietz, R.D., R.A. Woods, P. Manivasakam, and R.H. Schiestl. 1998. Growth and transformation of Saccharomyces cerevisiae. In Cells: A
laboratory manual. Vol. I. D. Spector, R. Goldman, and L. Leinwand, editors. Cold Spring Harbor Press, Cold Spring Harbor, NY.
Goding, J.W. 1996. Monoclonal antibodies: Principles and Practice.
Academic Press, San Diego. 492 pp.

Gorman, C.M., L.F. Moffat, and B.H. Howard. 1982. Recombinant genomes which express chloramphenicol acetyltransferase in mammalian cells.
Mol. Cell. Biol. 2:1044-1051.
Graham, F.L., and A.J. van der Eb. 1973. A new technique for the assay of infectivity of human adenovirus 5 DNA. Virology. 52:456-.
Griffin, H.G., and A.M. Griffin. 1993. DNA sequencing. Recent innovations and future trends. ApplBiochem Biotechnol. 38:147-59.
Grompe, M., D.M. Muzny, and C.T. Caskey. 1989. Scanning detection of mutations in human omithine transcarbamoylase by chemical mismatch cleavage. Proc Natl Acad Sci U S A. 86:5888-92.
Gruber, M., B.A. Schodin, E.R. Wilson, and D.M. Kranz. 1994. Efficient tumor cell lysis mediated by a bispecific single chain antibody expressed in Escherichia coli. J Immunol. 152:5368-74.
Grunfeld, C., and K.R. Feingold. 1992. Tumor necrosis factor, interleukin, and interferon induced changes in lipid metabolism as part of host defense.
Proc Soc Exp Biol Med. 200:224-7.
Guan, X.M., H. Yu, and L.H. Van der Ploeg. 1998. Evidence of altered hypothalamic pro-opiomelanocortin/ neuropeptide Y mRNA expression in tubby mice. Brain Res Mol Brain Res. 59:273-9.
Guatelli, J.C., K.M. Whitfield, D.Y. Kwoh, K.J. Barringer, et al. 1990.
Isothermal, in vitro amplification of nucleic acids by a multienzyme reaction modeled after retroviral replication. Proc Natl Acad Sci U S A.
87:1874-8.

Guebre-Xabier, M., S. Yang, H.A. Lin, R. Schwenk, et al. 2000. Altered hepatic lymphocyte subpopulations in obesity-related murine fatty livers: potential mechanism for sensitization to liver damage.
Hepatology. 31:633-640.
Gura, T. 1998. Uncoupling proteins provide new clue to obesity's causes [news]. Science. 280:1369-70.
Hanahan, D. 1983. Studies on transformation of Escherichia coli with plasmids. J. Mol. Biol. 166:557-580.
Hansen, G., and M.-D. Chilton. 1999. Lessons in gene transfer to plants by a gifted microbe. Curr. Top. Microbiol. Immunol. 240:21-57.
Hansen, G., and M.S. Wright. 1999. Recent advances in the transformation of plants. Trends Plant Sci. 4:226-231.
Hardman, J., A. Gilman, and L. Limbird. 1996. Goodman &Gilman's The pharmacological basis of therapeutics. McGraw-Hill, New York. 1905 pp.
Harlow, E., and D. Lane. 1988. Antibodies: A laboratory manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor. 726 pp.
Harlow, E., and D. Lane. 1999. Using antibodies: A laboratory manual. Cold Spring Harbor Laboratory PRess, Cold Spring Harbor, New York.
Haseloff, J., and W.L. Gerlach. 1988. Simple RNA enzymes with new and highly specific endoribonuclease activities. Nature. 334:585-91.
Hayashi, K. 1992. PCR-SSCP: A method for detection of mutations. Genetic and Analytical Technipues Applications. 9:73-79.

Helene, C. 1991. The anti-gene strategy: control of gene expression by triplex-forming- oligonucleotides. Anticancer Drug Des. 6:569-84.
Helene, C., N.T. Thuong, and A. Harel-Bellan. 1992. Control of gene expression by triple helix-forming oligonucleotides. The antigene strategy. Ann N Y Acad Sci. 660:27-36.
Himms-Hagen, J. 1969a. The effect of age and cold acclimation on the metabolism of brown adipose tissue in cold-exposed rats. Can J
Biochem. 47:251-6.
Himms-Hagen, J. 1969b. The role of brown adipose tissue in the calorigenic effect of adrenaline and noradrenaline in cold-acclimated rats. J
Physiol (Lond). 205:393-403.
Hinnen, A., J.B. Hicks, and G.R. Fink. 1978. Transformation of yeast. Proc.
Natl. Acad. Sci. USA. 75:1929-1933.
Hoffman, F. 1996. Laser microbeams for the manipulation of plant cells and subcellular structures. Plant Sci. 113:1-11.
Hogan, B., Beddington, R., Costantini, F., Lacy, E. 1994. Manipulating the Mouse Embryo: A Laboratory Manual. Cold Spring Harbor Laboratory Press. 500 pp.
Holliger, P., T. Prospero, and G. Winter. 1993. "Diabodies": small bivalent and bispecific antibody fragments. Proc Natl Acad Sci U S A. 90:6444-8.
Hoogenboom, H.R., A.D. Griffiths, K.S. Johnson, D.J. Chiswell, et al. 1991.
Multi-subunit proteins on the surface of filamentous phage:

methodologies for displaying antibody (Fab) heavy and light chains.
Nucleic Acids Res. 19:4133-7.
Houghten, R.A., J.R. Appel, S.E. Blondelle, J.H. Cuervo, et al. 1992. The use of synthetic peptide combinatorial libraries for the identification of bioactive peptides. Biotechnipues. 13:412-21.
Hsu, I.C., Q. Yang, M.W. Kahng, and J.F. Xu. 1994. Detection of DNA point mutations with DNA mismatch repair enzymes. Carcinogenesis.
15:1657-62.
Huang, J.Y., and D.L. Brutlag. 2001. The EMOTIF database. NucIeicAcids Res. 29:202-4.
Hwang, K.J., K.F. Luk, and P.L. Beaumier. 1980. Hepatic uptake and degradation of unilamellar sphingomyelin/cholesterol liposomes: a kinetic study. Proc Natl Acad Sci U S A. 77:4030-4.
Hyrup, B., and P.E. Nielsen. 1996. Peptide nucleic acids (PNA): synthesis, properties and potential applications. Bioorg Med Chem. 4:5-23.
Inoue, H., Y. Hayase, A. Imura, S. Iwai, et al. 1987a. Synthesis and hybridization studies on two complementary nona(2'-O-methyl)ribonucleotides. NucIeicAcids Res. 15:6131-48.
Inoue, H., Y. Hayase, S. Iwai, and E. Ohtsuka. 1987b. Sequence-dependent hydrolysis of RNA using modified oligonucleotide splints and RNase H.
FEBS Lett. 215:327-30.
Ishiura, M., S. Hirose, T. Uchida, Y. Hamada, et al. 1982. Phage particle-mediated gene transfer to cultured mammalian cells. Molecular and Cellular Biology. 2:607-616.

Ito, H., Y. Fukuda, K. Murata, and A. Kimura. 1983. Transformation of intact yeast cells treated with alkali cations. J. Bacteriol. 153:163-168.
Janeway, C., and P. Travers. 1997. Immunobiology : the immune system in health and disease. Current Biology ;
Garland Pub., London ; San Francisco New York. 1 v. (various pagings).
Jayasena, S.D. 1999. Aptamers: an emerging class of molecules that rival antibodies in diagnostics. Clin Chem. 45:1628-50.
Jones, P.T., P.H. Dear, J. Foote, M.S. Neuberger, et al. 1986. Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature. 321:522-5.
Junqueira, L.C., J. Carneiro, and R.O. Kelly. 1998. Basic Histology. Appleton & Lange, Stamford. 494 pp.
Kapur, S., B. Marcotte, and A. Marette. 1999. Mechanism of adipose tissue iNOS induction in endotoxemia. Am J Physiol. 276:E635-41.
Kato, T., M. Esumi, S. Yamashita, K. Abe, et al. 1992. Interferon-inducible gene expression in chimpanzee liver infected with hepatitis C virus.
Virology. 190:856-60.
Kaufman, R.J. 1990. Vectors used for expression in mammalian cells.
Methods Enzymol. 185:487-511.

Kaufman, R.J., P. Murtha, D.E. Ingolia, C.-Y. Yeung, et al. 1986. Selection and amplification of heterologous genes encoding adenosine deaminase in mammalian cells. Proc. Natl. Acad. Sci. USA. 83:3136-3140.
Kawai, S., and M. Nishizawa. 1984. New procedure for DNA transfection with polycation and dimethyl sulfoxide. MoL Cell. Biol. 4:1172.
Keen, J., D. Lester, C. Inglehearn, A. Curtis, et al. 1991. Rapid detection of single base mismatches as heteroduplexes on Hydrolink gels. Trends Genet. 7:5.
Kelly, J.M., A.C. Porter, Y. Chernajovsky, C.S. Gilbert, et al. 1986.
Characterization of a human gene inducible by alpha- and beta-interferons and its expression in mouse cells. Embo J. 5:1601-6.
Kostelny, S.A., M.S. Cole, and J.Y. Tso. 1992. Formation of a bispecific antibody by the use of leucine zippers. J Immunol. 148:1547-53.
W094/16101. DNA SEQUENCING BY MASS SPECTROMETRY. 1994.
Kozal, M.J., N. Shah, N. Shen, R. Yang, et al. 1996. Extensive polymorphisms observed in HIV-1 Glade B protease gene using high-density oligonucleotide arrays. Nat Med. 2:753-9.
Kozbor, D., P. Tripputi, J.C. Roder, and C.M. Croce. 1984. A human hybrid myeloma for production of human monoclonal antibodies. J Immunol.
133:3001-5.
Kriegler, M. 1990. Gene transfer and expression: A laboratory manual.
Stockton Press, New York. 242 pp.

WO 91/01140. HOMOLOGOUS RECOMBINATION FOR UNIVERSAL
DONOR CELLS AND CHIMERIC MAMMALIAN HOSTS. 1991.
Kwoh, D.Y., G.R. Davis, K.M. Whitfield, H.L. Chappelle, et al. 1989.
Transcription-based amplification system and detection of amplified human immunodeficiency virus type 1 with a bead-based sandwich hybridization format. Proc Natl Acad Sci U S A. 86:1173-7.
US Patent No. 5,223,409. Directed evolution of novel binding proteins. 1993.
Lakso, M., B. Sauer, B. Mosinger, E.J. Lee, et al. 1992. Targeted oncogene activation by site-specific recombination in transgenic mice. Proc Natl Acad Sci U S A. 89:6232-6.
Lam, K.S. 1997. Application of combinatorial library methods in cancer research and drug discovery. Anticancer Drug Design. 12:145-167.
Lam, K.S., S.E. Salmon, E.M. Hersh, V.J. Hruby, et al. 1991. General method for rapid synthesis of multicomponent peptide mixtures. Nature.
354:82-84.
Landegren, U., R. Kaiser, J. Sanders, and L. Hood. 1988. A ligase-mediated gene detection technique. Science. 241:1077-80.
Landolfo, S., M. Gariglio, G. Gribaudo, and D. Lembo. 1998. The Ifi 200 genes: an emerging family of IFN-inducible genes. Biochimie. 80:721-8.
WO 90/11354. Process for the specific replacement of a copy of a gene present in the receiver genome via the integration of a gene. 1990.
U.S. Patent No. 4,736,866. Transgenic non-human animals. 1988.

Leduc, N., and e. al. 1996. Isolated maize zygotes mimic in vivo embryogenic development and express microinjected genes when cultured in vitro.
Dev. Biol. 10:190-203.
Lee, J.S., D.A. Johnson, and A.R. Morgan. 1979. Complexes formed by (pyrimidine)n . (purine)n DNAs on lowering the pH are three-stranded.
Nucleic Acids Res. 6:3073-91.
Lee, V.H.L. 1990. Peptide and protein drug delivery. Marcel Dekker, New York, NY.
Lemaitre, M., B. Bayard, and B. Lebleu. 1987. Specific antiviral activity of a poly(L-lysine)-conjugated oligodeoxyribonucleotide sequence complementary to vesicular stomatitis virus N protein mRNA initiation site. Proc Natl Acad Sci U S A. 84:648-52.
Lembo, M., C. Sacchi, C. Zappador, G. Bellomo, et al. 1998. Inhibition of cell proliferation by the interferon-inducible 204 gene, a member of the Ifi 200 cluster. Oncogene. 16:1543-51.
Lemischka, I.R., D.H. Raulet, and R.C. Mulligan. 1986. Developmental potential and dynamic behavior of hematopoietic stem cells. Cell.
45:917-927.
Letsinger, R.L., G.R. Zhang, D. K. Sun, T. Ikeuchi, et al. 1989. Cholesteryl-conjugated oligonucleotides: synthesis, properties, and activity as inhibitors of replication of human immunodeficiency virus in cell culture.
Proc Natl Acad Sci U S A. 86:6553-6.
Li, E., T.H. Bestor, and R. Jaenisch. 1992. Targeted mutation of the DNA
methyltransferase gene results in embryonic lethality. Cell. 69:915-26.

Linder, M.W., R.A. Prough, and R. Valdes. 1997. Pharmacogenetics: a laboratory tool for optimizing therapeutic efficiency. Clin Chem. 43:254-66.
Littlefield, J.W. 1964. Selection of hybrids from matings of fibroblasts in vitro and their presumed recombinants. Science. 145:709-710.
Lizardi, P.M., C.E. Guerra, H. Lomeli, I. Tussle-Luna, et al. 1988.
Exponential amplification of recombinant-RNA hybridization probes. Biotechnology.
6:1197-1202.
Lonberg, N., and D. Huszar. 1995. Human antibodies from transgenic mice.
Int Rev Immunol. 13:65-93.
Lonberg, N., L.D. Taylor, F.A. Harding, M. Trounstine, et al. 1994. Antigen-specific human antibodies from mice comprising four distinct genetic modifications (see comments]. Nature. 368:856-9.
Lopata, M.A., D.W. Cleveland, and B. Sollner-Webb. 1984. High-level expression of a chloramphenicol acetyltransferase gene by DEAEdextran-mediated DNA traansfection couled with a dimethylsulfoxide or glycerol shock treatment. Nucleic Acids Research.
12:5707.
Lowell, B.B., S.S. V, A. Hamann, J.A. Lawitts, et al. 1993. Development of , obesity in transgenic mice after genetic ablation of brown adipose tissue [see comments]. Nature. 366:740-2.
Luckow, V.A. 1991. Cloning and expression of heterologous genes in insect cells with baculovirus vectors. In Recombinant DNA technology and applications. A. Prokop, R.K. Bajpai, and C. Ho, editors. McGraw-Hill, New York. 97-152.
Maher, L.J. 1992. DNA triple-helix formation: an approach to artificial gene repressors? Bioessays. 14:807-15.
Mandel, M., and A. Higa. 1970. Calcium-dependent bacteriophage DNA
infection. J. Mol. biol. 53:159-162.
Marasco, W.A., W.A. Haseltine, and S.Y. Chen. 1993. Design, intracellular expression, and activity of a human anti-human immunodeficiency virus type 1 gp120 single-chain antibody. Proc Natl Acad Sci U S A.
90:7889-93.
Marks, J.D., A.D. Griffiths, M. Malmqvist, T.P. Clackson, et al. 1992. By-passing immunization: building high affinity human antibodies by chain shuffling. Biotechnology (N Y). 10:779-83.
Marks, J.D., H.R. Hoogenboom, T.P. Bonnert, J. McCafferty, et al. 1991. By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J Mol Biol. 222:581-97.
Martin, F.J., and D. Papahadjopoulos. 1982. Irreversible coupling of immunoglobulin fragments to preformed vesicles. An improved method for liposome targeting. J Biol Chem. 257:286-8.
Maxam, A.M., and W. Gilbert. 1977. A new method for sequencing DNA. Proc Natl Acad Sci U S A. 74:560-4.
Miller, A.D., and C. Buttimore. 1986. Redesign of retrovirus packaging cell lines to avoid recombination leading to helper virus production. Mol.
Cell biol. 6:2895-2902.

Miller, L. K. 1988. Baculoviruses as gene expression vectors. Annu. Rev.
Microbiol. 42:177-199.
Milstein, C., and A.C. Cuello. 1983. Hybrid hybridomas and their use in immunohistochemistry. Nature. 305:537-40.
Min, W., S. Ghosh, and P. Lengyel. 1996. The interferon-inducible p202 protein as a modulator of transcription: inhibition of NF-kappa B, o-Fos, and c-Jun activities. Mol Cell Biol. 16:359-68.
Minvielle-Sebastia, L., P.J. Preker, and W. Kelley. 1994. RNA14 and RNA15 proteins as components of a yeast pre-mRNA 3'-end processing factor.
Science. 266:1702-5.
US Patent No. 5,459,039. Methods for mapping genetic mutations. 1995.
Montague, C.T., I.S. Farooqi, J.P. Whitehead, M.A. Soos, et al. 1997.
Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature. 387:903-8.
Mori, K., K. Fujimoto-Ouchi, T. Ishikawa, F. Sekiguchi, et al. 1996a. Murine interleukin-12 prevents the development of cancer cachexia in a murine model. Int J Cancer. 67:849-55.
Mori, K., K. Fujimoto-Ouchi, T. Ishikawa, F. Sekiguchi, et al. 1996b. Murine interleukin-12 prevents the development of cancer cachexia in a murine model. Int J Cancer. 67:849-55.
Morita, T., T. Sato, H. Nyunoya, A. Tsujimoto, et al. 1993. Isolation of a cDNA
clone encoding DNA-binding protein (TAXREB107) that binds specifically to domain C of the tax-responsive enhancer element in the long terminal repeat of human T-cell leukemia virus type I. AIDS Res Hum Retroviruses. 9:115-21.
Morrison, S.L., L. Wims, S. Wallick, L. Tan, et al. 1987. Genetically engineered antibody molecules and their application. Ann N YAcad Sci. 507:187-98.
US Patent No. 4,683,202. Process for amplifying nucleic acid sequences.
1987.
US Patent No. 4,683,195. Process for amplifying, detecting, and/or cloning nucleic acid sequences. 1987.
Munson, P.J., and D. Rodbard. 1980. Ligand: a versatile computerized approach for characterization of ligand-binding systems. Anal Biochem.
107:220-39.
Myers, R.M., Z. Larin, and T. Maniatis. 1985. Detection of single base substitutions by ribonuclease cleavage at mismatches in RNA:DNA
duplexes. Science. 230:1242-6.
US Patent No. 5,328,470. Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor. 1994.
Naeve, C.W., G.A. Buck, R.L. Niece, R.T. Pon, et al. 1995. Accuracy of automated DNA sequencing: a multi-laboratory comparison of sequencing results. Biotechnipues. 19:448-53.
Nakai, K., and P. Horton. 1999. PSORT: a program for detecting sorting signals in proteins and predicting their subcellular localization. Trends Biochem Sci. 24:34-6.

Nakamura, S., T. Otani, Y. Ijiri, R. Motoda, et al. 2000. IFN-gamma-dependent and -independent mechanisms in adverse effects caused by concomitant administration of IL-18 and IL-12. J Immunol. 164:3330-6.
Nakazawa, H., D. English, P.L. Randell, K. Nakazawa, et al. 1994. UV and skin cancer: specific p53 gene mutation in normal skin as a biologically relevant exposure measurement. Proc Natl Acad Sci U S A. 91:360-4.
Neumann, E., M. Schaefer-Ridder, Y. Wang, and P.H. Hofschneider. 1982.
Gene transfer into mouse lyoma cells by electroporation in high electric fields. EM80 J. 1:841-845.
Nevill-Manning, C.G., T.D. Wu, and D.L. Brutlag. 1998. Highly specific protein sequence motifs for genome analysis. Proc Natl Acad Sci U S A.
95:5865-71.
O'Gorman, S., D.T. Fox, and G.M. Wahl. 1991. Recombinase-mediated gene activation and site-specific integration in mammalian cells. Science.
251:1351-5.
Okano, H., J. Aruga, T. Nakagawa, C. Shiota, et al. 1991. Myelin basic protein gene and the function of antisense RNA in its repression in myelin-deficient mutant mouse. J Neurochem. 56:560-7.
O'Reilly, D.R., L.K. Miller, and V.A. Luckow. 1992. Baculovirus expression vectors. W.H. Freeman and Company, New York. .
Orita, M., H. Iwahana, H. Kanazawa, K. Hayashi, et al. 1989. Detection of polymorphisms of human DNA by gel electrophoresis as single-strand conformation polymorphisms. Proc Natl Acad Sci U S A. 86:2766-70.

Ou-Lee, T.M., R. Turgeon, and R. Wu. 1986. Uptake and expression of a foreign gene linked to either a plant virus or Drosophila promoter in protoplasts of rice, wheat and sorghum. Proc. Natl. Acad. Sci. USA.
83:6815-6819.
Pace, C.N., F. Vajdos, L. Fee, G. Grimsley, et al. 1995. How to measure and predict the molar absorption coefficient of a protein. Protein Sci.
4:2411-23.
Palmer, T.D., R.A. Hock, W.R.A. osborne, and A.D. Miller. 1987. Efficient retrovirus-mediated transfer and expression of a human adenosine deaminase gene in diploid skin fibroblasts from an adenosie-deficient human. Proc. Natl. Acad. Sci. USA. 84:1055-1059.
Palou, A., C. Pico, M.L. Bonet, and P. Oliver. 1998. The uncoupling protein, thermogenin. Int J Biochem Cell Biol. 30:7-11.
Pear, W., G. Nolan, M. Scott, and D. Baltimore. 1993. Production of high-titer helper-free retroviruses by transient transfection. Proc. Natl. Acad. Sci.
USA. 90:8392-8396.
Perry-O'Keefe, H., X.W. Yao, J.M. Coull, M. Fuchs, et al. 1996. Peptide nucleic acid pre-gel hybridization: an alternative to southern hybridization. Proc Natl Acad Sci U S A. 93:14670-5.
Perusse, L., and C. Bouchard. 1999. Role of genetic factors in childhood obesity and in susceptibility to dietary variations. Ann Med. 31 Suppl 1:19-25.
Petersen, K.H., D.K. Jensen, M. Egholm, O. Buchardt, et al. 1976. A PNA-DNA linker synthesis of N-((4,4'-dimethoxytrityloxy)ehtyl)-N-(thymin-1-ylacetyl)glycine. Biorganic and Medicianl Chemistry Letters. 5:1119-1124.
Pi-Sunjer, F., and N.O.E.I.E. Panel. 1998. Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults-The Evidence Report. National Institutes of Health.
Pomp, D. 1997. Genetic dissection of obesity in polygenic animal models.
Behav Genet. 27:285-306.
Potter, H. 1988. Electroporation in biology: Methods, applications" and instrumentation. Analytical Biochemistry. 174:361-373.
Potter, H., L. Weir, and P. Leder. 1984. Enhancer-dependent expression of human kappa immunoglobulin genes introduced into mouse pre-B
lymphocytes by electroporation. Proc. NatL Acad. Sci. USA. 81:7161-7165.
Presta, L.G. 1992. Antibody engineering. Curr Opin Biotechnol. 3:394-8.
Prosser, J. 1993. Detecting single-base mutations. Trends Biotechnol.
11:238-46.
Rasmussen, U.B., C. Wolf, M.G. Mattei, M.P. Chenard, et al. 1993.
Identification of a new interferon-alpha-inducible gene (p27) on human chromosome 14q32 and its expression in breast carcinoma. Cancer Res. 53:4096-101.
Rassoulzadegan, M., B. Binetruy, and F. Cuzin. 1982. High frequency of gene transfer after fusion befinreen bacteria and eukaryotic cells. Nature.
295:257.

Reisfeld, R.A., and S. Sell. ~~1985. Monoclonal antibodies and cancer therapy:
Proceedings of the Roche-UCLA symposium held in Park City, Utah, January 26-February 2, 1985. Alan R. Liss, New York. 609 pp.
Rhodes, C.A., D.A. Pierce, I.J. Mettler, D. Mascarenhas, et al. 1988.
Genetically transformed maize plants from protoplasts. Science.
240:204-207.
Riechmann, L., M. Clark, H. Waldmann, and G. Winter. 1988. Reshaping human antibodies for therapy. Nature. 332:323-7.
Rose, J.K., L. Buonocore, and M. Whitt. 1991. A new cationic liposome reagent mediating nearly quantitative transfection of animal cells.
BioTechniques. 10:520-525.
Rossi, J.J. 1994. Practical ribozymes. Making ribozymes work in cells. Curr Biol. 4:469-71.
Rossiter, B.J., and C.T. Caskey. 1990. Molecular scanning methods of mutation detection. J Biol Chem. 265:12753-6.
Saiki, R.K., T.L. Bugawan, G.T. Horn, K.B. Mullis, et al. 1986. Analysis of enzymatically amplified beta-globin and HLA-DQ alpha DNA with allele-specific oligonucleotide probes. Nature. 324:163-6.
Saiki, R.K., P.S. Walsh, C.H. Levenson, and H.A. Erlich. 1989. Genetic analysis of amplified DNA with immobilized sequence-specific oligonucleotide probes. Proc Natl Acad Sci U S A. 86:6230-4.
Saleeba, J.A., and R.G. Cotton. 1993. Chemical cleavage of mismatch to detect mutations. Methods Enzymol. 217:286-95.

Sambrook, J. 1989. Molecular cloning: a laboratory manual. Cold Spring Harbor Laboratory, Cold Spring Harbor.
Sandri-Goldin, R.M., A.L. Goldin, J.C. Glorioso, and M. Levine. 1981. High-frequency transfer of cloned herpes simjplex virus type I sequences to mammalian cells by protoplast fusion. Mol. Cell. Biol. 1:7453-752.
Sanger, F., S. Nicklen, and A.R. Coulson. 1977. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci U S A. 74:5463-7.
Saunders, J.A., B.F. Matthews, and P.D. Miller. 1989. Plant gene transfer using electrofusion and electroporation. In Electroporation and electrofusion in cell biology. E. Neumann, A.E. Sowers, and C.A.
Jordan, editors. Plenum Press, New York. 343-354.
Savontaus, E., J. Rouru, O. Boss, R. Huupponen, et al. 1998. Differential regulation of uncoupling proteins by chronic treatments with beta 3-adrenergic agonist BRL 35135 and metformin in obese fa/fa Zucker rats. 8iochem Biophys Res Commun. 246:899-904.
Sbarbati, A., F. Leclercq, F. Osculati, and I. Gresser. 1995. Interteron alpha/beta-induced abnormalities in adipocytes of suckling mice. Biol Cell. 83:163-7.
Schade, R., C. Staak, C. Hendriksen, M. Erhard, et al. 1996. The production of avian (egg yold) antibodies: IgY. The report and recommendations of ECVAM workshop. Alternatives to Laboratory Animals (ATLA).
24:925-934.
Schaffner, W. 1980. Direct transfer of cloned genes from bacteria to mammalian cells. Proc. Natl. Acad. Sci. USA. 77:2163.

Schook, L.B. 1987. Monoclonal antibody production techniques and applications. Marcel Dekker, Inc., New York. 336 pp.
Schrauwen, P., K. Walder, and E. Ravussin. 1999. Human uncoupling proteins and obesity. Obes Res. 7:97-105.
Scott, J.K., and G.P. Smith. 1990. Searching for peptide ligands with an epitope library. Science. 249:386-90.
Selden, R.F., K. Burke-Howie, M.E. Rowe, H.M. Goodman, et al. 1986.
Human growth hormone as a reporter gene in regulation studies employing transient gene expression. Molecular and Cellular Biololgy.
6:3173-3179.
Shalaby, M.R., H.M. Shepard, L. Presta, M.L. Rodrigues, et al. 1992.
Development of humanized bispecific antibodies reactive with cytotoxic lymphocytes and tumor cells overexpressing the HER2 protooncogene.
J Exp Med. 175:217-25.
Shigekawa, K., and W.J. Dower. 1988. Electroporation of eukaryotes and prokaryotes: A general approach to the introduction of macomolecules into cells. BioTechnipues. 6:742-751.
Shillito, R. 1999. Methods of genetic transformations: Electroporation and polyethylene glycol treatment. In Molecular improvement of cereal crop. I. Vasil, editor. Kluwer, Dordrecht, The Netherlands. 9-20.
Shilo, B.Z., and R.A. Weinberg. 1981. DNA sequences homologous to vertebrate oncogenes are conserved in Drosophila melanogaster. Proc Natl Acad Sci U S A. 78:6789-92.

Shopes, B. 1992. A genetically engineered human IgG mutant with enhanced cytolytic activity. J Immunol. 148:2918-22.
Simonsen, C.C., and A. D. Levinson. 1983. Isolation and expression of an altered mouse dihydrofolate reductase cDNA. Proc. Natl. Acid. Sci.
USA. 80:2495-2499.
US Patent No. 5,272,057. Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerise. 1993.
Southern, P.J., and P. Berg. 1982. Transformation of mammalian cells to antibiotic resistanced with a bacterial gene under control of the SV40 early region promoter. J. Mol. Appl. Gen. 1:327-341.
Spiegelman, B.M., and J.S. Flier. 1996. Adipogenesis and obesity: rounding out the big picture. Cell. 87:377-89.
Sreekrishna, K., R.H. Potenz, J.A. Cruze, W.R. McCombie, et al. 1988. High level expression of heterologous proteins in methylotrophic yeast Pichia pastoris. J Basic Microbiol. 28:265-78.
Stein, C.A., and J.S. Cohen. 1988. Oligodeoxynucleotides as inhibitors of gene expression: a review. Cancer Res. 48:2659-68.
Stein, L., L. Kruglyak, D. Slonim, and E. Lander. 1995. RHMAPPER, unpublished software. Whitehead Institute/Massachusetts Institute of Technology Center for Genome Research, Cambridge, MA.
Stevenson, G.T., A. Pindar, and C.J. Slide. 1989. A chimeric antibody with dual Fc regions (bisFabFc) prepared by manipulations at the IgG
hinge. Anticancer Drug Des. 3:219-30.

Suresh, M.R., A.C. Cuello, and C. Milstein. 1986. Bispecific monoclonal antibodies from hybrid hybridomas. Methods Enzymol. 121:210-28.
Tannenbaum, C.S., J. Major, Y. Ohmori, and T.A. Hamilton. 1993. A
lipopolysaccharide-inducible macrophage gene (D3) is a new member of an interferon-inducible gene cluster and is selectively expressed in mononuclear phagocytes. J Leukoc Biol. 53:563-8.
5,861,485. Polypeptides involved in body weight disorders, including obesity.' 1999.
Thomas, K.R., and M.R. Capecchi. 1987. Site-directed mutagenesis by gene targeting in mouse embryo-derived stem cells. Cell. 51:503-12.
Thompson, J.A., and e. al. 1995. Maize transformation utilizing silicon carbide whiskers: A review. Euphytica. 85:75-80.
Tisdale, M.J. 1999. Wasting in cancer. J Nutr. 129:243S-246S.
Touraev, A., and e. al. 1997. Plant male germ line transformation. Plant J.
12:949-956.
Traunecker, A., F. Oliveri, and K. Karjalainen. 1991. Myeloma based expression system for production of large mammalian proteins. Trends Biotechnol. 9:109-13.
Trick, H.N., and e. al. 1997. Recent advances in soybean transformation.
Plant Tissue Cult. Biotechnol. 3:9-26.

Tuerk, C., and L. Gold. 1990. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerise.
Science. 249: 505-10.
Turner, D.L., E.Y. Snyder, and C.L. Cepko. 1990. Lineage-independent determinationh of cell type in the embryonic mouse retina. Neuron.
4:833-845.
Tutt, A., G.T. Stevenson, and M.J. Glennie. 1991. Trispecific F(ab')3 derivatives that use cooperative signaling via the TCR/CD3 complex and CD2 to activate and redirect resting cytotoxic T cells. J Immunol.
147:60-9.
van der Krol, A.R., J.N. Mol, and A.R. Stuitje. 1988b. Modulation of eukaryotic gene expression by complementary RNA or DNA sequences.
Biotechniques. 6:958-76.
van der Krol, A.R., J.N. Mol, and A.R. Stuitje. 1988x. Modulation of eukaryotic gene expression by complementary RNA or DNA sequences.
Biotechniques. 6:958-76.
Van Zwieten, P.A., K. L. Kim, A.J. Pijl, M.G. Hendriks, et al. 1996.
Hypertensive diabetic rats in pharmacological studies. Pharmacol Res.
33:95-105.
Verhoeyen, M., C. Milstein, and G. Winter. 1988. Reshaping human antibodies: grafting an antilysozyme activity. Science. 239:1534-6.
Vitetta, E.S., R.J. Fulton, R.D. May, M. Till, et al. 1987. Redesigning nature's poisons to create anti-tumor reagents. Science. 238:1098-104.

Voelker, R.A., W. Gibson, J.P. Graves, J.F. Sterling, et al. 1991. The Drosophila suppressor of sable gene encodes a polypeptide with regions similar to those of RNA-binding proteins. Mol Cell Biol. 11:894-905.
US Patent No. 4,873,191. Genetic transformation of zygotes. 1989.
Wang, H., G. Chatterjee, J.J. Meyer, C.J. Liu, et al. 1999. Characteristics of three homologous 202 genes (Ifi202a, Ifi202b, and Ifi202c) from the murine interferon-activatable gene 200 cluster. Genomics. 60:281-94.
Weigle, D.S., and J.L. Kuijper. 1996. Obesity genes and the regulation of body fat content. Bioessays. 18:867-74.
Wells, J.A., M. Vasser, and D.B. Powers. 1985. Cassette mutagenesis: an efficient method for generation of multiple mutations at defined sites.
Gene. 34:315-23.
Wexler, B.C., S.G. lams, and J.P. McMurtry. 1980. Pathophysiological differences between obese and non-obese spontaneously hypertensive rats. Br J Exp Pathol. 61:195-207.
Whitt, M.A., L. Buonocore, J.K. Rose, V. Ciccarone, et al. 1990. TransfectACE
reagent promotes transient transfection frequencies greater than 90%.
Focus. 13:8-12.
Wigler, M., A. Pellicer, S. Silversttein, and R. Axel. 1978. Biochemical transfer of single-copy eucaryotic genes using total cellular DNA as donor. Cell.
14:725.

Williams, D.A., I.R. Lemischka, D.G. Nathan, and R.C. Mulligan. 1984.
Introduction of a new genetic material into pluripotent haematopoietic stem cells of the mouse. Nature. 310:476-480.
Wilmut, I., A.E. Schnieke, J. McWhir, A.J. Kind, et al. 1997. Viable offspring derived from fetal and adult mammalian cells. Nature. 385:810-3.
Wolff, E.A., G.J. Schreiber, W. L. Cosand, and H.V. Raff. 1993. Monoclonal antibody homodimers: enhanced antitumor activity in nude mice.
Cancer Res. 53:2560-5.
Wong, T.K., and E. Neumann. 1982. Electric field mediated gene transfer.
Biochemical and Biophysical Research Communications. 107:584-587.
Wyborski, D.L., L.C. DuCoeur, and J.M. Short. 1996. Parameters affecting the use of the lac repressor system in eukaryotic cells and transgenic animals. Environ Mol Mutagen. 28:447-58.
Wyborski, D.L., and J.M. Short. 1991. Analysis of inducers of the E.coli lac repressor system in mammalian cells and whole animals. Nucleic Acids Res. 19:4647-53.
Yaswen, L., N. Diehl, M.B. Brennan, and U. Hochgeschwender. 1999. Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin. Nat Med. 5:1066-1070.
Zhou, G., and et al. 1983. Introduction of exogenous DNA into cotton embryos. Methods Enzymol. 101:433-481.
Zoller, M.J., and M. Smith. 1987. Oligonucleotide-directed mutagenesis: a simple method using two oligonucleotide primers and a single-stranded DNA template. Methods Enzymol. 154:329-50.

Zon, G. 1988. Oligonucleotide analogues as potential chemotherapeutic agents. Pharm Res. 5:539-49.
Zuckermann, R.N., E.J. Martin, D.C. Spellmeyer, G.B. Stauber, et al. 1994.
Discovery of nanomolar ligands for 7-transmembrane G-protein-coupled receptors from a diverse N-(substituted)glycine peptoid library.
J Med Chem. 37:2678-85.

SEQUENCE LISTING
<110> Lewin, David Adams, Sean <120> IF1206, A NOVEL INTERFERON-INDUCED POLYPEPTIDE, AND
NUCLEIC ACIDS ENCODING THE SAME
<130> 10716/32 <160> 24 <170> PatentIn version 3.0 <210> 1 <211> 1879 <212> DNA
<213> . Interferon inducible polypeptide 206 (IFI206) nucleic acid sequence (mouse) <400> 1 cgattcgaat tcggccacactggccggatcctctagagatccctcgacctcgacccacgc60 gtccgagcac agtgagagacacccagtgctgctcaagaagtgaaacaactctgagagtat120 cctaaccact ggtgtcttcctttataccccatttttcactttctcagttactgaattatc180 tgcctaccta ctcaaaccaagcaggccacttctgttgttgaagatctcagcacctgtaca240 ttgctgccga aattccagggagtataaccaacaacttgaaagatggagaatgaatataag300 agacttgttc tgctggaaggacttgaatgtatcaataagcatca.attcaatttatttaag360 tcattgatgg tcaaagatttaaatctggaagaagacaaccaagagaaatataccacgttt420 cagattgcta acatgatggtaaagaaatttccagctgatgctggattggacaaactgatc480 aacttttgtg aacgtgtaccaactcttaaaaaacgtgctgaaattcttaaaaaagagaga540 tcagaagtaa atcactggaaataaataggcaagaagcaagtcctgcaaca600 caggagaaac cctacatcaa ctacaagccacatgttagcatctgaaagaggcaagacttccacaaccacc660 actgagaccc aggaagagacttccacagcccagtcggggacttccacagctcacgcgggg720 acttctacag caccggcggggactttcacaactcagaaaagaaaaagtaggagagaagaa780 gagactggag tgaaaaagagcaaggcgtctaaggaaccagatcagcctccctgttgtgaa840 gaacccacag ccaggtgccagtcaccaatactccacagctcatcttcagcttcatctaac900 attccttcag ctacgaaccaaaaaccacaaccccagaaccagaacattcccagaggtgct960 gttctccact cagagcccctgacagtgatggtgctcactgcaacagacccgtttgaatat1020 gaatcaccag aacatgaagtaaagaacatgtttcatgctacagtggctacagtgagccag1080 tatttccatg tgaaagttttcaacatcaacttgaaagagaagttcacaaaaaagaatttt1140 atcatcatat ccaattactttgagagcaaaggcatcctggagatcaatgagacttcctct1200 gtgttaaagg ctgatcctgaccaaatgattgaagtgcccaacaatattatcagaaatgca1260 aatgccagtc ctaagatctgtgatattcaaaagggtacttctggagcagtgttctatgga1320 gtgtttacat tacacaagaaaaaagtgaaaacacagaacacaagctatgaaataaaagat1380 ggttcaggaa gtatagaagtggaggggagtggacaatggcacaacatcaactgtaaggaa1440 ggagataagc tccacctcttctgctttcacctgaaaagagaaagaggacaaccaaagtta1500 gtgtgtggag accacagtttcgtcaagatcaaggtcaccaaggctgggaaaaaaaaggaa1560 gcatcaactg tcctgtcaagcacaaaaaatgaagaagaaaataattacccaaaagatgga1620 attaaggtag agatgccagactattcacgtctaaatgacagctttagtagtatatccaag1680 catttaataa ccttcatacctgatttctgattttgtattttcatttgaaaaaatttctta1740 ttgttctgtt tttctatgaaaataaaatttgatttaatttctctactgtaaaaataataa1800 acatgtcttt ttaaagggacatcaaaaaaaaagaaggagggaggggagggggttggtata1860 agaaaaaccg gggcggccg 1879 <210> 2 <211> 475 <212> PRT

<213> Interferon inducible polypeptide IFI206 (mouse) <400> 2 Met Glu Asn Glu 'I~rr Lys Arg Leu Leu Glu LeuGlu Leu Val Gly Cars Ile Asn Lys His Gln Phe Asn Lys Ser Leu ValLys Leu Phe Met Asp Leu Asn Leu Glu Glu Asp Asn Lys err Thr PheGln Gln Glu Thr Ile Ala Asn Met Met Val Lys Lys Ala Asp Ala LeuAsp Phe Pro Gly Lys Leu Ile Asn Phe Cps Glu Arg Thr Leu Lys ArgAla Val Pro Lys Glu Ile Leu Lys Lys Glu Arg Ser Thr Gly Glu SerLeu Glu Val Thr Glu Ile Asn Arg Gln Glu Ala Ser Thr Pro Thr Thr'I'hr Pro Ala Ser Ser His Met Leu Ala Ser Glu Arg Thr Ser Thr ThrThr Gly Lys Thr Glu Thr Gln Glu Glu Thr Ser Thr Ser Gly Thr ThrAla Ala Gln Ser His Ala Gly Thr Ser 'rhr Ala Pro T'hr Phe Thr GlnLys Ala Gly Thr Arg Lys Ser Arg Arg Glu Glu Glu Val Lys Lys LysAla Thr Gly Ser Ser Lys Glu Pro Asp Gln Pro Pro Glu Glu Pro AlaArg Cys Cars Thr Cys Gln Ser Pro Ile Leu His Ser Ser Ala Ser AsnIle Ser Ser Ser Pro Ser Ala Thr Asn Gln Lys Pro Gln Asn Gln IlePro Gln Pro Asn Arg Gly Ala Val Leu His Ser Glu Thr Val Met LeuThr Pro Leu Val Ala Thr Asp Pro Phe Glu Tyr Glu Glu His Glu LysAsn Ser Pro Val Met Phe His Ala Thr Val Ala Thr Gln 'I~rr ValLys Val Ser Phe His Val Phe Asn Ile Asn Leu Lys Glu Thr Lys Lys PheIle Lys Phe Asn Ile Ile Ser Asn Tyr Phe Glu Ser Lys Gly GluIle Asn Thr Ile Leu Glu Ser Ser Val Leu Lys Ala Asp Pro Asp IleGlu Val Asn Gln Met Pro Asn Ile Ile Arg Asn Ala Asn Ala Ser IleCps Asp Gln Pro Lys Ile Lys Gly Thr Ser Gly Ala Val Phe Tyr PheThr Leu Lys Gly Val His Lys Lys Val Lys Thr Gln Asn Thr Ser IleLys Asp Ser Tyr Glu Gly Gly Ser Ile Glu Val Glu Gly Ser Gly HisAsn Ile Cars Gln Trp Asn Lys Glu Gly Asp Lys Leu His Leu Phe HisLeu Lys Glu Cars Phe Arg Arg Gly Gln Pro Lys Leu Val Cys Gly SerPhe Val Ile Asp His Lys Lys Val Thr Lys Ala Gly Lys Lys Lys SerThr Val Ser Glu Ala Leu Ser Thr Lys Asn Glu Glu Glu Asn Asn LysAsp Gly Lys Tyr Pro Ile Val Glu Met Pro Asp Tyr Ser Arg Leu SerPhe Ser Ile Asn Asp Ser Ser Lys His Leu Ile Thr Phe Ile Pro Asp Phe <210> 3 <211> 1840 <212> DNA

<213> Interferon-inducible polypeptidevariant 206 (IFI206b) nucleic acid sequence (mouse) <400> 3 cgattcgaat tcggccacac tggccggatc ctctagagat ccctcgacct cgacccacgc 60 gtccgagcac agtgagagac acccagtgct gctcaagaag tgaaacaact ctgagagtat 120 cctaaccact ggtgtcttcc tttatacccc atttttcact ttctcagtta ctgaattatc 180 tgcctaccta ctcaaaccaa tctgttgttgaagatctcagcacctgtaca240 gcaggccact ttgctgccga aattccagggagtataaccaacaacttgaaagatggagaatgaatataag300 5 agacttgttc tgctggaaggacttgaatgtatcaataagcatcaattcaatttatttaag360 tcattgatgg tcaaagatttaaatctggaagaagacaaccaagagaaatataccacgttt420 cagattgcta acatgatggtaaagaaatttccagctgatgctggattggacaaactgatc480 aacttttgtg aacgtgtaccaactcttaaaaaacgtgctgaaattcttaaaaaagagaga540 tcagaagtaa caggagaaacatcactggaaataaataggcaagaagcaagtcctgcaaca600 cctacatcaa ctacaagccacatgttagcatctgaaagaggcgagacttccacaacccag660 gaagagactt ccacagccca gtccgggcct tcgacagctc ctgcgcggac tttaacagcc 720 cagaaaagaa agaagaagagactggagtgaaaaagagcaa ggcgtctaag780 aaagtaggag gaaccagatc agcctccctgttgtgaagaacccacagccaggtgccagtc accaatactc840 cacagctcat cttcagcttcatctaacattccttcagctacgaaccaaaa accacaaccc900 cagaaccaga acattcccagaggtgctgttctccactcagagcccctgac agtgatggtg960 ctcactgcaa cagacccgtttgaatatgaatcaccagaacatgaagtaaa gaacatgttt1020 catgctacag tggctacagtgagccagtatttccatgtgaaagttttcaa catcaacttg1080 aaagagaagt tcacaaaaaagaattttatcatcatatccaattactttga gagcaaaggc1140 atcctggaga tcaatgagacttcctctgtgttaaaggctgatcctgacca aatgattgaa1200 gtgcccaaca atattatcagaaatgcaaatgccagtcctaagatctgtga tattcaaaag1260 ggtacttctg gagcagtgttctatggagtgtttacattacacaagaaaaa agtgaaaaca1320 cagaacacaa gctatgaaataaaagatggttcaggaagtatagaagtgga ggggagtgga1380 caatggcaca acatcaactgtaaggaaggagataagctccacctcttctg ctttcacctg1440 aaaagagaaa gaggacaaccaaagttagtgtgtggagaccacagtttcgt caagatcaag1500 gtcaccaagg ctgggaaaaaaaaggaagcatcaactgtcctgtcaagcac aaaaaatgaa1560 gaagaaaata attacccaaaagatggaattaaggtagagatgccagacta tcacgtctaa1620 atgacagctt tagtagtatatccaagcatttaataaccttcatacctgat ttctgatttt1680 gtattttcat ttgaaaaaatttcttattgttctgtttttctatgaaaata aaatttgatt1740 taatttctct actgtaaaaataataaacatgtctttttaaagggacatca aaaaaaaaga1800 aggagggagg ggagggggttggtataagaaaaaccggggc 1840 <210> 4 <211> 445 <212> PRT
<213> Interferon inducible polypeptide IFI206 variant (IFI206b) <400> 4 Met Glu Asn Glu Tyr Lys Arg Leu'Val Leu Leu Glu Gly Leu Glu Cps Ile Asn Lys His Gln Phe Asn Leu Phe Lys Ser Leu Met Val Lys Asp 20 Leu Asn Leu Glu Glu Asp Asn Gln Glu Lys Tyr Thr Thr Phe Gln Ile Ala Asn Met Met Val Lys Lys Phe Pro Ala Asp Ala Gly Leu Asp Lys Leu Ile Asn Phe Cars Glu Arg Val Pro Thr Leu Lys Lys Arg Ala Glu Ile Leu Lys Lys Glu Arg Ser Glu Val Thr Gly Glu Thr Ser Leu Glu Ile Asn Arg Gln Glu Ala Ser Pro Ala Thr Pro Thr Ser Thr Thr Ser His Met Leu Ala Ser Glu Arg Gly Glu Thr Ser Thr Thr Gln Glu Glu Thr Ser Thr Ala Gln Ser Gly Pro Ser Thr Ala Pro Ala Arg Thr Leu Thr Ala Gln Lys Arg Lys Ser Arg Arg Glu Glu Glu Thr Gly Val Lys Lys Ser Lys Ala Ser Lys Glu Pro Asp Gln Pro Pro Cars Cars Glu Glu 16s 170 175 Pro Thr Ala Arg Cars Gln Ser Pro Ile Leu His Ser Ser Ser Ser Ala Ser Ser Asn Ile Pro Ser Ala Thr Asn Gln Lys Pro Gln Pro Gln Asn Gln Asn Ile Pro Arg Gly Ala Val Leu His Ser Glu Pro Leu Thr Val Met Val Leu Thr Ala Thr Asp Pro Phe Glu Tyr Glu Ser Pro Glu His Glu Val Lys Asn Met Phe His Ala Thr Val Ala Thr Val Ser Gln Tyr Phe His Val Lys Val Phe Asn Ile Asn Leu Lys Glu Lys Phe Thr Lys Lys Asn Phe Ile Ile Ile Ser Asn Tyr Phe Glu Ser Lys Gly Ile Leu Glu Ile Asn Glu Thr Ser Ser Val Leu Lys Ala Asp Pro Asp Gln Met Ile Glu Val Pro Asn Asn Ile Ile Arg Asn Ala Asn Ala Ser Pro Lys Ile Cps Asp Ile Gln Lys Gly Thr Ser Gly Ala Val Phe Tyr Gly Val Phe Thr Leu His Lys Lys Lys Val Lys Thr Gln Asn Thr Ser Tyr Glu Ile Lys Asp Gly Ser Gly Ser Ile Glu Val Glu Gly Ser Gly Gln Trp His Asn Ile Asn Cars Lys Glu Gly Asp Lys Leu His Leu Phe Cps Phe His Leu Lys Arg Glu Arg Gly Gln Pro Lys Leu Val C'ys Gly Asp His 385 390 395 400' Ser Phe Val Lys Ile Lys Val Thr Lys Ala Gly Lys Lys Lys Glu Ala Ser Thr Val Leu Ser Ser Thr Lys Asn Glu Glu Glu Asn Asn Tyr Pro Lys Asp Gly Ile Lys Val Glu Met Pro Asp Tyr His Val <210> 5 <211> 30 <212> DNA
<213> muIFIhlog.probel <400> 5 tcctgcacac ctacatcaac tacaagccac 30 <210> 6 <211> 18 <212> PRT
<213> IFI motif <220>
<221> X
<222> (2) . . (2) <223> Phe, Tyr, Trp, Leu, or Ile <220>
<221> X
<222> (8) . . (8) <223> Thr, Ala, Ser <220>
<221> X
<222> (9) . . (9) <223> Any <220>
<221> X
<222> (10) .. (10) <223> Ser, Thr, Lys, or Arg <220>
<221> X
<222> (11) .. (11) <223> Glu or Gln <220>
<221> X
<222> (12) .. (12) <223> Phe, Tyr, or Txp <220>
<221> X
<222> (13) .. (13) <223> Phe, Tyr, or Trp <220>
<221> X
<222> (14) .. (14) <223> His, Arg, Lys, Phe, Tyr, or Trp <220>
<221> X
<222> (16) .. (16) <223> Lys, Arg, Met, Leu, or Ile <220>
<zzl> x <2zz> (la) . . (1s) <223> Phe, Tyr, Leu, or Ile <400> 6 Met Xaa His Ala Thr Val Ala Xaa Xaa Xaa Xaa Xaa Xaa Xaa Val Xaa Val Xaa <210> 7 <211> 7 <212> PRT

<213> IFI motif <220>
<221> X
<222> (1)..(1) <223> Phe or Tyr <220>
<221> X
<222> (7)..(7) <223> Ser or Thr <400> 7 Xaa His Ala Thr Val Ala Xaa <210> s <211> 35 <212> PRT
<213> IFI motif <220>
<221> X

<222> (2)..(2) <223> Any <220>

<221> X

<222> (3)..(3) <223> Any <220>

<221> X

<222> (4) . . (4) <223> Glu, Lys, or Gln <220>

<221> X

<222> (7) . . (7) <223> Any <220>

<221> X

<222> (8)..(8) <223> Ile, Leu, or Val <220>

<221> X

<222> (9)..(9) <223> Ile, Leu, or Val <220>
<221> X
<222> (12) .. (12) <223> Any <z2o>
<221> X
<222> (14) .. (14) <223> Phe, Leu, or Tyr <220>
<221> X
<222> (15) .. (15) <223> Asp or Glu <220>
<221> X
<222> (16) .. (16) <223> Any <220>
<221> X
<222> (17) .. (17) <223> Ile, Leu, Met or Val <220>
<221> X
<222> (18) .. (18) <223> Any <2zo>
<221> X
<222> (19)..(19) <223> Any <220>
<221> X
<222> (20) .. (20) <223> Any <220>
<221> X
<222> (21) .. (21) <223> Any <220>
<221> X
<222> (22) .. (22) <223> Phe, Leu or err <220>
<221> X
<222> (23) .. (23) <223> Any <220>
<221> X
<222> (24) . . (24) <223> Any <220>
<221> X
<222> (25) .. (25) <223> Phe, Ile, Leu, Met, or Val <220>
<221> X
<222> (27) .. (27) <223> Any <220>
<z21> x <222> (28) .. (28) <223> Phe, Leu, or 'I~rr <220>
<221> X
<222> (29) .. (29) <223> Ile, Leu, Met, or Val <220>
<221> X
<222> (30) .. (30) <223> Any <220>
5 <221> X
<222> (31) .. (31) <223> Any <220>
<221> X
<222> (32) .. (32) <223> Asp or Glu <220>
<221> x <222> (33) .. (33) <223> Phe, Leu, or T~rr <220>
<221> x <222> (34) .. (34) <223> Any <220>
<221> x <222> (35) . . (35) <223> Ile, Leu, or Val <400> s Met Xaa Xaa Xaa Xaa Tyr Lys Xaa Xaa Xaa Leu Leu Xaa Gly Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Lys Xaa Xaa Xaa Xaa Xaa Xaa xaa xaa Xaa <210> 9 10 <211> 14 <212> PRT

<213> IFI motif <220>
20 <221> X
<222> (1) . . (1) <223> Phe, Leu, or Tyr <220>
<221> X
<222> (2)..(2) <223> Any <220>
<221> X
<222> (3)..(3) <223> Any <220>
<z21> x <222> (4) . . (4) <223> Phe, Ile, Leu, Met, or Val <z2o>
<221> X
<222> (6)..(6) <223> Any <220>
<221> X
<222> (7)..(7) <223> Phe, Leu, or 'I~r <220>
<221> x <222> (8) . . (8) <223> Ile, Leu, Met, or Val <220>
<221> X
<222> (9) . . (9) <223> Any <220>
<221> X
<222> (10) .. (10) <223> Any <220>
<221> X

<222> (11) .. (11) <223> Asp or Glu <220>
<221> X
<222> (12) .. (12) <223> Phe, Leu, or Tyr <220>
<221> X
<222> (13) .. (13) <223> Any <220>
<221> X
<222> (14) .. (14) <223> Ile, Leu, or Val <400> 9 Xaa Xaa Xaa Xaa Lys Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 1 s to <210> to <211> 48 <212> PRT
<213> IFI motif <220>
<221> X

<222> (1) . . (1) <223> Lys, Gln, or Arg <220>

<221> X

<222> (5)..(5) <223> Any <220>

<221> X

<222> (6)..(6) <223> Any <220>

<221> X

<222> (7) . . (7) <223> Phe, Ile, Leu, Met, or Val <220>

<221> X

<222> (8) . . (8) <223> Lys, Gln or Arg <220>
<221> X
<222> (9) . . (9) <223> Ile, Leu, or Val <220>
<221> X
5 <222> (10) . . (10) <223> Ala, Ser, or Thr <220>
<221> X
<222> (11) . . (11) <223> Asp or Asn <220>
<221> X
<222> (12) .. (12) <223> Any <220>
<221> X
<222> (14) .. (14) <223> Any <220>
<221> X
<222> (15) .. (15) <223> Any <220>
<221> X
<222> (18)..(18) <223> Any <220>
<221> X
<222> (19) .. (19) <223> Any <220>
<221> X
<222> (20) .. (20) <223> Any <220>
<221> X
<222> ~ (21) .. (21) <223> Ala or Ser <220>
<221> X
<222> (22) .. (22) <223> Any <2zo>
<221> X
<222> (24) .. (24) <223> Any <220>

<221> X
<222> (28) .. (28) <223> Any <220>
<221> X
<222> (29) .. (29) <223> Phe, Ile, Leu, Met, or Val <220>
<221> X
<222> (30) . . (30) <223> Any <220>
<221> X
<222> (31) . . (31) <223> Glu, Lys, or Gln <220>
<221> X
<222> (32) .. (32) <223> Any <220>
<221> X
<222> (33) .. (33) <223> Ile, Leu, Met or Val <220>
<221> X
<222> (34) .. (34) <223> Any <220>
<z21> x <222> (35) .. (35) <223> Any <220>
<221> x <222> (37) .. (37) <223> Glu, Lys, Gln or Arg <220>
<221> X
<222> (38) . . (38) <223> Any <zzo>
<221> x <222> (39) .. (39) <223> Any <220>
<221> x <222> (40) .. (40) <223> Any <220>
<221> X
<222> (41) .. (41) <223> Any <220>
<221> X
<222> (42) .. (42) <223> Any <220>
<221> X
<222> (43) .. (43) <223> Any <220>
<221> X
<222> (45) .. (45) <223> Lys or Arg <zzo>
<221> x <222> (46) .. (46) <223> Any <220>
<221> X
5 <222> (48) .. (48) <223> Lys or Arg <220>
<221> X
<222> (3) . . (3) <223> Any <400> 10 Xaa Glu Xaa Tyr Xaa Xaa XaaXaa Xaa Xaa Xaa Xaa Xaa Xaa Met Lys Phe Xaa Xaa Xaa Xaa Leu LysLeu Ile Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Leu Xaa Xaa XaaXaa Xaa Leu Xaa Glu Xaa Xaa Xaa Xaa <210> 11 <211> 11 <212> PRT

<213> IFI motif <220>

<221> X

<222> (1) . . (1) <223> Lys, Gln, or Arg <220>
<221> X

<222> (3) . . (3) <223> Any <220>
<221> X
<222> (5)..(5) <223> Any <220>

<221> X

<222> (6)..(6) <223> Any <220>

<221> X

<222> (7)..(7) <223> Phe, Ile, Leu, Met or Val <220>

<221> X

<222> (8) . . (8) <223> Lys, Gln, or Arg <220>

<221> X

<222> (9) . . (9) <223> Ile, Leu or Val <z2o>
<221> X
<222> (10) .. (10) <223> Ala, Ser, or Thr <z2o>
<221> X
<222> (11) . . (11) <223> Asp or Asn <400> 11 Xaa Glu Xaa Tyr Xaa Xaa Xaa Xaa Xaa Xaa Xaa <210> 12 <211> 18 <212> PRT
<213> IFI motif <220>
<221> X
<222> (2) . . (2) <223> Any <220>
<221> X
<222> (3) . . (3) <223> Thr, Gly, Val, or Ser <220>
<221> X
<222> (4) . . (4) <223> Thr, Ala, Ser, or Glu <220>
<221> X
<222> (5) . . (5) <223> Gln, Lys, Ala, Glu, or Arg <220>
<221> X
<222> (6) . . (6) <223> Lys or Arg <220>
<221> X
<222> (7)..(7) <223> Arg or Lys <220>
<221> X
<222> (8)..(8) <223> Lys, Arg, Val, or Asn <220>
<221> X
<222> (9) . . (9) <223> Any <220>
<221> X
<222> (10) .. (10) <223> Any <220>
<221> X
<222> (11) .. (11) <223> Any <220>
<221> X
<222> (12) .. (12) <223> Any <220>
<221> X
<222> (13) .. (13) <223> Glu, Gln, Lys, Arg, Ile or Leu <220>
<221> X
<222> (14)..(14) <223> Any <220>

<221> X
<222> (15) .. (15) 5 <223> Any <220>
<221> x <222> (16) .. (16) <223> Any <220>
<221> X
<222> (17) .. (17) <223> Any <400> 12 Thr Xaa Xaa Xaa Xaa Xaa Xaa xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Lys <210> 13 <211> 9 <212> PRT

<213> IFI motif <220>

<221> X

<222> (2)..(2) <223> Lys, Arg, Glu or Gln <220>
<221> X
<222> (3)..(3) <223> Any <220>
<221> X
<222> (5) . . (5) <223> Asp, Glu, Ser, Thr, Asn, or Gln <220>
<221> X
<222> (6) . . (6) <223> Lys, Arg, Thr, or Ser <220>
<221> X
<222> (7) . . (7) <223> Leu, Ile, or Val <220>
<221> X
<222> (8) . . (8) <223> Any <400> 13 Cys Xaa Xaa Gly Xaa Xaa Xaa Xaa Leu <210> 14 <211> 2118 < 212 > DIdA
<213> Interferon inducible polypeptide 206 variant (IFI206c) nucleotide sequence (mouse) <400> 14 agcacagtga gagacacccagtgctgctcaagaagtgaaacaactctgagagtatcctaa60 ccactggtgt cttcctttataccccatttttcactttctcagttactgaattatctgcct120 acctactcaa accaagcaggccacttctgttgttgaagatctcagcacctgtacattgct180 gccgaaattc cagggagtataaccaacaacttgaaaaatggagaatgaatataagagact240 tgttctgctg gaaggacttgaatgtatcaataagcatcaattcaatttatttaagtcatt300 gatggtcaaa gatttaaatctggaagaagacaaccaagagaaatataccacgtttcagat360 tgctaacatg atggtaaagaaatttccagctgatgctggattggacaaactgatcaactt420 ttgtgaacgt gtaccaactcttaaaaaacgtgcagaaattcttaaaaaagagagatcaga480 agtaacagga gaaacatcactggaaataaataggcaagaagcaggtcctgcaacacctac540 atcaactaca agccacatgttagcatctgaaagaggcgagacttccgcaacccaggaaga600 gacttccaca ggccagaaaaggaagccaggtggagagattaggtctgtctcccagccaag660 gccagtcagg aaccagaggggagctgggctggcaaggaaaggttggggtgtgctggctga720.

aggagagaaa ggagagaaaggagagaaaggaaagaaggaaggagagaaagaaagaaagaa780 agaaaggaag gaaggaaggaaggaagaaagaaaaagaaagaaagaaagaaagaaagaaag840 aaagaaagaa agaaagaaagaaagaaagaaagaaagaaagacagaccacaggtttgtcat900 CttCagCCtC CaggtttgtCatCttCagCCtccaggtttgtcatcttcagcctccaggtt960 tgtcatcttc agcctccaggtttgtcatcttcagcctccaggtttgtcatcttcagcctc1020 caggtttgtc atcttcagcctccaggtttgtcatcttcagcctccaggtttgtcatcttc1080 agcctccagg tttgtcatcttcagcctccacaggtttgtcatcttcagcctccaggtagg1140 tggggtaggc tctggctctgtgtcctgcctttagagactagcacaccagcaaaccaaatt1200 cccatctcgt cagagtagcagtaagggcaagcccaggggggtagtgtgccacccagtgac1260 ccattgatcc ttgggtaatggtcctctctgtccataaggctcaggagtcacagaaggtcc1320 i agctatctca accccacactcttgggaacacctccccgcctttttagaacagtaagttct1380 ctgtggcctc atgctgttctgagagccccttggtgctgccacttctccctgtgctctctc1440 attcccttct gcttcctgcacatctgctgaacccacgtcatttccggtactgcctagtta1500 gtcctggaaa aaactctcttggccattggcaggaatcagtgtagaaaagtttgcaggaca1560 tccctggctt tccagagcatgcagaatcagtgtagctcatgacactgtcagacactttag1620 acacgagaga aattcttaagagacctacgcctttgacctctcagatggcacggccgctgt1680 acacagggaa gtgttcactttccttgagacgggaagctggcttcaggttcctatggaata1740 gagttttctt tccttattcccttttcacctaacagttttgctcttcagacagctgcccat1800 tccctaagcc tcgcctagaaaccataacacagatgtacctagatgaatgagccaagcaac1860 tgagaaacag caaggaaactggaaggcttgaggtgggaatatgaaggtcaagacaagaat1920 tagggagctg aaaagatggctcatcagttgactgctcttccagaggtcctgagttcaatt1980 cccagcaacc acatgatggctcgcaaccatctataataggatccacacactcttctggtg2040 tgtctgaaga cagctacagtgtactcataataaataaagtaaataaatttaaaaaaaaaa2100 aaaaaatgga gaatgaat 2118 <210> 15 <211> 318 <212> PRT
<213> Interferon inducible polypeptide 206 variant (IFI206c) (mouse) <400> 15 Met Glu Asn Glu Tyr Lys Arg Leu Val Leu Leu Glu Gly Leu Glu Cps Ile Asn Lys His Gln Phe Asn Leu Phe Lys Ser Leu Met Val Lys Asp Leu Asn Leu Glu Glu Asp Asn Gln Glu Lys Tyr Thr Thr Phe Gln Ile Ala Asn Met Met Val Lys Lys Phe Pro Ala Asp Ala Gly Leu Asp Lys Leu Ile Asn Phe Cps Glu Arg Val Pro Thr Leu Lys Lys Arg Ala Glu Ile Leu Lys Lys Glu Arg Ser Glu Val Thr Gly Glu Thr Ser Leu Glu Ile Asn Arg Gln Glu Ala Gly Pro Ala Thr Ser Thr Thr Ser Thr Pro loo l05 llo His Met Leu Ala Ser Glu Arg Gly Glu Ala Thr Gln Glu Glu Thr Ser Thr Ser Thr Gly Gln Lys Arg Lys Pro Glu Ile Arg Ser Val Gly Gly Ser Gln Pro Arg Pro Val Arg Asn Gln Ala Gly Leu Ala Arg Arg Gly Lys Gly Trp Gly Val Leu Ala Glu Gly Gly Glu Lys Gly Glu Glu Lys Lys Gly Lys Lys Glu Gly Glu Lys Glu Lys Glu Arg Lys Glu Arg Lys Gly Arg Lys Glu Glu Arg Lys Arg Lys Arg Lys Lys Glu Arg Lys Glu Lys Lys Glu Arg Lys Lys Glu Arg Lys Arg Lys Thr Asp His Lys Glu Arg Phe Val Ile Phe Ser Leu Gln Val Leu Gln Pro Pro Gly Cars His Leu Ser Ser Ser Ala Ser Arg Phe Val Ser Leu Gln Val Cys Ile Phe His Leu Gln Pro Pro Gly Leu Ser Ser Ser Arg Phe Val Ile Ser Ala Phe Ser Leu Gln Val Cars His Leu Gln Gly Leu Ser Ser Ser Pro Pro Ala Ser Arg Phe Val Ile Phe Ser Leu Phe Val Ile Phe Ser His Arg Leu Gln Val Gly Gly Val Gly Ser Gly Ser Cys Leu Ser Val <210> 16 , <211> 35 <212> DNA

<213> IFI206.snr1 PCR oligo) ' <400> 16 catcatgtta gcaatctgaa acgtggtata tttet 35 <210> 17 <211> 31 <212> DNA
<213> IFI206.snr1 PCR oligo) <400> 17 15 gtaaagaaat ttccagctga tgctggattg g 31 <210> 18 20 <211> 36 <212> DNA
<213> IFI206.p1 probe <400> la cttcctgggt tgcggaagtc tcgcctcttt cagatg 36 <210> 19 <211> 24 <212> DNA
<213> muIFIhlog.forl <400> 19 tggaaataaa taggcaagaa agca 24 <210> 20 <211> 24 <212> DNA
<213> muIFIhlog.revl <400> 20 tctcgccttc tttcagatgt aaca 24 <210> 21 <211> 2353 <212> DNA

<213> Interferon (IFI204) inducible polypeptide <400> 21 tttctcattt actgacttatctgcctacctactcaagccaagcaggccacttcttgaccc60 ggtgaaggtc tcaggatctgtacatcactgcagaaatatccaggaaggctcagcaacaac120 ttcaaagatg gtgaatgaatacaagagaattgttctgctgagaggacttgaatgtatcaa180 taagcattat tttagcttatttaagtcattgctggccagagatttaaatctggaaagaga240 caaccaagag caatacaccacgattcagattgctaacatgatggaagagaaatttccagc300 tgattctgga ttgggcaaactgattgagttttgtgaagaagtaccagctcttagaaaacg360 agctgaaatt cttaaaaaagagagatcagaagtaacaggagaaacatcactggaaaaaaa420 tggtcaagaa gcaggtcctgcaacacctacatcaactacaagccacatgttagcatctga480 aagaggcgag acttctgcaacccaggaagagacttccacagctcaggcggggacttccac540 agctcaggcg aggacttccacagctcaggcgaggacttccacagctcaggcgaggacttc600 cacagctcag gcgaggacttccacagctcaggcggggacttccacagcccagaaaagaaa660 aagtatgaga gaagaagagactggagtgaaaaagagcaaggcggctaaggaaccagatca720 gcctccctgt tgtgaagaacccacagccatgtgccagtcaccaatactccacagctcatc780 ttcggcttca tctaacattccttcggctaagaaccaaaaatcacaaccccagaatcagaa840 tattcccaga ggtgctgttctccactcagagcccctgacagtgatggtgctcactgcaac900 agacccattt gaatatgaatcaccagaacatgaagtaaagaacatgcttcatgctacagt960 ggctacagtg agccagtatttccatgtgaaagttttcaacatcaacttgaaagaaaagtt1020 cacaaaaaag aattttatcatcatatccaattactttgagagcaaaggcatcctggagat1080 caatgagact tcctctgtgttagaggctgctcctgaccaaatgattgaagtgcccaacag1140 tattatcaga aatgcaaatg.ccagtcctaagatctgtgatattcaaaagggtacttctgg1200 agcagtgttc tatggagtgtttacattacacaagaaaacagtgaaccgaaagaacacaat1260 ctatgaaata caggaagcatagaagtggtggggagtggaa 1320 aaagatggtt aatggcacaa catcaactgc aaggaaggagataaactccacctcttctgctttcacctgaaaacaattga1380 caggcaacca aagttagtgtgtggagaacacagtttcatcaagatatcaaagagaggaaa1440 tgtaccaaag gagcctgctaaggaagaagatcaccatcatggtcccaaacaagtgatggt1500 gctgaaagta acagaaccatttacatatgacctgaaagaggataaaagaatgtttcatgc1560 taccgtggct actgaaactgagttcttcagagtgaaggtttttgacacagctctaaagag1620 caagttcatc ccaagaaatatcattgccatatcagattattttgggtgcaatgggtttct1680 ggagatatac agagcttcctgtgtctctgatgtgaacgttaatccaagaatggttatctc1740 aaatacactg agacaaagagctaatgcaactcctaaaatttcttatcttttctcacaagc1800 aagggggaca tttgtgagtggagagtacttagtaaataagaaaacggagaggaataaatt1860 catttactat ggaattggagatgatacagggaaaatggaagtggtggtttatggaagact1920 caccaatgtc aggtgtgaaccaggcagtaaactaagacttgtctgctttgaattgacttc1980 cactgaagat gggtggcagctgaggtctgtaaggcacagttacatgcaggtcatcaatgc2040 tagaaagtga aggaaagccactcaacccagactcagtcgggagaacctctctggaaccat2100 acttctgaaa acctgaatgccaatgatatttttttgtggagataagattcaattacagaa2160 aataaatgtg tagaagcctattgaaatatcagtcctataaagattatctcttaattctag2220 gaaatggtat tttcttatatctttacacattt~tctatatctaaattcatttgttgtctct2280 ataacttcta taactgttcaatttgcaatttttatgcctaaaacttataaaaataaattc2340 acacaatttc tgt 2353 <210> 22 <211> 640 <212> PRT
<213> Interferon inducible protein 204 (IFI204) <400> 22 Met Val Asn Glu Tyr Lys Arg Ile val Leu Leu Arg Gly Leu Glu Cps Ile Asn Lys His Tyr Phe Ser Leu Phe Lys Ser Leu Leu Ala Arg Asp Leu Asn Leu Glu Arg Asp Asn ThrIle Ile Gln Glu Gln Gln Tyr Thr Ala Asn Met Met Glu Glu Lys Ala Asp GlyLeu Lys Phe Pro Ser Gly Leu Ile Glu Phe Cars Glu Glu Ala Leu LysArg Glu Val Pro Arg Ala ' Ile Leu Lys Lys Glu Arg Ser Thr Gly ThrSer Glu Glu Val Glu Leu Lys Asn Gly Gln Glu Ala Gly Thr Pro SerThr Ser Pro Ala Thr Thr loo l05 llo His Met Leu Ala Ser Glu Arg Thr Ser ThrGln Glu Gly Glu Ala Glu Thr Ser Thr Ala Gln Ala Gly Thr Ala AlaArg Ser Thr Ser Gln Thr Thr Ala Gln Ala Arg Thr Ser Gln Ala ThrSer Ala Thr Ala Arg Thr Gln Ala Arg Thr Ser Thr Ala Gly Thr ThrAla Lys Gln Ala Ser Gln Arg Lys Ser Met Arg Glu Glu Gly Val LysSer Ala Glu Thr Lys Lys 180 1s5 190 Ala Lys Glu Pro Asp Gln Pro Cys Glu ProThr Met Pro Cys Glu Ala cys Gln Ser Pro Ile Leu His Ser Ser SerSer Ile Ser Ser Ala Asn Pro Ser Ala Lys Asn'Gln Lys Pro Gln GlnAsn Pro Ser Gln Asn Ile Arg Gly Ala Val Leu His Ser Leu Thr MetVal Thr Glu Pro Val Leu Ala Thr Asp Pro Phe Glu 'I~rr Pro Glu GluVal Asn Glu Ser His Lys Met Leu His Ala Thr Val Ala Ser Gln PheHis Lys Thr Val 'I~rr Val Val Phe Asn Ile Asn Leu Lys Phe Thr LysAsn Ile Glu Lys Lys Phe Ile Ile Ser Asn Tyr Phe Glu Gly Ile GluIle Glu Ser Lys Leu Asn Thr Ser Ser Val Leu Glu Ala Asp Gln IleGlu Pro Ala Pro Met Val Asn Ser Ile Ile Arg Asn Ala Asn Pro Ile Cars Ile Ala Ser Lys Asp ' Gln Lys Gly Thr Ser Gly Ala Val Gly Phe Thr His Phe Tyr Val Leu Lys Lys Thr Val Asn Arg Lys Asn Tyr Ile Lys Gly Thr Ile Glu Asp Ser Gly Ser Ile Glu Val Val Gly Lys His Asn Asn Ser Gly Trp Ile Cars Lys Glu Gly Asp Lys Leu His Cps His Leu Thr Leu Phe Phe Lys Ile Asp Arg Gln Pro Lys Leu Val Glu Ser Phe Lys Cys Gly His Ile 420 ' 425 430 Ile Ser Lys Arg Gly Asn Val Pro Pro Lys Glu Asp Lys Glu Ala Glu His His His Gly Pro Lys Gln Val Leu Val Thr Pro Met Val Lys Glu Phe Thr Tyr Asp Leu Lys Glu Asp Met ~:is Ala Val Lys Arg Phe Thr Ala Thr Glu Thr Glu Phe Phe Arg Val Asp Thr Leu Val Lys Phe Ala Lys Ser Lys Phe Ile Pro Arg Asn Ala Ser Asp Phe Ile Ile Ile Tyr Gly Cars Asn Gly Phe Leu Glu Ala Cars Val Asp Ile Tyr Arg Ser Ser Val Asn Val Asn Pro Arg Met Val Asn Leu Arg Arg Ile Ser Thr Gln Ala Asn Ala Thr Pro Lys Ile Ser Phe Gln Ala Gly Tyr Leu Ser Arg Thr Phe Val Ser Gly Glu Tyr Leu Lys Thr Glu Asn Val Asn Lys Arg Lys Phe Ile Tyr Tyr Gly Ile Gly Thr Lys Met Val Asp Asp Gly Glu Val Val Tyr Gly Arg Leu Thr Asn Cars Pro Gly Lys Val Arg Glu Ser Leu Arg Leu Val Cars Phe Glu Thr Asp Gly Gln Leu Thr Ser Glu Trp Leu Arg Ser Val Arg His Ser 'I~r Met Gln Val Ile Asn Ala Arg Lys <210> 23 <211> 1623 <212> DNA
10 <213> Interferon-inducible polypeptide 205D3 (IFI205D3) <400> 23 15 cctgcctacc tactcaagccaagcaggccacttcttgacccggtgaaggtctcaggatct60 gtacatcact gcagaaatatccaggaaggctcagcaacaacttcaaagatggtgaatgaa120 tacaagagaa ttgttctgctgagaggacttgaatgtatcaataagcattattttagctta180 tttaagtcat tgctggccagagatttaaatctggaaagagacaaccaagagcaatacacc240 acgattcaga ttgctaacatgatggaagagaaatttccagctgattctggattgggcaaa300 25 ctgattgagt tttgtgaagaagtaccagctcttagaaaacgagctgaaattcttaaaaaa360 gagagatcag aagtaacaggagaaacatcactggaaaaaaatggtcaagaagcaggtcct420 gcaacaccta catcaactacaagccacatgttagcatctgaaagaggcgagacttctgca480 acccaggaag agacttccacagctcaggcggggacttccacagctcaggcggggacttcc540 acagctcagg cggggacttccacagcccagaaaagaaaaagtatgagagaagaagagact600 35 ggagtgaaaa agagcaaggcggctaaggaaccagatcagcctccctgttgtgaagaaccc660 acagccatgt gccagtcaccaatactccacagctcatcttcggcttcatctaacattctt720 tcggctaaga accaaaaatcacaaccccagaaccagaacattcccagaggtgctgttctc780 cactcagagc ccctgacagtgatggtgctcactgcaacagacccgtttgaatatgaatca840 ccagaacatg aagtaaagaacatgtttcatgctacagtggctacagtgagccagtatttc900 catgtgaaag ttttcaacatcgatttgaaagagaagttcacaaaaaataattttatcacc960 atatccaatt actttgagagcaaaggcatcctggagatcaatgagacttcctctgtgtta1020 gaggctgctc ctaaacaaatgattgaagtgcccaactgtattaccagaaatgcaaatgcc1080 agtcctaaga tctgtgatattcaaaagggtacttctggaacagtgttctatggagtgttt1140 acattacaca agaaaaaagtgaaaacacagaacacaagctatgaaataaaagatggttca1200 ggaaggatag aagttgtggggagtggacaatggcacaacatcaactgtaaggaaggagat1260, aagctccacc tcttctgctt tcacctgaaaagagaaagaggacaaccaaa 1320 gttagtgtgt ggagaccaca gtttcgtcaa ggtcaccaaggctgggaaaaaaaaagaagcatcaactgtc1380 cagtgaagca caaaaaatga agaagaaaatgattacccaaaagttggaattaaggtagag1440 atgccaaaat agaaatgtca cctctaaatgacagctttagtagtatatccaaccattgat1500 taatcttcat acctgatttc tgattttgtgttttcatttgaaaaattcttattgttctgt1560 ttttctatga aaataaaatt tgatttcatttctctactgtaaaaataataaacatgtctt1620 ttt 1623 <210> 24 <211> 425 <212> PRT
<213> Interferon inducible protein 205D3 (IFI 205D3) <400> 24 Met Val Asn Glu Tyr Lys Arg Ile Leu Leu Arg LeuGlu Val Gly Cars Ile Asn Lys His Tyr Phe Ser Leu Lys Ser Leu AlaArg Phe Leu Asp Leu Asn Leu Glu Arg Asp Asn Gln Gln Tyr Thr IleGln Glu Thr Ile Ala Asn Met Met Glu Glu Lys Phe Ala Asp Ser LeuGly Pro Gly Lys Leu Ile Glu Phe Cps Glu Glu Val Ala Leu Arg ArgAla Pro Lys Glu Ile Leu Lys Lys Glu Arg Ser Glu Thr Gly Glu SerLeu Val Thr Glu Lys Asn Gly Gln Glu Ala Gly Pro Thr Pro Thr ThrThr Ala Ser Ser His Met Leu Ala Ser Glu Arg Gly Thr Ser Ala GlnGlu Glu Thr Glu Thr Ser Thr Ala Gln Ala Gly Thr Thr Ala Gln GlyThr Ser Ala Ser Thr Ala Gln Ala Gly Thr Ser Thr Gln Lys Arg SerMet Ala Lys Arg Glu Glu Glu Thr Ala Ala Asp Gly Val Lys Lys Glu Lys Ser Lys Pro Gln Pro Pro Cps Cars Glu Glu AlaMet Cys Ser Ile Pro Thr Gln Pro Leu His Ser Ser Ser Ser Ala AsnIle Leu Ala Asn Ser Ser Ser Lys Gln Lys Ser Gln Pro Gln Asn IlePro Arg Ala Leu Gln Asn Gly Val His Ser Glu Pro Leu Thr Val LeuThr Ala Asp Phe Met Val Thr Pro Glu Tyr Glu Ser Pro Glu His LysAsn Met His Thr Glu Val Phe Ala Val Ala Thr Val Ser Gln Tyr ValLys Val Asn Asp Phe His Phe Ile Leu Lys Glu Lys Phe Thr Lys PheIle Thr Ser Tyr Asn Asn Ile Asn Phe Glu Ser Lys Gly Ile Leu AsnGlu Thr Ser Leu Glu Ile Ser Val Glu Ala Ala Pro Lys Gln Met ValPro Asn Ile Arg Ile Glu Cuss Thr Asn Ala Asn Ala Ser Pro Lys AspIle Gln Gly Ser Ile Cps Lys Thr Gly Thr Val Phe Tyr Gly Val LeuHis Lys Lys Lys Phe Thr Lys Val Thr Gln Asn Thr Ser Tyr Glu AspGly Ser Arg Glu Ile Lys Gly Ile .

Val Val Gly Ser Gly Gln Tzp IleAsn Cars Glu Asp His Asn Lys Gly Lys Leu His Leu Phe Cys Phe LysArg Glu Gly Pro His Leu Arg Gln Lys Leu Val Cys Gly Asp His ValLys Val Lys Gly Ser Phe Thr Ala Lys Lys Lys Glu Ala Ser Thr Val Gln

Claims (58)

We claim:
1. An isolated polypeptide comprising an amino acid sequence having at least 89.2% sequence identity to the sequence SEQ ID NO:2, SEQ
ID NO:4 or SEQ ID N0:15.
2. The polypeptide of claim 1, wherein said sequence is the sequence of an active IFI206 polypeptide.
3. The polypeptide of claim 2, wherein said sequence has at least 90% sequence identity to the sequence SEQ ID NO:2, SEQ ID NO:4 or SEQ
ID NO:15.
4. The polypeptide of claim 2, wherein said sequence has at least 98% sequence identity to the sequence SEQ ID NO:2, SEQ ID NO:4 or SEQ
ID NO:15.
5. An isolated polynucleotide encoding the polypeptide of any one of claims 1-4, or a complement of said polynucleotide.
6. An isolated polynucleotide comprising a nucleotide sequence having at least 89.2% sequence identity to the sequence SEQ ID NO:1, SEQ
ID NO:3 or SEQ ID NO:14, or a complement of said polynucleotide.
7. The polynucleotide of claim 5 or 6, wherein said sequence has at least 90% sequence identity to the sequence SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:14, or a complement of said polynucleotide.
8. The polynucleotide of claim 5 or 6, wherein said sequence has at least 98% sequence identity to the sequence SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:14, or a complement of said polynucleotide.
9. An antibody that specifically binds to the polypeptide of the sequences of any one of claims 1-4.
10. A method of quantifying an amount of IFI206 in a composition, comprising:

contacting the antibody of claim 9 with said composition.
11. The method of claim 10, further comprising measuring the amount of said antibody bound to IFI206 in said composition.
12. A method of measuring IFI206 agonist or antagonist activity of a compound, comprising:

contacting said compound with a composition comprising nucleic acid and a polypeptide having at least 89.2% sequence identity to the sequence SEQ ID NO:2, SEQ ID NO:4 or SEQ ID NO:15.
13. The method of claim 12, wherein said nucleic acid is brown adipose tissue mRNA.
14. The method of any one of claims 12 and 13, wherein said polypeptide has at least 90% sequence identity to the sequence SEQ ID
NO:2, SEQ ID NO:4 or SEQ ID NO:15.
15. The method of any one of claims 12 and 13, wherein said polypeptide has at least 98% sequence identity to the sequence SEQ ID
NO:2, SEQ ID NO:4 or SEQ ID NO:15.
16. A method of measuring IF1206 transcription up-regulation or down-regulation activity of a compound, comprising:

measuring the amount of mRNA transcribed in a composition comprising the compound, a RNA polymerise and a polynucleotide of any one of claims 5-8.
17. The method of claim 16, wherein said composition is in a cell.
18. A method of measuring IFI206 translation up-regulation or down-regulation activity of a compound, comprising:

measuring the amount of polypeptide translated in a composition comprising the compound, a ribosome and a polynucleotide of any one of claims 5-8.
19. The method of claim 18, wherein said composition is in a cell.
20. A vector, comprising a polynucleotide of any one of claims 5-8.
21. A cell, comprising the vector of claim 20.
22. A method of screening a subject for an IFI206 related disorder, comprising:

measuring IFI206 gene expression in a tissue sample from the subject.
23. The method of claim 22, wherein said measuring IFI206 gene expression is measuring an amount of IFIO206 polypeptide.
24. The method of claim 22, wherein said measuring IFI206 gene expression is measuring an amount of mRNA encoding IFI206 polypeptide.
25. A method of screening a sample for an IFI206 mutation, comprising:

comparing the sequence of an at least a portion of an IFI206 gene in the sample with at least a corresponding portion of SEQ ID NLO:1, SEQ ID No:3 or SEQ ID NO:14.
26. A fusion polypeptide, comprising at least 2 sequences selected from the group consisting of SEQ ID NOS:7-13.
27. The fusion polypeptide of claim 26, comprising at least 4 sequences selected from the group consisting of SEQ ID NOS:7-13.
28. The fusion polypeptide of claim 26, comprising at least 6 sequences selected from the group consisting of SEQ ID NOS:7-13.
29. An isolated polynucleotide encoding the polypeptide of any one of claims 26-28, or a complement of said polynucleotide.
30. A method of measuring the presence of infection in a subject, comprising measuring an amount of IFI206 in a sample from the subject.
31. The method of claim 30, wherein said measuring comprises contacting the sample with an antibody that specifically binds to an IFI206 polypeptide.
32. The method of claim 30, wherein said measuring comprises measuring IFI206 gene expression in the sample.
33. A method of measuring the obesity-reducing activity of a modality, comprising:

administering to a subject the modality; and measuring the amount of IFI206 in the subject.
34. The method of claim 33, wherein the subject is selected from the group consisting of diabetic (db) mouse, agouti mouse, tub mouse, POMC
knockout mouse, ob/ob mouse, fatty rat, and spiny mouse.
35. A method of reducing obesity of a subject, comprising reducing the activity of IFI206 in the subject.
36. The method of claim 35, wherein said reducing activity comprises disrupting the IFI206 gene in the subject.
37. The method of claim 35, wherein said reducing activity comprises reducing IFI206 mRNA transcription in the subject.
38. A transgenic non-human animal, having a disrupted IFI206 gene.
39. A transgenic non-human animal, comprising an exogenous polynucleotide having at least 89.2% sequence identity to the sequence SEQ
ID NO:1, SEQ ID NO:3 or SEQ ID NO:14, or a complement of said polynucleotide.
40. The transgenic non-human animal of claim 39, wherein said exogenous polynucleotide has at least 90% sequence identity to the sequence SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:14, or a complement of said polynucleotide.
41. The transgenic non-human animal of claim 39, wherein said exogenous polynucleotide has at least 98% sequence identity to the sequence SEQ ID NO:1, SEQ ID NO:3 or SEQ ID NO:14, or a complement of said polynucleotide.
42. A method of altering expression of IFI206 in a subject, comprising controlling IFI206 gene expression in the subject with an exogenous promoter.
43. The method of claim 42, wherein said controlling comprises operably-linking the promoter to the endogenous IFI206 gene of the subject.
44. The method of claim 42, wherein said controlling comprises operably-linking the promoter to an anti-sense polynucleotide of the endogenous IFI206 gene of the subject.
45. The method of any one of claims 42-44, wherein said promoter is an inducible promoter.
46. A method of inhibiting adipocyte differentiation, comprising inhibiting the activity of IFIO206.
47. The method of claim 46, wherein said reducing activity comprises disrupting the IFI206 gene.
48. The method of claim 47, wherein said reducing activity comprises reducing IFI206 mRNA transcription.
49. The method of claim 47, wherein said reducing activity comprises reducing IFI206 gene translation.
50. A polypeptide, comprising sequences SEQ ID NOS:7-13, and having less than 98% sequence identity with SEQ ID NOS:22 and 23.
51. The polypeptide of claim 50, having less than 95% sequence identity with SEQ ID NOS:22 and 23.
52. The polypeptide of claim 50, having less than 90% sequence identity with SEQ ID NOS:22 and 23.
53. An isolated polynucleotide encoding the polypeptide of any one of claims 50-53, or a complement of said polynucleotide.
54. An isolated polypeptide comprising an amino acid sequence having at least 80% sequence identity to the sequence SEQ ID NO:2, SEQ ID
NO:4 or SEQ ID NO:15, wherein said isolated polypeptide does not have the sequence SEQ ID NO:22 nor SEQ ID NO:24.
55. The polypeptide of claim 54, wherein said sequence is the sequence of an active IFI206 polypeptide.
56. The polypeptide of claim 54 or 55, wherein said isolated polypeptide has at most 99% sequence identity with sequence SEQ ID NO:22 or SEQ ID No:24.
57. The polypeptide of claim 54 or 55, wherein said isolated polypeptide has at most 90% sequence identity with sequence SEQ ID NO:22 or SEQ ID NO:24.
58. An isolated polynucleotide encoding the polypeptide of any one of claims 54-57, or a complement of said polynucleotide.
CA002402877A 2000-03-13 2001-03-13 Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same Abandoned CA2402877A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US18871600P 2000-03-13 2000-03-13
US60/188,716 2000-03-13
PCT/US2001/008333 WO2001068830A1 (en) 2000-03-02 2001-03-13 Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same

Publications (1)

Publication Number Publication Date
CA2402877A1 true CA2402877A1 (en) 2001-09-20

Family

ID=22694231

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002402877A Abandoned CA2402877A1 (en) 2000-03-13 2001-03-13 Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same

Country Status (3)

Country Link
EP (1) EP1282696A4 (en)
JP (1) JP2004522405A (en)
CA (1) CA2402877A1 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002239543A1 (en) * 2000-12-07 2002-06-18 Incyte Genomics, Inc. Molecules for disease detection and treatment

Also Published As

Publication number Publication date
EP1282696A1 (en) 2003-02-12
JP2004522405A (en) 2004-07-29
EP1282696A4 (en) 2005-02-09

Similar Documents

Publication Publication Date Title
EP1265914B1 (en) Wnt-1 related polypeptides, and nucleic acids encoding the same
AU2001247781A1 (en) Novel polypeptides, and nucleic acids encoding the same
US20070015700A1 (en) Mammalian genes modulated during fasting and feeding
US20030207288A1 (en) GPCR-like retinoic acid-induced gene 1 protein and nucleic acid
EP1265920B9 (en) Angiogenesis-associated proteins, and nucleic acids encoding the same
US6872704B2 (en) Acidic mammalian proteins and polynucleotides encoding the same
AU2001245765B2 (en) IFI206, a novel interferon-induced polypeptide, and nucleic acids encoding the same
US20030021788A1 (en) Novel human STRA6-like protein and nucleic acids encoding the same
CA2402877A1 (en) Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same
AU2001245765A1 (en) IFI206, a novel interferon-induced polypeptide, and nucleic acids encoding the same
WO2001068830A1 (en) Ifi206, a novel interferon-induced polypeptide, and nucleic acids encoding the same
US20030104351A1 (en) FIZZ1 for metabolism regulation
CA2447209A1 (en) Compositions and methods for adipose abundant protein
AU2002348423A1 (en) Novel acidic mammalian proteins and polynucleotides encoding the same
AU2001297935A1 (en) Human Stra6-like protein and nucleic acids encoding the same
AU2002323284A1 (en) GPCR-like retinoic acid-induced gene 1 protein and nucleic acid
AU2002254486A1 (en) FIZZ1 for metabolism regulation
AU2002314808A1 (en) Compositions and methods for adipose abundant protein

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued