Methods and Compositions for the Treatment of Immune Disorders

The present invention provides a method of treating an immune-related disorder in a subject, comprising administering to the subject an effective amount of an inhibitor of plexin-A4 activity, which results in reducing the plexin-A4 activity in the subject, and thereby treating the immune-related disorder. Inhibitors of plexin-A4 activity include, for example, plexin-A4 antibodies and plexin-A4 fusion proteins. The present invention further provides a method of treating an immune-related disorder in a subject, comprising administering to the subject an effective amount of an inhibitor of semaphorin-3A (Sema3A) activity, which results in reducing the Sema3A activity in the subject, thereby treating the immune-related disorder. Inhibitors of Sema3A activity include, for example, Sema3A antibodies and Sema3A fusion proteins.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
STATEMENT OF PRIORITY

The present application is a continuation application of, and claims priority to, U.S. application Ser. No. 13/511,933, filed Jun. 12, 2012, now U.S. Pat. No. 8,871,205, which is a 35 U.S.C. §371 national phase application of International Application No. PCT/US2010/057807, filed Nov. 23, 2010, which claims the benefit, under 35 U.S.C. §119(e), of U.S. Provisional Application Ser. No. 61/264,338, filed Nov. 25, 2009, the entire contents of each of which are incorporated by reference herein.

STATEMENT OF GOVERNMENT SUPPORT

This invention was made with government support under Grant Nos. AI029564 and AI067798 awarded by The National Institutes of Health. The government has certain rights in the invention.

STATEMENT REGARDING ELECTRONIC FILING OF A SEQUENCE LISTING

A Sequence Listing in ASCII text format, submitted under 37 C.F.R. §1.821, entitled 5470-536TSCT_ST25.txt, 129,544 bytes in size, generated on Oct. 2, 2014 and filed via EFS-Web, is provided in lieu of a paper copy. This Sequence Listing is incorporated herein by reference into the specification for its disclosures.

FIELD OF THE INVENTION

The present invention relates to methods and compositions for treatment of immune related disorders and diseases.

BACKGROUND OF THE INVENTION

The functions of plexins and their ligands, semaphorins, have been extensively studied in the central nervous system (CNS). They represent two large families of molecules that can transduce signals essential for the regulation of neuronal repulsion and attraction, cell shape, motility and cell-cell interactions (Kruger et al., 2005; Tran et al., 2007). In addition to their roles in the CNS, the diverse functions of plexins and semaphorins have also been identified in cardiac development (Toyofuku et al., 2004), vascularization and angiogenesis (Gu et al., 2003a; Serini et al., 2003), and tumorigenesis (Neufeld and Kessler, 2008; Sierra et al., 2008). More recent data strongly indicate a role for these molecules in the immune system (Kikutani and Kumanogoh, 2003; Suzuki et al., 2008). For example, plexin-A1 is expressed by dendritic cells (DCs) and regulates DC interaction with T cells to affect adaptive immunity (Takegahara et al., 2006; Wong et al., 2003). Plexin-C1 is also found on DCs, although its role is less defined and it only mildly affects T cell activation (Walzer et al., 2005). A further paper showed the high expression of plexin-Dl in double-positive (DP) thymocytes and a role for this protein in the control of intrathymic migration of these cells from the cortical to medullary region (Choi et al., 2008). Therefore, plexins are involved in diverse functions in the immune system.

Plexin-A4 belongs to the plexin A-type group (Kruger et al., 2005) and serves as a guidance cue molecule in sensory and sympathetic neurons (Waimey et al., 2008; Yaron et al., 2005) and hippocampal mossy fibers (Suto et al., 2007). One study identified plexin-A4 as a negative regulator in T cell activation (Yamamoto et al., 2008). T cells lacking plexin-A4 (Plxna4−/−) exhibited hyperproliferative responses upon stimulations in vivo and in vitro. In addition, Plxna4−/− mice developed exacerbated experimental autoimmune encephalomyelitis (EAE) when immunized with myelin oligodendrocyte glycoprotein (MOG)-derived peptides. However, given the much higher expression of plexin-A4 in myeloid cells relative to lymphoid cells (Yamamoto et al., 2008), the role of plexin-A4 in cells of myeloid lineage such as macrophages and DCs needed to be elucidated.

The innate immune system constitutes the first line of defense by rapidly detecting invading pathogens and nonmicrobial danger signals through the pattern recognition receptors (PRRs). Several classes of PRRs have been identified; the best-characterized are the Toll-like receptors (TLRs) (Iwasaki and Medzhitov, 2004). TLR family members are localized either on the cell surface (TLRs 1, 2, 4, 5 and 6) or in endosomal compartments (TLRs 3, 7, 8, 9) to detect a multitude of pathogen-associated molecular patterns (PAMPs) (Akira et al., 2006; Iwasaki and Medzhitov, 2004). TLR activation leads to the direct interactions of the TLR toll-interleukin 1 receptor (TIR) domain with a cytoplasmic TIR-containing adaptive molecule such as Myd88, TRIF, TRAM or TIRAP. Activation of Myd88-dependent signaling pathway results in the activation of IRAK kinases, the ubiquitin ligase TRAF6, TAK1 kinase complex, NF-κB transcription factor, and mitogen-activated protein kinases (MAPKs) (Akira and Takeda, 2004; Akira et al., 2006; Iwasaki and Medzhitov, 2004). TRIF-dependent type I interferon (IFN) requires a cascade involving the adaptor TRAF3, the kinase TBK1, the inhibitor of κB kinase ε (IKKε), and the transcription factor interferon-regulatory factor 3 (IRF3) (Akira et al., 2006; Kawai and Akira, 2006).

The present invention addresses the shortcomings in the art by providing methods and compositions for the treatment of immune related and inflammatory disorders and diseases based on new therapeutic targets.

SUMMARY OF THE INVENTION

Provided herein is a method of treating an immune-related and/or inflammatory disorder in a subject (e.g., a subject in need thereof), comprising administering to the subject in need of treatment an effective amount of an inhibitor of plexin-A4 activity, whereby the plexin-A4 activity in said subject is reduced, thereby treating the immune-related disorder and/or inflammatory disorder. In some embodiments, the inhibitor of plexin-A4 activity is a plexin-A4 fusion protein, a plexin-A4 antibody or an active fragment thereof, or any combination thereof. In some embodiments, the plexin-A4 fusion protein comprises the extracellular domain of plexin-A4 and the Fc region of immunoglobulin G (IgG).

Also provided is a method of reducing cytokine production in a subject (e.g., a subject in need thereof), comprising administering to the subject an effective amount of an inhibitor of plexin-A4 activity. In some embodiments, the inhibitor of plexin-A4 activity is a plexin-A4 fusion protein, a plexin-A4 antibody or an active fragment thereof, or any combination thereof. In some embodiments, the plexin-A4 fusion protein comprises the extracellular domain of plexin-A4 and the Fc region of IgG.

Further provided is a method of identifying a substance having an inhibitory effect on plexin-A4 activity, comprising contacting the substance with macrophage/dendritic cells under conditions whereby plexin-A4 activity can occur and measuring the amount of plexin-A4 activity in the presence and absence of the substance, whereby a decrease in plexin-A4 activity in the presence of the substance as compared to the amount of plexin-A4 activity in the absence of the substance identifies a substance having an inhibitory effect on plexin-A4 activity.

BRIEF DESCRIPTION OF THE FIGURES

FIGS. 1A-C show that plexin-A4 is required for tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) production in macrophages. (FIG. 1A) Expression of Plxna4 mRNA in different immune cells was normalized to Actb mRNA level. T, B, NK, bone marrow (BM) plasmacytoid DCs (pDCs), and splenic myeloid DCs (mDCs) were isolated by fluorescence activated cell sorting (FACS) according to appropriate cell surface markers described in the Examples section. BM-derived macrophages (BMMs) and BM-derived dendritic cells (BMDCs) were respectively cultured from BM cells in the presence of L929 cell supernatant and granulocyte-macrophage colony-stimulating factor (GM-CSF) plus interleukin-4 (IL-4). Peritoneal and splenic macrophages were separated from other cells by adherence for 2 h at 37° C. FIG. 1B and FIG. 1C illustrate stimulation of peritoneal macrophages with Pam3Cys (5 μg/ml), poly(I:C) (10 μg/ml), ultrapure lipopolysaccharide (LPS) (1 μg/ml), R837 (10 μg/ml), or CpG-B (4 Wimp for 4 h. Messenger RNA (FIG. 1B) and protein (FIG. 1C) levels of TNF-α, IL-6 and interferon-beta (IFN-β) were measured by RT-PCR and ELISA, respectively. The results shown are representative of five independent experiments and are expressed as mean±s.d. *P<0.05 compared to WT peritoneal macrophages.

FIGS. 2A-B show that plexin-A4 is required for cytokine production in response to bacterial challenge. Peritoneal macrophages isolated from WT or Plxna4−/− mice were stimulated with Escherichia coli, Salmonella typhimurium, Listeria monocytogenes or Staphylococcus aureus at a multiplicity of infection (MOI) of 40. Messenger RNA (FIG. 2A) and protein (FIG. 2B) levels of TNF-α, IL-6 and IFN-β were measured by RT-PCR and ELISA, respectively. The results shown are representative of three independent experiments and are expressed as mean±s.d. * P<0.05 compared to WT peritoneal macrophages.

FIGS. 3A-E shows that plexin-A4 is required for LPS-induced activation of nuclease factor-kappa B (NF-κB), c-Jun-N-terminal kinase (JNK) and small GTPase Rac1. Immunoblot analysis of NF-κB and mitogen-activated protein kinase (MAPK) signaling molecules was performed with WT and Plxna4−/− peritoneal macrophages left untreated (0) or treated for 15, 30 or 60 min with 1 μg/ml ultrapure LPS (FIG. 3A). FIG. 3B and FIG. 3C show chromatin immunoprecipitation (ChIP) assays of the binding of p65 or c-Jun at the promoter regions of Tnfa and Il6 gene in WT and Plxna4−/− peritoneal macrophages left untreated or treated with 10 μg/ml poly(I:C), 1 μg/ml ultrapure LPS (B), 5 μg/ml Pam3Cys or 4 μg/ml CpG (C) for 1 h. Rho family GTPase (Rac1, Cdc42 and RhoA) pull-down analyses in WT and Plxna4−/− peritoneal macrophages is shown in FIG. 3D. Cells were left untreated (0) or treated for 15 or 30 min with 1 μg/ml ultrapure LPS. Rac1, Cdc42 and RhoA pull-down analysis were performed as described in the Examples section. Total Rac1, Cdc42 and RhoA in cell lysate were used as controls. WT peritoneal macrophages were pretreated with or without a Rac1 inhibitor, NSC23766 (200 μM) for 1 h, or left untreated, followed by stimulation with 5 μg/ml Pam3Cys, 10 μg/ml poly(I:C), 1 μg/ml ultrapure LPS, 10 μg/ml R837, or 4 μg/ml CpG for 4 h as shown in FIG. 3E. IFN-43 production in WT macrophages was not affected by Rac1 inhibition as shown in FIG. 3F. Protein levels of TNF-α and IL-6 were measured by ELISA. The results shown are representative of three independent experiments and are expressed as mean±s.d. * P<0.05 compared to WT peritoneal macrophages without NSC23766 pretreatment.

FIGS. 4A-C show that Plxna4−/− mice are protected from septic shock induced by poly(I:C) or LPS. FIG. 4A illustrates the survival of WT and Plxna4−/− mice (n=8 per group) given intraperitoneal injection of poly(I:C) (20 mg per kg body weight; left panel) or LPS (12.5 mg per kg body weight; right panel). FIG. 4B and FIG. 4C show the results of ELISA of inflammatory cytokines in peritoneal lavage (FIG. 4B) and serum (FIG. 4C) 4 h after the administration of poly(I:C), LPS, or PBS control (n=4 per group). The results shown are representative of two experiments and are expressed as mean±s.d. * P<0.05 compared to WT mice.

FIGS. 5A-D show that Plxna4−/− mice are protected from polymicrobial peritonitis induced by cecal ligation and puncture (CLP). FIG. 5A shows the survival of WT and Plxna4−/− mice (n=10 per group) after CLP procedure with two punctures (left panel) or nine punctures (right panel). Results are representative of three individual experiments. FIGS. 5B-D show the results of ELISA of inflammatory cytokines in peritoneal lavage (FIG. 5B), serum (FIG. 5C) and lung homogenate (FIG. 5D) before CLP or 4, 24, and 72 h after CLP (n=5 per group). The results are representative of at least two individual experiments. * P<0.05 compared with cytokine levels measured in peritoneal lavage, serum, or lung homogenate from WT mice.

FIGS. 6A-E shows that plexin-A4 is not required for macrophage phagocytosis and bacteria killing. WT and Plxna4−/− mice were subjected to CLP surgery (FIG. 6A). Bacterial loads in peritoneal lavage (left panel), blood (middle panel) and lung homogenate (right panel) were determined by counting the colony forming units. FIG. 6B provides FACS histograms showing phagocytosis of E. coli-GFP (MOI=100) by WT (black lines) or Plxna4−/− (tinted) peritoneal macrophages at 37° C. The two show a complete overlap. GFP fluorescence is plotted on the x axis, and cell number is plotted on they axis. FIG. 6C and FIG. 6D provide the mean fluorescence intensities (MFIs) of GFP showing phagocytosis of E. coli-GFP after various time periods (FIG. 6C) or at different MOI (FIG. 6D) by WT and Plxna4−/− peritoneal macrophages at 37° C. FIG. 6E shows the results of treatment of WT and Plxna4−/− peritoneal macrophages with E. coli (strain LF82) at a MOI of 10 (left panel) or 100 (right panel) for 1 h at 37° C., followed by the addition of 100 μg/ml gentamicin. Intracellular alive bacteria were determined by plate counting after various time periods (0, 1, 2 or 3 h).

FIGS. 7A-G show that Semaphorin 3A (Sema3A) enhances LPS-induced cytokine production in a plexin-A4-dependent manner. WT and Plxna4−/− peritoneal macrophages were stimulated with LPS (1 μg/ml) for 4 h in the absence or presence of either Sema3A-Fc or Sema6A-Fc fusion proteins (20 or 100 μg/ml) (FIG. 7A and FIG. 7B). Messenger RNA (FIG. 7A) or protein (FIG. 7B) levels of TNF-α and IL-6 were measured by RT-PCR and ELISA, respectively. The IgG Fc fragment (100 μg/ml) was used as a negative control. WT and Plxna4−/− mice (n=7˜9 per group) were pretreated with either Sema3A-Fc or IgG Fc control proteins at a dosage of 25 μg per kg body weight 1 h prior to CLP procedure (FIG. 7C). Survival rate was monitored for 5 days. τ P<0.05 compared with WT mice pretreated with Sema3A-Fc. WT mice (n=4 per group) were pretreated with either Sema3A-Fc or IgG Fc control proteins, followed by CLP surgery (FIG. 7D). Inflammatory cytokines in peritoneal lavage collected 4 h after CLP were determined by ELISA. * P<0.05 compared to peritoneal lavage from WT mice pretreated with Sema3A-Fc (FIG. 7E). FIG. 7F shows a schematic of the cellular interactions during cytokine storm. WT and Plxna4−/− mice (n=4 per group experiment) were pretreated with either Sema3A-Fc or IgG Fc control proteins at a dosage of 25 ug/kg body weight 1 hour before CLP procedure. Inflammatory cytokines in peritoneal lavage collected 4 h after CLP were determined by ELISA.*P<0.05 compared with peritoneal lavage from WT mice pretreated with Sema3A-Fc (FIG. 7G).

FIGS. 8A-B show that plexin-A4 does not modulate the presentation of OVA antigen by bone marrow-derived dendritic cells (BMDCs). BMDCs generated from WT or Plxna4−/− mice were pulsed with either 1 μg/ml OVA peptide323-339 (FIG. 8A) or 50 μg/ml OVA whole protein (FIG. 8B), and co-cultured with carboxyfluorescein succinimidyl ester (CFSE)-labeled splenic CD4+ T cells isolated from naïve TCR-transgenic OTII mice. Two or four days later, T cell proliferation was analyzed by the dilution of CFSE fluorescence.

FIG. 9 shows that plexin-A4 does not affect TLR-induced IFN-α production by plasmacytoid dendritic cells (pDCs). Plasmacytoid DCs were isolated from the bone marrows of WT and Plxna4−/− mice by FACS, and stimulated with 10 μg/ml R837 (TLR7) or 4 μg/ml CpG-B for 16 h. IFN-α protein levels in the supernatants were determined by ELISA.

FIGS. 10A-C show that plexin-A4 does not affect the number of peritoneal macrophages. Total peritoneal cells were harvested from WT and Plxna4−/− mice by peritoneal lavage. Cytospins were prepared and stained by hematoxylin and eosin (Panel A). The percent of peritoneal macrophages from WT and Plxna4−/− mice (Panel B) were multiplied by the total cell count to determine the absolute numbers (Panel C).

FIGS. 11A-F show that plexin-A4 does not affect immune cell composition in the spleen. Cells of myeloid or lymphoid lineage in the spleen were analyzed by FACS, including CD11b+CD11c+ myeloid DCs (FIG. 11A), B220+mPDCA1+CD11clow pDCs (FIG. 11B), CD11b+Ly6G+ neutrophils (FIG. 11C), CD11b+Ly6c+ monocytes and CD11b+F4/80+ macrophages (FIG. 11D), CD3+CD4+ T cells and CD3+CD8+ T cells (FIG. 11E), and CD19+ B cells and NK1.1+ NK cells (FIG. 11F).

FIGS. 12A-C show amino acid sequences for three plexin-A4 proteins. The residues indicated in bold show the sequence for the extracellular domain of the protein.

FIG. 13 shows the nucleotide sequence of the plexin-A4 coding sequence.

FIG. 14 shows the nucleotide sequence of the plexin-A4 cDNA. The nucleotides in bold are the primer sites and start (ATG) and stop codons (TGA).

FIGS. 15A-C show the amino acid sequence of the plexin-A4 fusion protein. In FIG. 15A, the portion of the sequence in bold indicates the extracellular domain of plexin-A4 and the non-bolded portion indicates the Fc region of IgG1. The two residues in bold and larger font between the plexin-A4 extracellular domain and the IgG Fc comprise the linker peptide of the fusion protein. In FIG. 15B is shown the amino acid sequence of the extracellular domain of plexin-A4 and in FIG. 15C is shown the amino acid sequence of the Fc region of IgG1.

FIGS. 16A-C show the nucleotide sequence of the plexin-A4 fusion protein. In FIG. 16A, the portion of the sequence in bold indicates the plexin-A4 extracellular domain and the non-bolded portion indicates the Fc region of IgG1. The six nucleotides in bold and larger font between the plexin-A4 extracellular domain and the IgG Fc show the nucleotide sequence encoding the linker peptide. FIG. 16B shows the nucleotide sequence of the extracellular domain of plexin-A4 and FIG. 16C shows the nucleotide sequence of the Fc region of IgG1.

FIGS. 17A-C. FIG. 17A shows plexin-A4 mRNA levels in a variety of different cell types. FIG. 17B shows the amino acid sequence for IgG with the Fc region of the sequence in bold. FIG. 17C shows a protein immunoblot for three different fusion proteins: plexin-A4/IgG Fc fusion protein (lanes 1, 2, 4, and 5), a Sema6D/IgG Fc fusion protein (lane 3) and a sema6D-IgG Fc fusion protein (lane 6). Lanes 1-3 were probed with antibody to plexin-A4 and lanes 5-6 were probed with antibody to hIgG.

FIGS. 18A-E. FIG. 18A shows the amino acid sequence for the Semaphorin 3A (Sema3A) polypeptide. FIG. 18B shows the nucleotide sequence of the Sema3A coding sequence. FIG. 18C shows nucleotide sequence of the Sema3A cDNA. The nucleotides in bold are the primer sites and start (ATG) and stop codons (TGA). FIG. 18D shows the amino acid sequence of the fusion protein between full-length human Sema3A and human IgG Fc. Full-length human Sema3A is in bold and the non-bolded portion of the sequence indicates the Fc region of IgG1. The two residues in bold and larger font between the Sema3A and the IgG Fc comprise a linker peptide. FIG. 18E shows the nucleotide sequence encoding the fusion protein between full-length human Sema3A and human IgG Fc. In FIG. 18E, the portion of the sequence in bold indicates the full length human Sema3A nucleotide sequence and the non-bolded portion indicates nucleotide sequence of the Fc region of IgG1. The six nucleotides in bold and larger font between Sema3A and IgG Fc show the nucleotide sequence encoding a linker peptide.

FIGS. 19A-B show that plexin-A4 is required for TLR-induced cytokine production in bone marrow-derived macrophages (BMMs) and bone marrow-derived dendritic cells (BMDCs). BMMs (FIG. 19A) and BMDCs (FIG. 19B) were generated from the bone marrow cells of WT and Plxna4−/− mice, and stimulated with various TLR agonists for 4 h. Protein levels of TNF-α and IL-6 in the supernatant were determined by ELISA.

FIGS. 20A-C show that plexin-A4 does not affect TNF-α, CD40, or PI3K/Akt signaling pathways. Peritoneal macrophages were isolated from WT and Plxna4−/− mice by peritoneal lavage. Cells were treated either with recombinant mouse TNF-α (20 ng/ml) or anti-CD40 (5 μg/ml) after TNF-α (1 ng/ml) priming for 3 h. TNF-α priming is required to upregulate CD40 expression (Lich et al., 2007). Immunoblot of MAPK signaling molecules was performed (FIG. 20A). Cells were treated with recombinant mouse IFN-γ, and STAT1 phosphorylation at tyrosine 701 was detected by immunoblot (FIG. 20B). Cells were treated with either Pam3Cys (5 μg/ml) or LPS (1 μg/ml). Akt phosphorylated at serine 473 was detected by immunoblot (FIG. 20C).

FIG. 21 shows that plexin-A4 does not affect toll-like receptor mRNA expression in peritoneal macrophages. Messenger RNA levels of Tlr2, 3, 4, 7 and 9 in WT and Plxna4−/− peritoneal macrophages were analyzed by RT-PCR using different primer sets, as described in Example 21, Table 1, below.

FIG. 22 shows the expression of Sema3a and Sema6a mRNA in different immune cells. Messenger RNA levels of Sema3a and Sema6a in different immune subpopulations were analyzed by RT-PCR and normalized to Actb mRNA level.

DETAILED DESCRIPTION

The present invention will now be described more fully hereinafter with reference to the accompanying drawings and specification, in which preferred embodiments of the invention are shown. This invention may, however, be embodied in different forms and should not be construed as limited to the embodiments set forth herein.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention.

All publications, patent applications, patents and other references cited herein are incorporated by reference in their entireties for the teachings relevant to the sentence and/or paragraph in which the reference is presented.

As used herein, “a,” “an” or “the” can mean one or more than one. For example, “a” cell can mean a single cell or a multiplicity of cells.

Also as used herein, “and/or” refers to and encompasses any and all possible combinations of one or more of the associated listed items, as well as the lack of combinations when interpreted in the alternative (“or”).

The term “about,” as used herein when referring to a measurable value such as an amount of dose (e.g., an amount of a non-viral vector) and the like, is meant to encompass variations of ±20%, ±10%, ±5%, ±1%, ±0.5%, or even ±0.1% of the specified amount.

As used herein, the transitional phrase “consisting essentially of” (and grammatical variants) means that the scope of a claim is to be interpreted to encompass the specified materials or steps recited in the claim, “and those that do not materially affect the basic and novel characteristic(s)” of the claimed invention. Thus, the term “consisting essentially of” when used in a claim of this invention is not intended to be interpreted to be equivalent to “comprising.”

Plexins are large (˜200 Kd) transmembrane glycoproteins with a conserved extracellular “Sema” domain. Plexin binds to its ligand, semaphorin, through Sema-Sema domain interaction, and transduces signals essential for cell migration in various tissue types (Kruger et al., 2005; Tran et al., 2007). The cytoplasmic domains of plexins have two highly conserved regions that share homology with GTPase-activating proteins (GAPs) that are known to activate the Ras superfamily of small GTPases. The present invention is based on the unexpected discovery that plexin-A4 in macrophages is required for optimal cytokine production upon TLR stimulation and bacterial challenge. Semaphorin 3A (Sema3A) serves as a ligand for plexin-A4 and enhances LPS-induced macrophage activation and cytokine production in a plexin-A4-dependent manner. Thus, through the inhibition of plexin-A4 activity, immune-related and/or inflammatory disorders including disorders/diseases that result in exacerbated production of proinflammatory cytokines and chemokines (those where TLR signaling is implicated) can be ameliorated.

Accordingly, in one embodiment, the present invention provides a method of treating an immune-related and/or inflammatory disorder in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor of plexin-A4 activity, whereby plexin-A4 activity in said subject is reduced, thereby treating the immune-related and/or inflammatory disorder.

Accordingly, in one embodiment, the present invention provides a method of treating an immune-related and/or inflammatory disorder in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor of plexin-A4 expression, whereby plexin-A4 expression in said subject is reduced, thereby treating the immune-related and/or inflammatory disorder.

Accordingly, in one embodiment, the present invention provides a method of treating an immune-related and/or inflammatory disorder in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor of Sema3A activity, whereby Sema3A activity in said subject is reduced, thereby treating the immune-related and/or inflammatory disorder.

Accordingly, in one embodiment, the present invention provides a method of treating an immune-related and/or inflammatory disorder in a subject in need thereof, comprising administering to the subject an effective amount of an inhibitor of Sema3A expression, whereby Sema3A expression in said subject is reduced, thereby treating the immune-related and/or inflammatory disorder.

As used herein, the terms “express,” “expressing,” or “expression” (or grammatical variants thereof) in reference to a gene or coding sequence can refer to transcription to produce an RNA and, optionally translation to produce a polypeptide. Thus, unless the context indicates otherwise, the terms “express,” “expressing,” “expression” and the like can refer to events at the transcriptional, post-transcriptional, translational and/or post-translational level.

In some embodiments of the present invention, the immune related and/or inflammatory disease or disorder can be, but is not limited to, sepsis; colitis; malignancies; systemic lupus erythematosis (SLE); arthritis, including, but not limited to, rheumatoid arthritis, osteoarthritis, juvenile chronic arthritis, and spondyloarthropathies; systemic sclerosis; idiopathic inflammatory myopathies; Sjogren's syndrome; systemic vasculitis; sarcoidosis; autoimmune hemolytic anemia; autoimmune thrombocytopenia; thyroiditis; diabetes mellitus; immune-mediated renal disease; demyelinating diseases of the central and peripheral nervous systems such as multiple sclerosis, idiopathic demyelinating polyneuropathy or Guillain-Barre syndrome, and chronic inflammatory demyelinating polyneuropathy; Alzheimer's disease; myocarditis; kidney disease; obesity; cardiovascular disease; hepatobiliary diseases such as infectious, autoimmune chronic active hepatitis, primary biliary cirrhosis, granulomatous hepatitis, and sclerosing cholangitis; inflammatory bowel disease; gluten-sensitive enteropathy; Whipple's disease; autoimmune or immune-mediated skin diseases including bullous skin diseases, erythema multiforme and contact dermatitis or hypersensitivity; psoriasis; allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and urticaria; immunologic diseases of the lung such as asthma, allergies, COPD (chronic obstructive pulmonary disease), eosinophilic pneumonias, idiopathic pulmonary fibrosis and hypersensitivity pneumonitis; transplantation associated diseases including graft rejection and graft-versus-host-disease; inflammation of the eye including but not limited to retinitis and uveitis; and any/or combination thereof.

In some embodiments, the immune related disease or disorder can be sepsis, arthritis, hepatitis, systemic lupus erythematosus (SLE), multiple sclerosis, Guillain-Barre syndrome, Alzheimer's disease, colitis, psoriasis, contact hypersensitivity, retinitis, uveitis, malignancies, systemic lupus erythematosis, asthma, myocarditis, hepatitis, kidney diseases, diabetes, obesity, cardiovascular diseases, inflammatory bowel disease and any/or combination thereof. In other embodiments, the immune-related disease or disorder is sepsis.

A “subject” of this invention includes any subject that is susceptible to the various diseases and/or disorders described herein. Nonlimiting examples of subjects of this invention include mammals, such as humans, nonhuman primates, domesticated mammals (e.g., dogs, cats, rabbits, guinea pigs, rats), livestock and agricultural mammals (e.g., horses, bovine, pigs, goats). In other embodiments, a subject may additionally be an animal such as a bird or reptile. Thus, in some embodiments, a subject can be any domestic, commercially or clinically valuable animal. Subjects may be male or female and may be any age including neonate, infant, juvenile, adolescent, adult, and geriatric subjects. In particular embodiments, the subject is a human. A human subject of this invention can be of any age, gender, race or ethnic group (e.g., Caucasian (white), Asian, African, black, African American, African European, Hispanic, Mideastern, etc.).

A “subject in need thereof” is a subject known to have, or suspected of having, diagnosed with, or at risk of having an immune related and/or inflammatory disease or disorder. A subject of this invention can also include a subject not previously known or suspected to have an immune related and/or inflammatory disease or disorder or in need of treatment for an immune related and/or inflammatory disease or disorder. For example, a subject of this invention can be administered the compositions of this invention even if it is not known or suspected that the subject has an immune related and/or inflammatory disease or disorder (e.g., prophylactically). A subject of this invention is also a subject known or believed to be at risk of developing an immune related and/or inflammatory disease or disorder. Accordingly, a subject in need thereof also includes a subject known to need, suspected of needing, or at risk of needing reduced cytokine production, reduced plexin-A4 activity or expression, or reduced Sema3A activity or expression. A subject of this invention can also include a subject not previously known or suspected to need reduced cytokine production, reduced plexin-A4 activity or expression, or reduced Sema3A activity or expression or to need treatment to reduce cytokine production, reduce plexin-A4 activity or expression, or reduce Sema3A activity or expression. For example, a subject of this invention can be administered the compositions of this invention even if it is not known or suspected that the subject needs reduced cytokine production or needs reduced plexin-A4 activity or expression or needs reduced Sema3A activity or expression (e.g., prophylactically). A subject of this invention is also a subject known or believed to be at risk of developing a need for reduced cytokine production, or for reduced plexin-A4 activity or expression, or for reduced Sema3A activity or expression.

A “subject in need thereof” is additionally a subject known to need, or suspected of needing, diagnosed with needing, or at risk of needing increased plexin-A4 activity or expression, or increased Sema3A activity or expression. A subject of this invention can also include a subject not previously known or suspected to need increased plexin-A4 activity or expression, or increased Sema3A activity or expression, or to need treatment to increase cytokine production, or to increase plexin-A4 activity or expression, or to increase Sema3A activity or expression. For example, a subject of this invention can be administered the compositions of this invention even if it is not known or suspected that the subject needs increased plexin-A4 activity or expression or increased Sema3A activity or expression (e.g., prophylactically). A subject of this invention is also a subject known or believed to be at risk of developing a need for increased plexin-A4 activity or expression or increased Sema3A activity or expression.

Also provided herein is a method of reducing cytokine production in a subject (e.g., a subject in need thereof), comprising administering to a subject an effective amount of an inhibitor of plexin-A4 activity. In other embodiments, a method of reducing plexin-A4 activity or expression in a subject is provided, comprising administering to a subject in need thereof an effective amount of an inhibitor of plexin-A4 activity. A subject in need thereof in these embodiments is a subject in need of reduced cytokine production or reduced plexin-A4 activity or expression, including, but not limited to, a subject having an immune-related and/or inflammatory disease or disorder including, but not limited to, those described herein.

Additionally provided herein is a method of reducing cytokine production in a subject (e.g., a subject in need thereof), comprising administering to a subject an effective amount of an inhibitor of Sema3A activity. In other embodiments, a method of reducing Sema3A activity or expression in a subject is provided, comprising administering to a subject in need thereof an effective amount of an inhibitor of Sema3A activity. A subject in need thereof in these embodiments is a subject in need of reduced cytokine production or reduced Sema3A activity or expression, including, but not limited to, a subject having an immune-related and/or inflammatory disease or disorder including, but not limited to, those described herein.

In further embodiments, an inhibitor of plexin-A4 activity includes, but is not limited to, a plexin-A4 fusion protein, a plexin-A4 antibody or an active fragment thereof or any combination thereof. In some embodiments, the plexin-A4 antibody is a monoclonal antibody and/or is derived from a monoclonal antibody. In other embodiments, the plexin-A4 antibody or active fragment thereof is a polyclonal antibody and/or is derived from a polyclonal antibody. In yet other embodiments, the present invention provides a composition comprising an inhibitor of plexin-A4 activity in an admixture with a pharmaceutically acceptable carrier. Thus, in some embodiments, a plexin-A4 antibody or active fragment thereof or a plexin-A4 fusion protein is provided in a composition further comprising a pharmaceutically acceptable carrier.

In still further embodiments, an inhibitor of Sema3A activity includes, but is not limited to, a Sema3A fusion protein, a Sema3A antibody or an active fragment thereof or any combination thereof. In some embodiments, the Sema3A antibody is a monoclonal antibody and/or is derived from a monoclonal antibody. In other embodiments, the Sema3A antibody or active fragment thereof is a polyclonal antibody and/or is derived from a polyclonal antibody. In yet other embodiments, the present invention provides a composition comprising an inhibitor of Sema3A activity in an admixture with a pharmaceutically acceptable carrier. Thus, in some embodiments, a Sema3A antibody or active fragment thereof or a Sema3A fusion protein is provided in a composition further comprising a pharmaceutically acceptable carrier.

In yet further embodiments, an inhibitor of plexin-A4 expression or Sema3A expression includes, but is not limited to, siRNA, shRNA, miRNA, antisense RNA and ribozymes.

The present invention further contemplates that enhancers or activators of plexin-A4 or Sema3A activity may be useful for treating subjects having immune deficiencies. Immune deficiencies can be inherited, or acquired through infection and/or other illness, and/or produced as an inadvertent side effect of particular drug treatments. Non-limiting examples of immune deficiencies include severe combined immunodeficiency disease (SCID), chronic fatigue and immune dysfunction syndrome (CFIDS), acquired immune deficiency syndrome (AIDS), and/or immune deficiencies resulting from chemotherapeutics administered to subjects (e.g., cancer patients) undergoing chemotherapy.

Accordingly, the present invention provides a method of treating an immune deficiency in a subject, comprising administering to a subject in need thereof an effective amount of an enhancer and/or activator of plexin-A4 or Sema3A activity or expression, whereby the plexin-A4 or Sema3A activity or expression in said subject is increased, thereby treating the immune deficiency. In some embodiments, the present invention provides a method for increasing cytokine production in a subject, comprising administering to a subject in need thereof an effective amount of an enhancer and/or activator of plexin-A4 activity or expression, or an effective amount of an enhancer and/or activator of Sema3A activity or expression. In other embodiments, a method is provided for increasing plexin-A4 activity or expression in a subject, comprising administering to a subject in need thereof an effective amount of an enhancer and/or activator of plexin-A4 activity or expression. In still other embodiments, a method is provided for increasing Sema3A activity or expression in a subject, comprising administering to a subject in need thereof an effective amount of an enhancer and/or activator of Sema3A activity or expression. The increase in cytokine production and/or in plexin-A4 activity and/or expression and/or in Sema3A activity and/or expression is any increase as compared to the level of cytokine production, and/or plexin-A4 activity and/or expression and/or in Sema3A activity and/or expression in the absence of the enhancer and/or activator.

As used herein an activator and/or enhancer of plexin-A4 activity and/or expression is a substance, or combination of substances that increase plexin-A4 activity and/or expression over the level of the plexin-A4 activity and/or expression in the absence of the substance. As used herein an activator and/or enhancer of Sema3A activity and/or expression is a substance, or combination of substances that increase Sema3A activity and/or expression over the level of the Sema3A activity and/or expression in the absence of the substance.

In some particular embodiments, an enhancer and/or activator of plexin-A4 activity and/or expression includes, but is not limited to, a plexin-A4 antibody or an active fragment thereof, or any combination thereof. In other particular embodiments, an enhancer and/or activator of Sema3A activity and/or expression includes, but is not limited to, a Sema3A antibody or an active fragment thereof, or any combination thereof.

As used herein, the term “antibody” includes intact immunoglobin molecules as well as active fragments thereof, such as Fab, F(ab′)2, and Fc, which are capable of binding the epitopic determinant of an antigen (i.e., antigenic determinant). Antibodies that bind the polypeptides of this invention are prepared using intact polypeptides and/or fragments containing small peptides of interest as the immunizing antigen. The polypeptide or fragment used to immunize an animal can be derived from enzymatic cleavage, recombinant expression, isolation from biological materials, synthesis, etc., and can be conjugated to a carrier protein, if desired. Commonly used carriers that are chemically coupled to peptides and proteins for the production of antibody include, but are not limited to, bovine serum albumin, thyroglobulin and keyhole limpet hemocyanin. The coupled peptide or protein is then used to immunize a host animal (e.g., a mouse, rat, goat, sheep, human or rabbit). The polypeptide or peptide antigens can also be administered with an adjuvant, as described herein and as otherwise known in the art.

The terms “antibody” and “antibodies” as used herein refer to all types of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE. The antibody can be monoclonal or polyclonal and can be of any species of origin, including, for example, mouse, rat, rabbit, horse, goat, sheep or human, and/or can be a chimeric or humanized antibody. See, e.g., Walker et al., Molec. Immunol. 26:403-11 (1989). The antibodies can be recombinant monoclonal antibodies produced according to the methods disclosed in U.S. Pat. No. 4,474,893 or U.S. Pat. No. 4,816,567. The antibodies can also be chemically constructed according to the method disclosed in U.S. Pat. No. 4,676,980. The antibody can further be a single chain antibody or bispecific antibody.

Techniques for the production of chimeric antibodies or humanized antibodies by splicing mouse antibody genes to human antibody genes to obtain a molecule with appropriate antigen specificity and biological activity can be used (Morrison et al. 1984. Proc. Natl. Acad. Sci. 81:6851-6855; Neuberger et al. 1984. Nature 312:604-608; Takeda et al. 1985. Nature 314:452-454). Alternatively, techniques described for the production of single chain antibodies can be adapted, using methods known in the art, to produce single chain antibodies specific for the polypeptides and/or fragments and/or epitopes of this invention. Antibodies with related specificity, but of distinct idiotypic composition, can be generated by chain shuffling from random combinatorial immunoglobin libraries (Burton 1991. Proc. Natl. Acad. Sci. 88:11120-3).

Active antibody fragments included within the scope of the present invention include, for example, Fab, F(ab′)2, and Fc fragments, and the corresponding fragments obtained from antibodies other than IgG. Such fragments can be produced by known techniques. For example, F(ab′)2 fragments can be produced by pepsin digestion of the antibody molecule, and Fab fragments can be generated by reducing the disulfide bridges of the F(ab′)2 fragments. Alternatively, Fab expression libraries can be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity (Huse et al., (1989) Science 254:1275-1281).

Monoclonal antibodies can be produced in a hybridoma cell line according to the technique of Kohler and Milstein (Nature 265:495-97 (1975)). For example, a solution containing the appropriate antigen can be injected into a mouse and, after a sufficient time, the mouse sacrificed and spleen cells obtained. The spleen cells are then immortalized by fusing them with myeloma cells or with lymphoma cells, typically in the presence of polyethylene glycol, to produce hybridoma cells. The hybridoma cells are then grown in a suitable medium and the supernatant screened for monoclonal antibodies having the desired specificity. Monoclonal Fab fragments can be produced in bacterial cell such as E. coli by recombinant techniques known to those skilled in the art. See, e.g., W. Huse, (1989) Science 246:1275-81.

Antibodies can also be obtained by phage display techniques known in the art or by immunizing a heterologous host with a cell containing an epitope or immunogen of interest.

In some embodiments of the present invention, the plexin-A4 fusion protein comprises, consists essentially of, or consists of an extracellular domain of plexin-A4 and an Fc region of IgG. The extracellular domain of plexin-A4 and the Fc region of IgG can be derived from a plexin-A4 or IgG from any species (e.g., mouse, human). In particular embodiments, the extracellular domain of plexin-A4 and the IgG Fc region are derived from human.

A plexin-A4 polypeptide of this invention includes, but is not limited to, the amino acid sequence of SEQ ID NO:1, SEQ ID NO:2 and SEQ ID NO:3. It is further contemplated that plexin-A4 polypeptides useful for the present invention include those known in the art, including the non-limiting examples of the plexin-A4 polypeptides having NCBI Accession Nos. NM020911, NM181775, and NM001105543.

As discussed above, in some embodiments of the invention, the plexin-A4 fusion protein comprises the extracellular domain of a plexin-A4 polypeptide. Thus, in some embodiments, the extracellular domain of a plexin-A4 polypeptide includes, but is not limited to, the amino acid sequence of SEQ ID NO:7.

In some embodiments, the plexin-A4 fusion protein further comprises a region of an immunoglobulin, for example, IgG, IgM, IgA, IgD, and IgE. In further embodiments, the plexin-A4 fusion protein comprises a region of IgG. The amino acid sequence of a representative IgG is SEQ ID NO:12 (FIG. 17B). In other embodiments of the invention, the plexin-A4 fusion protein comprises the Fc region of an immunoglobulin. In yet other embodiments of the invention, the plexin-A4 fusion protein comprises the Fc region of IgG. Thus, in some embodiments of the present invention, the plexin-A4 fusion protein comprises IgG Fc having the amino acid sequence of SEQ ID NO:8 (FIG. 15C).

Accordingly, in some embodiments of the present invention a plexin-A4 fusion protein is provided comprising the amino acid sequences of SEQ ID NO:7 (FIG. 15B) and SEQ ID NO:8 (FIG. 15C).

In further embodiments of the present invention, the fusion protein comprises an extracellular domain of plexin-A4 and the Fc region of IgG, wherein the plexin-A4 extracellular domain and the IgG Fc region are linked by a peptide or peptide fragment. In some embodiments, the peptide or peptide fragment linker is a length from about 1 amino acid residue to about 20 amino acid residues. Thus, the peptide linker can be a length of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in length, Thus, in further embodiments, the length of the peptide linker can be from about 1 to about 5 amino acid residues, from about 1 to about 10 amino acid residues, from about 1 to about 15 amino acid residues, from about 2 to about 5 amino acid residues, from about 2 to about 10 amino acid residues, from about 2 to about 15 amino acid residues, from about 2 to about 20 amino acid residues, from about 5 to about 10 amino acid residues, from about 5 to about 15 amino acid residues, from about 5 to about 20 amino acid residues, and the like. In some embodiments, the peptide linker comprises, consists essentially of, and/or consists of 2 amino acids in length. The peptide linker can be any amino acid residue or combination of amino acid residues in any order and/or in any multiplicity of the same amino acid residues. In some embodiments, the peptide linker of the present invention comprises glycine and/or cysteine. In particular embodiments, the peptide linker comprises a glycine residue and a cysteine residue (i.e., G-C; gly-cys).

Accordingly, the present invention further provides embodiments wherein the plexin-A4 fusion protein comprises the amino acid sequence of SEQ ID NO:6 (FIG. 15A).

In some embodiments of the present invention, the Sema3A fusion protein comprises, consists essentially of, or consists of full length Sema3A and an Fc region of IgG. The Sema3A and the Fc region of IgG can be derived from a Sema3A or IgG from any species (e.g., mouse, human). In particular embodiments, the Sema3A and the IgG Fc region are derived from human.

A Sema3A polypeptide of this invention includes, but is not limited to, the amino acid sequence of SEQ ID NO:13 (FIG. 18A). It is further contemplated that Sema3A polypeptides useful for the present invention include those known in the art, including the non-limiting example of a Sema3A polypeptide having NCBI Accession No. NM006080.

As discussed above, in some embodiments of the invention, the Sema3A fusion protein comprises the full length polypeptide. In other embodiments, the Sema3A fusion protein comprises fragments of the full length polypeptide. Thus, in some embodiments, the Sema3A fusion protein includes, but is not limited to, fragments of the amino acid sequence of SEQ ID NO:13 (FIG. 18A).

In some embodiments, the fusion protein further comprises a region of an immunoglobulin, for example, IgG, IgM, IgA, IgD, and IgE. In further embodiments, the fusion protein comprises a region of IgG. The amino acid sequence of a representative IgG is SEQ ID NO:12 (FIG. 17B). In other embodiments of the invention, the fusion protein comprises the Fc region of an immunoglobulin. In yet other embodiments of the invention, the fusion protein comprises the Fc region of IgG. Thus, in some embodiments of the present invention, the fusion protein comprises IgG Fc region having the amino acid sequence of SEQ ID NO:8 (FIG. 15).

Accordingly, in some embodiments of the present invention a Sema3A fusion protein is provided comprising the amino acid sequences of SEQ ID NO:13 and SEQ ID NO:8.

In further embodiments of the present invention, the Sema3A fusion protein comprises a full length Sema3A and the Fc region of IgG, wherein the Sema3A and the IgG Fc region are linked by a peptide or peptide fragment. In some embodiments, the peptide or peptide fragment linker is a length from about 1 amino acid residue to about 20 amino acid residues. Thus, the peptide linker can be a length of about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in length, Thus, in further embodiments, the length of the peptide linker can be from about 1 to about 5 amino acid residues, from about 1 to about 10 amino acid residues, from about 1 to about 15 amino acid residues, from about 2 to about 5 amino acid residues, from about 2 to about 10 amino acid residues, from about 2 to about 15 amino acid residues, from about 2 to about 20 amino acid residues, from about 5 to about 10 amino acid residues, from about 5 to about 15 amino acid residues, from about 5 to about 20 amino acid residues, and the like. In some embodiments, the peptide linker comprises, consists essentially of and/or consists of 2 amino acids in length. The peptide linker can be any amino acid residue or combination of amino acid residues in any order and/or in any multiplicity of the same amino acid residues. In some embodiments, the peptide linker of the present invention comprises glycine and/or cysteine. In particular embodiments, the peptide linker comprises a glycine residue and a cysteine residue (i.e., G-C; gly-cys).

Accordingly, the present invention further provides embodiments wherein the plexin-A4 fusion protein comprises the amino acid sequence of SEQ ID NO:16 (FIG. 18D).

As used herein, the term “polypeptide” encompasses both peptides and proteins, unless indicated otherwise.

The terms “polypeptide,” “protein,” and “peptide” refer to a chain of covalently linked amino acids. In general, the term “peptide” can refer to shorter chains of amino acids (e.g., 2-50 amino acids); however, all three terms overlap with respect to the length of the amino acid chain. Polypeptides, proteins, and peptides may comprise naturally occurring amino acids, non-naturally occurring amino acids, or a combination of both. The polypeptides, proteins, and peptides may be isolated from sources (e.g., cells or tissues) in which they naturally occur, produced recombinantly in cells in vivo or in vitro or in a test tube in vitro, and/or synthesized chemically. Such techniques are known to those skilled in the art. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual 2nd Ed. (Cold Spring Harbor, N.Y., 1989); Ausubel et al. Current Protocols in Molecular Biology (Green Publishing Associates, Inc. and John Wiley & Sons, Inc., New York).

The term “fragment,” as applied to a polypeptide, will be understood to mean an amino acid sequence of reduced length relative to a reference polypeptide or amino acid sequence and comprising, consisting essentially of, and/or consisting of an amino acid sequence of contiguous amino acids identical, or substantially identical, to the reference polypeptide or amino acid sequence. Such a polypeptide fragment according to the invention may be, where appropriate, included in a larger polypeptide of which it is a constituent. In some embodiments, such fragments can comprise, consist essentially of, and/or consist of peptides having a length of at least about 4, 6, 8, 10, 12, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 150, 200, or more consecutive amino acids of a polypeptide or amino acid sequence according to the invention.

A fragment of a polypeptide or protein of this invention can be produced by methods well known and routine in the art. Fragments of this invention can be produced, for example, by enzymatic or other cleavage of naturally occurring peptides or polypeptides or by synthetic protocols that are well known. Such fragments can be tested for one or more of the biological activities of this invention (e.g., plexin-A4 binding to Sema3A) according to the methods described herein, which are routine methods for testing activities of polypeptides, and/or according to any art-known and routine methods for identifying such activities. Such production and testing to identify biologically active fragments of the polypeptides described herein would be well within the scope of one of ordinary skill in the art and would be routine.

Ribozymes are RNA-protein complexes that cleave nucleic acids in a site-specific fashion. Ribozymes have specific catalytic domains that possess endonuclease activity (Kim et al. (1987); Gerlach et al. (1987); Forster et al. (1987)). For example, a large number of ribozymes accelerate phosphoester transfer reactions with a high degree of specificity, often cleaving only one of several phosphoesters in an oligonucleotide substrate (Michel et al. (1990); Reinhold-Hurek et al. (1992)). This specificity has been attributed to the requirement that the substrate bind via specific base-pairing interactions to the internal guide sequence (“IGS”) of the ribozyme prior to chemical reaction.

Ribozyme catalysis has primarily been observed as part of sequence-specific cleavage/ligation reactions involving nucleic acids (Joyce (1989)). For example, U.S. Pat. No. 5,354,855 reports that certain ribozymes can act as endonucleases with a sequence specificity greater than that of known ribonucleases and approaching that of the DNA restriction enzymes. Thus, sequence-specific ribozyme-mediated inhibition of nucleic acid expression may be particularly suited to therapeutic applications (Scanlon et al. (1991); Sarver et al. (1990); Sioud, M. et al. (1992)).

MicroRNAs (miRNA) are RNA molecules, generally 21-23 nucleotides long, that can down-regulate gene expression by hybridizing to miRNA. Over-expression or diminution of a particular miRNA can be used to treat a dysfunction and has been shown to be effective in a number of disease states and animal models of disease (Couzin 2008). Mature miRNAs are produced from a primary transcript (pri-miRNA) that is processed into a short stem-loop structure (a pre-miRNA) that then forms the final miRNA product.

The term “antisense oligonucleotide” (including “antisense RNA”) as used herein, refers to a nucleic acid that is complementary to and specifically hybridizes to a specified DNA or RNA sequence such as a plexin-A4 or Sema3A DNA or RNA sequence. Antisense oligonucleotides and nucleic acids that encode the same can be made in accordance with conventional techniques. See, e.g., U.S. Pat. No. 5,023,243 to Tullis; U.S. Pat. No. 5,149,797 to Pederson et al.

Those skilled in the art will appreciate that it is not necessary that the antisense oligonucleotide be fully complementary to the target sequence as long as the degree of sequence similarity is sufficient for the antisense nucleotide sequence to specifically hybridize to its target (as defined above) and reduces production of the protein product (e.g., by at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more).

To determine the specificity of hybridization, hybridization of such oligonucleotides to target sequences can be carried out under conditions of reduced stringency, medium stringency or even high stringency conditions, as described herein. Exemplary conditions for reduced, medium and high stringency hybridization are as follows: (e.g., conditions represented by a wash stringency of 35-40% Formamide with 5×Denhardt's solution, 0.5% SDS and 1×SSPE at 37° C.; conditions represented by a wash stringency of 40-45% Formamide with 5×Denhardt's solution, 0.5% SDS, and 1×SSPE at 42° C.; and conditions represented by a wash stringency of 50% Formamide with 5×Denhardt's solution, 0.5% SDS and 1×SSPE at 42° C., respectively). See, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual (2d Ed. 1989) (Cold Spring Harbor Laboratory).

Alternatively stated, in particular embodiments, the antisense oligonucleotide has at least about 60%, 70%, 80%, 90%, 95%, 97%, 98% or higher sequence similarity with the complement of the target sequence and reduce production of the protein product (as defined above). In some embodiments, the antisense sequence contains 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 or more mismatches as compared with the target sequence.

Methods of determining percent identity of nucleic acid sequences are described in more detail elsewhere herein.

The length of the antisense oligonucleotide is not critical as long as it specifically hybridizes to the intended target and reduces production of the protein product and can be determined in accordance with routine procedures. In general, the antisense oligonucleotide is at least about eight, ten or twelve or fifteen nucleotides in length and/or less than about 20, 30, 40, 50, 60, 70, 80, 100 or 150 nucleotides in length.

An antisense oligonucleotide can be constructed using chemical synthesis and enzymatic ligation reactions by procedures known in the art. For example, an antisense oligonucleotide can be chemically synthesized using naturally occurring nucleotides or various modified nucleotides designed to increase the biological stability of the molecules and/or to increase the physical stability of the duplex formed between the antisense and sense nucleotide sequences, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.

Examples of modified nucleotides which can be used to generate the antisense oligonucleotide include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomet-hyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopenten-yladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine.

The antisense oligonucleotides can further include nucleotide sequences wherein at least one, or all, of the internucleotide bridging phosphate residues are modified phosphates, such as methyl phosphonates, methyl phosphonothioates, phosphoromorpholidates, phosphoropiperazidates and phosphoramidates. For example, every other one of the internucleotide bridging phosphate residues can be modified as described.

As another non-limiting example, one or all of the nucleotides in the oligonucleotide can contain a 2′ loweralkyl moiety (e.g., C1-C4, linear or branched, saturated or unsaturated alkyl, such as methyl, ethyl, ethenyl, propyl, 1-propenyl, 2-propenyl, and isopropyl). For example, every other one of the nucleotides can be modified as described. See also, Furdon et al. (1989) Nucleic Acids Res. 17, 9193-9204; Agrawal et al. (1990) Proc. Natl. Acad. Sci. USA 87, 1401-1405; Baker et al. (1990) Nucleic Acids Res. 18, 3537-3543; Sproat et al. (1989) Nucleic Acids Res. 17, 3373-3386; Walder and Walder (1988) Proc. Natl. Acad. Sci. USA 85, 5011-5015.

The antisense oligonucleotide can be chemically modified (e.g., at the 3′ and/or 5′ end) to be covalently conjugated to another molecule. To illustrate, the antisense oligonucleotide can be conjugated to a molecule that facilitates delivery to a cell of interest, enhances absorption by the nasal mucosa (e.g., by conjugation to a lipophilic moiety such as a fatty acid), provides a detectable marker, increases the bioavailability of the oligonucleotide, increases the stability of the oligonucleotide, improves the formulation or pharmacokinetic characteristics, and the like. Examples of conjugated molecules include but are not limited to cholesterol, lipids, polyamines, polyamides, polyesters, intercalators, reporter molecules, biotin, dyes, polyethylene glycol, human serum albumin, an enzyme, an antibody or antibody fragment, or a ligand for a cellular receptor.

Other modifications to nucleic acids to improve the stability, nuclease-resistance, bioavailability, formulation characteristics and/or pharmacokinetic properties are known in the art.

RNA interference (RNAi) is another useful approach for reducing production of a protein product (e.g., shRNA or siRNA). RNAi is a mechanism of post-transcriptional gene silencing in which double-stranded RNA (dsRNA) corresponding to a target sequence of interest (e.g., plexin-A4 and/or Sema3A) is introduced into a cell or an organism, resulting in degradation of the corresponding mRNA. The mechanism by which RNAi achieves gene silencing has been reviewed in Sharp et al. (2001) Genes Dev 15: 485-490; and Hammond et al. (2001) Nature Rev Gen 2:110-119). The RNAi effect persists for multiple cell divisions before gene expression is regained. RNAi is therefore a powerful method for making targeted knockouts or “knockdowns” at the RNA level. RNAi has proven successful in human cells, including human embryonic kidney and HeLa cells (see, e.g., Elbashir et al., Nature (2001) 411:494-8).

The RNAi molecule (including an siRNA molecule) can be a short hairpin RNA (shRNA; see Paddison et al. (2002) PNAS USA 99:1443-1448), which is believed to be processed in the cell by the action of the RNase III like enzyme Dicer into 20-25 mer siRNA molecules. The shRNAs generally have a stem-loop structure in which two inverted repeat sequences are separated by a short spacer sequence that loops out. There have been reports of shRNAs with loops ranging from 3 to 23 nucleotides in length. The loop sequence is generally not critical. Exemplary loop sequences include the following motifs: AUG, CCC, UUCG, CCACC, CTCGAG, AAGCUU, CCACACC and UUCAAGAGA.

The RNAi can further comprise a circular molecule comprising sense and antisense regions with two loop regions on either side to form a “dumbbell” shaped structure upon dsRNA formation between the sense and antisense regions. This molecule can be processed in vitro or in vivo to release the dsRNA portion, e.g., a siRNA.

International patent publication WO 01/77350 describes a vector for bi-directional transcription to generate both sense and antisense transcripts of a heterologous sequence in a eukaryotic cell. This technique can be employed to produce RNAi for use according to the invention.

Shinagawa et al. (2003) Genes & Dev. 17:1340 reported a method of expressing long dsRNAs from a CMV promoter (a pol II promoter), which method is also applicable to tissue specific pol II promoters. Likewise, the approach of Xia et al. (2002) Nature Biotech. 20:1006, avoids poly(A) tailing and can be used in connection with tissue-specific promoters.

Methods of generating RNAi include chemical synthesis, in vitro transcription, digestion of long dsRNA by Dicer (in vitro or in vivo), expression in vivo from a delivery vector, and expression in vivo from a PCR-derived RNAi expression cassette (see, e.g., TechNotes 10(3) “Five Ways to Produce siRNAs,” from Ambion, Inc., Austin Tex.).

Guidelines for designing siRNA molecules are available (see e.g., literature from Ambion, Inc., Austin Tex.). In particular embodiments, the siRNA sequence has about 30-50% G/C content. Further, long stretches of greater than four T or A residues are generally avoided if RNA polymerase III is used to transcribe the RNA. Online siRNA target finders are available, e.g., from Ambion, Inc., through the Whitehead Institute of Biomedical Research or from Dharmacon Research, Inc.

The antisense region of the RNAi molecule can be completely complementary to the target sequence (e.g., plexin-A4 and/or Sema3A), but need not be as long as it specifically hybridizes to the target sequence and reduces production of the protein product (e.g., by at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more). In some embodiments, hybridization of such oligonucleotides to target sequences can be carried out under conditions of reduced stringency, medium stringency or even high stringency conditions, as defined herein.

In other embodiments, the antisense region of the RNAi has at least about 60%, 70%, 80%, 90%, 95%, 97%, 98% or higher sequence identity with the complement of the target sequence and reduces production of the protein product (e.g., by at least about 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more). In some embodiments, the antisense region contains 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 mismatches as compared with the target sequence. Mismatches are generally tolerated better at the ends of the dsRNA than in the center portion.

In particular embodiments, the RNAi is formed by intermolecular complexing between two separate sense and antisense molecules. The RNAi comprises a ds region formed by the intermolecular basepairing between the two separate strands. In other embodiments, the RNAi comprises a ds region formed by intramolecular basepairing within a single nucleic acid molecule comprising both sense and antisense regions, typically as an inverted repeat (e.g., a shRNA or other stem loop structure, or a circular RNAi molecule). The RNAi can further comprise a spacer region between the sense and antisense regions.

The RNAi molecule can contain modified sugars, nucleotides, backbone linkages and other modifications as described above for antisense oligonucleotides.

Generally, RNAi molecules are highly selective. If desired, those skilled in the art can readily eliminate candidate RNAi that are likely to interfere with expression of nucleic acids other than the target by searching relevant databases to identify RNAi sequences that do not have substantial sequence homology with other known sequences, for example, using BLAST.

Kits for the production of RNAi are commercially available, e.g., from New England Biolabs, Inc. and Ambion, Inc.

A further aspect of the present invention is an isolated nucleic acid encoding a plexin-A4 polypeptide, an extracellular domain of a plexin-A4 polypeptide, an immunoglobulin Fc fragment, and/or a plexin-A4 fusion protein (e.g., a plexin-A4/Fc IgG fusion protein). Examples include (a) nucleic acids as disclosed herein, such as isolated nucleic acids having the nucleotide sequence as set forth in SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:9, SEQ ID NO:10, or SEQ ID NO:11 (FIGS. 13, 14, 16A-C); (b) nucleic acids that hybridize to isolated nucleic acids of (a) above or the complement thereof (e.g., under stringent conditions), and/or have substantial sequence identity to nucleic acids of (a) above (e.g., are about 75, 80, 85, 90 95 or 99% identical to nucleic acids of (a) above), and encode a plexin-A4 polypeptide, an extracellular domain of a plexin-A4 polypeptide, an immunoglobulin Fc fragment, or a plexin-A4 fusion protein; and (c) nucleic acids that differ from the nucleic acids of (a) or (b) above due to the degeneracy of the genetic code, and code for a plexin-A4, an extracellular domain of plexin-A4, an immunoglobulin Fc fragment, or a plexin-A4 fusion protein.

Thus, in particular embodiments, the present invention provides an isolated plexin-A4 polypeptide, a plexin-A4 fusion protein, a nucleic acid comprising a nucleotide sequence encoding a plexin-A4 polypeptide or a plexin-A4 fusion protein of this invention, a vector comprising said nucleic acid and a cell containing said vector. The plexin-A4 polypeptide and fusion protein, the nucleic acid, the vector and/or the cell can be present singly and/or in any combination in a composition comprising a pharmaceutically acceptable carrier.

Accordingly, in some embodiments, a plexin-A4 coding sequence includes, but is not limited to, the nucleotide sequence of SEQ ID NO:4. In other embodiments, a plexin-A4 cDNA includes, but is not limited to, the nucleotide sequence of SEQ ID NO:5. The genomic sequence of the plexin-A4 gene is also contemplated to be a part of the present invention.

The present invention further provides a nucleic acid comprising an isolated nucleotide sequence encoding the extracellular domain of a plexin-4A polypeptide. Thus, in some embodiments, the extracellular domain of a plexin-4A polypeptide is encoded by the nucleotide sequence of SEQ ID NO:10. The present invention additionally provides a nucleic acid comprising a nucleotide sequence encoding the Fc region of IgG. In some embodiments, the IgG Fc region is encoded by the nucleotide sequence of SEQ ID NO:11. In still further embodiments, the present invention provides a nucleic acid comprising, consisting essentially of, and/or consisting of a nucleotide sequence encoding a plexin-A4/Fc IgG fusion protein. In additional embodiments of the invention, the nucleic acid comprising a nucleotide sequence encoding a plexin-A4/Fc IgG fusion protein comprises the nucleotide sequences of SEQ ID NO:10 and SEQ ID NO:11.

The present invention further provides a nucleic acid comprising a nucleotide sequence encoding a plexin-A4/Fc IgG fusion protein, wherein the plexin-A4/IgG Fc fusion protein comprises an extracellular domain of plexin-A4 fused to the Fc region of an IgG via a peptide linker. Thus, in some embodiments of the invention, the plexin-A4/IgG Fc fusion protein is encoded by the nucleotide sequence of SEQ ID NO:9.

An additional aspect of the present invention is an isolated nucleic acid encoding a Sema3A polypeptide, an immunoglobulin Fc fragment, and/or a Sema3A fusion protein (e.g., a Sema3A/Fc IgG fusion protein). Examples include (a) nucleic acids as disclosed herein, such as isolated nucleic acids having the nucleotide sequence as set forth in SEQ ID NO:11 (FIG. 16C), SEQ ID NO:14, SEQ ID NO:15, or SEQ ID NO: 17 (FIGS. 18B,C and E); (b) nucleic acids that hybridize to isolated nucleic acids of (a) above or the complement thereof (e.g., under stringent conditions), and/or have substantial sequence identity to nucleic acids of (a) above (e.g., are about 75, 80, 85, 90 95 or 99% identical to nucleic acids of (a) above), and encode a Sema3A polypeptide, an immunoglobulin Fc fragment, or a Sema3A fusion protein; and (c) nucleic acids that differ from the nucleic acids of (a) or (b) above due to the degeneracy of the genetic code, and code for a Sema3A polypeptide, an immunoglobulin Fc fragment, or a Sema3A fusion protein.

Thus, in particular embodiments, the present invention provides an isolated Sema3A polypeptide, a Sema3A fusion protein, a nucleic acid comprising a nucleotide sequence encoding a Sema3A polypeptide or a Sema3A fusion protein of this invention, a vector comprising said nucleic acid and a cell containing said vector. The Sema3A polypeptide and fusion protein, the nucleic acid, the vector and/or the cell can be present singly and/or in any combination in a composition comprising a pharmaceutically acceptable carrier.

Accordingly, in some embodiments, a Sema3A coding sequence includes, but is not limited to, the nucleotide sequence of SEQ ID NO:14 (FIG. 18B). In other embodiments, a Sema3A cDNA includes, but is not limited to, the nucleotide sequence of SEQ ID NO:15 (FIG. 18C). The genomic sequence of the Sema3A gene is also contemplated to be a part of the present invention.

As discussed above, the present invention additionally provides a nucleic acid comprising a nucleotide sequence encoding the Fc region of IgG. In some embodiments, the IgG Fc region is encoded by the nucleotide sequence of SEQ ID NO:11. In still further embodiments, the present invention provides a nucleic acid comprising, consisting essentially of, and/or consisting of a nucleotide sequence encoding a Sema3A/Fc IgG fusion protein. In additional embodiments of the invention, the nucleic acid comprising a nucleotide sequence encoding a Sema3A/Fc IgG fusion protein comprises the nucleotide sequences of SEQ ID NO:14 and SEQ ID NO:11.

The present invention further provides a nucleic acid comprising a nucleotide sequence encoding a Sema3A/Fc IgG fusion protein, wherein the Sema3A/IgG Fc fusion protein comprises Sema3A fused to the Fc region of an IgG via a peptide linker. Thus, in some embodiments of the invention, the Sema3A/IgG Fc fusion protein is encoded by the nucleotide sequence of SEQ ID NO:17.

As used herein, “nucleic acid,” “nucleic acid molecule,” “nucleotide sequence” and “polynucleotide” encompass both RNA and DNA, including cDNA, genomic DNA, mRNA, synthetic (e.g., chemically synthesized) DNA and chimeras of RNA and DNA [e.g., DNA-RNA hybrid sequences (including both naturally occurring and non-naturally occurring nucleotides)], but are typically either single or double stranded DNA or RNA sequences.

The term polynucleotide, nucleotide sequence, or nucleic acid refers to a chain of nucleotides without regard to length of the chain. The nucleic acid can be double-stranded or single-stranded (i.e., including the complementary nucleic acid). Where single-stranded, the nucleic acid can be a sense strand or an antisense strand. The nucleic acid can be synthesized using oligonucleotide analogs or derivatives (e.g., inosine or phosphorothioate nucleotides). Such oligonucleotides can be used, for example, to prepare nucleic acids that have altered base-pairing abilities or increased resistance to nucleases. The present invention further provides a nucleic acid that is the complement (which can be either a full complement or a partial complement) of a nucleic acid or nucleotide sequence of this invention.

The term “nucleic acid fragment” will be understood to mean a nucleotide sequence of reduced length relative to a reference nucleic acid or nucleotide sequence and comprising, consisting essentially of and/or consisting of a nucleotide sequence of contiguous nucleotides identical or almost identical (e.g., about 75%, 80%, 82%, 85%, 88%, 90%, 92%, 95%, 98%, 99% identical) to the reference nucleic acid or nucleotide sequence. Such a nucleic acid fragment according to the invention may be, where appropriate, included in a larger polynucleotide of which it is a constituent. In some embodiments, such fragments can comprise, consist essentially of and/or consist of, oligonucleotides having a length of at least about 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25. 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 750, 1000 or more, consecutive nucleotides of a nucleic acid or nucleotide sequence according to the invention.

The terms “complementary” or “complementarity,” as used herein, refer to the natural binding of polynucleotides under permissive salt and temperature conditions by base-pairing. For example, the sequence “A-G-T” binds to the complementary sequence “T-C-A.” Complementarity between two single-stranded molecules may be “partial,” in which only some of the nucleotides bind, or it may be complete when total complementarity exists between the single stranded molecules. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. Similarly, a sequence “homologous” to a target nucleic acid can mean that the sequence is of exact, or nearly exact, identity to the nucleic acid, such that it would specifically hybridize to a nucleic acid exactly complementary to the target nucleic acid. As used herein, stringent hybridization conditions are those conditions that provide selective hybridization for a selected target(s). By “selective hybridization” as used herein is meant that a nucleic acid can hybridize to a target nucleic acid under sufficient stringency conditions without significant hybridization to a nucleic acid likely to be present in the reaction but not of interest. Conditions can be selected to allow for detection of, for example, one or more related nucleic acids having, e.g., one, two, or three mismatches, as in, for example, related strains of the same organism, or they can be selected to provide that hybridization only occurs with one selected target, e.g., such that it differentiates between two strains, depending upon the purpose of the hybridization, amplification and detection. Such conditions are well known to persons of ordinary skill in the art.

Hybridization is typically performed under stringent hybridization conditions. Stringent hybridization conditions are described in, e.g., Sambrook, et. al, Molecular Cloning: A Laboratory Manual, 2nd Ed. Cold Spring Lab. Press, December 1989. However, hybridization conditions can be modified and selected to suit the particular reaction design, including the Tms of the selected primers and the temperature at which the amplification reaction is to be performed. One of skill in the art can suitably design the appropriate reaction conditions for the selected type of amplification and for the selected combination of targets and primers.

Thus, stringency conditions can be adjusted to screen for moderately similar fragments, such as homologous sequences from distantly related organisms, to highly similar fragments, such as genes that duplicate functional enzymes from closely related organisms. Post-hybridization washes determine stringency conditions. Higher stringency conditions utilize buffers with lower ionic strength and/or a higher reaction temperature, and tend to require a more perfect match between probe/primer and a target sequence in order to form a stable duplex. If the stringency is too high, however, hybridization may not occur at all. In contrast, lower stringency conditions utilize buffers with higher ionic strength and/or a lower reaction temperature, and permit the formation of stable duplexes with more mismatched bases between a probe/primer and a target sequence.

Non-limiting examples of high stringency hybridization conditions include: (1) prehybridization with a solution containing 5× standard saline phosphate EDTA (SSPE), 0.5% NaDodSO4 (SDS) at 55° C., and incubating probe with target nucleic acid molecules in the same solution at the same temperature, followed by washing with a solution containing 2×.SSPE, and 0.1% SDS at 55° C. or room temperature; (2) a series of washes starting with 6×SSC, 0.5% SDS at room temperature for 15 min, then repeated with 2×SSC, 0.5% SDS at 45° C. for 30 min, and then repeated twice with 0.2×SSC, 0.5% SDS at 60° C. for 30 min; (3) alternatively, the last two washes of (2) can be carried out at a higher temperature of 65° C.; and/or (4) a single wash at 0.1×SSC, 65° C.

Non-limiting examples of moderate stringency hybridization conditions include: (1) a series of washes starting with 6×SSC, 0.5% SDS at room temperature for 15 min, then repeated with 2×SSC, 0.5% SDS at 45° C. for 30 min, and then repeated twice with 0.2×SSC, 0.5% SDS at 50° C. for 30 min; (2) a wash with a solution of about 50 mM KCl at about 46° C.; (3) a wash with a solution of about 100 mM KCl at about 46° C.; and/or (4) a wash with a solution of 2×SSC, 65° C.

Further exemplary conditions for reduced, medium and high stringency hybridization are as follows: (e.g., conditions represented by a wash stringency of 35-40% Formamide with 5×Denhardt's solution, 0.5% SDS and 1×SSPE at 37° C.; conditions represented by a wash stringency of 40-45% Formamide with 5×Denhardt's solution, 0.5% SDS, and 1×SSPE at 42° C.; and conditions represented by a wash stringency of 50% Formamide with 5×Denhardt's solution, 0.5% SDS and 1×SSPE at 42° C., respectively). See, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual (2d Ed. 1989) (Cold Spring Harbor Laboratory).

The term “isolated” can refer to a nucleic acid, nucleotide sequence, polypeptide, peptide or fragment that is substantially free of cellular material, viral material, and/or culture medium (e.g., when produced by recombinant DNA techniques), or chemical precursors or other chemicals (e.g., when chemically synthesized). Moreover, an “isolated fragment” is a fragment of a nucleic acid, nucleotide sequence or polypeptide that is not naturally occurring as a fragment and would not be found as such in the natural state. “Isolated” does not mean that the preparation is technically pure (homogeneous), but it is sufficiently pure to provide the polypeptide or nucleic acid in a form in which it can be used for the intended purpose.

An “isolated nucleic acid” is a nucleotide sequence (e.g., DNA or RNA) that is not immediately contiguous with nucleotide sequences with which it is immediately contiguous (one on the 5′ end and one on the 3′ end) in the naturally occurring genome of the organism from which it is derived. Thus, in one embodiment, an isolated nucleic acid includes some or all of the 5′ non-coding (e.g., promoter) sequences that are immediately contiguous to a coding sequence. The term therefore includes, for example, a recombinant DNA that is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., a cDNA or a genomic DNA fragment produced by PCR or restriction endonuclease treatment), independent of other sequences. It also includes a recombinant DNA that is part of a hybrid nucleic acid encoding an additional polypeptide or peptide sequence.

Different nucleic acids or proteins having homology are referred to herein as “homologues.” The term homologue includes homologous sequences from the same and other species and orthologous sequences from the same and other species. “Homology” refers to the level of similarity between two or more nucleic acid and/or amino acid sequences in terms of percent of positional identity (i.e., sequence similarity or identity). Homology also refers to the concept of similar functional properties among different nucleic acids or proteins.

As is well known in the art, nucleic acid sequences can have changes in one or more nucleotide bases that results in substitution of one or more amino acids, but which do not affect the functional properties of the polypeptide encoded by the nucleotide sequence. It is therefore understood that the invention encompasses more than the specific exemplary nucleotide or amino acid sequences and includes functional equivalents thereof.

For example, alterations in the nucleotide sequence of a gene which result in the production of a chemically equivalent amino acid at a given site, but do not affect the functional properties of the encoded protein, are well known in the art. Such alterations include “conservative” changes, wherein a substituted amino acid has similar structural or chemical properties. In particular, such changes can be guided by known similarities between amino acids in physical features such as charge density, hydrophobicity/hydrophilicity, size and configuration, so that amino acids are substituted with other amino acids having essentially the same functional properties. For example: Ala may be replaced with Val or Ser; Val may be replaced with Ala, Leu, Met, or Ile, preferably Ala or Leu; Leu may be replaced with Ala, Val or Ile, preferably Val or Ile; Gly may be replaced with Pro or Cys, preferably Pro; Pro may be replaced with Gly, Cys, Ser, or Met, preferably Gly, Cys, or Ser; Cys may be replaced with Gly, Pro, Ser, or Met, preferably Pro or Met; Met may be replaced with Pro or Cys, preferably Cys; His may be replaced with Phe or Gln, preferably Phe; Phe may be replaced with His, Tyr, or Trp, preferably His or Tyr; Tyr may be replaced with His, Phe or Trp, preferably Phe or Trp; Trp may be replaced with Phe or Tyr, preferably Tyr; Asn may be replaced with Gln or Ser, preferably Gln; Gln may be replaced with His, Lys, Glu, Asn, or Ser, preferably Asn or Ser; Ser may be replaced with Gln, Thr, Pro, Cys or Ala; Thr may be replaced with Gln or Ser, preferably Ser; Lys may be replaced with Gln or Arg; Arg may be replaced with Lys, Asp or Glu, preferably Lys or Asp; Asp may be replaced with Lys, Arg, or Glu, preferably Arg or Glu; and Glu may be replaced with Arg or Asp, preferably Asp. Once made, changes can be routinely screened to determine their effects on function. Each of the proposed modifications is well within the routine skill in the art, as is determination of retention of biological activity of the encoded products.

Alternatively, alterations in the coding sequence of a gene may involve “nonconservative” changes (e.g., replacement of glycine with tryptophan). Analogous minor variations may also include amino acid deletions or insertions, or both. Guidance in determining which amino acid residues may be substituted, inserted, or deleted without abolishing biological activity may be found using computer programs well known in the art, such as for example, LASERGENE™ software.

In particular embodiments, a resultant variant polypeptide has at least about 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more amino acid sequence similarity or identity with the amino acid sequence of a naturally occurring protein or reference amino acid sequence (e.g., plexin-A4, Sema3A or IgG-Fc).

Thus, the invention further provides homologues, as well as methods of obtaining homologues, of the polypeptides and/or fragments of this invention from other organisms included in this invention. As used herein, an amino acid sequence or protein is defined as a homologue of a polypeptide or fragment of the present invention if it shares significant homology or identity to a polypeptide, peptide and/or fragment of the present invention. Significant homology or identity means at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, and/or 100% homology or identity with another amino acid sequence. Specifically, by using the nucleic acids that encode the proteins, peptides and fragments of this invention, as a probe or primer, and techniques such as PCR amplification and colony/plaque hybridization, one skilled in the art can identify homologues of the polypeptides, peptides and/or fragments of this invention in other organisms on the basis of information available in the art.

The term “percent identity,” as known in the art, describes a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between polypeptide or polynucleotide sequences as determined by the match between strings of such sequences. “Identity” and “similarity” can be readily calculated by known methods including, but not limited to, those described in: Computational Molecular Biology (Lesk, A. M., ed.) Oxford University Press, New York (1988); Biocomputing: Informatics and Genome Projects (Smith, D. W., ed.) Academic Press, New York (1993); Computer Analysis of Sequence Data, Part I (Griffin, A. M., and Griffin, H. G., eds.) Humana Press, New Jersey (1994); Sequence Analysis in Molecular Biology (von Heinje, G., ed.) Academic Press (1987); and Sequence Analysis Primer (Gribskov, M. and Devereux, J., eds.) Stockton Press, New York (1991).

Accordingly, the present invention further provides nucleotide sequences having significant sequence similarity or identity to the nucleotide sequences of the present invention. Significant sequence similarity or identity means at least 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, and/or 100% similarity or identity with another nucleotide sequence.

Exemplary methods to determine identity are designed to give the best match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Sequence alignments and percent identity calculations can be performed using the Megalign program of the LASERGENE bioinformatics computing suite (DNASTAR Inc., Madison, Wis.). Multiple alignment of the sequences may be performed using the Clustal method of alignment (Higgins and Sharp (1989) CABIOS. 5:151-153) with the default parameters (GAP PENALTY=10, GAP LENGTH PENALTY=10). Exemplary default parameters for pairwise alignments using the Clustal method can be selected: KTUPLE 1, GAP PENALTY=3, WINDOW=5 and DIAGONALS SAVED=5.

The term “sequence analysis software” refers to any computer algorithm or software program that is useful for the analysis of nucleotide and/or amino acid sequences. Sequence analysis software is commercially available or can be independently developed. Typical sequence analysis software will include but is not limited to the GCG suite of programs (Wisconsin Package Version 9.0, Genetics Computer Group (GCG), Madison, Wis.), BLASTP, BLASTN, BLASTX (Altschul et al., J. Mol. Biol. 215:403-410 (1990), and DNASTAR (DNASTAR, Inc. 1228 S. Park St. Madison, Wis. 53715 USA). Within the context of this application, it will be understood that where sequence analysis software is used for analysis, the results of the analysis will be based on the “default values” of the program referenced, unless otherwise specified. As used herein “default values” will mean any set of values or parameters, which originally load with the software when first initialized.

A percentage amino acid sequence identity value is determined by the number of matching identical residues divided by the total number of residues of the “longer” sequence in the aligned region. The “longer” sequence is the one having the most actual residues in the aligned region (gaps introduced by WU-Blast-2 to maximize the alignment score are ignored).

The alignment can include the introduction of gaps in the sequences to be aligned. In addition, for sequences which contain either more or fewer amino acids than the polypeptides specifically disclosed herein, it is understood that in one embodiment, the percentage of sequence identity will be determined based on the number of identical amino acids in relation to the total number of amino acids. Thus, for example, in one embodiment, sequence identity of sequences shorter than a sequence specifically disclosed herein, will be determined using the number of amino acids in the shorter sequence. In percent identity calculations relative weight is not assigned to various manifestations of sequence variation, such as insertions, deletions, substitutions, etc.

In other embodiments, only identities are scored positively (+1) and all forms of sequence variation including gaps are assigned a value of “0,” which obviates the need for a weighted scale or parameters as described below for sequence similarity calculations. Percent sequence identity can be calculated, for example, by dividing the number of matching identical residues by the total number of residues of the “shorter” sequence in the aligned region and multiplying by 100. The “longer” sequence is the one having the most actual residues in the aligned region.

The nucleotide sequences and/or nucleic acid fragments of the instant invention may be used to isolate cDNAs and genes encoding homologous proteins from the same or different organisms. Isolation of homologous genes using sequence-dependent protocols is well known in the art. Examples of sequence-dependent protocols include, but are not limited to, methods of nucleic acid hybridization, and methods of DNA and RNA amplification as exemplified by various uses of nucleic acid amplification technologies (e.g., polymerase chain reaction, ligase chain I reaction, etc.).

For example, genes encoding other plexin-A4 and/or Sema3A polypeptides, either as cDNAs or genomic DNAs, could be isolated directly by using all or a substantial portion of the nucleic acid sequences or fragments of the present invention as DNA hybridization probes to screen libraries from any desired organism employing methodology well known to those skilled in the art. Specific oligonucleotide probes based upon the instant nucleic acid sequences can be designed and synthesized by methods known in the art (See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual 2nd Ed. (Cold Spring Harbor, N.Y., 1989); Ausubel et al. Current Protocols in Molecular Biology (Green Publishing Associates, Inc. and John Wiley & Sons, Inc., New York). Moreover, the entire sequence(s) can be used directly to synthesize DNA probes by methods known to the skilled artisan such as random primer DNA labeling, nick translation, end-labeling techniques, or RNA probes using available in vitro transcription systems. In addition, specific primers can be designed and used to amplify a part or all of the instant sequences. The resulting amplification products can be labeled directly during amplification reactions or labeled after amplification reactions, and used as probes to isolate full length cDNA or genomic fragments under conditions of appropriate stringency.

In addition, two short segments of the instant nucleic acid fragments may be used in polymerase chain reaction protocols to amplify longer nucleic acid fragments encoding homologous genes from DNA or RNA. The polymerase chain reaction may also be performed on a library of cloned nucleic acid fragments wherein the sequence of one primer is derived from the instant nucleic acid fragments, and the sequence of the other primer takes advantage of the presence of the polyadenylic acid tracts to the 3′ end of the mRNA precursor encoding the plexin-A4 genes or the Sema3A genes. Alternatively, the second primer sequence may be based upon sequences derived from the cloning vector. For example, the skilled artisan can follow the RACE protocol (Frohman et al. (1988) Proc. Natl. Acad. Sci. USA 85:8998-9002) to generate cDNAs by using PCR to amplify copies of the region between a single point in the transcript and the 3′ or 5′ end. Primers oriented in the 3′ and 5′ directions can be designed from the instant sequences. Using commercially available 3′ RACE or 5′ RACE systems (BRL), specific 3′ or 5′ cDNA fragments can be isolated (Ohara et al. (1989) Proc. Natl. Acad. Sci. USA 86:5673-5677; Loh et al. (1989) Science 243:217-220). Products generated by the 3′ and 5′ RACE procedures can be combined to generate full-length cDNAs (Frohman and Martin (1989) Techniques 1:165).

The term “therapeutically effective amount” or “effective amount,” as used herein, refers to that amount of a composition of this invention that imparts a modulating effect, which, for example, can be a beneficial effect, to a subject afflicted with a disorder, disease or cosmetic condition, including improvement in the disease or disorder of the subject (e.g., in one or more symptoms), delay or reduction in the progression of the disease or disorder, prevention or delay of the onset of the disease or disorder, and/or change in clinical parameters of the disease or disorder, as would be well known in the art. The effective amount will vary with the age, general condition of the subject, the severity of the disease, disorder or condition being treated, the particular agent or composition administered, the duration of the treatment, the nature of any concurrent treatment, the pharmaceutically acceptable carrier used, and like factors within the knowledge and expertise of those skilled in the art. As appropriate, an “effective amount” in any individual case can be determined by one of ordinary skill in the art by reference to the pertinent texts and literature and/or by using routine experimentation. (See, for example, Remington, The Science and Practice of Pharmacy (20th ed. 2000)). For example, a therapeutically effective amount or effective amount can refer to the amount of a composition, compound, or agent that improves a disease or disorder in a subject by at least 5%, e.g., at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 100%.

Although individual needs may vary, the determination of optimal ranges for effective amounts of a composition of this invention is within the skill of the art. Human doses can also readily be extrapolated from animal studies (Katocs et al., Chapter 27 In: Remington's Pharmaceutical Sciences, 18.sup.th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990). Generally, the dosage required to provide an effective amount of a composition of this invention, which can be adjusted by one skilled in the art, will vary depending on several factors, including the age, health, physical condition, weight, type and extent of the disease or disorder of the recipient, frequency of treatment, the nature of concurrent therapy, if required, and the nature and scope of the desired effect(s) (Nies et al., Chapter 3 In: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9.sup.th Ed., Hardman et al., eds., McGraw-Hill, New York, N.Y., 1996). In some embodiments, a daily dosage of fusion protein can be about 1 μg to 100 milligrams per kilogram of body weight. In other embodiments, the dosage can be 0.5 μg to 50 milligrams per kilogram of body weight, and preferably 1 to 10 milligrams per kilogram per day given in divided doses 1 to 6 times a day or in sustained release form is effective to obtain desired results.

The pharmaceutical compositions according to the present invention may be administered as a single dose or in multiple doses. The pharmaceutical compositions of the present invention may be administered either as individual therapeutic agents or in combination with other therapeutic agents, which when combined may be administered sequentially or simultaneously.

For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight. In some aspects of the invention, the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight. In other aspects of the invention, the dosage can be 1 mg/kg to 10 mg/kg of the patient's body weight. Generally, human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible. Further, the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.

By the terms “treat,” “treating” or “treatment of” (or grammatically equivalent terms) it is meant that the severity of the subject's disease or disorder is reduced or at least partially improved or ameliorated and/or that some alleviation, mitigation or decrease in at least one clinical symptom is achieved and/or there is a delay in the progression of the disease or disorder, as would be well known in the art. Thus, in some embodiments, the terms “treat,” “treating” or “treatment of” refer only to therapeutic regimens. In other embodiments, the terms “treat,” “treating” or “treatment of” (or grammatically equivalent terms) refer to both prophylactic and therapeutic regimens. As an example, a patient exhibiting bacteremia may be treated with the compositions of the present invention prior to the onset of sepsis in order to prevent the onset of said disease.

The terms “prevent,” “preventing” and “prevention” (and grammatical variations thereof) refer to avoidance, prevention and/or delay of the onset of a disease, disorder and/or a clinical symptom(s) in a subject and/or a reduction in the severity of the onset of the disease, disorder and/or clinical symptom(s) relative to what would occur in the absence of the methods of the invention. The prevention can be complete, e.g., the total absence of the disease, disorder and/or clinical symptom(s). The prevention can also be partial, such that the occurrence of the disease, disorder and/or clinical symptom(s) in the subject and/or the severity of onset is delayed and/or is less than what would occur in the absence of the method of the present invention.

An “effective amount,” as used herein, refers to an amount that imparts a desired effect, which is optionally a therapeutic or prophylactic effect.

A “treatment effective” amount as used herein is an amount that is sufficient to provide some improvement or benefit to the subject. Alternatively stated, a “treatment effective” amount is an amount that will provide some alleviation, mitigation, decrease or stabilization in at least one clinical symptom in the subject. Those skilled in the art will appreciate that the therapeutic effects need not be complete or curative, as long as some benefit is provided to the subject.

A “prevention effective” amount as used herein is an amount that is sufficient to prevent and/or delay the onset of a disease, disorder and/or clinical symptoms in a subject and/or to reduce and/or delay the severity of the onset of a disease, disorder and/or clinical symptoms in a subject relative to what would occur in the absence of the methods of the invention. Those skilled in the art will appreciate that the level of prevention need not be complete, as long as some benefit is provided to the subject.

Pharmaceutical compositions comprising the plexin-A4/Fc IgG fusion proteins, plexin-A4 antibodies and/or fragments thereof of this invention, and a pharmaceutically acceptable carrier are also provided. Additionally provided herein are pharmaceutical compositions comprising the Sema3A/Fc IgG fusion proteins, Sema3A antibodies and/or fragments thereof of this invention, and a pharmaceutically acceptable carrier. The compositions described herein can be formulated for administration in a pharmaceutical carrier in accordance with known techniques. See, e.g., Remington, The Science And Practice of Pharmacy (latest edition). In the manufacture of a pharmaceutical composition according to embodiments of the present invention, the composition of this invention is typically admixed with, inter alia, a pharmaceutically acceptable carrier. By “pharmaceutically acceptable carrier” is meant a carrier that is compatible with other ingredients in the pharmaceutical composition and that is not harmful or deleterious to the subject. The carrier may be a solid or a liquid, or both, and is preferably formulated with the composition of this invention as a unit-dose formulation, for example, a tablet, which may contain from about 0.01 or 0.5% to about 95% or 99% by weight of the composition. The pharmaceutical compositions are prepared by any of the well-known techniques of pharmacy including, but not limited to, admixing the components, optionally including one or more accessory ingredients. In certain embodiments, the pharmaceutically acceptable carrier is sterile and would be deemed suitable for administration into human subjects according to regulatory guidelines for pharmaceutical compositions comprising the carrier.

Furthermore, a “pharmaceutically acceptable” component such as a salt, carrier, excipient or diluent of a composition according to the present invention is a component that (i) is compatible with the other ingredients of the composition in that it can be combined with the compositions of the present invention without rendering the composition unsuitable for its intended purpose, and (ii) is suitable for use with subjects as provided herein without undue adverse side effects (such as toxicity, irritation, and allergic response). Side effects are “undue” when their risk outweighs the benefit provided by the composition. Non-limiting examples of pharmaceutically acceptable components include any of the standard pharmaceutical carriers such as phosphate buffered saline solutions, water, emulsions such as oil/water emulsion, microemulsions and various types of wetting agents.

Exemplary modes of administration of the proteins, peptides, fragments, nucleic acids and/or vectors of this invention can include oral, rectal, transmucosal, topical, intranasal, inhalation (e.g., via an aerosol), buccal (e.g., sublingual), vaginal, intrathecal, intraocular, transdermal, in utero (or in ovo), parenteral (e.g., intravenous, subcutaneous, intradermal, intramuscular [including administration to skeletal, diaphragm and/or cardiac muscle], intraperitoneal, intradermal, intrapleural, intracerebral, intracranial, and intraarticular), topical (e.g., to both skin and mucosal surfaces, including airway surfaces, and transdermal administration, and the like, as well as direct tissue or organ injection (e.g., to liver, skeletal muscle, cardiac muscle, diaphragm muscle or brain). The most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular protein, peptide, fragment, nucleic acid and/or vector that is being used.

The compositions of the present invention may be administered to a subject in need of treatment prior to, during or after onset of the disease or disorder. For instance, a patient exhibiting bacteremia may be treated with a composition of the present invention prior to onset of sepsis to prevent progression to sepsis. Thus, the compositions of the present invention can be used to treat ongoing immune-related and/or inflammatory diseases or disorders or to prevent diseases or delay the development of immune-related and/or inflammatory diseases or disorders.

In some embodiments, an effective dose or effective amount can comprise one or more (e.g., two or three or four or more) doses of the composition of this invention at any time interval (e.g., hourly, daily, weekly, monthly, yearly, as needed, etc.) so as to achieve and/or maintain the desired therapeutic benefit.

Several methods known in the art may be used to produce a polynucleotide and/or vector according to this invention. A “vector” is any nucleic acid molecule for the cloning and/or amplification of nucleic acid as well as for the transfer of nucleic acid into a subject (e.g., a cell of the subject). A vector may be a replicon to which another nucleotide sequence may be attached to allow for replication of the attached nucleotide sequence. A “replicon” can be any genetic element (e.g., plasmid, phage, cosmid, chromosome, viral genome) that functions as an autonomous unit of nucleic acid replication in vivo, i.e., capable of replication under its own control. The term “vector” includes both viral and nonviral nucleic acid molecules for introducing a nucleic acid into a cell in vitro, ex vivo, and/or in vivo.

A large number of vectors known in the art may be used to manipulate nucleic acids, incorporate response elements and promoters into genes, etc. Such vectors include, for example, plasmids or modified viruses including, for example bacteriophages such as lambda derivatives, or plasmids such as pBR322 or pUC plasmid derivatives, or the Bluescript vector. For example, the insertion of the nucleic acid fragments corresponding to response elements and promoters into a suitable vector can be accomplished by ligating the appropriate nucleic acid fragments into a chosen vector that has complementary cohesive termini. Alternatively, the ends of the nucleic acid molecules may be enzymatically modified or any site may be produced by ligating nucleotide sequences (linkers) to the nucleic acid termini. Such vectors may be engineered to contain sequences encoding selectable markers that provide for the selection of cells that contain the vector and/or have incorporated the nucleic acid of the vector into the cellular genome. Such markers allow identification and/or selection of host cells that incorporate and express the proteins encoded by the marker.

Vectors have been used in a wide variety of gene delivery applications in cells, as well as in living animal subjects. Viral vectors that can be used include, but are not limited to, retrovirus, lentivirus, adeno-associated virus, poxvirus, alphavirus, baculovirus, vaccinia virus, herpes virus, Epstein-Barr virus, and adenovirus vectors, as well as any combination thereof. Nonlimiting examples of non-viral vectors include plasmids, liposomes, electrically charged lipids (cytofectins), nucleic acid-protein complexes, and biopolymers, as well as any combination thereof. In addition to a nucleic acid of interest, a vector may also comprise one or more regulatory regions (e.g., promoters, enhancers, termination sequences, etc.), and/or selectable markers useful in selecting, measuring, and monitoring nucleic acid transfer results (delivery to specific tissues, duration of expression, etc.).

In embodiments in which a nucleic acid of this invention is delivered in a viral vector (e.g., a virus particle), the dosage of virus particles to be administered to a subject will depend upon the mode of administration, the disease or disorder to be treated, the individual subject's condition, the particular virus vector, and the nucleic acid to be delivered, and can be determined in a routine manner. Exemplary doses are virus titers of at least about 105, 106, 107, 108, 109, 1010, 1011, 1012, 103, 1014, 1015 transducing units or more, preferably about 108-1013 transducing units, yet more preferably 1012 transducing units.

The present invention further provides a method of identifying a substance having an inhibitory effect on plexin-A4 activity, comprising contacting the substance with macrophage/dendritic cells under conditions whereby plexin-A4 activity can occur and measuring the amount of plexin-A4 activity in the presence and absence of the substance, whereby a decrease in plexin-A4 activity in the presence of the substance as compared to the amount of plexin-A4 activity in the absence of the substance identifies a substance having an inhibitory effect on plexin-A4 activity.

In addition, the present invention provides a method of identifying a substance having an inhibitory effect on plexin-A4 expression, comprising contacting the substance with macrophage/dendritic cells under conditions whereby plexin-A4 expression can occur and measuring the amount of plexin-A4 expression in the presence and absence of the substance, whereby a decrease in plexin-A4 expression in the presence of the substance as compared to the amount of plexin-A4 expression in the absence of the substance identifies a substance having an inhibitory effect on plexin-A4 expression.

Plexin-A4 activity and expression can be measured by many means as known in the art. For example, plexin-A4 expression can be measured at the RNA level by conventional protocols including, but not limited to, Northern blots, real-time PCR, reverse transcribed PCR, RNase protection protocols, and/or in situ hybridization. Plexin-A4 protein can be measured by protocols including, but not limited to, immunodetection such as ELISA, flow cytometric analysis, binding to Sema3A, and/or a combination of microscopic and immunolabelling and immunoblot methods. The activity of plexin-A4 protein can be measured by its ability optimize TLR activation by TLR agonists in the absence or presence of plexin-A4 fusion protein. TLR activation can be measured by events such as the secretion of proinflammatory cytokines (non-limiting examples of which are IL-6 and TNF) and chemokines, and by the activation of Rac, NF-kB and MAP kinases.

The present invention further provides a method of identifying a substance having an inhibitory effect on Sema3A activity, comprising contacting the substance with macrophage/dendritic cells under conditions whereby SemaA3 activity can occur and measuring the amount of Sema3A activity in the presence and absence of the substance, whereby a decrease in Sema3A activity in the presence of the substance as compared to the amount of Sema3A activity in the absence of the substance identifies a substance having an inhibitory effect on Sema3A activity.

In addition, the present invention provides a method of identifying a substance having an inhibitory effect on Sema3A expression, comprising contacting the substance with macrophage/dendritic cells under conditions whereby Sema3A expression can occur and measuring the amount of Sema3A expression in the presence and absence of the substance, whereby a decrease in Sema3A expression in the presence of the substance as compared to the amount of Sema3A expression in the absence of the substance identifies a substance having an inhibitory effect on Sema3A expression.

Sema3A activity and expression can be measured by many means as known in the art. For example, Sema3A expression can be measured at the RNA level by conventional protocols including, but not limited to, Northern blots, real-time PCR, reverse transcribed PCR, RNase protection protocols, and/or in situ hybridization. Sema3A protein can be measured by protocols including, but not limited to, immunodetection such as ELISA, flow cytometric analysis, binding to plexin-A4, and/or a combination of microscopic and immunolabelling and immunoblot methods. The activity of Sema3A protein can be measured by its ability optimize TLR activation by TLR agonists in the absence or presence of Sema3A fusion protein. TLR activation can be measured by events such as the secretion of proinflammatory cytokines (non-limiting examples of which are IL-6 and TNF) and chemokines, and by the activation of Rac, NF-kB and MAP kinases.

The present invention further provides a method of identifying a substance having an enhancing effect on plexin-A4 activity or expression, said method comprising contacting the substance with macrophage/dendritic cells under conditions whereby plexin A4 activity or expression can occur and measuring the amount of plexin-A4 activity or expression in the presence and absence of the substance, whereby an increase in plexin-A4 activity or expression in the presence of the substance as compared to the amount of plexin-A4 activity or expression in the absence of the substance identifies a substance having an enhancing effect on plexin-A4 activity or expression. Methods for detecting plexin-A4 activity or expression are known in the art and discussed above.

The present invention additionally provides a method of identifying a substance having an enhancing effect on Sema3A activity or expression, said method comprising contacting the substance with macrophage/dendritic cells under conditions whereby Sema3A activity or expression can occur and measuring the amount of Sema3A activity or expression in the presence and absence of the substance, whereby an increase in Sema3A activity or expression in the presence of the substance as compared to the amount of Sema3A activity or expression in the absence of the substance identifies a substance having an enhancing effect on Sema3A activity or expression. Methods for detecting Sema3A activity or expression are known in the art and discussed above.

The following examples are included to demonstrate various embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples that follow represent techniques discovered by the inventors to function well in the practice of the invention. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.

EXAMPLES Example 1 Mice

C57BL/6 mice were purchased from the Jackson Laboratory. Plxna4−/− mice were provided by M. Tessier-Lavigne (Stanford University) (Yaron et al., 2005) and were backcrossed for nine generations onto the C57BL/6 background. OT-II TCR-transgenic mice were obtained from M. Croft (La Jolla Institute of Allergy and Immunology). Mice were treated in accordance with the National Institute of Health Guide for the Care and Use of Laboratory Animals. The Institutional Animal Care and Use Committee (IACUC) of the University of North Carolina at Chapel Hill approved all experimental procedures.

Reagents.

Pam3Cys, poly(I:C), Ultrapure LPS, imiquimod (R837) and mouse CpG-B (ODN 1826) were from InvivoGen. LPS (E. coli 0111:B4) used for the in vivo injections was purchased from Sigma. Recombinant human Sema3A-Fc (1250-S3), human Sema6A-Fc (1146-S6) and human IgG1 Fc (110-HG) were from R&D. The Rac1 inhibitor NSC23766 (Gao et al., 2004) was from Calbiochem. In immunoblotting experiments, the primary antibodies specific for NF-κB p65 (3034), NF-κB p65 phosphorylated at Ser536 (93H1; 3033), IκBα phosphorylated at Ser32 (14D4; 2859), phospho-ERK1/2 (Thr202/Thr204) (197G2; 4377), phospho-JNK1/2 (Thr183/Tyr185) (81E11; 4668) and phospho-p38 (Thr180/Tyr182) (9211) were from Cell Signaling Technology. Anti-β-actin (C-11; sc-1615 HRP) was from Santa Cruz Biotechnology. Antibody specific for mouse plexin-A4 has been previously described (Suto et al., 2007) and was kindly provided by F. Suto (Tohoku University). In ChIP analysis, the antibodies specific for NF-κB p65 (A) X (sc-109) and c-Jun phosphorylated at Ser63 (9261) were from Santa Cruz Biotechnology (Santa Cruz, Calif.) and Cell Signaling Technology (Danvers, Mass.), respectively. In assays for GTP-bound Rac1, Cdc42 and RhoA, glutathione-SEPHAROSE™ (crosslinked polysaccharide polymer) beads conjugated with either GST-Pak1 PBD (for GTP-bound Rac1 and Cdc42) or GST-rhotekin RBD (for GTP-bound RhoA) were kindly provided by K. Burridge (University of North Carolina at Chapel Hill). The primary antibodies specific for Rac1 (C-14; sc-217), Cdc42 (B-8; sc-8401) and RhoA (26C4; sc-418) were from Santa Cruz Biotechnology.

Example 2 Peritoneal Macrophage Isolation and In Vitro Stimulation

Total peritoneal cells were harvested from the peritoneal cavities of naïve WT or Plxna44″ mice with two 10 ml cold sterile PBS wash. Lavage was pooled for mice in the same group. RBCs were lysed in ammonium chloride buffer (150 mM NH4CL, 10 mM NaHCO3, 1 mM EDTA-tetrasodium). The remaining cells were thoroughly washed with PBS and counted in a hemocytometer. Cytospins were prepared and stained with Diff-Quik solutions, and the number of peritoneal macrophages was determined. Cells were resuspended at a concentration of 106 macrophages/ml in RPMI 1640 medium supplemented with 10% FCS, 2 mM L-glutamine, 100 U/ml penicillin, and 100 μg streptomycin. Macrophages were plated in 96-well cell culture plate (2×105/well) and incubated for 2 h at 37° C. Non-adherent cells were removed, and adherent cells were washed with complete RPMI 1640 medium, followed by stimulation with various reagents. In some experiments, cells were pretreated with 200 μM NSC23766 for 1 h followed by the stimulation with TLR agonists.

Example 3 Bacterial Infection In Vitro

L. monocytogenes and S. typhimurium were from the ATCC (ATCC 43251 and ATCC14028, respectively). E. coli strain LF82 has been previously described (Carvalho et al., 2008) and was provided by R. Balfour Sartor (University of North Carolina at Chapel Hill). S. aureus strain RN6390 has been previously described (Labandeira-Rey et al., 2007) and was provided by M. Gabriela Bowden (Texas A&M University). All bacterial strains were grown to the late exponential phase. The number of viable bacteria used in each experiment was calibrated by colony counting. After three washes with sterile cold PBS, bacteria were added to peritoneal macrophages at a multiplicity of infection (MOI) of 40. After incubation for 1 h at 37° C., cells were treated with 100 μg/ml gentamicin to kill extracellular bacteria and were further incubated for various time periods (S. typhimurium for 1 h; L. monocytogenes, E. coli and S. aureus for 3 h).

Example 4 ELISA

Cytokines generated by in vitro cultured peritoneal macrophages were quantified using the ELISA Set for mouse TNF-α or IL-6 (BD Biosciences) and mouse IFN-β ELISA Kit (PBL InterferonSource). For the in vivo experiments, concentrations of TNF-α, IL-1β, IL-6, IL-12p70, CCL2 and CCL3 were measured in cell-free peritoneal lavage fluid, serum, and lung homogenate using a Customized Mouse Cytokine 6-plex Panel (BioLegend) and a Luminex Bio-Plex 200 system (Bio-Rad Laboratories).

Example 5 Immunoblotting

Electrophoresis of proteins was performed by using the NuPAGE system (Invitrogen) according to the manufacturer's protocol. Briefly, peritoneal macrophage stimulated with LPS or left unstimulated were collected and lysed with buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 2 mM EDTA, 1% TRITON™ X-100 (a non-ionic surfactant), 0.1% SDS, and protease inhibitor cocktail). Proteins were separated on a NuPAGE® SDS PAGE gel and were transferred onto nitrocellulose membranes (Bio-Rad Laboratories). The membranes were blocked with 10% milk proteins in 1×PBS and 0.1% Tween-20 and then probed with the primary antibodies. Appropriate HRP-conjugated secondary antibodies were used and proteins were detected using an Enhanced Chemiluminescent (ECL) Western Blotting Substrate reagent (peroxidase substrate) from Thermo Scientific.

Example 6 Chromatin Immunoprecipitation (ChIP) Assay

ChIP assays were performed as described previously (Wen et al., 2008). Briefly, resident peritoneal macrophages were stimulated with various TLR agonists for 1 hour. DNA-protein structure was then crosslinked by 1% formaldehyde for 10 min at 37° C. Cells were collected and lysed in 400 μl SDS lysis buffer. The resulting lysates were sonicated to obtain DNA fragments ranging from 200-1,000 bp using a Sonics VCX 750 (Sonics & Materials, Inc.) 4× for periods of 15 s each. After centrifugation, the supernatant containing chromatin was diluted and an aliquot (2% volume) was saved to quantitate the input DNA in each sample. The remaining chromatin fractions were pre-cleared with salmon sperm DNA/protein A agarose beads (Invitrogen) followed by immunoprecipitation overnight with primary antibodies at 4° C. with gentle rotation. Cross-linking was reversed for 4 h at 65° C. and was followed by proteinase K digestion. DNA was purified by standard phenol/chloroform and ethanol precipitation, and subjected to real-time PCR. Primers for mouse Tnfa promoter κB binding site: forward, 5′-TGAAAGGAGAAGGCTTGTGA (SEQ ID NO:18); reverse, 5′-TAATGGGATGAGTATGGGGCA (SEQ ID NO:19). Primers for mouse Tnfa promoter c-Jun binding site: forward, 5′-CCCAACTTTCCAAACCCTCT (SEQ ID NO:20); reverse, 5′-ACCATGATCTCATGTGGAGGA (SEQ ID NO:21). Primers for mouse Il6 promoter: forward, 5′-GCCTCAAGGATGACTTAAGCA (SEQ ID NO:22); reverse, 5′-AGATTGCACAATGTGACGTCG (SEQ ID NO:23).

Example 7 Detection of GTP-Bound Rac1, Cdc42 and RhoA

The assays for GTP-bound Rac1, Cdc42 and RhoA were performed as described (Noren et al., 2000). Briefly, peritoneal macrophages were stimulated with 1 μg/ml LPS for various time periods. Cells were lysed and GTP-bound Rac1 and Cdc42 were affinity precipitated using Rac1/Cdc42-binding domain of PAK (PBD) fused with GST. GTP-bound RhoA was affinity precipitated using RhoA-binding domain of rhotekin (RBD) fused with GST. Bound proteins were resolved on a NuPAGE® SDS PAGE gel and were transferred onto nitrocellulose membranes. Membranes were immunoblotted with the appropriate primary antibodies against Rac1, Cdc42 or RhoA.

Example 8 Experimental Sepsis Induced by CLP

CLP surgery was performed on mice as previously described with minor modifications (Wen et al., 2008). In brief, mice were anesthetized with an intraperitoneal injection of 8 mg of 2,2,2-tribromoethanol (Avertin) (Sigma). Under sterile surgical conditions, a 1-cm midline incision was made to the ventral surface of the abdomen and the cecum was exposed. The cecum was partially ligated at its base with a 3.0 silk suture and punctured two or nine times with 21-gauge needle. The cecum was returned to the peritoneal cavity and the abdominal incision was closed using surgical staples. Mice were rehydrated with 1 ml saline subcutaneously and placed on a heating pad until they recovered from anesthetic. Sham-operated mice were subjected to a similar laparotomy without ligation and puncture. For survival studies, mice subjected to surgery were monitored for 5 days. For analysis of septic inflammatory responses, CLP mice were anesthetized and bled at 4, 24 and 72 h after surgery. Peritoneal lavage was performed with 1 ml of cold sterile PBS wash. Serum and cell-free peritoneal lavage fluid were collected for cytokine protein analyses. To determine bacterial loads after CLP surgery, peritoneal lavage fluid, EDTA-treated blood and lung homogenate from 24 h post-CLP were placed on ice and serially diluted in sterile PBS. A 10 μl aliquot of each dilution was spread on LB agar plates without antibiotics and incubated at 37° C. overnight. Colonies were counted and expressed as CFU/ml. In some experiments, recombinant human Sema3A-Fc or human IgG1 Fc control protein were injected intraperitoneally at a dosage of 25 μg/kg 1 h prior to CLP procedure.

Example 9 Statistics

Statistical analysis was carried out with Prism 4 for Macintosh. In survival studies a log-rank test was used to test for significance. For all other studies results were presented as the mean±s.d., and unpaired Student's t-test (one tailed) was applied to evaluate significance. P values less than 0.05 were considered statistically significant.

Example 10 Antigen Presenting Assay

Bone marrow-derived dendritic cells (BMDCs) were generated from WT and Plxna4−/− mice as described previously (Eun et al., 2006). BMDCs were incubated with either 1 μg/ml OVA peptide323-339(ISQAVHAAHAEINEAGR (SEQ ID NO:60); New England Peptide) or 50 μg/ml OVA whole protein (Worthington Biochemical Corp.) at 37° C. overnight. Soluble antigen was removed from DC culture by washing cells two times with medium. CD4+ T cells isolated from spleens of OT II mice with Mouse CD4 Subset Column (R&D) were labeled with 2 μM carboxyfluorescein succinimidyl ester (CFSE) and then added to the DC culture. Two or four days later, cells were collected, and T cell proliferation was analyzed by the dilution of CFSE fluorescence detected on a CyAn ADP flow cytometer (DAKO).

Example 11 Sorting and Stimulation of Plasmacytoid DCs

Total bone marrow cells were harvested from the femurs and tibiae of mice. Single cell suspensions were prepared, and RBCs were lysed. Plasmacytoid DCs were sorted to more than 98% purity by FACS by labeling cells with 3 colors: anti-mPDCA1-PE, antiCD11c-APC and anti-B220-pacific blue (all from BD Biosciences). Sorted pDCs (B220+CD11cintmPDCA1+) were stimulated with either TLR7 ligand Imiquimod (R837) or TLR9 ligand B-type CpG oligodeoxynucleotide (ODN1826) in 96-well plates for 16 h. IFN-α protein levels in cell-free supernatant were determined using a mouse IFN-α ELISA kit (PBL InterferonSource).

Example 12 Flow Cytometry Analysis

Spleens were collected from naïve WT and Plxna4−/− mice and dispersed in 0.5% collagenase A (Roche). After lysing RBCs with ammonium chloride buffer, total cell numbers were determined using a hemocytometer. Fc binding was blocked via a 10 min incubation with purified rat anti-mouse CD16/CD32 (FcγIII/II receptor). The cells were stained with the following monoclonal antibodies to identify different mouse cell types: anti-CD11b-APC, anti-F4/80-PE-Cy5, anti-Ly6C-PE, anti-Ly6G-FITC, anti-B220-Pacific Blue, anti-CD11c-PE-Cy7, anti-mPDCA1-PE, anti-NK1.1-FITC, anti-CD3-PE, anti-CD4-PerCP, anti-CD8-Pacific Blue, anti-CD19-PE-Cy7. All antibodies were purchased from eBioscience, except for anti-F4/80 (Serotec). The cells were fixed in 1% paraformaldehyde and kept in the dark at 4° C. until analysis with a CyAn ADP flow cytometer (DAKO).

Example 13 Plexin-A4 is Expressed by Myeloid and Monocytic Cells

Plexin-A4 is highly expressed in the nervous system and functions as an axon guidance factor during neuronal development (Suto et al., 2007; Yaron et al., 2005). To investigate the role of plexin-A4 in the immune system, the Plxna4 mRNA expression level was analyzed in different immune subpopulations. Cells of the lymphoid lineage (T, B and NK cells) showed no detectable expression, while cells of the myeloid lineage including macrophages and conventional DCs expressed relatively high levels of the gene with the exception of plasmacytoid DCs (pDCs), which expressed a low level of the gene (FIG. 1A). The highest level of Plxna4 mRNA was observed in peritoneal macrophages. Plexin-A4 protein was detected on the surface of wild type (WT), but not of Plxna4−/− peritoneal macrophages as determined by immunofluorescence staining and flow cytometry analysis.

Due to the selective expression of plexin-A4 in myeloid cells, the role of plexin-A4 was investigated in functions that are well studied in cells of this lineage. Previous work has shown that plexin-A1 on DCs mediates the activation of T cells (Takegahara et al., 2006; Wong et al., 2003). In contrast, plexin-A4 is not involved in DC antigen presentation, since OVA323-339-specific OTII CD4+ T cell proliferation was comparable when stimulated with either WT or Plxna4−/− bone marrow-derived DCs (BMDCs) that have been loaded with either OVA323-339 peptide or whole OVA protein (FIG. 8). The role of plexin-A4 in type I IFN production by pDCs was also evaluated. WT and Plxna4−/− pDCs isolated from the bone marrow generated comparable amounts of IFN-α in response to stimulation with TLR7 (R837) or TLR9 (B-type CpG oligodeoxynucleotide, CpG-B) agonists. This indicates that plexin-A4 does not play a role in type 1 IFN production by pDCs during TLR stimulation (FIG. 9).

Example 14 Plexin-A4 is Required for TNF-α and IL-6 Production in Macrophages

The role of plexin-A4 in cytokine production in response to TLR activation was examined next. Plxna4−/− and WT mice showed no difference in the percent or absolute number of peritoneal macrophages as determined by the analysis of a cytospin sample (FIGS. 10A-10C). A variety of TLR agonists were used to stimulate naïve peritoneal macrophages, including Pam3Cys (TLR2), polyinosinic-polycytidylic acid (poly(I:C), TLR3), lipopolysaccharide (LPS, TLR4), R837 and CpG-B. During TLR stimulation with these diverse agonists, Plxna4−/− peritoneal macrophages generated significantly lower amount of TNF-α and IL-6 mRNA and protein compared to similarly treated WT controls (FIGS. 1B-C). In contrast, there was no difference in IFN-β production between WT and Plxna4−/− peritoneal macrophages upon TLR activation. In addition to peritoneal macrophages, Plxna4−/− bone marrow-derived macrophages (BMMs) (FIG. 19A) and BMDCs (FIG. 19B) exhibited defective TNF-α and IL-6 production upon TLR stimulation when compared to their WT counterparts. These findings show that plexin-A4 is required for TLR-initiated proinflammatory cytokines but not type 1 IFN production.

The role of macrophage plexin-A4 in response to bacterial challenge was further tested. Both Gram-positive (Listeria monocytogenes, Staphylococcus aureus) and -negative (Escherichia coli, Salmonella typhimurium) bacteria were used to stimulate peritoneal macrophages. In all cases, bacteria-challenged Plxna4−/− macrophages produced significantly reduced levels of TNF-α and IL-6 mRNA and protein, but a similar level of IFN-β when compared to WT macrophages (FIGS. 2A and 2B). These data suggest that plexin-A4 is important in the bacteria-induced, inflammatory cytokine response.

Example 15 Plexin-A4 Mediates the Activation of NF-κB and JNK but not ERK or p38

NF-κB and MAPK signaling pathways control the production of inflammatory cytokines during macrophage activation in response to TLR agonists and microbial pathogens (Vallabhapurapu and Karin, 2009). The defective production of TNF-α and IL-6 by Plxna4−/− macrophages prompted us to investigate the function of plexin-A4 in these signaling pathways. After LPS stimulation, naïve WT peritoneal macrophages exhibited enhanced phosphorylation of p65, JNK, p38 and ERK1/2 MAPKs as expected. However, Plxna4−/− macrophages showed a dramatic defect in the phosphorylation of p65 and JNK, but not ERK1/2 and p38, suggesting a specific regulatory function of plexin-A4 in the activation of NF-κB and JNK kinase (FIG. 3A). Phosphorylation and activation of p65 requires phosphorylation of the inhibitor of κB (IκB). In accordance with defective p65 phosphorylation during LPS stimulation, Plxna4−/− macrophages showed a defect in IκB phosphorylation compared with WT macrophages (FIG. 3A). In contrast, Plxna4−/− macrophages were not defective in response to TNF-α, anti-CD40 and IFN-γ treatment. The phosphorylation of MAPKs upon TNF-α or anti-CD40 treatment (FIG. 20A) and of STAT1 (signal transducer activators of transcription 1) upon IFN-γ treatment (FIG. 20B) was not altered in Plxna4−/− macrophages. This indicates that plexin-A4 specifically mediates TLR activation. Additionally, Plxna4−/− peritoneal macrophages showed no defect in Akt phosphorylation in response to TLR activation by Pam3Cys and LPS stimulation (FIG. 20C). These findings indicate that plexin-A4 deficiency leads to a TLR-specific defect in NF-κB and JNK activation. Furthermore, this defect is not due to decreased TLR expression, since the mRNA levels of Tlr2, 3, 4, 7 or 9 were similar between WT and Plxna4−/− peritoneal macrophages (FIG. 21).

To determine if impaired NF-κB and JNK activation in Plxna4−/− macrophages causes defective binding of the downstream transcription factors, p65 and c-Jun, at the promoter regions of Tnfa and Il6, chromatin immunoprecipitation (ChIP) assays were performed by using specific anti-p65 and anti-phospho-c-Jun antibodies. Appropriate primer sets were designed that flanked the well-defined binding sites of κB and c-Jun at the promoter regions of Tnfa and Il6 (Galvez et al., 2009; Leng et al., 2009). One hour after stimulation with various TLR agonists, WT macrophages showed enhanced binding of p65 and c-Jun at both Tnfa and Il6 promoters, which correlated with the increased expression of these inflammatory genes (FIGS. 3B and 3C). Conversely, Plxna4−/− macrophages showed significantly decreased p65 and c-Jun binding at these promoters compared with similarly treated WT controls. This data suggests that plexin-A4 mediates TLR-induced TNF-α and IL-6 production by inducing the activities of NF-κB and JNK to cause the epigenetic activation of these genes.

Example 16 Plexin-a4 Causes the Activation of the Small GTPase, Rac1, but not RhoA or Cdc42

Several members of the plexin family, such as plexin-B1 (Perrot et al., 2002; Swiercz et al., 2002) and plexin-A1 (Turner et al., 2004), mediate their downstream effects through the activation of Rho family of small GTPases such as RhoA, Rac1 and Cdc42. More importantly, Rac1 has been shown to serve an essential role upstream of NF-κB activation and cytokine production in response to either TLR2 (Arbibe et al., 2000) or TLR4 (Sanlioglu et al., 2001) agonists. To determine if plexin-A4 regulates the activation of small GTPases during TLR activation, a small GTPase pull-down assay was used in which the binding domain of the downstream effector of a particular GTPase was used to pull down the GTP-bound and hence activated form of that GTPase. LPS-stimulated WT macrophages displayed an increase in the GTP-bound form of Rac1 and Cdc42 when compared to unstimulated controls (FIG. 3D). LPS-stimulated Plxna4−/− macrophages exhibited a dramatic reduction in the quantity of GTP-bound Rac1 when compared to WT cells, while GTP-bound Cdc42 and RhoA were indistinguishable. Thus, plexin-A4 affects the activation of Rac1, which is known to promote NF-κB activation and cytokine production in TLR-stimulated macrophages (Arbibe et al., 2000; Sanlioglu et al., 2001). In support of the importance of Rac1 in TLR-stimulated gene expression, a specific Rac1 inhibitor NSC23766 (Gao et al., 2004) was used and found to significantly attenuate TNF-α and IL-6 production in WT peritoneal macrophages stimulated by various TLR agonists (FIG. 3E). However, NSC23766 showed no inhibitory effect on cytokine production by TLR-activated Plxna4−/− macrophages. This is compatible with the conclusion that Rac1 activation is significantly attenuated in Plxna4 macrophages such that a Rac1 inhibitor has no further suppressive effect on these cells. IFN-β production in WT macrophages was not affected by Rac1 inhibition (FIG. 3F). This is consistent with the earlier observation that plexin-A4 does not affect TLR-induced type I IFN production.

Example 17 Plxna4−/− Mice Exhibit Reduced Cytokine Storm and are Protected from Lethal Challenge with LPS and Poly(I:C)

Next, the physiological relevance of these findings was evaluated. The number of immune cells in the Plxna4−/− mice was first determined. No apparent defect was detected for all immune cell types tested, including myeloid DCs (mDCs), pDCs, neutrophils, monocytes, macrophages, CD4+ or CD8+ T cells, B cells, NK cells (FIG. 11). Thus, any physiological difference observed between WT and Plxna4−/− mice is not due to a change in immune subpopulations.

Since Plxna4−/− macrophages showed decreased TNF-α and IL-6 production during TLR stimulation in vitro, the in vivo role of plexin-A4 was examined next. Mice were injected intraperitoneally with either LPS (12.5 mg per kg body weight) or poly(I:C) (20 mg per kg body weight), and the peritoneal lavage fluid was analyzed for inflammatory cytokines by the Multi-plex ELISA assay. Four hours after the injection of LPS or poly(I:C), both localized (FIG. 4B) and systemic (FIG. 4C) levels of inflammatory cytokines, including TNF-α, IL-1β, IL-6, IL-12p70, CCL2 and CCL3, were attenuated in Plxna4−/− mice compared to WT mice.

The production of a plethora of inflammatory cytokines is frequently detrimental to the animals and referred to as the cytokine storm (FIG. 7F). Thus, morbidity among animals injected with TLR agonists was examined. While WT mice succumbed to high doses of TLR agonists, Plxna4−/− mice were significantly protected from LPS or poly(I:C)-induced lethality (FIG. 4A). These findings indicate that plexin-A4 enhances the in vivo inflammatory responses induced by TLR agonists.

Example 18 Plxna4−/− Mice are Resistant to Septic Inflammation Induced by Cecal Ligation and Puncture (CLP)

To further evaluate the role of plexin-A4 during in vivo bacterial infection, a well-defined CLP-induced polymicrobial peritonitis model was utilized, which closely replicates the nature and course of clinical sepsis (Hubbard et al., 2005). Acute sepsis results in a high mortality rate, which is primarily due to the overzealous production of proinflammatory cytokines. Given that the number of cecal punctures may determine the severity of sepsis, both moderate sepsis (two punctures) and severe sepsis (nine puncture) models were employed. WT mice with two or nine punctures respectively showed 80% and 100% mortality rate as early as 3 days after CLP (FIG. 5A). However, Plxna4−/− mice were significantly protected from CLP-induced lethality in both moderate sepsis (p=0.0285) and severe sepsis (p=0.0012). Hence the absence of plexin-A4 provided a clear survival benefit. Inflammatory cytokine levels were also examined in the peritoneal lavage, serum, and lung homogenate before and after CLP-induced peritonitis. At 4 h and 24 h after CLP, Plxna4−/− mice produced significantly reduced levels of inflammatory cytokines such as TNF-α, IL-1β, IL-6, IL-12p70, CCL2 and CCL3 in the peritoneal lavage fluid (FIG. 5B), serum (FIG. 5C) and lung homogenate (FIG. 5D) compared to WT mice. Therefore, plexin-A4 enhances the septic inflammatory response and promotes a cytokine storm in an experimental peritonitis model.

Example 19 Plexin-A4 Shows No Role in Macrophage Phagocytosis and Bacteria Killing

Another essential factor that contributes to host survival during acute sepsis is efficient phagocytosis and killing of invading bacteria (Matsukawa et al., 2000). The dramatic survival benefit in Plxna44″ mice prompted an investigation of the function of plexin-A4 in macrophage phagocytosis and bacteria killing. Twenty-four hours after CLP surgery, there were similar levels of bacterial loads in the peritoneal lavage, blood and lung homogenate from WT and Plxna4−/− mice, indicating that plexin-A4 is not required for bacterial clearance (FIG. 6A). WT and Plxna4−/− peritoneal macrophages did not differ in the in vitro phagocytosis of GFP-expressing E. coli following different points of incubation (30, 60 or 120 min) (FIG. 6B and FIG. 6C) or infection with different MOIs (10 or 100) (FIG. 6D). Furthermore, there was no difference in the bacterial killing capacity of WT and Plxna4−/− peritoneal macrophages, because similar numbers of E. coli were recovered from macrophages of both genotypes after the extracellular bacteria were killed by antibiotics and the E. coli-loaded macrophages were incubated for different time periods (FIG. 6E). Taken together, these data suggest that plexin-A4 does not play a role in bacterial phagocytosis and killing in macrophages.

Example 20 Sema3A Enhances LPS-Induced Cytokine Production in a Plexin-A4-Dependent Manner

Previous studies in the CNS have identified Sema3A and Sema6A as the ligands for plexin-A4 (Suto et al., 2007; Yaron et al., 2005). Sema3a and Sema6a mRNA expression levels were first analyzed in immune subpopulations (FIG. 22). The highest level of Sema3a mRNA was observed in cells of the lymphoid lineage such as T, B and NK cells. Myeloid and monocytic cells also express Sema3a, but at a lower level. Sema6a mRNA is expressed at a relatively low level in all immune subpopulations that we analyzed. The level of Sema6a was nearly 100× lower than that of Sema3a when normalized to the Actb signal.

To determine if soluble Sema3A or Sema6A also functions in the immune system as a ligand for plexin-A4, WT or Plxna4−/− peritoneal macrophages were stimulated with increasing concentrations of Sema3A or Sema6A in the absence or presence of LPS. These ligands were expressed as chimeric proteins consisting of the human IgG1 Fc fragment fused to either Sema3A or Sema6A. The IgG1 Fc fragment alone was used as a negative control. Neither Sema3A nor Sema6A alone induced TNF-α and IL-6 production in macrophages. In the presence of a suboptimal concentration of LPS (50 ng/ml), Sema3A, but not Sema6A or IgG Fc control, enhanced LPS-induced TNF-α and IL-6 production in WT peritoneal macrophages in a dose-dependent manner (FIG. 7A and FIG. 7B). However, this enhancement in LPS-responsiveness was completely abolished in Plxna4−/− macrophages (FIG. 7A and FIG. 7B). The absence of a functional effect of Sema6A agrees with its low expression in different immune populations shown earlier.

The data presented suggest that LPS via TLR and Sema3A via plexin-A4 might synergistically induce macrophage activation. To assess a converging point for TLR and plexin-A4, we assayed for GTP-bound Rac1. WT macrophages stimulated by LPS or Sema3A resulted in Rac1 activation, while the two stimuli together enhanced this activation. However, Rac1 activation was not observed upon stimulation by either LPS or Sema3A in Plxna4−/− macrophages, suggesting that plexin-A4 is required for signals that emanate from the binding of TLR agonists and Sema3A (FIG. 7C). Similarly, neither Sema3A nor LPS activated Rac1 in Myd88−/− macrophages. This finding suggests that MyD88 lies downstream of the Sema3A/plexin-A4 axis. These findings further suggest that Sema3A and LPS both require intact MyD88 signaling to synergistically promote Rac1 activation.

We next assessed if Sema3A engagement of plexin-A4 could synergistically exacerbate CLP-induced septic inflammation. A single peritoneal injection of Sema3A or control IgG Fc at a dosage of 25 μg/kg body weight was administrated to WT and Plxna4 mice 1 h prior to CLP procedure. Sema3A, but not IgG control, significantly exacerbated CLP-induced lethality in WT mice (FIG. 7D). In contrast, Sema3A did not affect the survival rate of Plxna4−/− mice in the same experiment. This indicates that the observed biologic effect of Sema3A is dependent on plexin-A4. To further determine if decreased survival observed in Sema3A-pretreated WT mice was associated with dysregulated cytokine production, cytokine production in the peritoneal lavage was measured 4 h after CLP. Indeed, Sema3A-pretreated mice had significantly higher levels of inflammatory cytokines than IgG Fc-treated controls (FIG. 7E).

The above experiments were performed in cell culture with highly enriched macrophage preparations, thus it was important to assess the physiologic relevance of these findings. To address this, we assessed if Sema3A engagement of plexin-A4 could synergistically exacerbate CLP-induced septic inflammation. A single peritoneal injection of Sema3A or control IgG Fc at a dosage of 25 μg/kg body weight was administrated to WT and Plxna4−/− mice 1 h prior to CLP procedure. We measured cytokine production in the peritoneal lavage 4 h after CLP. Sema3A-pretreated mice had significantly higher levels of inflammatory cytokines than IgG Fc-treated controls in WT mice, but not in Plxna4−/− mice (FIG. 8G). Collectively, these in vitro and in vivo results suggest that plexin-A4 binding by Sema3A synergizes with TLR engagement by its agonist(s) to amplify innate inflammatory responses. Strategies to reduce Sema3A engagement of plexin-A4 should be beneficial in controlling an adverse inflammatory response associated with endotoxin shock or sepsis.

Example 21 Preparation of a Fusion Protein Between Human Plexin-A4 Extracellular Fragment and Human IgG1 Fc

Total RNA was isolated from human myeloid leukemia cell line KG1 cells, which expresses a relatively high PLXNA4 mRNA level (FIG. 17A). Reverse transcription was performed to yield complementary DNA. Human plexin-A4 extracellular fragment (hPLXNA4EF) (bold in FIG. 12A) was cloned using a primer set that contained HindIII (forward primer) digestion site or BamHI (reverse primer) digestion site: forward, 5′-AAGCTTGCCACCCATGAAAGCCATGCCCTGGAACT (SEQ ID NO:24); reverse, 5′-GGATCCGTCCGGGGCAATGTACACCATC (SEQ ID NO:25). Using these enzyme digestion sites, human plexin-A4 extracellular fragment was subcloned into pCDNA3 vector containing human IgG1 Fc fragment (hIgG Fc) (bold in FIG. 17B). Then human plexin-A4 extracellular fragment is upstream of human IgG1 Fc with a linker of two amino acids between them. Human PLXNA4 EF-hIgG Fc fragment was further subcloned into pCEP4 vector to generate fusion protein between human plexin-A4 extracellular fragment and human IgG1 Fc (FIG. 15A; FIG. 17C).

Example 22 Primers Amplifying Primers:

Full-Length hPLXNA4:

Forward (Hind3): (SEQ ID NO: 24) AAGCTTGCCACCCATGAAAGCCATGCCCTGGAACT Reverse (BamHI): (SEQ ID NO: 26) GGATCCAAGGACGGTTCTCAGCTGTCTA.

hPLXNA4 Extracellular Fragment:

Forward (Hind3): (SEQ ID NO: 24) AAGCTTGCCACCCATGAAAGCCATGCCCTGGAACT Reverse (BamHI): (SEQ ID NO: 25) GGATCCGTCCGGGGCAATGTACACCATC

Sequencing Primers:

(SEQ ID NO: 27)  1. GATTGCCTGTGGGAGCCTGTA (SEQ ID NO: 28)  2. TATGTAGAGGTGCCCATTGGCT (SEQ ID NO: 29)  3. TATGAGACGGTGCAGGTGGT (SEQ ID NO: 30)  4. TTCGCCAGCACCAGCTTT (SEQ ID NO: 31)  5. TGACCCAGACTTCGCATGT (SEQ ID NO: 32)  6. AACCTGAATGCCGGAAGCAA (SEQ ID NO: 33)  7. GTGTTTGAGGCCTTTGGTCC (SEQ ID NO: 34)  8. GGATTCCGTTCCTGGACTATA (SEQ ID NO: 35)  9. TTGAGCGAGGACAAGCTCAT (SEQ ID NO: 36) 10. TGACCTGGAGAGTGGAGTCAA Forward-Primer 19 SEQ ID NO: 37 GCCTCTGCCATGAAAGCCA Reverse-Primer 23 SEQ ID NO: 38 AAGGACGGTTCTCAGCTGTCTAA

TABLE 1 Sequences of RT-PCR primers Mouse genes Forward Reverse Sema3a GCCTGCAGAAGAAGGATTCA TCAGGTTGGGGTGGTTAATG (SEQ ID NOs: 39, 40) Sema6a AATGGCCAGATGCCCT CCGAGTAGAGTTTTCCATTGCA TATG (SEQ ID NOs: 41, 42) Tlr2 CTCCTGAAGCTGTTGCGTTAC TACTTTACCCAGCTCGCTC ACTAC (SEQ ID NOs: 43, 44) Tlr3 TCTTCTTTACGAAA TTGCCAATTGTCTGGAAACACC GTTGGACTTGTC (SEQ ID NOs: 45, 46) Tlr4 ATGGCATGGCTTACACCACC GAGGCCAATTTTGTCTCCACA (SEQ ID NOs: 47, 48) Tlr7 CTGGAGTTCAGAGGCAAC GTTATCACCGGCTCTCCATAGAA CATT (SEQ ID NOs: 49, 50) Tlr9 AGCTGAACATGAACGGCATCT TGAGCGTGTACTTGTTGAGCG (SEQ ID NOs: 51, 52) Ifnb1 ATGAGTGGTGGTT TGACCTTTCAAATGCAGTAGA GCAGGC TTCA (SEQ ID NOs: 53, 54)

Example 23 Preparation of a Fusion Protein Between Human Semaphorin 3A and Human IgG1 Fc

An expression vector pCR-Blunt-TOPO containing full-length human SEMA3A was purchased from Mammalian Gene Collection (MGC). SEMA3A sequence was amplified by a primer set that contained HindIII (forward primer) digestion site or BamHI (reverse primer) digestion site: forward, 5′-AAGCTTGCCACCCATGGGCTGGTTAACTAGGATTGT (SEQ ID NO:55); reverse, 5′-GGATCCGACACTCCTGGGTGCCCTCTCA (SEQ ID NO:56). Using these enzyme digestion sites, human SEMA3A was subcloned into pCDNA3 vector containing human IgG1 Fc fragment (hIgG Fc) (bold portion in FIG. 17B). Then human SEMA3A is upstream of human IgG1 Fc with a linker of two amino acids between them. Human SEMA3A-hIgG Fc fragment was further subcloned into pCEP4 vector to generate fusion protein between human SEMA3A and human IgG1 Fc (FIG. 16A).

Sequencing Primers:

(SEQ ID NO: 57) 1 TGGGAAGAGTCCATATGACCCT (SEQ ID NO: 58) 2 ATTTTCAAGGGATCAGCCGT (SEQ ID NO: 59) 3 CTATATATTGGTTCAACGGC

Example 24 Preparation and Testing of Monoclonal Antibodies to Plexin-A4

Monoclonal antibodies were prepared against plexin-A4 using standard protocols (See, e.g., Monoclonal Antibodies. Shepherd and Dean, eds. New York: Oxford University Press, 2000; Monoclonal Antibodies: Preparation and Use of Monoclonal Antibodies and Engineered Antibody Derivatives. Heddy Zola, ed. Oxford: BIOS Scientific Publishers Limited, 2000). Western blot analysis was used to assay sixteen of the monoclonal antibodies for the ability to bind to plexin-A4. Briefly, expression vector encoding full-length human PLXNA4 was transfected into 293T cell line by using FuGENE® 6 Transfection Reagent (Roche). Sixteen hours later, cell were collected and lysed with buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 2 mM EDTA, % Triton X-100, 0.1% SDS, and protease inhibitor cocktail). Proteins were separated on a NuPAGE gel (Invitrogen) and were transferred onto nitrocellulose membranes (Bio-Rad Laboratories). The membranes were blocked with 10% milk proteins in 1×PBS and 0.1% Tween-20 and then probed with each of sixteen anti-human plexin-A4 antibodies. HRP-conjugated anti-rat secondary antibodies were used and overexpressed human plexin-A4 were detected using an Enhanced Chemiluminescent (ECL) reagent (Thermo Scientific).

Example 25 Functional Testing of Monoclonal Anti-Plexin-A4 Antibodies in Macrophages

We have identified plexin-A4 as an essential cell surface molecule involved in macrophage activation and cytokine production. Therefore, we plan to test the efficacy of monoclonal anti-plexin-A4 antibody in inhibiting macrophage cytokine production such as TNF and IL-6. Briefly, resident peritoneal macrophages are isolated and stimulated with a series of TLR agonists in the absence or presence of each of sixteen anti-plexin-A4 antibodies. The supernatants are collected and TNF and IL-6 will be measured by ELISA. Furthermore, the efficacy of anti-plexin-A4 in the treatment of experimental sepsis is tested by using polymicrobial peritonitis animal model, which has been described above.

Example 26

Prior to the present study, none of the plexin molecules had been shown to regulate TLR signaling. The present invention shows the novel intersection of the plexin-A4 and TLR pathways through the intracellular activation of Rac1, JNK and NF-κB (FIG. 7F). This intersection results in epigenetic changes at the promoters of proinflammatory genes, followed by a pan-cytokine inflammatory response in response to TLR stimulation and bacterial challenge. It cumulates in a cytokine storm that precipitates endotoxin shock induced by TLR agonists and sepsis induced by bacterial infection. The data presented herein show that the plexin-A4 ligand, Sema3A, together with endotoxin can synergistically induce cytokine production in a plexin-A4-dependent manner. Collectively, these data reveal a pivotal role of plexin-A4 in TLR-induced macrophage cytokine production through activation of Rac-1, NF-κB and JNK.

Whereas it is generally believed that TLR engagement by TLR agonists is able to fully activate immune cells, the present study identifies another requirement during this process involving the engagement of membrane plexin-A4 by Sema3A. Without wishing to be bound by any particular theory of the invention, it appears that TLR and plexin-A4 constitute components of a “two-signal” model for TLR activation. Signal 1 is initiated upon the binding of TLRs with their corresponding ligands and transduced sequentially through MyD88, IRAK, TRAF6, TAK1 kinase complex, IKK and MAPK. Meanwhile, signal 2 is initiated by plexin-A4 binding to its soluble ligand, Sema3A, which triggers Rac1, leading to the activation of NF-κB and JNK. In the absence of TLR stimulation, plexin-A4 activation by Sema3A failed to induce cytokine production, indicating that plexin-A4-mediated signal 2 is not sufficient for the induction of cytokine production. In the absence of plexin-A4, TLR signaling is significantly attenuated but not abolished, indicating that TLR signaling is amplified by plexin-A4 activation but is not initiated by this activation. The data presented herein show that in macrophages lacking MyD88, which is the adaptor for all TLRs except for TLR3, Rac1 activation is attenuated in response to Sema3A stimulation. This indicates that Sema3A/plexin-A4 signaling also requires MyD88. Whether this is achieved via direct binding of plexin-A4 and MyD88 or an indirect route remains to be elucidated. In contrast to its role in the promotion of proinflammatory cytokines, plexin-A4 is not required for TLR-induced TRIF-dependent type 1 IFN production, suggesting that plexin-A4 does not affect TRIF signaling.

The findings of the present invention show that while plexin-A4 is most highly expressed by monocytic and myeloid cell lineages and significantly reduced on lymphoid cells, Sema3A is highly expressed by lymphoid cells with reduced expression by myeloid/monocytic cell types. Thus it is possible that during an early inflammatory response, the smaller amount of Sema3A produced by myeloid and monocytic cells together with TLR ligands is sufficient to activate the higher density of plexin-A4 on macrophage membrane. Later when T cells are called in, the higher amounts of Sema3A expressed by activated T cells then serve as a feed-back-loop to downregulate T cells, which express less plexin-A4 receptors.

The biologic function of plexin-A4 in the activation of a pan-cytokine response has broad clinical implications. New anti-inflammatory therapeutic methods are needed in the treatment of immune-related and/or inflammatory diseases. In addition, pathogen-induced sepsis represents a major burden to the US health care system and has become increasingly significant over the past decades (Martin et al., 2003). The hallmark of sepsis in the acute phase is an exacerbated production of proinflammatory cytokines and chemokines, leading to the cytokine storm. While these inflammatory mediators are essential in providing an immediate host defense, their overzealous production can be deleterious to the host if left uncontrolled. The present invention suggests a role for plexin-A4 in mediating the production of proinflammatory cytokines, which affects host survival in models of sepsis. Therefore, the interaction of plexin-A4 and Sema3A presents a new therapeutic target for anti-inflammatory and anti-sepsis treatment.

REFERENCES

  • Akira, S., and Takeda, K. (2004). Toll-like receptor signalling. Nat Rev Immunol 4, 499-511.
  • Akira, S., Uematsu, S., and Takeuchi, O. (2006). Pathogen recognition and innate immunity. Cell 124, 783-801.
  • Arbibe, L., Mira, J. P., Teusch, N., Kline, L., Guha, M., Mackman, N., Godowski, P. J., Ulevitch, R. J., and Knaus, U. G. (2000). Toll-like receptor 2-mediated NF-kappa B activation requires a Rac1-dependent pathway. Nat Immunol 1, 533-540.
  • Carvalho, F. A., Barnich, N., Sauvanet, P., Darcha, C., Gelot, A., and Darfeuille-Michaud, A. (2008). Crohn's disease-associated Escherichia coli LF82 aggravates colitis in injured mouse colon via signaling by flagellin. Inflammatory Bowel Diseases 14, 1051-1060.
  • Choi, Y. I., Duke-Cohan, J. S., Ahmed, W. B., Handley, M. A., Mann, F., Epstein, J. A., Clayton, L. K., and Reinherz, E. L. (2008). PlexinD1 glycoprotein controls migration of positively selected thymocytes into the medulla. Immunity 29, 888-898.
  • Couzin, J. MicroRNAs Make Big Impression in Disease After Disease. Science 319, 1782-1784 (2008).
  • Eun, S. Y., O'Connor, B. P., Wong, A. W., van Deventer, H. W., Taxman, D. J., Reed, W., Li, P., Blum, J. S., McKinnon, K. P., and Ting, J. P. (2006). Cutting edge: rho activation and actin polarization are dependent on plexin-A1 in dendritic cells. J Immunol 177, 4271-4275.
  • Forster, A. C. and R. H. Symons. Self-cleavage of plus and minus RNAs of a virusoid and a structural model for the active sites. Cell 49, 211-220 (1987).
  • Galvez, A. S., Duran, A., Linares, J. F., Pathrose, P., Castilla, E. A., Abu-Baker, S., Leitges, M., Diaz-Meco, M. T., and Moscat, J. (2009). Protein kinase Czeta represses the interleukin-6 promoter and impairs tumorigenesis in vivo. Mol Cell Biol 29, 104-115.
  • Gao, Y., Dickerson, J. B., Guo, F., Zheng, J., and Zheng, Y. (2004). Rational design and characterization of a Rac GTPase-specific small molecule inhibitor. Proc Natl Acad Sci USA 101, 7618-7623.
  • Gerlach, W. L. et al. Construction of a plant disease resistance gene from the satellite RNA of tobacco ringspot virus. Nature 328, 802-805 (1987).
  • Gu, C., Rodriguez, E. R., Reimert, D. V., Shu, T., Fritzsch, B., Richards, L. J., Kolodkin, A. L., and Ginty, D. D. (2003a). Neuropilin-1 conveys semaphorin and VEGF signaling during neural and cardiovascular development. Developmental Cell 5, 45-57.
  • Hubbard, W. J., Choudhry, M., Schwacha, M. G., Kerby, J. D., Rue, L. W., 3rd, Bland, K. I., and Chaudry, I. H. (2005). Cecal ligation and puncture. Shock 24 Suppl 1, 52-57.
  • Iwasaki, A., and Medzhitov, R. (2004). Toll-like receptor control of the adaptive immune responses. Nat Immunol 5, 987-995.
  • Joyce, G. F. RNA evolution and the origins of life. Nature 338. 217-224 (1989).
  • Kawai, T., and Akira, S. (2006). Innate immune recognition of viral infection. Nat Immunol 7, 131-137.
  • Kikutani, H., and Kumanogoh, A. (2003). Semaphorins in interactions between T cells and antigen-presenting cells. Nat Rev Immunol 3, 159-167.
  • Kim, S.-H. and T. R. Cech. Three-dimensional model of the active site of the self-splicing rRNA precursor of Tetrahymena. Proc Natl Acad Sci USA 84, 8788-8792 (1987).
  • Kruger, R. P., Aurandt, J., and Guan, K. L. (2005). Semaphorins command cells to move. Nat Rev Mol Cell Biol 6, 789-800.
  • Labandeira-Rey, M., Couzon, F., Boisset, S., Brown, E. L., Bes, M., Benito, Y., Barbu, E. M., Vazquez, V., Hook, M., Etienne, J., et al. (2007). Staphylococcus aureus Panton-Valentine leukocidin causes necrotizing pneumonia. Science 315, 1130-1133.
  • Leng, J., Butcher, B. A., Egan, C. E., Abdallah, D. S., and Denkers, E. Y. (2009). Toxoplasma gondii prevents chromatin remodeling initiated by TLR-triggered macrophage activation. J Immunol 182, 489-497.
  • Lich, J. D., Williams, K. L., Moore, C. B., Arthur, J. C., Davis, B. K., Taxman, D. J., and Ting, J. P. (2007). Monarch-1 suppresses non-canonical NF-kappaB activation and p52-dependent chemokine expression in monocytes. J Immunol 178, 1256-1260.
  • Martin, G. S., Mannino, D. M., Eaton, S., and Moss, M. (2003). The epidemiology of sepsis in the United States from 1979 through 2000. N Engl J Med 348, 1546-1554.
  • Matsukawa, A., Hogaboam, C. M., Lukacs, N. W., Lincoln, P. M., Evanoff, H. L., and Kunkel, S. L. (2000). Pivotal role of the CC chemokine, macrophage-derived chemokine, in the innate immune response. J Immunol 164, 5362-5368.
  • Michel F. and E. Westhof. Modelling of the three-dimensional architecture of group I catalytic introns based on comparative sequence analysis. J Biol Chem 216, 585-610 (1990).
  • Neufeld, G., and Kessler, O. (2008). The semaphorins: versatile regulators of tumour progression and tumour angiogenesis. Nature Reviews 8, 632-645.
  • Noren, N. K., Liu, B. P., Burridge, K., and Kreft, B. (2000). p120 catenin regulates the actin cytoskeleton via Rho family GTPases. J Cell Biol 150, 567-580.
  • Perrot, V., Vazquez-Prado, J., and Gutkind, J. S. (2002). Plexin B regulates Rho through the guanine nucleotide exchange factors leukemia-associated Rho GEF (LARG) and PDZ-RhoGEF. J Biol Chem 277, 43115-43120.
  • Reinhold-Hurek, B. and D. A. Shub. Self-splicing introns in tRNA genes of widely divergent bacteria. Nature 357, 173-176 (1992).
  • Sanlioglu, S., Williams, C. M., Samavati, L., Butler, N. S., Wang, G., McCray, P. B., Jr., Ritchie, T. C., Hunninghake, G. W., Zandi, E., and Engelhardt, J. F. (2001). Lipopolysaccharide induces Rac1-dependent reactive oxygen species formation and coordinates tumor necrosis factor-alpha secretion through IKK regulation of NF-kappa B. J Biol Chem 276, 30188-30198.
  • Sarver, N. et al. Ribozymes as potential anti-HIV-1 therapeutic agents. Science 247, 1222-1225 (1990).
  • Scanlon, K. J. et al. Ribozyme-mediated cleavage of c-fos mRNA reduces gene expression of DNA synthesis enzymes and metallothionein. Proc Natl Acad Sci USA 88, 10591-10595 (1991).
  • Serini, G., Valdembri, D., Zanivan, S., Morterra, G., Burkhardt, C., Caccavari, F., Zammataro, L., Primo, L., Tamagnone, L., Logan, M., et al. (2003). Class 3 semaphorins control vascular morphogenesis by inhibiting integrin function. Nature 424, 391-397.
  • Sierra, J. R., Corso, S., Caione, L., Cepero, V., Conrotto, P., Cignetti, A., Piacibello, W., Kumanogoh, A., Kikutani, H., Comoglio, P. M., et al. (2008). Tumor angiogenesis and progression are enhanced by Sema4D produced by tumor-associated macrophages. J Exp Med 205, 1673-1685.
  • Sioud, M. et al. Preformed Ribozyme Destroys Tumor Necrosis Factor mRNA in Human Cells. J Mol Biol 223, 831-835 (1992).
  • Suto, F., Tsuboi, M., Kamiya, H., Mizuno, H., Kiyama, Y., Komai, S., Shimizu, M., Sanbo, M., Yagi, T., Hiromi, Y., et al. (2007). Interactions between plexin-A2, plexin-A4, and semaphorin 6A control lamina-restricted projection of hippocampal mossy fibers. Neuron 53, 535-547.
  • Suzuki, K., Kumanogoh, A., and Kikutani, H. (2008). Semaphorins and their receptors in immune cell interactions. Nat Immunol 9, 17-23.
  • Swiercz, J. M., Kuner, R., Behrens, J., and Offermanns, S. (2002). Plexin-B1 directly interacts with PDZ-RhoGEF/LARG to regulate RhoA and growth cone morphology. Neuron 35, 51-63.
  • Takegahara, N., Takamatsu, H., Toyofuku, T., Tsujimura, T., Okuno, T., Yukawa, K., Mizui, M., Yamamoto, M., Prasad, D. V., Suzuki, K., et al. (2006). Plexin-A1 and its interaction with DAP12 in immune responses and bone homeostasis. Nat Cell Biol 8, 615-622.
  • Toyofuku, T., Zhang, H., Kumanogoh, A., Takegahara, N., Yabuki, M., Harada, K., Hori, M., and Kikutani, H. (2004). Guidance of myocardial patterning in cardiac development by Sema6D reverse signalling. Nat Cell Biol 6, 1204-1211.
  • Tran, T. S., Kolodkin, A. L., and Bharadwaj, R. (2007). Semaphorin regulation of cellular morphology. Annu Rev Cell Developmental Biol 23, 263-292.
  • Turner, L. J., Nicholls, S., and Hall, A. (2004). The activity of the plexin-A1 receptor is regulated by Rac. J Biol Chem 279, 33199-33205.
  • Vallabhapurapu, S., and Karin, M. (2009). Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol 27, 693-733.
  • Waimey, K. E., Huang, P. H., Chen, M., and Cheng, H. J. (2008). Plexin-A3 and plexin-A4 restrict the migration of sympathetic neurons but not their neural crest precursors. Developmental Biology 315, 448-458.
  • van Deventer, H. W., Serody, J. S., McKinnon, K. P., Clements, C., Brickey, W. J., and Ting, J. P. (2002). Transfection of macrophage inflammatory protein 1 alpha into B16 F10 melanoma cells inhibits growth of pulmonary metastases but not subcutaneous tumors. J Immunol 169, 1634-1639.
  • Walzer, T., Galibert, L., and De Smedt, T. (2005). Dendritic cell function in mice lacking Plexin C1. Int Immunol 17, 943-950.
  • Wen, H., Dou, Y., Hogaboam, C. M., and Kunkel, S. L. (2008). Epigenetic regulation of dendritic cell-derived interleukin-12 facilitates immunosuppression after a severe innate immune response. Blood 111, 1797-1804.
  • Wong, A. W., Brickey, W. J., Taxman, D. J., van Deventer, H. W., Reed, W., Gao, J. X., Zheng, P., Liu, Y., Li, P., Blum, J. S., et al. (2003). CIITA-regulated plexin-A1 affects T-cell-dendritic cell interactions. Nat Immunol 4, 891-898.
  • Yamamoto, M., Suzuki, K., Okuno, T., Ogata, T., Takegahara, N., Takamatsu, H., Mizui, M., Taniguchi, M., Chedotal, A., Suto, F., et al. (2008). Plexin-A4 negatively regulates T lymphocyte responses. Int Immunol 20, 413-420.
  • Yaron, A., Huang, P. H., Cheng, H. J., and Tessier-Lavigne, M. (2005). Differential requirement for Plexin-A3 and -A4 in mediating responses of sensory and sympathetic neurons to distinct class 3 Semaphorins. Neuron 45, 513-523.

Claims

1. A method of treating an immune-related and/or inflammatory disorder in a subject, comprising administering to the subject an effective amount of an inhibitor of plexin-A4 activity, whereby the plexin-A4 activity in said subject is reduced, thereby treating the immune-related disorder and/or inflammatory disorder.

2. A method of reducing cytokine production in a subject, comprising administering to the subject an effective amount of an inhibitor of plexin-A4 activity, thereby reducing the cytokine production the subject.

3. The method of claim 1, wherein the inhibitor of plexin-A4 activity is selected from the group consisting of a plexin-A4 fusion protein, a plexin-A4 antibody or an active fragment thereof, or any combination thereof.

4. The method of claim 3, wherein the plexin-A4 antibody or active fragment thereof is a monoclonal antibody or is derived from a monoclonal antibody.

5. The method of claim 3, wherein the plexin-A4 fusion protein comprises the extracellular domain of plexin-A4 and the Fc region of IgG.

6. The method of claim 5, wherein the extracellular domain of plexin-A4 and the IgG1 Fc region is human.

7. The method of claim 5, wherein the extracellular domain of plexin-A4 comprises the amino acid sequence of SEQ ID NO:7 and the IgG Fc region comprises the amino acid sequence of SEQ ID NO:8.

8. The method of claim 3, wherein the plexin-A4 fusion protein comprises the amino acid sequence of SEQ ID NO:6.

9. The method of claim 1, wherein the immune-related and/or inflammatory disorder is selected from the group consisting of sepsis, arthritis, hepatitis, systemic lupus erythematosus (SLE), multiple sclerosis, Guillain-Barre syndrome, Alzheimer's disease, colitis, psoriasis, contact hypersensitivity, retinitis, uveitis, malignancies, systemic lupus erythematosis, asthma, myocarditis, hepatitis, kidney diseases, diabetes, obesity, cardiovascular diseases, and inflammatory bowel disease, or any combination thereof.

10. The method of claim 3, wherein the inhibitor is administered subcutaneously, intramuscularly, intraperitoneally, topically, intravenously, and any combination thereof.

11. (canceled)

12. A method of treating an immune-related and/or inflammatory disorder in a subject, comprising administering to the subject an effective amount of an inhibitor of semaphorin-3A (Sema3A) activity, whereby the Sema3A activity in said subject is reduced, thereby treating the immune-related disorder and/or inflammatory disorder.

13. A method of reducing cytokine production in a subject, comprising administering to the subject an effective amount of an inhibitor of Sema3A activity, thereby reducing the cytokine production the subject.

14. The method of claim 12, wherein the inhibitor of Sema3A activity is selected from the group consisting of a Sema3A fusion protein, a Sema3A antibody or an active fragment thereof, or any combination thereof.

15. The method of claim 14, wherein the plexin-A4 antibody or active fragment thereof is a monoclonal antibody or is derived from a monoclonal antibody.

16. The method of claim 14, wherein the Sema3A fusion protein comprises the amino acid sequence encoding Sema3A and the Fe region of IgG.

17. The method of claim 16, wherein the amino acid sequence encoding Sema3A and the IgG1 Fc region is human.

18. The method of claim 16, wherein the amino acid sequence encoding Sema3A comprises the amino acid sequence of SEQ ID NO:13 and the IgG Fe region comprises the amino acid sequence of SEQ ID NO:8.

19. The method of claim 14, wherein the Sema3A fusion protein comprises the amino acid sequence of SEQ ID NO:16.

20. The method of claim 12, wherein the immune-related and/or inflammatory disorder is selected from the group consisting of sepsis, arthritis, hepatitis, systemic lupus erythematosus (SLE), multiple sclerosis, Guillain-Barre syndrome, Alzheimer's disease, colitis, psoriasis, contact hypersensitivity, retinitis, uveitis, malignancies, systemic lupus erythematosis, asthma, myocarditis, hepatitis, kidney diseases, diabetes, obesity, cardiovascular diseases, and inflammatory bowel disease, or any combination thereof.

21. The method of claim 14, wherein the inhibitor is administered subcutaneously, intramuscularly, intraperitoneally, topically, intravenously, and any combination thereof.

22. (canceled)

Patent History
Publication number: 20150158939
Type: Application
Filed: Oct 3, 2014
Publication Date: Jun 11, 2015
Inventors: JENNY P.-Y. TING (CHAPEL HILL, NC), HAITAO WEN (CARRBORO, NC)
Application Number: 14/506,332
Classifications
International Classification: C07K 16/18 (20060101); C07K 14/47 (20060101);