Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS FOR TREATING AND PREVENTING MULTIPLE SCLEROSIS
Document Type and Number:
WIPO Patent Application WO/2008/134767
Kind Code:
A2
Abstract:
We have discovered that LRG-47 (also called p47 GTPase), plays a central role in the pathogenesis of multiple sclerosis, and that inhibition of LRG-47 activity by anti-LRG-47 antibodies or of LRG-47 expression by siRNA dramatically reduce the pathology and symptoms of multiple sclerosis. Certain embodiments of the invention are directed to the therapeutic use of anti-LRG-47 antibodies (mouse or rabbit or other antibodies that are humanized or human antibodies to LRG-47, preferably antibodies made against human LRG-47) or siRNA or antisense nucleotides that specifically hybridize with the gene or mRNA or cDNA encoding human LRG-47 to treat or prevent multiple sclerosis and other autoimmune diseases that are T-cell -mediated. Other embodiments are directed to methods for the diagnosis of multiple sclerosis or to determining the aggressiveness of multiple sclerosis by determining the amount of human LRG-47 or LRG-47 mRNA in a biological sample from the patient.

Inventors:
YAN SHI DU (US)
Application Number:
PCT/US2008/062158
Publication Date:
November 06, 2008
Filing Date:
May 01, 2008
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV COLUMBIA (US)
YAN SHI DU (US)
International Classes:
A61K39/395; A61K48/00
Foreign References:
US20030017985A1
Other References:
SORACE ET AL.: 'Identification of an endotoxin and IFN-inducible cDNA: possible identification of a novel proteiin family' J. LEUKOC. BIOL. vol. 58, no. 4, 1995, pages 477 - 484
LECLAIRE ET AL.: 'Characterization and use of monoclonal and polyclonal antibodies against the mouse interferon-gamma receptor' J. LEUKOC. BIOL. vol. 51, no. 5, 1992, pages 507 - 516
Attorney, Agent or Firm:
EVANS, Judith et al. (P. O. Box 7024Fairfax Station, Virginia, US)
Download PDF:
Claims:

CLAIMS

What is claimed is:

1. A method for treating or preventing multiple sclerosis in a patient, comprising administering a therapeutically effective amount of an isolated anti-LRG-47 antibody or biologically active fragment thereof.

2. A method for treating or preventing multiple sclerosis in a patient, comprising administering a therapeutically effective amount of a compound that decreases expression of human LRG-47.

3. A method for treating or preventing an autoimmune disease in a patient, comprising administering a therapeutically effective amount of an isolated anti-LRG-47 antibody or biologically active fragment thereof.

4. A method for treating or preventing an autoimmune disease in a patient, comprising administering to the animal a therapeutically effective amount of a compound that decreases expression of human LRG-47.

5. The method of claim 1 or claim 3, wherein the isolated antibody is the polyclonal anti- human LRG-47 antibody 138AB or fragment or variant thereof, preferably a humanized form thereof.

6. The method of claim 1 or claim 3 wherein the isolated antibody is monoclonal, polyclonal, chimeric, humanized or bispecific.

7. The method of claim 2 or claim 4, wherein the compound is an antisense nucleic acid or siRNA.

8. The method of claim 7, wherein antisense nucleic acid is sufficiently complementary to the human gene or mRNA encoding LRG-47 to hybridize to it thereby forming a stable duplex.

9. The method of clam 8, wherein the gene encoding human LRG-47 is identified by SEQ ID NO: !.

10. A method for treating or preventing multiple sclerosis in a patient, comprising

a. determining a pre-treatment level of human LRG-47 protein in a pre-treatment biological sample taken from the animal,

b. administering an amount of a compound that reduces human LRG-47 expression levels in animal cells,

c. determining a post-treatment level of HUMAN LRG-47 protein in a post- treatment biological sample taken from the animal,

d. comparing the pre-treatment and post-treatment levels of LRG-47 protein in the respective biological samples, and

e. determining that treatment is effective if the post- treatment level of human LRG-47 is significantly lower than the pre-treatment level.

11. The method of claim 10, wherein the compound is an antisense nucleic acid or siRNA.

12. The method of claim 11, wherein antisense nucleic acid or siRNA are each sufficiently complementary to the human gene or mRNA encoding LRG-47 to hybridize to it thereby forming a stable duplex.

13. The method of clam 12, wherein the gene encoding human LRG-47 is identified by SEQ ID NO: 1.

14. The method of claim 10, further comprising

f. if the difference between the pre-treatment and post-treatment levels of human LRG-47 are not significantly different, then increasing the amount or frequency of administration of the compound until the post-treatment level of human LRG-47 is significantly lower than the pre-treatment level..

15. A method for treating or preventing an autoimmune disease in a patient, comprising

a. determining a pre-treatment level of human LRG-47 protein in a pre-treatment biological sample taken from the animal,

b. administering an amount of a compound that reduces human LRG-47 expression levels in animal cells,

c. determining a post-treatment level of human LRG-47 protein in a post-treatment biological sample taken from the animal,

d. comparing the pre-treatment and post-treatment levels of human LRG-47 protein in the respective biological samples, and

e. determining that treatment is effective if the post- treatment level of human LRG-47 is significantly lower than the pre-treatment level.

16. The method of claim 15, further comprising

f. if the difference between the pre-treatment and post-treatment levels of human LRG-47 are not significantly different, then increasing the amount or frequency of administration of the compound until the post-treatment level of human LRG-47 is significantly lower than the pre-treatment level..

17. The method of claim 15, wherein the compound is an antisense nucleic acid or siRNA.

18. The method of claim 17, wherein antisense nucleic acid or siRNA are each sufficiently complementary to the human gene or mRNA encoding LRG-47 to hybridize to it thereby forming a stable duplex.

19. The method of clam 18, wherein the gene encoding LRG-47 is identified by SEQ ID NO: 1.

20. The method of claim 10 or claim 15, wherein the biological sample is a member selected from the group comprising a nerve sample, serum, blood, plasma, cerebral spinal fluid, fibroblasts, leukocytes, skin, and urine.

21. A method for diagnosing a patient who is at risk of developing multiple sclerosis, comprising

a. determining the level of human LRG-47 protein in a biological sample taken from the patient and in a corresponding sample taken from a control subject that is not affected with multiple sclerosis,

b. comparing the level of human LRG-47 protein in the biological samples and forming a diagnosis that the patient is at risk of developing multiple sclerosis if the level of human LRG-47 protein in the patient sample is significantly higher than the level of human LRG-47 protein in the control sample.

22. A method for diagnosing a patient who is at risk of developing an autoimmune disease or who has an autoimmune disease, comprising

a. determining the level of human LRG-47 protein in a biological sample taken from the patient and in a corresponding sample taken from a control animal that is not affected with an autoimmune disease,

b. comparing the level of human LRG-47 protein in the biological samples and forming a diagnosis the patient as being at risk of developing an autoimmune disease if the level of human LRG-47 protein in the patient sample is significantly higher than the level of human LRG-47 protein in the control sample.

23. A method for determining the progression of multiple sclerosis, comprising

a. determining the level of human LRG-47 protein in a first biological sample taken from the patient at a first time,

b. determining the level of human LRG-47 protein in a second biological sample taken from the patient at a second later time ,

c. comparing the level of human LRG-47 protein in the first and second biological samples and diagnosing progression of multiple sclerosis if the level of human LRG- 47 protein in the second sample is significantly higher than the level of human LRG- 47 protein in the first sample.

24. A method for determining if a patient is responding to treatment for MS, comprising

a. determining the level of human LRG-47 protein in a first biological sample taken from the patient at a first time,

b. determining the level of human LRG-47 protein in a second biological sample taken from the patient at a second later time ,

c. comparing the level of human LRG-47 protein in the first and second biological samples and determining that the patient is responding to treatment if the level of human LRG-47 protein in the second sample is significantly lower than the level of human LRG-47 protein in the first sample.

25. The anti-human LRG-47 antibody 138AB, preferably in a humanized form.

26. A pharmaceutical composition comprising the anti-human LRG-47 antibody 138AB or fragment or variant thereof, preferably in a humanized form.

27. A pharmaceutical composition comprising a humanized isolated anti-LRG-47 antibody or fragment thereof selected from the group comprising LRG-47 (A- 19) Antibody , LRG-47 (M-95) Antibody , LRG-47 (M- 16) Antibody , and LRG-47 (P- 20) Antibody.

28. A diagnostic kit comprising: a) a) a vessel or vessels for receiving a blood/serum or CSF or cell sample from the subject; b) an agent that specifically detects human LRG- 47 protein or amplifies human LRG-47 mRNA; and c) printed instructions for detecting the LRG-47 protein or the amplified LRG-47 mRNA in the sample.

29. The method of claim 28, wherein the agent that specifically detects LRG-47 protein is isolated anti-LRG-47 antibody selected from the group comprising LRG-47 (A- 19) Antibody, LRG-47 (M-95) Antibody, LRG-47 (M- 16) Antibody , LRG-47 (P-20) Antibody, and anti-human LRG-47 138ABAntibody or fragment or variant thereof.

30. The method of claim 1 or claim 3, wherein the isolated anti-LRG-47 antibody is a humanized antibody selected from the group comprising LRG-47 (A- 19) Antibody , LRG-47 (M-95) Antibody , LRG-47 (M- 16) Antibody , and LRG-47 (P-20) Antibody, or fragment or variant thereof.

31. The method as in one of claims 21, 22, 23 and 24, wherein the biological sample is a member selected from the group comprising a nerve sample, serum, blood, plasma, cerebral spinal fluid, fibroblasts, leukocytes, skin, and urine

32. The method of claim 21, further comprising the step of

c. determining that the patient has multiple sclerosis if the patient shows other objective or subjective indicia of multiple sclerosis.

33. The method of claim 22, further comprising the step of

c. determining that the patient has an autoimmune disease if the patient shows other objective or subjective indicia of an autoimmune disease.

Description:

METHODS FOR TREATING AND PREVENTING MULTIPLE SCLEROSIS

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims benefit of Provisional Appln. 60/915,127, filed 5/1/2007, the entire contents of which are hereby incorporated by reference as if fully set forth herein, under 35 U.S.C. §119(e).

STATEMENT OF GOVERNMENTAL INTEREST

[0002] This invention was made with Government support under US Public Health Service grant NO: NINDS 042855). The Government has certain rights in the invention.

BACKGROUND OF THE INVENTION

1. Field of the Invention

[0003] This invention is in the field of methods and pharmaceutical compositions for treating or preventing Multiple Sclerosis, Encephalitis, and other autoimmune diseases that are associated with an abnormal increase in the number of activated T-cells.

2. Description of the Related Art

[0004] Multiple sclerosis (MS) is a severe, chronic disabling disease that affects approximately 1 out of every 1,600 people. The majority of the affected individuals develop symptoms as young adults between 20 and 40 years of age, with roughly 60% of the cases occurring in women. The disease is characterized by neuron deterioration in the central nervous system (CNS) with the associated loss of the insulating myelin sheath from around the axons of the nerve cells, referred to as demyelination. The disease presents itself in the white matter of the brain and spinal cord as a number of sclerotic lesions or plaques (Prineas (1985) Demyelinating Diseases, Elsvevier: Amsterdam; Raine (1983) Multiple Sclerosis, Williams and Wilkins: Baltimore; Raine et al. (1988) J. Neuroimmunol. 20:189-201; and Martin (1997) J. Neural Transmission (Suppl) 49:53-67). The characteristic MS lesion is inflamed, exhibits axonal demyelination, axonal degeneration, and is found around small

venules. These characteristics typically evolve early in plaque development and are hypothesized to occur as a result of a breakdown in the blood-brain barrier (BBB). As a consequence of BBB breakdown, infiltrates consisting of various lymphocytes and macrophages enter the brain or spinal cord. This inflammatory infiltrate ultimately leads to axonal degeneration and scar tissue formation, and in many instances, is associated with incomplete remyelination (Martin (1997) J. Neural Transmission (Suppl) 49:53-67). Further, it is hypothesized that this apparent immunologic attack targets not only the myelin sheath, but also the oligodendrocytes imperative to CNS myelin production. As a result, not only is the nerve-insulating myelin damaged, but the ability of oligodendroglial cells to repair damaged myelin is seriously compromised (Scientific American 269 (1993):106-114). Development of multiple areas of scar tissue (sclerosis) along the covering of the nerve cells slows or blocks the transmission of nerve impulses in the affected area, resulting in the development of the symptoms characteristic of MS. These symptoms include pain and tingling in the arms and legs; localized and generalized numbness, muscle spasm and weakness; difficulty with balance when standing or walking; difficulty with speech and swallowing; cognitive deficits; fatigue; and bowel and bladder dysfunction. [0005] \Approximately half of the people with this disease suffer from relapsing- remitting MS. In these cases, the afflicted individual experiences repeated unpredictable attacks, due to episodes of inflammation, axonal demyelination, axonal degeneration, and development of glial scar tissue. These attacks are separated by periods of remission, during which the symptoms stabilize or diminish. Acute neurological deficits occur with each attack, and in many cases, the accumulation of residual deficits as a result of these attacks eventually leads to worsening disability and impairment in quality of life. Approximately 30- 40% of the afflicted population have chronic progressive MS (either primary or secondary) in which neurological deterioration occurs in the absence of clinically apparent attacks. Recently, immunomodulatory therapy with interferon-beta (IFN-beta) has proven to be successful in reducing the severity of the underlying disease in patients with relapsing- remitting MS. FDA-approved IFN-beta therapies for the treatment of relapsing-remitting MS in the United States include interferon beta- Ia (marketed as Avonex.RTM, available from Biogen, Inc.) and interferon-beta- Ib (marketed as Betaseron.RTM, available from Chiron Corporation). Both of these therapeutic agents are partially effective in reducing the frequency and severity of relapses, slowing the rate of disease progression, or reducing the degree of brain inflammation as measured by a variety of magnetic resonance imaging (MRI) techniques.

[0006] MS is considered to be a T cell-mediated autoimmune disease of the brain and spinal cord [Traugott et al. 1983; Vizier et al. 1999]. The failed apoptosis of auto-reactive T cells has been implicated in MS pathogenesis. While there appears to be a localized CNS immune response, peripheral immune cell abnormalities appear to correlate with central disease activity [Hafler and Weiner 1989] and may precede MRI activity. Apoptosis is an important mechanism in immune system regulation, responsible for elimination of autoreactive T- lymphocytes (T cells), B-lymphocytes (B cells) and monocytes from the circulation and prevention of their entry into the CNS [Mahoney and Rosen 2005; Todaro et al. 2004]. It has been hypothesized that a genetic predisposition exists in MS patients whereby a failure of autoreactive T cells and B cells as well as activated macrophages to undergo apoptosis contributes to the pathogenesis of MS [Bernard and Derosbo 1992; Pender 1998; Pender and Rist 2001].

[0007] Prescribing decisions seem to be driven by evidence-based medicine and a recent report by the American Association of Neurologists (Goodin D S et al; Neurology Jan. 22, 2002;58(2): 169-78) is a key document. The consensus amongst many neurologists is that early, aggressive therapy with beta-interferons was desirable in increasing the time to first relapse and limiting the overall disease load, although it was recognized that there was no evidence that this approach showed long-term benefit on EDSS score (a measure of disease-related disability). There is currently no satisfactory diagnostic marker for multiple sclerosis. There is therefore a need for new diagnostic methods and for new disease-modifying therapies for MS.

SUMMARY OF THE INVENTION

[0008] A certain embodiment of the invention is directed to a method for treating or preventing multiple sclerosis (or another autoimmune disease) in a patient, by administering a therapeutically effective amount of an isolated anti-, antibody or biologically active fragment thereof. The isolated antibody can be the polyclonal anti-human LRG-47 antibody 138AB made in our laboratory and described herein, or a fragment or variant thereof, preferably a humanized form. Wherever a fragment of an antibody is mentioned herein it is understood that this fragment is a biologically active fragment or variant of the antibody. The antibody can also be a monoclonal, polyclonal, chimeric, humanized or bispecific anti-LRG-47 antibody or humanized antibody selected from the group comprising LRG-47 (A- 19) Antibody , LRG-47 (M-95) Antibody , LRG-47 (M- 16) Antibody , and LRG-47 (P-20) Antibody, anti-human LRG-47 138AB or fragment or variant thereof.

[0009] Another embodiment is directed to a method for treating or preventing multiple sclerosis (or another autoimmune disease) in a patient, by administering a therapeutically effective amount of a compound that decreases expression of human LRG-47. The compound can be an antisense nucleic acid or siRNA that is sufficiently complementary to the human gene or mRNA encoding LRG-47 to hybridize to it thereby forming a stable duplex.

[0010] Another embodiment is directed to a method for treating or preventing multiple sclerosis (or another autoimmune disease) in a patient, by a. determining a pre-treatment level of human LRG-47 protein in a pre-treatment biological sample taken from the animal, b. administering an amount of a compound that reduces human LRG-47 expression levels in animal cells (the above described siRNA or antisense nucleotide), c. determining a post- treatment level of human LRG-47 protein in a post-treatment biological sample taken from the animal, d. comparing the pre-treatment and post-treatment levels of LRG-47 protein in the respective biological samples, and e. determining that treatment is effective if the post- treatment level of LRG-47 is significantly lower than the pre-treatment level. This method optionally further includes the step f. if the difference between the pre-treatment and post- treatment levels of LRG-47 are not significantly different, then increasing the amount or frequency of administration of the compound until the post-treatment level of LRG-47 is significantly lower than the pre-treatment level. A similar method can be used for an autoimmune disease.

[0011] A biological sample for the purpose of the present inventions can be a nerve sample, serum, blood, plasma, cerebral spinal fluid, fibroblasts, leukocytes, skin, or urine. [0012] Another embodiment is directed to method for diagnosing a patient who is at risk of developing multiple sclerosis, by a. determining the level of human LRG-47 protein in a biological sample taken from the patient and in a corresponding sample taken from a control subject that is not affected with multiple sclerosis, b. comparing the level of LRG-47 protein in the biological samples and forming a diagnosis that the patient is at risk of developing multiple sclerosis if the level of LRG-47 protein in the patient sample is significantly higher than the level of LRG-47 protein in the control sample. A similar diagnostic method can be used to diagnose a patient at risk of developing an autoimmune disease. The method can have the further step of c. determining that the patient has multiple sclerosis if the patient shows other objective or subjective indicia of multiple sclerosis.

[0013] Another embodiment is directed to a method for determining the progression of multiple sclerosis, by a. determining the level of human LRG-47 protein in a first biological sample taken from the patient at a first time, b. determining the level of LRG-47 protein in a second biological sample taken from the patient at a second later time, c. comparing the level of LRG-47 protein in the first and second biological samples and diagnosing progression of multiple sclerosis if the level of LRG-47 protein in the second sample is significantly higher than the level of LRG-47 protein in the first sample. A similar method can be used to determine the progression of an autoimmune disease.

[0014] Another embodiment is directed to a method for determining if a patient is responding to treatment for MS, by a. determining the level of human LRG-47 protein in a first biological sample taken from the patient at a first time, b. determining the level of LRG- 47 protein in a second biological sample taken from the patient at a second later time, c. comparing the level of LRG-47 protein in the first and second biological samples and determining that the patient is responding to treatment if the level of LRG-47 protein in the second sample is significantly lower than the level of LRG-47 protein in the first sample. [0015] Another embodiment is directed to the new anti-human LRG-47 antibody 138AB and a fragment thereof, preferably a humanized form of the antibody, and to a pharmaceutical composition that includes the anti-human LRG-47 antibody 138AB or humanized fragment. Another embodiment is directed to a pharmaceutical composition that includes a humanized isolated anti-LRG-47 antibody or fragment thereof selected from the group including LRG- 47 (A- 19) Antibody , LRG-47 (M-95) Antibody , LRG-47 (M- 16) Antibody , and LRG-47 (P-20) Antibody.

[0016] Another embodiment is directed to a diagnostic kit for determining the presence of human LRG-47 protein or human LRG-47 mRNA in a biological sample including: a) a) a vessel or vessels for receiving a blood, serum or CSF or cell sample from the subject; b) an agent that specifically detects LRG-47 protein or amplifies LRG-47 mRNA; and c) printed instructions for detecting the LRG-47 protein or the amplified LRG-47 mRNA in the sample. The agent that specifically detects LRG-47 protein can be an isolated anti-LRG-47 antibody selected from the group including LRG-47 (A- 19) Antibody, LRG-47 (M-95) Antibody, LRG-47 (M- 16) Antibody , LRG-47 (P-20) Antibody, and anti-human LRG-47 138AB Antibody, preferably humanized.

DESCRIPTION OF THE DRAWINGS

[0017] The present invention is illustrated by way of example, and not by way of limitation, in the figures of the accompanying drawings.

[0018] FIG. 1. Expression of LRG-47 in EAE mice (A-B).Total RNA was isolated from spinal cords of naϊve, CFA (complete Freund's adjuvant)-treated and MBP- induced EAE mice. Quantitative real time PCR (A) shows transcripts of mouse LRG-47 in spinal cord tissues from each group on day 14 after induction of EAE (in the MBP-treated group; peak symptoms). * P < 0.001, n = 3-5 mice/group B. Northern analysis of spinal cord tissue for LRG-47 transcripts. Time points indicate days after immunization with MBP- derived peptide. C. Quantification of Northern blot images. * P <0.001, N = 3/group. (D-E) Effect of gene deletion of LRG-47 on EAE. (D) LRG-47 hemizygous (LRG-47 +/~ ), homozygous (LRG-47 " ' " ) mice, and WT littermates (LRG-47 +/+ ) were immunized with 1- 9NAc MBP-derived peptide. Symptoms were assessed according to a clinical scoring system (*P<0.01 vs. LRG " ' " mice). (E). Lower panel, representative H & E-stained spinal cord sections from EAE-induced mice of the indicated genotype (LRG-47 +/+ , LRG-47 +/" , and LRG-47 " ' " ) on day 21. Upper panel, analysis of area occupied by nuclei from samples derived from the above groups of mice (B-D). * P< 0.01, n = 3-4 /group.

[0019] FIG 2. LRG-47 " ' " mice display reduced accumulation and enhanced apoptosis of activated CD4 T-cells in the CNS during EAE (A-G). (A). Infiltrating cells were isolated from the CNS of LRG-47 " ' " mice and WT littermates on day 18-20 (peak symptomatic EAE, clinical score 3-4, in WT animals). Infiltrating cells isolated from the CNS were immunostained with antibodies to CD4 conjugated with PerCP, CD44 conjugated with PE, or Annexin V conjugated with FITC. FACS analyses were used to determine the percentage of cells staining positively for each of the markers, CD4, CD44 hl , and annexin V. CD44 hl and annexin V were gated for CD4. * P < 0.05, n = 3-5/group. (B). FACS analysis of activated CD4 T-cells isolated from lymph nodes of mice induced to develop EAE with MBP. Lymphocytes isolated from lymph nodes of LRG " ' " mice and WT littermates were stained with anti-CD4 conjugated with PerCP and CD44 conjugated with PE. The percentage of CD44 hl -positive cells was determined by FACS gated for CD4 cells. *P < 0.01, n = 3- 5/group. (C). Lymphcytes in lymph nodes from EAE mice were subjected to TUNEL staining. TUNEL-positive cells were quantitated using UNIVERSAL image software. (D). In vivo proliferation of CD4 + T-cells isolated from mice induced to develop EAE. Mononuclear cells isolated from the CNS were stained with anti-CD4 PerCP and anti-BrdU

followed by FACS analysis. Cells were pooled from 5 mice of each group. BrdU incorporation was expressed as the percentage of BrdU-positive CD4 + T-cells gated in CD4 + T-cells. (E-G) Effect of LRG-47 gene deletion on T-cell proliferation and apoptosis of CD4 T-cells in response to MBP: in vitro studies. (E). 3 H-thymidine incorporation was performed on lymphocytes and splenocytes isolated from the indicated mice on day 7 after immunization in the presence of MBP (2.5 μg/ml). * P < 0.01 vs. other groups of mice, n = 3-5/group. (F). Splenocytes from LRG-47 " ' " and WT littermates were stained with anti-CD4 conjugated to PE and annexin V conjugated to FITC after stimulation with MBP (2.5 μg/ml) for 48 and 72 hrs, respectively. FACS analyses show the percentage of annexin-positive cells gated in the CD4 + T-cell population. (G). TUNEL-positive CD4 T- cells following exposure to MBP. CD4 T-cells isolated from spleens of LRG "A and WT mice were incubated with MBP (2.5 μg/ml ) for 48 hours and subjected to TUNEL staining. #P<0.05, *P<0.01, n = 3- 5/group.

[0020] FIG. 3. Expression and translocation of LRG-47 in encephalitogenic CD4 T- cell (IAElO) and its effect on cell proliferation and apoptosis. (A) Northern blotting of IAElO cells shows elevated levels of LRG-47 transcripts in MBP-induced IAElO cells in a time-dependent manner (left panel). Image analysis of the intensity of bands is shown in the right panel. (B-C) Effect of MBP on LRG localization in IAElO cells. LRG-47 was localized in the Golgi compartment of unstimulated IAElO cells (panel B). When IAElO cells were stimulated with MBP (5 μg/ml) for 4 hours, LRG-47 was redistributed from Golgi apparatus to the cell membrane (panels C1-C3). Bl & C1 :GSK65 (marker for Golgi) staining; B2 & C2: LRG-47 staining; B3 & C3 merge images from B1/B2 and C1/C2, respectively. Addition of anti-LRG-47 IgG attenuates 3 H-thymidine incorporation (D) and proliferation (E) in MBP-treated IAE 10 cells. (E) CFES-labeled IAElO cells were stimulated by the presence of MBP (2.5 μg/ml) for the indicated times (24 to 96 hours) and stained by anti-CD44 conjugated to PE. The CFSE fluorescence profile was analyzed by FACS gated for CD44 + T-cells.

[0021] (F-I) Blockade of LRG-47 enhances apoptosis of encephalitogenic CD4 + T-cells (IAE 10) in a dose- and time-dependent manner. (F) IAElO cells were activated with MBP (2.5 μg/ml) for 72 hours in the presence of anti-LRG-47 IgG (A-19, 0-20μg/ml) or NI IgG (10 μg/ml). Apoptotic cells, as shown by annexin V staining, were assessed. #P <0.05, *P<0.01, n = 3-5/group. Percentage of annexin V-positive cells (G), PI (propidium iodide)- positive cells (H), and live cells (I) were determined in the population of MBP-treated IAElO

cells exposed to anti-LRG-47 IgG (A-19, 5μg/ml) or NI IgG (5 μg/ml) for the indicated times. *P<0.01, n=3-5/group.

[0022] FIG. 4. Effect of blockade of LRG-47 on induction of EAE, apoptosis, and IFN-γ production. (A) Activated IAElO cells were adoptively transferred into appropriately prepared (see text) wild-type (WT) BlOPL mice. Animals received anti-LRG-47 IgG (blue), nonimmune IgG (green) or vehicle (black) as indicated. Clinical symptoms were scored. *P < 0.01 vs. vehicle-treated or NI IgG-treated IAElO cells adoptively transferred into WT mice. (B) Apoptosis in the spinal cord of mice subject to adoptive transfer of activated IAElO cells. TUNEL-positive cells were quantified in spinal cords from WT mice after adoptive transfer of activated IAElO cells alone, transfer of IAElO cells and treatment with anti-LRG-47 IgG , or transfer of IAE 10 cells and treatment with NI IgG . * P < 0.01, n = 3- 5 mice/group. (C-E). Reduced accumulation and enhanced apoptosis of CD4 T-cells following adoptive transfer of IAElO cells into mice treated with anti-LRG-47 IgG. Mononuclear cells isolated from brain and spinal cord of mice subject to adoptive transfer of activated IAElO cells 16 and 28 days earlier were stained with anti-CD4-PE and annexin V- FITC followed by FACS analysis. FACS analyses show infiltrating CD4 T-cells (C, E) and annexin V-positive cells (D) in the CNS of the latter mice. FACS data shown in panels C-D (panels 1-3) are representative of 3-5 mice/group. Panels of C4, D4 and E4 display results of 3-4 experiments. Panels of D 1-3 denote the percentage of apoptotic cells as shown by annexin V-positives in the right upper frame. (F-H). Blockade of LRG-47 increases IFN-γ production. (F) Levels of IFN-γ were determined in supernatants from cultured IAElO cells exposed to MBP in the presence of anit-LRG-47 IgG or NI IgG by ELISA. (G) Cytokine production in CD4 T-cells from WT and LRG-47 " ' " mice. CD4 T-cells were isolated from spleens of WT mice and LRG-47 " ' " mice, and cultured with MBP (2.5 μg/ml) for the indicated time. Levels of IFN-γ in culture supernatants were determined by ELISA. (H) Levels of IFN-γ in plasma from WT and LRG-47 " ' " mice immunized with MBP to develop EAE (day 14-18). * P < 0.01, n = 12-16 mice/group.

[0023] FIG. 5. SiRNA inhibition of LRG-47 expression in IAElO Cells. Panel 5a shows the level of LRG-47 mRNA in cells measured using RT-PCT. FIG. 7b shows the level of LRG-47 measured by Real-Time PCR. After stimulation with MBP (5 micrograms/ml) for 48 hours, IAElO T-cells were transfected with siRNA targeted to mouse LRG-47.

[0024] FIG. 6. Blockade of LRG-47 expression by siRNA attenuates the clinical symptoms in EAE mice. MBP-activated IAElO cells were treated with either siRNA-LRG

47 or vehicle, and adoptively transferred into BlOPL mice. In wild- type (WT) BlOPL mice treated with IAElO cells alone or IAElO cells, symptomatic EAE progressed rapidly, occurring in all animals in the 15-20 day window, and reaching a clinical score/severity of 3- 4. In contrast, WT mice treated with IAElO cells exposed to siRNA-LRG 47 displayed a delay in the onset of EAE and attenuation of symptomatic EAE , a clinical score of only 1- 2.5. Thus, clinical symptomatology mirrors what might be expected if LRG-47 was an essential cofactor for pathogenicity of IAElO cells.

[0025] FIG. 7 Expression of LRG-47 antigen in spinal cord tissue from MS patients. A. LRG-47 antigen was prominently elevated in MS-affected spinal cord tissue as compared with age-matched normal control by immunoblotting with specific anti-LRG-47 IgG. B-C. By immunostaining with specific anti-LRG 47 IgG, LRG-47 (B2 & C2) was especially present in mononuclear monocytes (Bl) and CD4+ T cell (Cl) as shown by co-stained with CD68 and CD4+, respectively (FIG. 7Bl & Cl). There was no positive staining when anti- LRG IgG was replaced by nonimmune IgG (data not shown). These data indicate the significance of LRG-47 in pathogenesis of MS and its relevance beyond EAE. [0026] FIG. 8 Localization of LRG-47 antigen in spinal cords of EAE mice. Spinal cord sections were co-stained with goat anti-LRG-47 (1 micrograms/ml) and rat anti-CD4 or rat anti-CD 1 Ib followed by rabbit anti-goat IgG conjugated with FITC and anti-rat- conjugated with TRITC. LRG-47 antigen (panels S2 and S5) was detected in CD4+ T-cells (Sl) and CDl lb-positive mononuclear cells (S4). S3 and S6 are merged with S1/S2 and S3/S4, respectively. Magnification : Sl-3, x600; S4-6, x400.

[0027] FIG. 9 Immuno-dot blotting with antibody to mouse LRG-47 of mouse serum from MBP-induced EAE mice at day 7 and 12 and naϊve mice demonstrated an increase in LRG-47 in serum of EAE mice compared to serum from naϊve mice. Levels of LRG-47 were quantified by NIH image program.

[0028] FIG. 10 Immuno-dot blotting with antibody to human LRG-47 of cerebral spinal fluid (CSF) and serum from MS patients demonstrated an increase in LRG-47 in MS patients compared to age-matched control patients. Levels of LRG-47 were quantified by NIH image program.

DETAILED DESCRIPTION

[0029] We have discovered that LRG-47 (also called p47 GTPase), plays a central role in the pathogenesis of multiple sclerosis, and that inhibition of LRG-47 activity by anti-LRG-47 antibodies or of LRG-47 expression by siRNA dramatically reduce the pathology and symptoms of multiple sclerosis. Therefore certain embodiments of the invention are directed to the therapeutic use of anti-LRG-47 antibodies (mouse or rabbit or other antibodies that are humanized or human antibodies to LRG-47, preferably antibodies made against human LRG- 47) or siRNA or antisense nucleotides that specifically hybridize with the gene or mRNA or cDNA encoding human LRG-47 to treat or prevent multiple sclerosis and other autoimmune diseases that are T-cell -mediated. Other embodiments are directed to methods for the diagnosis of multiple sclerosis or to determining the aggressiveness of multiple sclerosis by determining the amount of human LRG-47 or LRG-47 mRNA in a biological sample from the patient. Other embodiments are directed to transgenic LRG-47 -/- mice. Certain embodiments are directed to various humanized anti-LRG-47 antibodies and to their diagnostic and therapeutic use.

[0030] We have discovered that LRG-47 which is known for its essential contribution to host resistance to certain intracellular pathogens, plays a central role in the pathogenesis of Experimental Autoimmune Encephalitis (EAE) that is a model for Multiple Sclerosis in animals. The experiments herein show that LRG-47 increases the survival of autoreactive T- cells, the levels of which are known to be increased in animals having EAE. We show that expression of LRG-47 occurs early in the course of myelin basic protein (MBP)-induced EAE in the central nervous system (CNS), especially in cells of lymphoid and mononuclear phagocytic origin. Using classical genetics we discovered that homozygous LRG-47 null mice (hereafter "LRG-47 null mice") were resistant to MBP-induced EAE, and CD4 T-cells in the spleen and CNS of these animals displayed decreased proliferative capacity and underwent increased apoptosis. LRG-47 null mice also displayed dysregulation of interferon- γ. Certain embodiments of the invention are directed to LRG-47 -/- transgenic mice. [0031] The results of experiments presented below further show that an encephalitogenic CD4 ThI clone (1AE10 cells) showed rapid upregulation of LRG-47 after exposure to MBP in vitro, while blockade of LRG-47 expression in the presence of MBP suppressed proliferation and induced cell death. Adoptive transfer of IAElO cells treated with anti-LRG IgG into naϊve WT mice attenuated EAE when the mice were treated with MBP. These data

indicate that one way in which the blockade of LRG-47 alleviates EAE is by suppressing expansion of activated CD4 T-cells. Importantly, we also discovered that LRG-47 expression levels are dramatically increased in spinal cord tissues taken from a patient diagnosed with Multiple Sclerosis (MS). The fluorescence detected by immunoblotting spinal cord tissue from an MS patient using an isolated antibody against LRG-47 was 16 times more intense than in normal animals. Further, we were able to block LRG-47 expression using small interfering RNA in IAElO cells in the EAE model and thereby reduce the severity of the disease and delay its onset.

[0032] These discoveries enable a therapy for treating or preventing MS by reducing the level of LRG-47 expression in an animal preferably a human that has the disease or is at risk of developing MS; preferably by reducing the level of expression in the central nervous system, more particularly in the spinal cord. Such inhibition can be accomplished by administering siRNA to an animal in an amount that either reduces LRG-47 expression in a biological sample taken from the animal, or in an amount that reduces the symptoms of MS in the animal. Since increased levels of activated CD4 T-cells are implicated in the pathogenesis of autoimmune disorders in general, certain embodiments of the invention are directed to methods of treating or preventing an autoimmune disorder in an animal preferably a human having elevated levels of activated CD4 T-cells by administering a therapeutic agent that reduces human LRG-47 expression.

[0033] Interferon-gamma (IFN-γ) plays a central role in host resistance to infection and regulation of the immune system. Thus, it is not surprising that INF-γ is an essential factor in CD4 T-cell homeostasis during the immune response, including activation-induced cell death [1-3]. In the setting of Experimental Autoimmune Encephalitis (EAE), INF-γ-deficient mice display a progressive and fatal clinical course associated with increased proliferation and decreased apoptosis of activated CD4 T-cells in response to antigen [3]. This pattern is also seen in other models of autoimmune diseases. These observations indicated to us the likelihood that downstream effectors of IFN- γ would also have important roles in immunobiology. Specifically, an IFN-γ-inducible family of intracellular 47 kDa GTPases has been shown to contribute to resistance to intracellular pathogens [4-9]. One member of this family, LRG-47, is essential in resistance to Mycobacterial tuberculosis and Toxoplasma gondii, whereas other members are involved in the response to different pathogens [4-9]. The results below demonstrate a role for LRG-47 in survival of autoreactive CD4 T-cells. In vivo and in vitro evidence shows that LRG-47 deficient mice show suppressed EAE because they do not show expansion of activated CD4 T-cells and decreased apoptosis of activated T cells.

[0034] We have recently demonstrated that receptor for advanced glycation endproducts (RAGE), a multiligand member of the immunoglobulin superfamily, can mediate induction of EAE under certain conditions. In the myelin basic protein (MBP) model used in the experiments described blow, blockade of RAGE prevents induction of symptomatic EAE, at least in part by inhibiting migration of immunocytes into the central nervous system [10]. In order to probe the underlying mechanisms, we performed microarray analysis to identify differences in gene expression between mice-induced by MBP and naϊve mice. We discovered that LRG-47 was significantly upregulated in EAE-induced animals treated with MBP compared to naϊve mice. The studies below that LRG-47 plays a key role in the pathogenesis of EAE and other autoimmune diseases associated with increased levels of activated T cells.

Results

Expression of LRG-47 in mouse EAE

[0035] PCR demonstrated ~30-fold upregulation of LRG-47 mRNA in spinal cord tissue in mice 14 days after treatment with MBP, when symptomatology was approaching a peak (15-17 days; see below), compared with controls (untreated animals or those receiving only complete Quantitative Freund's adjuvant, CFA; p<0.00I)(FIG. IA). Northern analysis on spinal cord from animals subjected to MBP-induced EAE was performed at several time points. A trend towards higher levels of LRG-47 transcripts was first noted on day 10 (post- MBP treatment), and a statistically significant increase was seen by day 14 (FIG. IB-C). These data indicate a close correspondence between LRG-47 expression and symptomatic EAE. LRG-47 antigen was detected in CD4 T-cells (FIG. S 1-3) and mononuclear phagocytes (FIG. S4-6) in spinal cords from EAE-induced mice by confocal microscopy. LRG-47 antigen was colocalized in a subpopulation of cells also staining for CD4 (FIG. S3) and CDl Ib (FIG. S6). These observations are consistent with a potential role of p47 GTPases in cells critical to the evolving host response.

Deficiency of LRG-47 protects animal from EAE

[0036] The close association of EAE with LRG-47 mRNA/protein expression led us to explore whether this mediator had a pathogenic role in EAE. To do this we used homozygous mutant mice devoid of LRG-47 (LRG-47 " ' " ) in the BlOPL strain. For these

studies, we employed the MPB model in which LRG-47 " ' " or age/strain- matched BlOPL littermate mice were immunized with an acylated N-terminal peptide fragment comprised of nine residues from myelin basic protein (1-9NAc MBP)[IO]. Compared to BlOPL controls, LRG-47 " ' " mice exhibited strikingly reduced incidence and severity of disease (FIG. ID, Table 1). Hemizygous LRG-47 +/" mice showed a slight reduction in the severity of symptomatic disease compared with strain-matched wild-type (WT) littermates, though there was no significance between WT controls and LRG-47 +/" mice (FIG. ID). Histologic analysis of spinal cord tissue harvested at the time of peak symptoms (day 18) after treatment with MBP demonstrated a striking reduction in the inflammatory cell infiltrates in LRG-47 " ' " mice, compared with other genotypes (FIG. IE). Thus, pathologic events correlate closely with symptomatology in terms of protection from disease in LRG-47 " ' " mice immunized with MBP.

Genotype N Incidence %

LRG+/+ 12/12 100

LRG+/- 12/12 100

LRG-/- 2/12 16.7

Table 1 The percentage of each group of mice developing disease

[0037] To begin to analyze mechanisms through which LRG-47 contributes to the pathogenesis of EAE, we analyzed the CD4 T-cell population in LRG-47 " ' " and control mice. The percentage of CD4 T-cells in the central nervous system (CNS), including brain and spinal cord eluates, from LRG-47 " ' " mice 20 days after MBP treatment was reduced -50%, compared with strain-matched controls (22.7% and 46.6% for LRG-47 " ' " and controls, respectivelyXFIG. 2A). The latter reduction was especially evident in the activated CD4 T- cell population that expresses high levels of CD44 (CD44 hl ). Activated CD4 T-cells comprised only 8-9% of cells in spinal cord eluates from EAE-induced LRG-47 " ' " mice versus 32% in EAE-induced WT animals (FIG. 2A). Most striking was the ~3-fold increase in apoptotic cells, based on annexin V staining, in the CD4 T-cell population harvested on day 20 from the CNS of LRG-47 " ' " mice compared with controls (30% versus 9%, for LRP-47 " ' " and controls, respectively; FIG. 2A). Similarly, the percentage of CD44 hl activated CD4 T- cells was significantly reduced in the lymphocyte population from lymph nodes of LRG-47 " ' "

mice compared to non-Tg (transgenic) littermate controls (FIG. 2B). A non-Tg littermate is an age-matched normal mouse in the same litters expressing endogenous levels of LRG-47 without transgene manipulation. Apoptosis of CD4 T cells, which includes activated T cells (shown as TUNEL-positive cells) was increased in the lymph nodes from LRG-47 " ' " mice, compared with WT littermates (FIG. 2C). To better understand the relationship between LRG-47 expression and cell proliferation in CD4 cells in vivo, mice received intraperitoneal BrdU, and FACS analysis was performed on cells isolated from the CNS using anti-CD4 and anti-BrdU IgG. Compared with WT littermate controls, cellular eluates from the CNS of LRG-47 " ' " mice displayed a strong reduction in the percentage of CD4 T-cells incorporating BrdU (4.69% in LRG-47 " ' " vs. 33% in WT mice; FIG. 2D). These data show a central role for LRG-47 in activation and/or expansion of autoreactive T-cells in EAE. Without being bound by theory, potentially, LRG-47 might promote survival of autoreactive T-cells in EAE.

[0038] These in vivo observations concerning a possible relationship between LRG-47 expression and the proliferative potential of CD4 T-cells led us to perform in vitro experiments to directly address this issue. T-cells were purified from spleens and lymph nodes of mice immunized 7 days previously with MBP, and cultured in the presence of MBP. T-cells from LRG-47 " ' " mice showed diminished proliferation in response to MBP, compared with T-cells from WT littermate controls (FIG. 2E). Correspondingly, the percentage of annexin V-positive cells was increased in LRG-47 " ' " mice indicating increased apoptosis, versus WT (wild type) littermate controls (FIG. 2F). Further analysis of CD4 T-cells isolated from spleens of EAE mice after MBP treatment displayed an increase in the percentage of TUNEL-positive CD4 T-cells from LRG-47 " ' " mice compared with WT mice (FIG. 2G). These experiments provide further support the role of LRG-47 in activation of autoreactive T cells, and decreased apoptosis of autoreactive T cells during EAE.

Induction of EAE by encephalitogenic T cells: effect of LRG-47

[0039] We next focused our experiments on an encephalitogenic CD4 ThI T-cell clone (IAElO)[IO] in order to assess the contribution of LRG-47 to EAE pathogenicity. Two approaches were utilized in these studies; 1) evaluation of the effect of LRG-47 on activation, proliferation and apoptosis of IAElO cells in vitro; and 2) determination of the impact of blocking LRG-47 activity on induction of EAE following adoptive transfer of activated IAElO cells in vivo. In order to characterize LRG-47 expression in IAElO cells, cultures were activated by addition of 1-9NAc MBP. LRG-47 transcripts increased by ~6-8-fold

comparing day 0 to days 4-10 after stimulation with MBP (FIG. 3A). High levels of LRG-47 transcripts were maintained from days 4-10, and thereafter decreased. In view of heightened expression of LRG-47 in activated IAElO cells, we assessed whether inhibition of LRG-47 would modulate properties of IAElO cells.

[0040] LRG-47 mainly localizes to the Golgi apparatus [11, 12] as shown by colocalization with Golgi marker GRAS65. Unstimuated IAElO cells were stained with antibodies to LRG-47 (FIG. 5B2) or to GRAS65 (FIG. 5Bl). LRG-47 was detected in Golgi apparatus (FIG. 3B3). However, after stimulation of IAElO cells with MBP (5μg/ml) for 4 hours, LRG-47 translocated from Golgi to the plasma membrane of cell surface (FIG. 3C2-3) as shown on non-permeable cells. Therefore, LRG-47 appears to redistribute to the cell surface during activation of IAElO cells induced by MBP. These data led us to determine the effect of inhibiting LRG-47 on properties of IAElO cells using specific antibodies to LRG-47. The location of LRG-47 on the cell surface of the activated cells makes it accessible to antibodies for therapeutic intervention to reduce its effect.

[0041] Activation of IAElO cells for 4 days in culture with MBP resulted in a corresponding increase in 3 H-thymidine incorporation which was blocked in a dose- dependent manner by anti-LRG-47 IgG (A- 19 ), but not by nonimmune (NI) IgG (FIG. 3D). Consistent with these data, FACS analysis of CFSE-FITC-labeled IAElO cells activated by exposure to MBP displayed time-dependent proliferation; cell labeling increased from 0.37% to 30.6% corresponding to the interval from 24 hour to 96 hours. The latter proliferative response was inhibited by ~10-fold at the 96 hour time point on addition of anti-LRG-47 IgG (Al 9). In contrast, NI IgG had no effect (FIG. 3E). These data indicate that blockade of LRG-47 suppresses antigen induced proliferation of encephalitogenic T-cells.

[0042] Apoptosis of lymphocytes is one of the major homeostatic mechanisms in the immune system. In view of the enhanced susceptibility of LRG-47 " ' " mice to protozoan and mycobaterial infection [7, 13, 14], we sought to determine whether LRG-47 functioned as a survival factor in MBP-primed encephalitogenic T-cells. To address this issue, we examined if the lack of proliferative activity in IAElO cells activated in the presence of MBP actually reflected an enhanced degree of apoptosis. MBP-treated IAElO cells displayed a dose- dependent increase in the population of annexin V-positive cells as the concentration of anti- LRG-47 IgG was increased from 5 to 20 μg/ml reaching up to ~70% of the cells, whereas NI IgG was without effect (72 hr time point; FIG. 3F). These results were paralleled by those

observed on time-dependent assessment of annexin V staining (FIG. 3G), propidium iodide uptake (FIG. 3H), and live cell assay (FIG. 31) in IAElO cells. Addition of anti-LRG-47 IgG showed evidence of increased cell death/apoptosis in each case. These data show that LRG- 47 is a key factor in expansion of encephalitogenic CD4 T-cells in response to antigen, in this case MBP, by inhibiting apoptosis.

[0043] The ultimate confirmation of this result concerns the encephalitogenic potential of IAElO cells in vivo. MBP-activated IAElO cells were treated with either anti-LRG-47 IgG or NI IgG, and adoptively transferred into BlOPL mice. Wild- type (WT) BlOPL mice treated with IAElO cells alone or with IAElO cells/NI IgG, showed symptomatic EAE that progressed rapidly, occurring in all animals in the 12-14 day window, and reaching a clinical score/severity of 3-4 (FIG. 4A, Table 2). In contrast, WT mice treated with IAElO cells exposed to anti-LRG-47 IgG (A- 19) displayed only 77.8% incidence of symptomatic EAE, with a quite different course (delayed) and a dramatically lower clinical score of only 1-1.5. Thus, clinical symptomatology mirrors what is expected if LRG-47 was an essential cofactor for pathogenicity of IAElO cells.

Table 2

Transfer EAE N Incidence %

WT. 9/9 100

Wt./A19 7/9 77.8

Wt./NI-IgG 6/6 100

[0044] Our clinical findings in mice indicated the importance of analyzing the nature of cells infiltrating the spinal cord under each of these situations following induction of EAE by infusion of IAElO cells. At day 14-16, during the peak of EAE symptoms, there were no significant differences in the number of CD4 T-cells in spinal cord eluates among WT mice adoptively transferred with IAElO cells that were treated with anti-LRG-47 IgG, control NI IgG, or in IAElO cells alone without any treatment. (FIG. 4C). However, apoptotic cells in spinal cord eluates, as shown by annexin V and TUNEL staining, showed substantial differences in each of these situations. Whereas there were fewer apoptotic cells in eluates from WT mice transferred with IAElO cells alone or treated with control NI IgG, there were

significantly more apoptotic cells in samples from WT mice transferred with IAElO cells treated with anti-LRG-47 IgG (A19)(FIG. 4B, D). Thus, death of CD4 T-cells in the spinal cord between days 14 and 28, was the likely explanation for their reduction in spinal cord eluates in WT mice transferred with IAElO cells treated with anti-LRG-47 IgG, versus WT mice treated with IAElO cells alone observed at days 28 (FIG. 4E).

Effect ofLRG-47 on IFN-γ induction

[0045] To further understand the role of LRG-47 in regulating CD4 T-cell function/survival in the setting of pathogenic auto-immunity, we examined how LRG-47 expression affects the expression of IFN-γ in IAElO cells. Cultured IAElO cells were stimulated with MBP-derived peptide alone or with MBP-derived peptide in the presence of either anti-LRG-47 IgG or NI IgG. IFN-γ was measured in supernatants (FIG. 4F). We found that LRG-47 is a negative regulator of IFN- γ. Inhibition of LRG-47 in cultured IAElO cells with anti-LRG-47 IgG resulted in dose- and time-dependent increases in levels of IFN-γ (FIG. 4F) secreted into the medium. Moreover, IFN-γ levels were increased in cultured CD4 T-cells isolated from lymph nodes/spleens of LRG-47 " ' " mice at 7 days after immunization with MBP-derived peptide (FIG. 4G). In addition, levels of IFN-γ were elevated in the sera of LRG-47 " ' " mice compared to WT mice induced with MBP to develop EAE (FIG. 4H).

[0046] Multiple sclerosis is the most common autoimmune disease of the central nerve system (CNS). T lymphocytes that are reactive with components of myelin sheaths are thought to play key roles in disease pathogenesis. Experimental autoimmune encephalitis (EAE) is the best animal model of multiple sclerosis. During EAE, the interaction of active CD4+ T cells and myelin antigens apparently provoke a massive destructive inflammatory response that promotes continuing proliferation of T cells and macrophage activation that escalates to intensive inflammation and dymyelination of nerves in the CNS. The presence of LRG-47 in auto-reactive CD4+ T cells and macrophages, and increased expression of LRG- 47 in EAE-affected spinal cord tissue indicate a key role of LRG-47 on the induction of EAE. Blockade of LRG-47 expression promotes apoptosis, which diminishes expansion of activation and proliferation of MBP-reactive CD4+ T cells. Furthermore, activation of an encephalitogenic CD4+ ThI T-cell clone with MBP was clearly suppressed in the presence of anti-LRG-47 specific IgG but not by non-immune IgG. At the same time, apoptotic cells were significantly increased in response to LRG-47 blockade. Also, LRG-47 blockade

prevented induction of EAE when fully activated IAElO cells were transferred into B 10. PL mice. Blockade of LRG-47 also increased apoptotic encephalitogenic CD4+ T cells in EAE- affected spinal cord tissues. These data show a requirement of LRG-47 protein for persistence of CD4 T-cells in the CNS to propagate the immune/inflammatory response. Thus it is likely that death of encephalitogenic CD4 T-cells and their decreased proliferation account for the relatively mild course of EAE in WT animals adoptively transferred with IAElO cells treated with anti-LRG-47.

[0047] IFN-γ has been shown to function as a negative regulator of lymphocyte expansion, eliminating activated CD4 T-cells by promoting apoptosis during the immune response [1-3, 17]. Without being bound by theory, we propose that blockade of LRG-47 interrupts a negative feedback loop that is triggered after the immune response is initiated, thereby limiting subsequent expression of IFN-γ. As a result, re-expression of IFN-γ leads to levels which attenuate the ongoing immune response.

[0048] While LRG-47 has been shown to contribute to the host response to intracellular bacterial infection [9, 12, 16] and has been suggested to function as a survival factor in the context of mycobacterial infection [14, 18]. Our data indicate that it is also present in CD4 T-cells where it promotes survival of pathogenic self-reactive T-cells at least in part by preventing T-cell apoptosis. Without being bound by theory, we propose that induction of EAE by activation of autoreactive MBP-specific T-cells leads to early (day 7) secretion of IFN-γ initiating a DTH (Delayed Type Hypersensitivity) response in the CNS. Induction of LRG-47 by IFN-γ facilitates the survival of the expanded self -reactive T-cell population, thus LRG-47 also plays an important role in the pathogenesis of autoimmune disease including MS and EAE. This protective role of LRG-47 for encephalitogenic CD4 T-cells in EAE (thereby enhancing their pathogenicity) contrasts with its role in buttressing a protective host response to certain pathogens [4, 7, 12, 14]. The translocation of LRG-47 to the outer cell membrane adds a new and unexpected dimension to the biology of this GTPase. Its location on the outer cell membrane means that it is an accessible target for immune therapy (i.e. blocking LRG-47 activity with anti-LRG-47 antibodies.) Certain embodiments of the invention are directed to treating or preventing an autoimmune disease including MS by administering anti-LRG47 antibodies or fragments or variants thereof, preferably in humanized form and preferably against human LRG-47, in order to reduce LRG-47 expression.

SiRNA Effectively Blocks LRG-47 Expression in IAElO T-cells

[0049] To test the ability of SiRNA to block LRG-47 expression, IAElO T-cells were stimulated with MBP (5 micrograms per ml) for 48 hours. IAElO T cells were transfected with siRNA targeted to mouse LRG-47 (sense sequence: 5'- GGUUACCUGAGGUCAGUAGtt-3', antisense sequence: 5'-

CUACUGACCUCAGGUAACCtg-3', : antisense 3'- GGUUACCUGAGGUC AGUAGtt-5 ' , SEQ ID NO; 5) using siPORT Amine (Ambion) for additional 24. Levels of LRG-47 mRNA in cells were determined by real-time quantitative PCR and RT-PCR. The level of LRG-47 mRNA was measured using real-time quantitative PCR (right) and RT-PCR (left). The results shown in FIG.5 show that LRG-47 expression was reduced by about 40% by transfection with the siRNA. In addition to reviewing the level of LRG-47 mRNA expression, we also looked at the clinical symptoms in EAE in vivo. Blockade of LRG-47 expression by siRNA attenuates the clinical symptoms in EAE mice. MBP-activated IAElO cells were treated with either siRNA-LRG 47 or vehicle, and adoptively transferred into BlOPL mice. In wild-type (WT) BlOPL mice treated with IAElO cells alone or IAElO cells, symptomatic EAE progressed rapidly, occurring in all animals in the 15-20 day window, and reaching a clinical score/severity of 3-4. In contrast, WT mice treated with IAElO cells exposed to siRNA-LRG 47 displayed a delay in the onset of EAE and attenuation of symptomatic EAE, a clinical score of only 1-2.5. Thus, clinical symptomatology mirrors what might be expected if LRG-47 was an essential cofactor for pathogenicity of IAElO cells.

[0050] As is discussed below certain embodiments of the invention are directed to methods for treating or preventing MS by administering an agent that reduces LRG-47 expression, preferably human LRG-47 for human use, including siRNAs. Although we tested only one siRNA against a murine LRG-47, the field of antisense technology is well established and it is routine in the art to develop antisense or siRNA molecules that inhibit expression of a given target gene or mRNA.

LRG-47 Levels are Elevated about Spinal Cord, CSF and Serum from MS Patients

[0051] In order to determine whether LRG-47 levels are elevated in a human patient having MS, we analyzed MS-affected spinal cord tissues. LRG-47 levels were measured using quantitative real-time PCR, which showed an approximately 16-fold increase in LRG- 47 in the MS-affected patient sample, compared to a spinal cord sample from an age-matched

unafflicted control. FIG. 7 shows expression of LRG-47 antigen in spinal cord tissue from MS patients. FIG. 7A. LRG-47 antigen was prominently elevated in MS-affected spinal cord tissue as compared with age-matched normal control by immunoblotting with specific anti- LRG-47 IgG. FIG. 7B-C. Immunostaining with specific anti-LRG 47 IgG, showed that LRG-47 (B2 & C2) was especially intense in mononuclear monocytes (Bl) and CD4+ T cell (Cl), as shown by co-staining with CD68 and CD4+, respectively (FIG. 7Bl & Cl). There was no positive staining when anti-LRG IgG was replaced by nonimmune IgG (data not shown). These results taken together show the significance of LRG-47 expression in the pathogenesis of MS and its relevance to autoimmune diseases other than EAE.

[0052] FIG. 8 shows the localization of LRG-47 antigen in spinal cords of EAE mice. Spinal cord sections were co- stained with goat anti-LRG-47 (1 micrograms/ml) and rat anti- CD4 or rat anti-CDl lb, followed by rabbit anti-goat IgG conjugated with FITC, and anti-rat- conjugated with TRITC. LRG-47 antigen (panels S2 and S5) was detected in CD4+ T-cells (Sl) and CDl lb-positive mononuclear cells (S4). S3 and S6 are merged with S1/S2 and S3/S4, respectively. Magnification: Sl-3, x600; S4-6, x400.

[0053] Recently, we showed that level of LRG-47 was increased in the serum of EAE mice compared to the serum of naϊve mice. Similarly, LRG-47 was up-regulated in the cerebral spinal fluid (CSF) or serum taken from MS patients at the activated stage. FIG. 9 shows immuno-dot blotting with antibody to mouse LRG-47 of mouse serum demonstrating a significant increase in LRG-47 in the serum of EAE mice compared to serum from naϊve mice. Levels of LRG-47 were quantified by NIH image program. Naϊve mice showed about 25 units of intensity compared to about 85 for EAE mice at day 7 and about 80 for day 12, indicating more than a 3 -fold increase in serum LRG-47. We next looked at human CSF and serum from MS patients compared to age-matched control patients using the to anti -human - LRG-47 AB antibody that was made in our laboratory. The results an approximate 4-fold increase in the levels of LRG-47 in the serum and CSF of MS patients. Based on these results we describe diagnostic methods below based on measuring elevated levels of secreted LRG-47 in serum or CSF of a patient compared to controls. Certain embodiments are also directed to the new anti-human -LRG-47 138AB antibody itself, or fragments or variants thereof, preferably in humanized form. Embodiments are also directed to pharmaceutical formulations comprising the new anti-human -LRG-47 138AB antibody itself, or fragments

or variants thereof, preferably in humanized form; and to its use to treat, prevent or diagnose MS or other autoimmune disease.

[0054] We made rabbit anti human LRG-47 antibody, called 138AB as follows: Synthetic peptides [Ac-VGHEGKASPPTELVKATQR-amide (54-71) and Ac- EDMGKKFYIVWTKLDMDLC-amide (132-149))] were immunized to rabbit, and then antiserum was affinity purified to IgG. This affinity purified antibody specific for human LRG was used for our study. Certain embodiments are directed to this antibody or a fragment or variant thereof preferably in humanized form, and to its therapeutic use to treat, prevent or diagnose MS or other autoimmune disease.

Treatment and Prevention of MS using Anti-LRG-47 Antibodies

[0055] Certain embodiments of the invention are directed to methods to treat or prevent MS in an animal, preferably a human, by administering a therapeutically effective amount of an isolated anti-LRG-47 antibody or biologically active fragment thereof, including polyclonal and monoclonal antibodies preferably humanized. In a preferred embodiment the isolated antibody is a polyclonal anti-human LRG47 antibody named 138AB or fragment or variant thereof that was made in our laboratory as described herein. An embodiment of the invention is also directed to this isolated polyclonal anti-human LRG47 138AB or fragment or variant thereof, preferably a humanized form, and to its therapeutic use as described herein. In another preferred embodiment the isolated antibody for use in treating or preventing MS includes humanized forms of the polyclonal anti-LRG47 antibodies commercially available from Santa Cruz Biotechnology, Inc. including those listed below, or a fragment or variant thereof. These antibodies can also be used to measure the level of secreted LRG-47 in a biological sample as described herein for diagnostic testing, and can be included in the kits described below. Non humanized forms can be used for the diagnostic claims herein.

[0056] For the purpose of this invention, a therapeutically effective amount of a compound is an amount that achieves the desired biologic or therapeutic effect, namely an amount that prevents, reduces or ameliorates one or more symptoms of the enumerated diseases being treated or prevented. In some embodiments the starting point for determining a therapeutic amount is an amount that significantly reduces the amount of secreted LRG-47 protein or LRG-47 mRNA in a patient biological sample, (i.e. a statistically significant reduction, preferably a two fold reduction).

[0057] LRG-47 specific siRNA gene silencers are also available. These include: LRG-47 siRNA (m): sc-41794.

[0058] The location of LRG-47 protein on the surface of activated T cells is particularly important for treating or preventing MS using antibody therapy because this target protein is accessible to the antibodies. Anti-LRG-47 therapy targets circulating activated T cells, so the antibody does not need to cross the BBB. The amount of anti-LRG-47 to be administered therapeutically ranges from about lug to 100 ug /ml. This amount typically varies and can be an amount sufficient to achieve serum therapeutic agent levels typically of between about 1 microgram per milliliter and about 10 micrograms per milliliter in the subject. In the context of the present invention, anti-LRG-47 antibodies are a type of neutralizing antibody that prevents LRG-47 from activating T cells.

[0059] An "antibody" refers to an intact immunoglobulin or to an antigen-binding portion thereof that competes with the intact antibody for specific binding. Antigen-binding portions may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. Antigen-binding portions include, inter alia, Fab, Fab', F(ab').sub.2, Fv, dAb, and complementarity determining region (CDR) fragments, single-chain antibodies (scFv), chimeric antibodies, diabodies and polypeptides that contain at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding to the polypeptide.

[0060] An "immunoglobulin" is a tetrameric molecule. In a naturally-occurring immunoglobulin, each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa) and one "heavy" chain (about 50 70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more

amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as .kappa, and .lamda. light chains. Heavy chains are classified as .mu., .DELTA., .gamma., .alpha., or .epsilon., and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D" region of about 10 more amino acids. See generally, Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N. Y. (1989)) (incorporated by reference in its entirety for all purposes). The variable regions of each light/heavy chain pair form the antibody binding site such that an intact immunoglobulin has two binding sites.

[0061] Immunoglobulin chains exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminus to C-terminus, both light and heavy chains comprise the domains FRl, CDRl, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk J. MoI. Biol. 196:901 917 (1987); Chothia et al. Nature 342:878 883 (1989).

[0062] An Fab fragment is a monovalent fragment consisting of the VL, VH, CL and CH I domains; a F(ab').sub.2 fragment is a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; a Fd fragment consists of the VH and CHl domains; an Fv fragment consists of the VL and VH domains of a single arm of an antibody; and a dAb fragment (Ward et al., Nature 341:544 546, 1989) consists of a VH domain.

[0063] A single-chain antibody (scFv) is an antibody in which a VL and VH regions are paired to form a monovalent molecules via a synthetic linker that enables them to be made as a single protein chain (Bird et al., Science 242:423 426, 1988 and Huston et al., Proc. Natl. Acad. Sci. USA 85:5879 5883, 1988). Diabodies are bivalent, bispecific antibodies in which VH and VL domains are expressed on a single polypeptide chain, but using a linker that is too short to allow for pairing between the two domains on the same chain, thereby forcing the domains to pair with complementary domains of another chain and creating two antigen binding sites (see e.g., Holliger, P., et al., Proc. Natl. Acad. Sci. USA 90:6444 6448, 1993,

and PoIj ak, R. J., et al., Structure 2:1121 1123, 1994). One or more CDRs may be incorporated into a molecule either covalently or noncovalently to make it an immunoadhesin. An immunoadhesin may incorporate the CDR(s) as part of a larger polypeptide chain, may covalently link the CDR(s) to another polypeptide chain, or may incorporate the CDR(s) noncovalently. The CDRs permit the immunoadhesin to specifically bind to a particular antigen of interest.

[0064] An antibody may have one or more binding sites. If there is more than one binding site, the binding sites may be identical to one another or may be different. For instance, a naturally-occurring immunoglobulin has two identical binding sites, a single-chain antibody or Fab fragment has one binding site, while a "bispecific" or "bifunctional" antibody has two different binding sites.

[0065] An "isolated antibody" is an antibody that (1) is not associated with naturally- associated components, including other naturally-associated antibodies, that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature.

[0066] The term "human antibody" includes all antibodies that have one or more variable and constant regions derived from human immunoglobulin sequences. In a preferred embodiment, all of the variable and constant domains are derived from human immunoglobulin sequences (a fully human antibody). These antibodies may be prepared in a variety of ways, as described below.

[0067] A humanized antibody is an antibody that is derived from a non-human species, in which certain amino acids in the framework and constant domains of the heavy and light chains have been mutated so as to avoid or abrogate an immune response in humans. Alternatively, a humanized antibody may be produced by fusing the constant domains from a human antibody to the variable domains of a non-human species. Examples of how to make humanized antibodies may be found in U.S. Pat. Nos. 6,054,297, 5,886,152 and 5,877,293, incorporated herein by reference.

[0068] The term "chimeric antibody" refers to an antibody that contains one or more regions from one antibody and one or more regions from one or more other antibodies.

[0069] Fragments or analogs of antibodies can be readily prepared by those of ordinary skill in the art following the teachings of this specification. Preferred amino- and carboxy- termini of fragments or analogs occur near boundaries of functional domains. Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases. Preferably, computerized comparison methods are used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known structure and/or function. Methods to identify protein sequences that fold into a known three-dimensional structure are known. Bowie et al. Science 253:164 (1991).

Antisense nucleotides and siRNA

[0070] Certain embodiments of the invention are directed to the use of antisense or siRNA to block expression of LRG-47 thereby treating or preventing MS or other autoimmune or T-cell mediated diseases, preferably in humans. Antisense nucleotides can be designed using routine skill in the art to target human DNA or mRNA encoding LRG-47 as is described in more detail below. The full cDNA sequence for the human LRG-47 gene is not yet known; the partial sequence known at the present time is public and is set forth in BC128168, incorporated herein by reference as SEQ ID NO: 1. When the full sequence becomes known and published, persons of skill in the art will be able to construct appropriate antisense nucleotides and siRNA using methods known in the art. The gene and mRNA sequence for mouse LRG-47 is public, is identified by accession NO: U19119, and is incorporated herein by reference as SEQ ID. No:3 (gene sequence) and SEQ ID NO: 4 (amino acid sequence).

[0071] Another embodiment for treating or preventing multiple sclerosis in an animal preferably a human, has the steps:

a. determining a pre-treatment level of human LRG-47 protein in a pre-treatment biological sample taken from the animal,

b. administering an amount of an antisense nucleotide or siRNA that specifically hybridizes with the gene or mRNA encoding LRG-47,

c. determining a post-treatment level of LRG-47 protein in a post-treatment biological sample taken from the animal,

d. comparing the pre-treatment and post-treatment levels of LRG-47 protein in the respective biological samples, and

e. determining that treatment is effective if the post-treatment level of LRG-47 is significantly lower than the pre-treatment level.

[0072] The method includes further step f. if the treatment is not effective: if the difference between the pre-treatment and post-treatment levels of LRG-47 protein is not significantly different, then increasing the amount or frequency of administration of the antisense or siRNA until the post-treatment level of LRG-47 is significantly lower than the pre-treatment level. These same basic methods can be used to treat any autoimmune disease because all are associated with elevated levels of activated T cells.

[0073] Any compound that inhibits LRG-47 expression can be used to treat or prevent MS or an autoimmune disease.

[0074] Any method known in the art for measuring LRG-47 expression (measuring LRG- 47 protein levels or mRNA, including using PCR and Northern blot hybridization) in the biological sample can be used including those described herein. By "significantly lower" is meant that there is a statistically significant difference between pre- and post-treatment levels of LRG-47 protein or mRNA. LRG-47 can be measured in any biological sample, preferably serum or cerebrospinal fluid (CSF), or blood using any method known in the art. Biopsy samples (for example of skin and fibroblasts), if available, can also be analyzed. The siRNA or antisense compounds directed to LRG-47 DNA or mRNA can be formulated into a pharmaceutical carrier using any method known in the art, and administered by any method known in the art.

[0075] The isolated antisense or siRNA nucleic acid molecules for use in the invention comprise a nucleic acid molecule which is a complement of the partial cDNA (confirm it is cDNA) sequence shown in SEQ ID NO:1, encoding human LRG-47, or a portion thereof. Antisense can also be complementary to mRNA encoding LRG-47. A nucleic acid molecule which is complementary to a given nucleotide sequence is one which is sufficiently complementary to the given nucleotide sequence that it can hybridize to the given nucleotide sequence thereby forming a stable duplex.

[0076] Antisense nucleic acid molecules, i.e., molecules which are complementary to a sense nucleic acid encoding a protein, e.g., complementary to the coding strand of a double- stranded DNA molecule (or cDNA) or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid. The antisense nucleic acid can be complementary to an entire LRG-47 coding strand, or to only a portion thereof, e.g., all or part of the protein coding region (or open reading frame). An antisense nucleic acid molecule can be antisense to a noncoding region of the coding strand of a nucleotide sequence encoding LRG-47. The noncoding regions ("5' and 3' untranslated regions") are the 5' and 3' sequences which flank the coding region and are not translated into amino acids.

[0077] Given the coding strand sequences encoding LRG-47 disclosed herein (e.g., SEQ ID NO:1), antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of LRG-47 mRNA, but more preferably is an oligonucleotide which is antisense to only a portion of the coding or noncoding region of LRG-47 mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of LRG-47mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Examples of modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-hodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl- 2-thouridine, 5-carboxymethylaminometh- yluracil, dihydrouracil, beta-D- galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1 -methylinosine, 2,2- dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-metnylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2- thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2- methylthio-N6-isopenten- yladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thlouracil, 4-thiouracil, 5-methyluracil,

uracil-5-oxyacetic acid methylester, uracil-5-cxyacetic acid (v), 5-methyl-2-thiouracil, 3- (3- amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).

[0078] The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding the protein of interest to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementary to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention include direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens. The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.

[0079] An antisense nucleic acid molecule of the invention can be an alpha- anomeric nucleic acid molecule. An .alpha. -anomeric nucleic acid molecule forms specific double- stranded hybrids with complementary RNA in which, contrary to the usual .beta. -units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids. Res. 15:6625-6641). The antisense nucleic acid molecule can also comprise a 2'-o -methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1987) FEBS Lett. 215:327-330). All of the methods described in the above articles regarding antisense technology are incorporated herein by reference.

[0080] The invention also encompasses ribozymes. Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave LRG-47mRNA transcripts thereby inhibit translation of LRG-47mRNA. A ribozyme having specificity for an LRG-47-encoding nucleic acid can be designed based upon the nucleotide sequence of a LRG-47cDNA disclosed herein. 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 LRG-47-encoding mRNA. See, e.g., Cech et al. U.S. Pat. No. 4,987,071; and Cech et al. U.S. Pat. No. 5,116,742. Alternatively, LRG-47mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel and Szostak (1993) Science 261:1411-1418, incorporated herein by reference.

[0081] As used herein, the term "nucleic acid" refers to both RNA and DNA, including cDNA, genomic DNA, and synthetic (e.g., chemically synthesized) DNA. The nucleic acid can be double-stranded or single- stranded (i.e., a sense or an antisense single strand). As used herein, "isolated nucleic acid" refers to a nucleic acid that is separated from other nucleic acid molecules that are present in a mammalian genome, including nucleic acids that normally flank one or both sides of the nucleic acid in a mammalian genome (e.g., nucleic acids that flank an ARPKD gene). The term "isolated" as used herein with respect to nucleic acids also includes any non-naturally-occurring nucleic acid sequence, since such non- naturally-occurring sequences are not found in nature and do not have immediately contiguous sequences in a naturally-occurring genome.

[0082] An isolated nucleic acid can be, for example, a DNA molecule, provided one of the nucleic acid sequences normally found immediately flanking that DNA molecule in a naturally-occurring genome is removed or absent. Thus, an isolated nucleic acid includes, without limitation, a DNA molecule that exists as a separate molecule (e.g., a chemically synthesized nucleic acid, or a cDNA or genomic DNA fragment produced by PCR or restriction endonuclease treatment) independent of other sequences as well as DNA that is incorporated into a vector, an autonomously replicating plasmid, a virus (e.g., a retrovirus, lenti virus, adenovirus, or herpes virus), or into the genomic DNA of a prokaryote or eukaryote. In addition, an isolated nucleic acid can include an engineered nucleic acid such

as a DNA molecule that is part of a hybrid or fusion nucleic acid. A nucleic acid existing among hundreds to millions of other nucleic acids within, for example, cDNA libraries or genomic libraries, or gel slices containing a genomic DNA restriction digest, is not to be considered an isolated nucleic acid.

Small Interfering RNA

[0083] A person of skill in the art can make any number of different siRNAs based on the partial human cDNA gene sequence SEQ ID NO: 1 for LRG-47. When the full gene sequence becomes available, other siRNAs can be designed to block expression, i.e. translation o LRG-47 using routine methods known in the art.

[0084] Patent Application 20040023390 (the entire contents of which are hereby incorporated by reference as if fully set forth herein ) teaches that double-stranded RNA (dsRNA) can induce sequence-specific posttranscriptional gene silencing in many organisms by a process known as RNA interference (RNAi). However, in mammalian cells, dsRNA that is 30 base pairs or longer can induce sequence-nonspecific responses that trigger a shutdown of protein synthesis and even cell death through apoptosis. Recent work shows that RNA fragments are the sequence- specific mediators of RNAi (Elbashir et al., 2001). Interference of gene expression by these small interfering RNA (siRNA) is now recognized as a naturally occurring strategy for silencing genes in C. elegans, Drosophila, plants, and in mouse embryonic stem cells, oocytes and early embryos (Cogoni et al., 1994; Baulcombe, 1996; Kennerdell, 1998; Timmons, 1998; Waterhouse et al., 1998; Wianny and Zernicka- Goetz, 2000; Yang et al., 2001 ; Svoboda et al., 2000).

[0085] In mammalian cell culture, a siRNA-mediated reduction in gene expression has been accomplished by transfecting cells with synthetic RNA nucleic acids (Caplan et al., 2001 ; Elbashir et al., 2001). The 20040023390 application, the entire contents of which are hereby incorporated by reference as if fully set forth herein, provides methods using a viral vector containing an expression cassette containing a pol II promoter operably-linked to a nucleic acid sequence encoding a small interfering RNA molecule (siRNA) targeted against a gene of interest.

[0086] As used herein RNAi is the process of RNA interference. A typical mRNA produces approximately 5,000 copies of a protein. RNAi is a process that interferes with or significantly reduces the number of protein copies made by an mRNA. For example, a

double- stranded short interfering RNA (siRNA) molecule is engineered to complement and match the protein-encoding nucleotide sequence of the target mRNA to be interfered with. Following intracellular delivery, the siRNA molecule associates with an RNA-induced silencing complex (RISC). The siRNA-associated RISC binds the target mRNA (such as mRNA encoding LRG-47) through a base-pairing interaction and degrades it. The RISC remains capable of degrading additional copies of the targeted mRNA. Other forms of RNA can be used such as short hairpin RNA and longer RNA molecules. Longer molecules cause cell death, for example by instigating apoptosis and inducing an interferon response. Cell death was the major hurdle to achieving RNAi in mammals because dsRNAs longer than 30 nucleotides activated defense mechanisms that resulted in non-specific degradation of RNA transcripts and a general shutdown of the host cell. Using from about 20 to about 29 nucleotide siRNAs to mediate gene-specific suppression in mammalian cells has apparently overcome this obstacle. These siRNAs are long enough to cause gene suppression but not of a length that induces an interferon response.

Methods for Diagnosing MS\

[0087] Certain other embodiments are also directed to a method for diagnosing multiple sclerosis in an animal preferably a human by assessing the level of human LRG-47 protein in any biological sample taken from the animal, preferably serum, cerebrospinal fluid (CSF) or blood, but also in a tissue sample including fibroblasts or skin.

[0088] A method for diagnosing a patient having MS or at risk of developing MS or having MS, involves the steps of

a. determining a level of LRG-47 protein in a biological sample taken from the patient and in a corresponding control sample taken from a control subject that is not affected with multiple sclerosis,

b. comparing the level of LRG-47 protein in the two biological samples and

c. and concluding that the patient has or is at risk of developing multiple sclerosis if the LRG-47 level in the patient sample is significantly higher than the level in the control sample. This method can also be used to detect an autoimmune disease associated with elevated T cells. It is hoped that early detection of changes in LRG-47 expression will facilitate early diagnosis and treatment.

[0089] If blood leukocytes or other cells are used in the assay then one can monitor either LRG-47 protein or mRNa levels to make the diagnosis. If the difference between the control and patient levels is very high, and the patient displays other indicia of MS, this test can confirm a diagnosis of MS. Another embodiment is directed to a method for monitoring the level of advancement or progression of MS in a patient by determining changes over time in the LRG-47 protein levels, preferably with regular assessments even during periods of apparent remission. If the level increases over time then the diseases is progressing or becoming more advanced. Monitoring the level of LRG-47 or LRG-47 mRNA in a patient sample can also be sued to measure the efficacy of MS therapy; if the levels are going down the therapy is effective. The level of LRG-47 may rise before any symptoms of MS have noticeably worsened, making it possible to intervene early in treatment to prevent a serious relapse.

[0090] Other embodiments are directed to a diagnostic kit for determining the level of LRG-47 in a biological sample that includes : a) a vessel or vessels for receiving a blood/serum or CSF or cell sample from the subject; b) an agent that specifically detects LRG-47 protein or amplifies LRG-47 mRNA; and c) printed instructions for detecting the LRG-47 protein or the amplified LRG-47 mRNA in the sample. The agent can be a primary anti-LRG-47 antibody for quantifying LRG-47, and a secondary antibody for detecting binding of the primary antibody can also be included in the kit. In another embodiment the antibody is already labeled for easy detection. Any antibody described herein or new antibodies that become known that target LRG-47 can be used, preferably those that target human LRG-47.

Pharmaceutical preparations

[0091] The present invention also includes pharmaceutical compositions and formulations which include the antibodies and antisense compounds of the invention that are administered to treat or prevent multiple sclerosis. The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. The preferred rout of administration is intravenous, but administration can also parenteral including intra-arterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration.

[0092] Pharmaceutical compositions of the present invention contain the therapeutic agent (antibodies or antisense nucleic acids or si RNA that reduce the expression of LRG-47) in an amount sufficient to prevent or treat MS or an autoimmune disease in a subject. The therapeutic agent can be formulated with an acceptable carrier using methods well known in the art. The actual amount of therapeutic agent will necessarily vary according to the particular formulation, route of administration, and dosage of the pharmaceutical composition, the specific nature of the condition to be treated, and possibly the individual subject. The dosage for the pharmaceutical compositions of the present invention can range broadly depending upon the desired effects, the therapeutic indication, the route of administration, regime, and purity and activity of the composition.

[0093] Pharmaceutical compositions of the present invention are suitable for administration to a subject in need of prophylaxis or therapy of MS or an autoimmune disease. The subject is preferably a human but can be non-human as well. A suitable subject can be an individual who is suspected of having, has been diagnosed as having, or is at risk of developing MS or an autoimmune disease, and like conditions as can be determined by one knowledgeable in the art.

[0094] Techniques for formulation and administration can be found in "Remington: The Science and Practice of Pharmacy" (20.sup.th edition, Gennaro (ed.) and Gennaro, Lippincott, Williams & Wilkins, 2000), incorporated herein by reference. The pharmaceutical compositions of the present invention can be administered to the subject by a medical device, such as, but not limited to, catheters, balloons, implantable devices, biodegradable implants, prostheses, grafts, sutures, patches, shunts, or stents.

[0095] The invention has been described in the foregoing specification with reference to specific embodiments. It will however be evident that various modifications and changes may be made to the embodiments without departing from the broader spirit and scope of the invention. The specification and drawings are to be regarded in an illustrative rather than a restrictive sense.

EXAMPLES

[0096] Mice. LRG-47 knockout mice (LRG-47 " ' " ) were generated as described previously [9]. Mice were backcrossed into the BlOPL background (N=IO generations). Wild-type (WT) littermates (in BlOPL) served as controls.

[0097] Induction of EAE. EAE was induced as previously described [10] using a protocol approved by Columbia University's Institutional Animal Care and Use Committee. Induction of EAE using the MBP model was performed by immunizing BlOPL mice subcutaneously with 1-9NAc myelin basic protein (MBP) in complete Freund's adjuvant (CFA) on day 0. For adoptive transfer of EAE, an activated encephalitogenic CD4 ThI clone (1AE10; 10 x 10 6 cells/animal) was administered intravenously to sublethally irradiated (350 R) mice. Pertussis toxin (100 μg/mouse) was given via the tail vein on days 1 and 3. Clinical symptoms were scored from 1 to 5 as described [10].

Isolation of mononuclear cell from the CNS.

[0098] To analyze cellular infiltrates in the central nerve system (CNS), mice were deeply anesthetized and perfused intracardially with PBS (30 ml). Brain and spinal cord were chopped into pieces and digested for 45 min at 37 0 C in Hanks balanced salt solution containing 0.2 units of liberase Rl (Roche) and 50μg ~l DNase I. Softened tissue fragments were forced through a 40 μm cell strainer, and homogenates were resuspended in 70% Percoll (Sigma) and overlaid with 30% Percoll. Gradients were centrifuged at 2000 x g for 30 min. Mononuclear cells in the CNS were collected from the Percoll interface.

[0099] Flow Cytometry. Cells were washed with FACS buffer [PBS containing 0.1 % (w/v) sodium azide and 2% (v/v) fetal calf serum] and preincubated with CD16/CD32 monoclonal antibody (clone 2.4G2, Pharmingen) for 15 min at 4 0 C to block non-specific binding to Fc receptors. Fluorochrome-conjugated monoclonal antibodies (rat anti-mouse CD4-PerCP, rat anti-mouse CD44-PE and appropriate isotype controls) were purchased from Pharmingen. After incubation, cells were washed twice with FACS buffer and data were acquired on a FACSCalibur flow cytometer (Becton Dickison, Franklin Lakes,NJ), analyzed by using FLOWJO software (Treestar, San Carlos, CA).

[0100] Proliferation assays. For T-cell proliferation, cells isolated from spleens and lymph nodes were cultured in the serum free medium with 1-9NAc MBP (2.5 μg/ml) for 72 hours. 3 H-thymindine (1 μCi/well) was added 16 hours prior to harvesting. For the in vivo 5- Bromo-2'-Deoxyuridine (BrdU, Sigma) uptake assay, mice were injected intraperitoneally with 1 mg of BrdU twice on days 13 and 14 during the course of EAE. CNS mononuclear cells were isolated and stained with anti-CD4-PerCP (Pharmingen). BrdU staining was performed according to instructions of the manufacturer (Pharmingen). BrdU incorporation was analyzed on gated CD4 + T cells.

[0101] IAElO cell labeling with carboxyfluorescein succinimidyl ester (CFSE) and assessment of proliferation. MBP-specific CD4 T-cell clones (IAElO) were incubated with 2 μM CFSE (Molecular Probes) in T-cell medium for 15 min at 37 0 C. CFSE staining was stopped by addition of excess medium, and cells were washed twice in medium. CFSE-labeled IAElO cells (I x 10 6 /ml) were cultured with wild-type spleen cells, the latter as antigen presenting cells (APCs)(2 x 10 6 /ml), in T-cell medium containing MBPi_ 9 (2.5 μg/ml) and with/without anti-LRG-47 antibody or non-immune IgG. Cells were harvested at the time points indicated, and stained with anti-CD4-PerCP and anti-CD44-PE IgG. The CFSE fluorescence profile was analyzed on gated CD4 + T-cells.

[0102] Assays for apoptosis. Apoptosis was measured in vitro using three different methods: (i) The Annexin V assay was used to detect cells in early stages of apoptosis. PI (propidium iodide) staining was detected in the dead cells. 1AE10, splenic and CNS cells were stained with anti-CD4-allophycocyanin and anti-CD44-PE IgG, and resuspended in annexin binding buffer. Cells were stained for 15 min with 5 μl of FITC-labeled annexin V (PharMingen) and 10 μl of PI at room temperature in the dark Analysis was performed by flow cytometry within 1 hr. (ii) In situ detection of apoptotic cells was performed on MBP-activated CD4 T-cells cultured on chamber slides using the in situ Cell Death Detection Kit (Roche, Germany). Briefly, air-dried cell samples were fixed with a freshly prepared fixation solution for 1 h at 15-25 0 C, and then incubated in permeabilization solution for 2 min on ice. TUNEL- positive cells were detected according to the manufacturer's instructions (Roche, CA). The percentage of TUNEL-positive cells is described as the percentage of the number of TUNEL- positive cells divided by the total number of cells in each field.

[0103] Quantitative real-time PCR. Total RNA was extracted from IAElO cells, lymph nodes and spinal cord using the TRIzol method (Invitrogen Life Technologies)

according to the manufacturer's protocol. cDNA synthesis was performed using Random Hexamers primers and the TaqMan Reverse Transcription kit (Applied Biosystems). Samples were subjected to real-time PCR analysis on an ABI Prism 7700 Sequence Detection System under standard conditions. Relative mRNA abundance was normalized against GAPDH (the endogenous control). The primers and probe for LRG were designed using Primer Express software (Applied Biosystems) and purchased from Applied Biosystems (5'- TGG CAA TGG CAT GTC ATC TT-3', reverse primer 5'-AGT ACT CAG TCC GCG TCT TCG T-3', and probe 6FAMACT TCG AGT CAT CGG CMGBNFQ).

[0104] Northern blotting. Samples for Northern blotting were collected from IAElO cells or MBP- immunized wild- type mice. Total RNA was isolated using Trizol (Invitrogen). For each sample, 5-10 μg of total RNA was electrophoresed through 1% agarose-6% formaldehyde gels. RNAs were transferred to Hybond N+ membranes and hybridized with the 32 P-labeled mouse LRG-47 probes.

[0105] ELISA for the cytokine detection. CD4 T-cells were isolated from MBP- immunized mouse spleen / lymph nodes and purified using magnetic beads conjugated with anti-CD4 mAb and a magnetic column (Milten Biotec). IAElO cells or CD4 T-cells were cultured in T-cell medium (Click's medium with 10% FCS, 0.01M Hepes, 5 x 10 "5 M 2-ME, 8 mM L-glutamine, and antibiotics) at 2 x 10 6 /ml (0.5ml/well) in 48-well plates with MBP (2.5 μg/ml), and irradiated splenic cells (4 x 10 6 AnI) as APCs. Supernatants were collected at the time points indicated. Analyses of murine IFNy were performed on both supernatants and serum samples, collected at day 14 post induction of EAE using commercially available ELISA kits (R&D Systems, MN). Sensitivity of the assays was < 2 pg/ml for IFNy.

[0106] Histology and immunostaining. Paraffin-embedded sections were stained with

H&E and the area occupied by nuclei per high-power field was evaluated using the Universal Imaging System. To localize LRG-47 antigen in spinal cord, double immunostaining with anti- LRG-47 and anti-CD4 or CDl Ib IgG was performed. Cryopreserved spinal cord sections were fixed with 4% paraformadehyde in PBS for 30 min. Slides were permeabilized and blocked in PBS containing 5% horse serum and 0.2% saponin for 60 min. They were then incubated with goat anti-LRG-47 IgG (A-19, Santa Cruze, CA), rat anti-CD4, or rat anti-CDl Ib mAbs (BD Pharmingen) followed by secondary antibodies (donkey anti-goat IgG conjugated-FITC and rabbit anti-rat IgG conjugated-TRITC, Sigma). Sections were examined by confocal microscopy.

[0107] To localize LRG-47 in MBP-stimulated IAElO cells as compared to non- stimulated IAElO cells, IAElO cells were exposed to MBP (5 μg/ml) for 4 hours, and then were fixed in 4% paraformadehyde alone or plus 0.1% NP-40 for 30 min. Sites of LRG-47 were detected using goat anti-LRG-47 IgG (1 :300) followed by donkey anti-goat IgG conjugated with FITC (1 :100). To determine the localization of LRG-47 in Golgi apparatus, IAElO cells were co-stained with GRASP65 IgG antibody, a maker for the Golgi apparatus (Abeam, Cambridge, MA), followed by sheep anti-rabbit IgG conjugated with TRITC (1:200, Sigma, CA). Sections were examined by confocal microscopy.

[0108] Statistic analysis. When two groups were compared, a two-tailed student t-test was used. When multiple groups were compared, analysis of variance (ANOVA) will be employed Fish's t-test was used for post-hoc comparisons. Values of p<0.05 was considered statistically significant.

[0109] SiRNA After stimulated with MBP (5μ/ml) for 48 hour, IAElO T cells were transfected with siRNA targeted to mouse LRG-47 (sense sequence: 5'- GGUUACCUGAGGUCAGUAGtt-3', antisense sequence: 5'-

CUACUGACCUCAGGUAACCtg-3') using siPORT Amine (Ambion) for additional 24. Levels of LRG-47 mRNA in cells were determined by real-time quantitative PCR and RT-PCR.

[0110] Multiple Sclerosis Patient The spinal cord tissues were obtained from the human Brain and spinal Fluid resource Center, VAMC, Los Angeles, CA 90073.

HSB# Age Gender Neuropathology diagnosis Coronal Structure

2696 86 F Multiple Sclerosis CXL-I Cervical Cord

2771 64 F Multiple sclerosis CXL-I Cervical Cord

2800 64 F Multiple Sclerosis CXL-I Cervical Cord

2932 57 F Multiple Sclerosis CXL-I Cervical Cord

2935 79 F Multiple Sclerosis CXL- 1 Cervical Cord

3175 54 F Normal CXL- 1 Cervical Cord

3216 79 M Normal Aging CXL-I Cervical Cord

3348 76 F Normal CXL-I Cervical Cord

3371 52 M Normal CXL- 1 Cervical Cord

2348 53 M Normal CXL- 1 Cervical Cord

BC128168 Homo sapiens gi:118764008;

Incorporated herein by reference

SEQ ID NO: 1

Partial cDNA sequence (sense strand) for LRG-47 atggaagcca tgaatgttga gaaagcctca gcagatggga acttgccaga ggtgatctct aacatcaagg agactctgaa gatagtgtcc aggacaccag ttaacatcac tatggcaggg gactctggca atgggatgtc caccttcatc agtgcccttc gaaacacagg acatgagggt aaggcctcac ctcctactga gctggtaaaa gctacccaaa gatgtgcctc ctatttctct tcccactttt caaatgtggt gttgtgggac ctgcctggca cagggtctgc caccacaacc ctggagaact acctgatgga aatgcagttc aaccggtatg acttcatcat ggttgcatct gcacaattca gcatgaatca tgtgatgctt gccaaaaccg ctgaggacat gggaaagaag ttctacattg tctggaccaa gctagacatg gacctcagca caggtgccct cccagaagtg cagctactgc agatcagaga aaatgtcctg gaaaatctcc agaaggagcg ggtatgtgaa tactaa

BC128168 Homo sapiens gi:118764008 Incorporated herein by reference SEQ ID NO 2 AMINO ACID SEQUENCE for LRG-47

MNVEKASADGNLPEVISNIKETLKIVSRTPVNITMAGDSGNGMS TFISALRNTGHEGKASPPTELVKATQRCASYFSSHFSNVVLWDLPGTGSATTTLENYL MEMQFNRYDFIMVASAQFSMNHVMLAKTAEDMGKKFYIVWTKLDMDLSTGALPEVQLL QIRENVLENLQKERVCEY

U19119 Mus musculus G-[gi:633753] Incorporated herein by reference SEQ ID NO 3. DNA SEQUENCE FOR for LRG-47 gcaaggtctg ctcgaagacc agaagctgaa agaaaatcca cgcgatcaga cctcctcttg gttcgtctcc tctcagaagg actccagacc tctgcatctc atctctcaac atccgggtct atattccagt tttggatctc tacataggga acttctgccg gaggacagca acgtttttgt cctaggaaga aaggggtgac gttccaggaa ggccactaac atcgaatcac acataataac tcctctggat cagggtttga ggagtattaa gtgagataag gcattcgaag gaaccaactc agattcacag acagaggacc tgtgtgctta aagtctaaga gtggaggaag aacctgagga gcggcttcct cagagaccct aataaaacca gagagcctca ccagggagct gaaaggtcca cagacagcgt cactcggatc ttatcatgaa accatcacac agttcctgcg aggctgctcc actactcccc aacatggcag agacccatta tgctcccctg agctcagcct tcccctttgt cacgtcatac caaacaggct ccagcaggtt acctgaggtc agtaggagca ccgaaagagc tttaagagaa ggaaaactac tggaactggt ctacggaatc aaggagactg tggcaacatt gtcccagatt ccagtgagca tctttgtgac tggggactct ggcaatggca tgtcatcttt catcaatgca cttcgagtca tcggccatga tgaagatgcc tcggctccca ctggggtggt gaggaccacg aagacgcgga ctgagtactc ttcatcccac tttcccaatg tggtgctgtg ggacttacct ggattggggg ccacagccca aaccgtagag gactatgtgg aagagatgaa atttagcaca tgtgacttat tcatcatcat tgcctctgag cagttcagct cgaatcatgt

gaagctgtcc aaaattatcc agagcatggg aaagaggttc tatattgtct ggaccaagct ggacagggac ctcagcacca gtgtcctatc agaggtccgg ctcctacaga atatccagga gaatatccga gagaatctgc agaaggagaa agtgaagtac ccccccgtgt tcctggtatc cagtctagac cctttactat atgacttccc gaagcttagg gacacacttc ataaagatct ctccaacatc aggtgctgtg aacccttaaa gaccctttat ggcacttatg agaagatcgt tggtgataaa gtagcagtct ggaagcagag aatagccaac gagtccttga agaattctct cggtgtcaga gatgatgaca acatgggcga gtgtctgaaa gtgtaccgac tgatatttgg tgtagatgac gaatcagttc agcaggtagc ccagagtatg gggacagtag tcatggagta caaggacaac atgaagtccc aaaactttta tactctccgc agagaggact ggaaactgag gctgatgaca tgtgcaattg tgaatgcatt cttccgtttg ttgagatttc tcccatgcgt atgctgctgt ttaagacgct tgagacataa acgcatgctt ttcttagttg cccaggacac caagaacatc ctagagaaaa tcctgaggga ctccatcttc cctccgcaga tctagtataa gggcagcctg gtacccttct tcttccacag aagccaggtt accttagatc tctttcctag atccctattt ctccaccaga aatcaagaga tacaaaaatg cttcctgtaa gggttttaga ttctctgaga ggagttaaaa tcactcatct cccctgtctc gattctaatg cattgttcca ctgagggaca gggacaagta gtgattaaaa ttcattgacc atgattctta gatttggaat atagaaattt tgtttttggg ctggagagat ggcttagcag ttaagaacac caactgcttt tccgaaggtc atgagttcaa atcccagcaa ccacgtgatg gctcacaacc atccgtagtg agatctgatg ccctcttctg agatgtctga agacagctac agtgtactta catataataa ataaataaat aaataaataa ataaataaat ctttgggaaa aaaattagaa attttgtttt cagctattaa atgtgatata tgtccaaatc aattttcctc tgaaacaata aagtcggttc ctcttca

U19119 Mus musculus G-[gi:633753]

Incorporated herein by reference

SEQ ID NO:4

AMINO ACID SEQUENCE FOR LRG-47

MKPSHSSCEAAPLLPNMAETHYAPLSSAFPFVTSYQTGSSRLPE

VSRSTERALREGKLLELVYGIKETVATLSQIPVSIFVTGDSGNGMSSFINALRVIGH D

EDASAPTGVVRTTKTRTEYSSSHFPNVVLWDLPGLGATAQTVEDYVEEMKFSTCDLF I

IIASEQFSSNHVKLSKIIQSMGKRFYIVWTKLDRDLSTSVLSEVRLLQNIQENIREN L

QKEKVKYPPVFLVSSLDPLL YDFPKLRDTLHKDLSNIRCCEPLKTL YGTYEKIVGDKV

AVWKQRIANESLKNSLGVRDDDNMGECLKVYRLIFGVDDESVQQVAQSMGTVVMEYK D

NMKSQNFYTLRREDWKLRLMTCAIVNAFFRLLRFLPCVCCCLRRLRHKRMLFLVAQD T

KNILEKILRDSIFPPQI

References

1. Refaeli, Y., L. Van Parijs, S.I. Alexander, and A.K. Abbas, Interferon gamma is required for activation-induced death of T lymphocytes. J Exp Med, 2002. 196(7): p. 999-1005.

2. Dalton, D.K., L. Haynes, CQ. Chu, S. L. Swain, and S. Wittmer, Interferon gamma eliminates responding CD4 T cells during mycobacterial infection by inducing apoptosis of activated CD4 T cells. J Exp Med, 2000. 192(1): p. 117-22.

3. Chu, C.Q., S. Wittmer, and D. K. Dalton, Failure to suppress the expansion of the activated CD4 T cell population in interferon gamma-deficient mice leads to exacerbation of experimental autoimmune encephalomyelitis. J Exp Med, 2000. 192(1): p. 123-8.

4. Taylor, G.A., CG. Feng, and A. Sher, p47 GTPases: regulators of immunity to intracellular pathogens. Nat Rev Immunol, 2004. 4(2): p. 100-9.

5. Taylor, G.A., M. Jeffers, D. A. Largaespada, N.A. Jenkins, N.G. Copeland, and G.F. Woude, Identification of a novel GTPase, the inducibly expressed GTPase, that accumulates in response to interferon gamma. J Biol Chem, 1996. 271(34): p. 20399-405.

6. MacMicking, J.D., IFN-inducible GTPases and immunity to intracellular pathogens. Trends Immunol, 2004. 25(11): p. 601-9.

7. Santiago, H.C, CG. Feng, A. Bafica, E. Roffe, R.M. Arantes, A. Cheever, G. Taylor, L.Q. Vierira, J. Aliberti, R.T. Gazzinelli, and A. Sher, Mice Deficient in LRG-47 Display Enhanced Susceptibility to Trypanosoma cruzi Infection Associated with Defective Hemopoiesis and Intracellular Control of Parasite Growth. J Immunol, 2005. 175(12): p. 8165-

72.

8. Gutierrez, M.G., S.S. Master, S.B. Singh, G.A. Taylor, M.I. Colombo, and V. Deretic, Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell, 2004. 119(6): p. 753-66.

9. MacMicking, J.D., G.A. Taylor, and J.D. McKinney, Immune control of tuberculosis by IFN-gamma-inducible LRG-47. Science, 2003. 302(5645): p. 654-9.

10. Yan, S.S., Z. Y. Wu, H.P. Zhang, G. Furtado, X. Chen, S.F. Yan, A.M. Schmidt, C. Brown, A. Stern, J. LaFaille, L. Chess, D.M. Stern, and H. Jiang, Suppression of experimental autoimmune encephalomyelitis by selective blockade of encephalitogenic T-cell infiltration of the central nervous system. Nat Med, 2003. 9(3): p. 287-93.

11. Martens, S., K. Sabel, R. Lange, R. Uthaiah, E. Wolf, and J.C. Howard, Mechanisms regulating the positioning of mouse p47 resistance GTPases LRG-47 and IIGP1 on cellular membranes: retargeting to plasma membrane induced by phagocytosis. J Immunol, 2004. 173(4): p. 2594-606.

12. Butcher, B.A., R.I. Greene, S. C. Henry, K.L. Annecharico, J.B. Weinberg, E. Y. Denkers, A. Sher, and G.A. Taylor, p47 GTPases regulate Toxoplasma gondii survival in activated macrophages. Infect Immun, 2005. 73(6): p. 3278-86.

13. MacMicking, J.D., Immune control of phagosomal bacteria by p47 GTPases. Curr Opin Microbiol, 2005. 8(1): p. 74-82.

14. Feng, C.G., CM. Collazo-Custodio, M. Eckhaus, S. Hieny, Y. Belkaid, K. Elkins, D. Jankovic, G.A. Taylor, and A. Sher, Mice deficient in LRG-47 display increased susceptibility to mycobacterial infection associated with the induction of lymphopenia. J Immunol, 2004. 172(2): p. 1163-8.

15. Sorace, J.M., RJ. Johnson, D. L. Howard, and B. E. Drysdale, Identification of an endotoxin and IFN-inducible cDNA: possible identification of a novel protein family. J Leukoc Biol, 1995. 58(4): p. 477-84.

16. Collazo, CM., G.S. Yap, G.D. Sempowski, K.C. Lusby, L. Tessarollo, G.F. Woude, A. Sher, and G.A. Taylor, Inactivation of LRG-47 and IRG-47 reveals a family of interferon gamma-inducible genes with essential, pathogen-specific roles in resistance to infection. J Exp Med, 2001. 194(2): p. 181-8.

17. O'Connor, R.A., S. Wittmer, and D.K. Dalton, Infection-induced apoptosis deletes bystander CD4+ T cells: a mechanism for suppression of autoimmunity during BCG infection. J Autoimmun, 2005. 24(2): p. 93-100.

18. Zhang, Y., J.N. Blattman, NJ. Kennedy, J. Duong, T. Nguyen, Y. Wang, RJ. Davis, P.D. Greenberg, R. A. Flavell, and C. Dong, Regulation of innate and adaptive immune responses by MAP kinase phosphatase 5. Nature, 2004. 430(7001): p. 793-7.