MXPA05006086A - Methods and compositions relating to gradient exposed cells - Google Patents

Methods and compositions relating to gradient exposed cells

Info

Publication number
MXPA05006086A
MXPA05006086A MXPA/A/2005/006086A MXPA05006086A MXPA05006086A MX PA05006086 A MXPA05006086 A MX PA05006086A MX PA05006086 A MXPA05006086 A MX PA05006086A MX PA05006086 A MXPA05006086 A MX PA05006086A
Authority
MX
Mexico
Prior art keywords
cell
protein
gradient
cells
concentration
Prior art date
Application number
MXPA/A/2005/006086A
Other languages
Spanish (es)
Inventor
Poznansky Mark
Rutishauser Rachel
Original Assignee
Poznansky Mark
Rutishauser Rachel
The General Hospital Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Poznansky Mark, Rutishauser Rachel, The General Hospital Corporation filed Critical Poznansky Mark
Publication of MXPA05006086A publication Critical patent/MXPA05006086A/en

Links

Abstract

The invention relates to methods and compositions for stimulating or inhibiting directed cell migration.

Description

METHODS AND COMPOSITIONS RELATED TO CELLS EXPOSED TO A GRADIENT FIELD OF THE INVENTION The invention is directed to methods and compositions related to the modulation of gene expression in cells in chemotactic and fugactic gradients.
BACKGROUND OF THE INVENTION Cellular movement in response to specific stimulation occurs in prokaryotes and eukaryotes (Doetsch RN and Seymour WF., 1970; Bailey GB et al., 1985). The cellular movement by these organisms has been classified into three types; chemotaxis, which is the cell movement along a gradient toward an increased concentration of an agent (eg, a chemical); negative chemotaxis, which is the cellular movement towards a decreasing concentration of an agent, and chemokinesis, which is the random movement of the cells. The receptors and signal transduction pathways affected by the actions of specific, chemotactically active compounds have been extensively defined in prokaryotic cells. The study of chemotaxis of E. coli has revealed that a chemical that attracts the bacteria in some concentrations and conditions can also act as a repellent to others (ie, a "chemical chemotactic negative" or "chemorepellant") (Tsang N et al. ., 1973, Repaske D. and Adler J. 1981, Tisa LS and Adler J., 1995, Taylor BL and Johnson MS., 1998). Chemotaxis and chemokinesis have been observed to occur in mammalian cells (McCutcheon MW, Wartman W and HM Dixon, 1934; Lotz M and Harris H; 1956; Boyden SV 1962) in response to the class of proteins, called chemokines (Ward SG and Westwick J; 1998; Kim CH et al., 1998; Baggiolini M 1998; Farber JM; 1997). Chemokines induce cell movement by signaling through receptors coupled to the G protein (Wells TN et al., 1998). The G protein-coupled receptors include a wide range of biologically active receptors, such as hormone, viral growth factor and neuroreceptors. The G protein family of coupled receptors includes dopamine receptors, which bind to neuroleptic drugs used to treat psychotic and neurological disorders. Other examples of members of this family include calcitonin, adrenergics, endothelin, cAMP, adenosine, muscarinic, acetylcholine, serotonin, histamine, thrombin, quinine, follicle stimulating hormone, opsins, endothelial differentiation gene-1 receptor, rhodopsins, odorants , cytomegalovirus receptors, etc. The G protein-coupled receptors have been characterized as having seven putative transmembrane domains, designated as the transmembrane domains 1-7 ("TM1", "TM2", "TM3", "TM4", "TM5", "TM6"). "and" TM7"). The domains are believed to represent transmembrane a-helices connected by extracellular or cytoplasmic loops. In each of the first two extracellular loops, the majority of the G protein-coupled receptors have conserved single cysteine residues that form disulfide linkages that are believed to stabilize the functional protein structure. Phosphorylation (as well as lipidation, eg, palmitylation or farnesylation) can affect signal transduction and potential phosphorylation sites fall within the third cytoplasmic loop and / or the carboxy terminus. For various G-protein coupled receptors, such as adrenoreceptor-β, phosphorylation by protein kinase A and / or specific receptor kinases mediates receptor desensitization. Phosphorylation of the cytoplasmic residues of coupled G protein receptors has been identified as an important mechanism for the regulation of G protein coupling. G protein coupled receptors can be coupled intracellularly by heterotrimeric G proteins to several intracellular enzymes, ion and transporters (see, Jonson et al., Endoc Rev, 1989, 10: 317-331). Different α-subunits of G protein preferably stimulate particular effectors to modulate various biological functions in a cell. This signaling path can be blocked, for example, by pertussis toxin (PTX) (Luter AD, 1998, Baggiolini, 1998).
As discussed above, chemokine-induced cellular chemotaxis is mediated through a signal transduction path linked to Ga1. The chemokine, SDF-1a, provides an example of this signaling model. SDF-1a causes the migration of leukocyte subpopulations at sites of inflammation (Aiuti A et al., 1997, Bleul CC et al., 1996, Bleul CC et al., 1996, Oberlin E et al., 1996). In addition, mice designed to be deficient in SDF-1a or its receptor, CXCR-4, have abnormal development of hematopoietic tissues and B cells due to the failure of fetal liver stem cells to migrate to the spinal cord (Friedland JS, 1995, Tan J and Thestrup-Pedersen K, 1995, Corrigan CJ and Kay AB, 1996, Qing M, et al., 1998, Ward SG et al., 1998). This movement is concentration dependent and mediated through the CXCR4 receptor, Gai protein and PI-3 kinase (Nature Medicine 2000; 6.543). The switch of a chemotactic or fugectic response in T cells is associated with intracytoplasmic levels of cyclic nucleotides and a differential sensitivity to tyrosine kinase inhibitors. Methods for the identification of genes involved in the modulation of cell movement across a gradient (eg, genes involved in the relevant Gai-coupled signal transduction pathways) have not been performed. Such methods would be useful for the identification of new therapeutic targets in diseases characterized by aberrant cell movement.
BRIEF DESCRIPTION OF THE INVENTION The invention is theorized, in part, in the discovery that exposure of cells to a gradient results in changes in the gene expression profile of such cells. Furthermore, it has unexpectedly been found that the movement of a cell through a gradient also induces changes in gene expression. In some cases, the gradients exist across the diameter of a cell, so that the most extreme conduction of a cell is exposed to a different concentration which is the trailing end of the cell. Thus, in one aspect, the invention provides a method for identifying a nucleic acid expressed in a concentration dependent manner, comprising determining a first expression profile of nucleic acid from a first cell in a first position in a concentration gradient. of agent, determining a second nucleic acid expression profile of a second cell in a second position in the agent concentration gradient, and determining a difference between the first and second expression profiles of the nucleic acid. The first position in the concentration gradient of the agent corresponds to a first concentration of the agent, and the second position in the concentration gradient of the agent corresponds to a second concentration of the agent. Preferably, the second cell was genetically identical to the first cell before migration through the concentration gradient of the agent. In some embodiments, at least the second cell has migrated through the concentration gradient of the agent. Therefore, the invention provides a method for identifying a nucleic acid expressed in a concentration dependent manner, comprising determining a first expression profile of nucleic acid from a first cell in a first position in a gradient of agent concentration, determining a second nucleic acid expression profile of a second cell that has migrated through the concentration gradient of the agent, and determining a difference between the first and second expression profiles of the nucleic acid. In other embodiments, none of the cells has migrated through the agent concentration gradient, but at least the second cell is presented in a gradient such that the concentration of the agent at one end of the cell is different from the concentration of the agent. at the opposite end of the cell. In a modality, the expression profile of nucleic acid is an expression profile of mRNA. In another embodiment, the mRNA expression profile is determined using PCR, RDA, Northern analysis, subtractive hybridization, or microarray analysis. In one embodiment, the concentration gradient of the agent is a gradient of ligand concentration. In another embodiment, the concentration gradient of agent is a chemokine concentration gradient. In yet another embodiment, the chemokine concentration gradient is selected from the group consisting of SDF-1a, SDF-1β, IP-10, MIG, GROa, GROβ, GRO ?, IL-8, PF4, MCP, MIP-1a , MIP-1ß, MIP-1? (mouse), MCP-2, MCP-3, MCP-4, MCP-5 (mouse), RANTES, fractalkine, lymphotactin, CXC, IL-8, GCP-2, ENA-78, NAP-2, IP-10 , MIG, l-TAC, SDF-1a, BCA-1, PF4, Bolequina, HCC-1, Leucotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin-3 (TSC) ), MDC, TARC, SLC (Exodus-2, 6Ckine), MIP-3a (LARC, Exodus-1), ELC (MIP-3ß), I-309, DC-CK1 (PARC, AMAC-1), TECK, CTAK, MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), C-10 (mouse), Linfotactin C and Fractelquina CX3C (Neurotactin) and ITAC concentration gradients. The concentration gradient of the agent may be a gradient of cytosine concentration. The cytosine concentration gradient can be selected from the group consisting of PAF, N-formylated peptides, C5a, LTB4 and LXA, chemokines: CXC, IL-8, GCP-2, GRO, GROa, GROß, GRO ?, ENA -78, NAP-2, IP-10, MIG, l-TAC, SDF-1a, BCA-1, PF4, Bolequin, MIP-1a, MIP-1β, RANTES, HCC-1, MCP-1, MCP-2 , MCP-3, MCP-4, MCP-5 (mouse), Leucotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2); Eotaxin-3 (TSC), MDC, TARC, SLC (Exodus-2, 6CKina), MIP-3a (LARC, Exodus-1), ELC (MIP-3ß), I-309, DC-CK1 (PARC, AMAC- 1), TECK, CTAK, MPIF1 (MIP-3); MIP-5 (HCC-2), HCC-4 (NCC-4), MIP-l? (mouse), gradients of concentration of C-10 (mouse), lymphotactin C, and Fractelquina CX3C (Neurotactin). Cytosine can be a member of the Cys-X-Cys family of chemokines (for example, the chemokines that bind to the CXCR-4 receptor). Preferred cytokines of the invention include SDF-1a, SDF-1β, met-SDF-1β, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL -10, IL-12, IL-15, IL-18, TNF, IFN-a, IFN-β, IFN-α, a factor that stimulates the granulocyte macrophage colony (GM-CSF), a factor that stimulates the colony of granulocyte (G-CSF), a factor that stimulates the macrophage colony (M-CSF), TGF-β, ligand FLT-3, VEGF, DMDA, endothelin, and CD40 ligand. In one embodiment, the first concentration of the agent is a zero concentration of! agent, and the second concentration of the agent is a concentration that does not equal the agent's zero. In another embodiment, the first concentration of the agent is greater than the second concentration of the agent. In one embodiment, the first cell has migrated through the agent concentration gradient. The migration through the concentration gradient of the agent may be fugactic migration or chemotactic migration. In one embodiment, the gradient is a gradient of stages. In another modality, the gradient is a continuous gradient. In yet another embodiment, the method further comprises a combination gradient, wherein at least one additional gradient coexists with the first gradient. In one embodiment, the first and second cells are adult cells. In preferred embodiments, the first and second cells are human cells. In one embodiment, the first and second cells are primary cells. In another preferred embodiment, the first and second cells are hemopoietic cells, such as, but not limited to, T lymphocytes. In another aspect, the invention provides a method for identifying a compound that can modulate cell migration at one or more concentration gradients. of agent comprising contacting a migrating cell in a gradient of agent concentration with a test compound, determining the expression profile of nucleic acid in the cell and identifying a change in the expression of a gene expression product. The cell movement can be chemotaxis and fugetaxis and therefore, the gene expression product can be a specific genetic product of chemotaxis or fugetaxis. In another aspect, the invention provides a method for inhibiting cellular fugetaxis comprising contacting a cell undergoing or likely experiencing fugetaxis with an agent that inhibits a gene expression product of fugetaxis in an amount effective to inhibit fugetaxis. In one embodiment, the specific genetic production product of fugetaxis is a nucleic acid or a peptide. In another embodiment, the product of specific genetic production of fugetaxis is a signaling molecule. The signaling molecule can be selected from the group consisting of the cell division cycle 42, annexin A3, guanine nucleotide exchange factor Rap1, adenylate cyclase 1, JAK binding protein, and alpha inhibitor Rho GDP dissociation, but not It is so limited. In another embodiment, the signaling molecule is a cell division cycle 42 (cdc42), the ribosomal protein of S6 kinase, the protein 2 associated with BAI1, the GTPase regulator associated with FAK, protein kinase C-beta 1, phospholipase-specific phosphoinositide C-beta 1, nitric oxide synthase 1, phosphatidylinositol-4-phosphate 5-kinase, and MAP kinase kinase kinase kinase 4. In another embodiment, the product of specific gene production of fugetaxis is a molecule related to the extracellular matrix. In a related embodiment, the molecule related to extracellular matrix can be selected from the group consisting of chitinase 3-like 1 (cartilage glycoprotein-39), cell adhesion molecule 6 related to carcinoembryonic antigen, matrix metalloproteinase 8 (neutrophil collagenase), protein 1 associated with cytoplasmic integrin domain, ficolin (containing the fibrinogen domain of collagen) 1, and membrane-associated protein 1 lysosomal, antigen 1 similar to epithelial V, vascular endothelial growth factor (VEGF), fibulin 1, cell adhesion molecule 3 related to carcinoembryonic antigen, but not so limited. In yet another embodiment, the specific genetic production product of fugetaxis is a cytoskeleton-related molecule. The cytoskeleton related molecule can be selected from the group consisting of ankyrin 1 (erythrocytic), calcium binding protein S12 (calgranulin C) plectin 1 (intermediate filament binding protein, 500kD), and ankyrin 2 (neuronal), RPEB3 of protein associated with microtubule, protein similar to 1A protein associated with microtubule (MILP), sealed protein (actin filament, similar to gasoline), but without being so limited. In yet another embodiment, the specific genetic production product of fugetaxis is a cell cycle molecule. The cell cycle molecule can be selected from the group consisting of feline sarcoma viral oncogene 4 v-ki Ardí-Zuckerman, lipocalin 2 (24p3 oncogene), lectin, (galactoside binding, galectin 3), RAB31 (family oncogene of the RAS member), incapacitated (Drosophila) homolog 2 (mitogen response phosphoprotein), RAB9 (oncogene family of the RAS member, psudógeno 1), and growth factor 8, but not so limited. In a further embodiment, the product of specific gene production of fugetaxis is a molecule related to the immune response. The molecule related to immune response can be selected from the group consisting of the highest histocompatibility complex (class II, DR alpha), calcium binding protein A8 S100 (calgranulin A), small subfamily A of inducible cytosine (Cys-Cys), factor 5A initiation of eukaryotic translation, small subfamily B of inducible cytosine (Cys-X-Cys) (member 6, granulocyte chemoattractant protein 2), Fc fragment or IgG binding protein, CD24 antigen (lung carcinoma clustering antigen) small cell), cytochrome P450 (subfamily IVF, polypeptide 3, leukotriene B4 omega hydroxylase), transactivator of MHC class II, T cell receptor (alpha chain), T cell activation (increased late expression), protein tyrosine phosphatase similar to MKP-1, T-cell gamma receptor constant 2, T-cell gamma receptor site, but not so limited. In a further embodiment, the specific gene production product of fugetaxis is chemokine receptor 1 (C-X3-C). In another aspect, the invention provides a method of inhibiting cellular chemotaxis comprising contacting a cell undergoing or likely undergoing chemotaxis with an agent that inhibits a specific gene-production product of chemotaxis in an amount effective to inhibit chemotaxis. In one embodiment, the specific genetic production product of chemotaxis is a nucleic acid or peptide. In another embodiment, the cell is an immune cell. In one embodiment, contact occurs in vivo in a subject who has, or is at risk of having an abnormal immune response. In one modality, the abnormal immune response is an inflammatory response. In another modality, the abnormal immune response is an immune response. The autoimmune response can be selected from the group consisting of rheumatoid arthritis, Crohn's disease, multiple sclerosis, systemic lupus erythematosus (SLE), autoimmune encephalomyelitis, myasthenia gravis (MG), Hashimoto's thyroiditis, Goodpasture's syndrome, penfigus (e.g., pemphigus) vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis, pernicious anemia, idiopathic Addison's disease, autoimmune associated infertility, glomerulonephritis (eg, growing glomerulonephritis, proliferative glomerulonephritis), pemphigoid bolus, Sjögren's syndrome, resistance to insulin, and autoimmune diabetes mellitus, but it is not so limited. In yet another embodiment, the abnormal immune response is a host versus graft response. In one embodiment, the specific gene expression product of chemotaxis is a signaling molecule. In a related embodiment, the signaling molecule is selected from the group consisting of kinase 6 of the G protein coupled receptor, kinase related to vaccine 1, tyrosine kinase 2 of protein PTK2, STAM-like protein containing SH3 and domains 2 ITAM, gene 1 associated with signal-induced proliferation, CD47 antigen (Rh-related antigen, integrin-associated signal transducer), and protein tyrosine phosphatase (type 12 without receptor). In another related embodiment, the signaling molecule is selected from the group consisting of PTK2 (focal adhesion kinase), MPA kinase kinase kinase kinase 2, guanine nucleotide binding protein, PT phosphatase (receptor), binding protein kinase beta cdc42, Ral GEF (RalGPSIA), MAP kinase 7, autotaxin, inositol, 1, 4,5-triphosphate receptor, phosphoinositide-3-kinase gamma, PT phosphatase (without receptor), RAS protein activator p21 (GAP), protein 2 that releases RAS guanil, and 20kDa subunit of the Arp23 complex. In one embodiment, the specific genetic production product of chemotaxis is a molecule related to the extracellular matrix. In a related embodiment, the molecule related to the extracellular matrix is selected from the group consisting of spondine 1 (f-spondine, extracellular matrix protein), collagen of type XVlll (alpha 1), adhesion molecule CD31, tetraspan 3, glycoprotein A33, and angio-associated migratory cell protein. In one embodiment, the specific gene expression product of chemotaxis is a molecule related to the cytoskeleton. In a related embodiment, the molecule related to the cytoskeleton is selected from the group consisting of the protein complex 23 related to actin (subunit 4, 20 kD), tropomyosin 2 (beta), actin-dependent regulator associated with actin associated with related matrix with chromatin SWISNF (subfamily to a 5 member), beta spectrin (non-erythrocytic 1), myosin (light polypeptide 5, regulator), keratin 1, placofilin 4, and sealed protein (actin filament, muscle Z line, alpha 2) . In one embodiment, the specific gene expression product of chemotaxis is a cell cycle molecule. In a related embodiment, the cell cycle molecule is selected from the group consisting of protein 1 that activates the FGF receptor, the oncogene homolog of musculoaponeurotic fibrosarcoma v-maf (avian), cyclin-dependent kinase (such as CDC2) 10, response 2 of early growth inducible by TFGB, retinoic acid alpha receptor, subunit 10 of anaphase-promoting complex, activator of RAS p21 protein (GPTase-activating protein, binding protein 3-lns-1, 3, 4, 5-P4) , cell division cycle 27, programmed cell death 2, c-myc binding protein, protein kinase kinase kinase 1 activated with mitogen, receptor III beta TGF (betaglycan, 300 kDa), and transition 1 from phase G1 to S In one embodiment, the specific gene expression product of chemotaxis is a molecule related to immune response. In a related embodiment, the molecule related to immune response is selected from the group consisting of class II DQ beta 1 of the highest histocompatibility complex, spinal cord stromal cell antigen 2, Burkitt lymphoma receptor 1 (GTP binding protein) , CXCR5), CD7 antigen (p41), Stat2 of type a, immune regulator 1 of T cell, and interleukin receptor 21. In another aspect, the invention provides a method for promoting cellular fugetaxis comprising contacting a cell with an agent without chemokine that promotes fugetaxis in an effective amount to promote fugetaxis. In one embodiment, contact occurs in vivo in a subject who has a disorder characterized by lack of fugetaxis. In one embodiment, the cell is a hematopoietic cell, such as a T lymphocyte. In another embodiment, the cell is a neural cell. In another aspect, the invention provides a method for promoting cellular chemotaxis comprising contacting a cell with an agent without chemokine that promotes chemotaxis in an amount sufficient to promote chemotaxis. In one embodiment, contact occurs in vivo in a subject who has a disorder characterized by a lack of chemotaxis. In another embodiment, the cell is a hematopoietic cell, such as a T lymphocyte. In another embodiment, the cell is a neural cell. The invention is also partially theorized, in several other findings. These include the finding that neutrophils migrate bi-directionally in response to IL-8. That is, neutrophils respond to low concentrations of IL-8 (for example, 10 ng / ml to 500 ng / ml) undergoing chemotaxis. Neutrophils respond to high concentration of IL-8 (eg, 1 microgram / ml to 10 micrograms / ml) undergoing fugetaxis. Accordingly, the invention provides methods for modulating the migration of neutrophils by modulating the concentration of IL-8. In one embodiment, the invention provides a method for promoting chemotaxis in a neutrophil comprising contacting a cell with LL-8 in an amount effective to promote chemotaxis by the neutrophil. In one embodiment, contact occurs live in a subject who has a disorder characterized by a lack of neutrophil chemotaxis. Disorders characterized by lack of neutrophil chemotaxis include, but are not limited to, bacterial infections and granulomatous diseases (eg, tuberculosis). In one embodiment, the invention provides a method for promoting fugetaxis in a neutrophil comprising contacting a cell with IL-8 in an amount effective to promote neutrophil fugetaxis. In one embodiment, contact occurs in vivo in a subject who has a disorder characterized by a lack of neutrophil fugetaxis. Disorders characterized by lack of neutrophil fugetaxis include, but are not limited to, inflammatory or immune mediated diseases, rejection of a transplanted organ or tissue, rheumatoid arthritis, autoimmune diseases and asthma. The invention further provides methods for identifying genetic products that are modulated (i.e., either upregulated or deregulated) in response to fugetaxis or chemotaxis induced by IL-8. Thus, in a further aspect, the invention also provides methods to modulate the effects of IL-8 on neutrophils by inhibiting or enhancing the effects of specific gene products of fugetaxis induced by IL-8 or chemically-specific gene products induced by IL. -8. In another embodiment, the invention provides a method of inhibiting neutrophil chemotaxis comprising contacting a neutrophil that undergoes or is likely to undergo chemotaxis with IL-8 in an amount effective to inhibit or enhance the expression of a specific gene expression product of chemotaxis. . In one embodiment, contact occurs live in a subject who has or is at risk of having an abnormal immune response. In a further embodiment, the gene expression product specific for chemotaxis is a molecule related to the immune response. The molecule related to the immune response can be selected from the group consisting of IL-8, GCP-2, Gro-a, Gro-ß, Gro- ?, CINC-1, CINC-2, ENA-78, NAP-2, LIX, SDF-1, IL-1a and I L-1 ß, C3a , C5a and leukotrienes.
The invention is further theorized in part by the finding that neutrophil-induced IL-8 chemotaxis is selectively inhibited by PIK3-inhibiting wortmannin, causing the cells to undergo fugetaxis at all IL-8 concentrations. Accordingly, in one embodiment, the invention provides methods for inhibiting chemotaxis induced by IL-8 neutrophils (by inversely improving the neutrophil fugetaxis induced by IL-8) by administering to a subject in need thereof an effective amount of wortmanin. The effective amount of the wortmanin is that amount effective to selectively inhibit neutrophil-induced IL-8 chemotaxis and optionally enhance neutrophil fugetaxis in the presence of IL-8. The method can also be performed with other species of this genus. In another embodiment, the invention provides a method for inhibiting neutrophil fugetaxis comprising contacting a neutrophil that undergoes or is likely to undergo fugetaxis with IL-8 in an amount effective to inhibit or enhance the expression of a specific gene expression product of fugetaxis. . In one embodiment, contact occurs in vivo in a subject who has or is at risk of having an abnormal immune response. In a further embodiment, the specific gene expression product of fugetaxis is a molecule related to the immune response. The molecule related to immune response can be selected from the group consisting of IL-8, GCP-2, Gro-a, Gro ß, Gro ?, CINC-1, CIN-2, ENA-78, NAP-2, LIX, SDF -1, IL-1a and IL-1ß, C3a, C5a and leukotrienes. The invention is further theorized in part in the finding that neutrophil-induced IL-8 fugetaxis is selectively inhibited by the alternative P13K inhibitor LY294002, causing the cell to produce chemotaxis at all concentrations of IL-8. Accordingly, in one embodiment, the invention provides methods for inhibiting neutrophil-induced IL-8 fugetaxis (and conversely improving neutrophil-induced IL-8 chemotaxis) by administering to a subject in need thereof an effective amount of the LY294002 P13K inhibitor. The effective amount of LY294002 is that amount effective to selectively inhibit neutrophil-induced IL-8 fugetaxis and optionally enhance chemotaxis in the presence of IL-8. The method can also be performed with other species of this genus. These and other objects of the invention will be described in further detail together with the detailed description of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS The following Detailed Description, given by way of example, but not intended to limit the invention to the specific embodiments described, may be understood in conjunction with the accompanying drawings, incorporated herein by reference. Several features and preferred embodiments of the present invention will now be described by way of non-limiting example and with reference to the accompanying drawings, wherein: Figure 1 is a schematic showing chemotaxis, chemokinesis, and fugetaxis in a cell migration assay T. A) T cell transmigration assays; B) Migration to SDF-1 (chemotaxis); C) Random movement in response to SDF-1 (chemokinesis); D) Migration out of SDF-1 (fugetaxis). Figures 2A and 2B are diagrams showing putative downstream events that result after chemokine coupling at the cell surface. FIGURE 2A: A) Chemokinesis; B) Eicosanoids; C) Tir Kinase Ser & Phosphatases / Trh Kinases & Phosphatases; D) Phosphorylation protein; E) Transcription Factors; F) Other Ser / Thr Kinase; G) Oxidase; H) Sealed protein; l) Activation; J) Actin polymerization; K) Cytoskeletal redisposition; L) Adhesion; M) Residence; N) Differentiation; Ñ) Chemotaxis Chemokinesis Fugetaxis. FIGURE 2B A) Protein kinase C, beta 1, Phospholipase C, beta Phosphatidinisitol-4-phosphate 5-kinase receptor Inositol 1,4,5-triphosphate; B) Phosphorylation protein; C) Ras guanil release protein; D) Other kinases, phosphatases; E) Transcription factors; F) CDC 42, protein 2 associated with BAII, CDC42 binding protein, beta kinase; G) Phosphoinositide 3-kinase, gamma; H) Focal adhesion molecules (PTK2 / FAK, Paxilin), GTPase regulator, asoc. FAK FAK, PT phosphatase, without receptor (acts in paxilina); I) Activation; J) Actin polymerization; K) Redisposition of cytoskeletal; L) Migration; M) Adhesion. Figures 3 to 8 indicate the genes that are significantly differentially regulated (p <0.05, fold change> 1.7) under different gradient conditions of SDF-1. The Gen Bank Access Numbers are provided to further describe the identical genetic products. Figure 3 depicts Table 1, indicating genes that are differentially regulated in the gradients of Medium against Chemokinesis of SDF-1. Positive values are upregulated in chemokinesis; negative values are deregulated in chemokinesis; p < _ 0.05.
FIGURE 3 Table 1 Expression of the Differential Gene in the Gradients of Chemokinesis VS Medium SDF-1 REGULATED INCREASE IN THE CHIMIOCINESIS COMPARED TO THE GRADIENTS OF MEDIUM SDF-1 Leaf 1 of 17 3.52 Hs. 171 814 parathymosin 3.45 Hs.2969 homologue of the viral oncogene of the avian sarcoma v-ski 3.39 Hs.321223 keratin 6B 3.33 Hs.1 39648 product of the KIAA0706 gene 3.32 Hs.66309 Homo sapiens, similar to the RI gene KEN cDNA 231 0034L04, clone MGC: 1 1 061, mRNA, complete cds 3.22 Hs.54505 aquaporin 6, specific kidney 3.1 3 Hs.84285 enzyme E2I conjugated by ubiquitin (homologue for yeast UBC9) 3.05 Hs.1 35626 cimasa 1, mast cell 3.00 Hs.249216 H2B family histone, member J 3.00 Hs.288650 aquaporin 4 2.95 Hs.82085 proteinase serine inhibitor (or cysteine), subtype E (nexin, plaminogen activator inhibitor type 1), member 1 2.93 Hs.287763 sequence Human DNA from clone RP 1 -23021 on chromosome 6. Contents of pseudogene calponin 3 acidic (CNN3) 2. 92 Hs.322680 Homo sapiens cDNA: FLJ21547 fis, clone COL06206 2.91 Hs.301667 Homo sapiens mRNA; CDNA DKFZp566l043 (from clone DKFZp5661 043) 2.90 gb: BC0061 14.1 / DEF = Homo sapiens, similar to the cholinergic receptor, nicotinic, alpha polypeptide 3, with MGC: 1 2991, mRNA 2.88 Hs.173594 inhibitor of proteinase serine (or cysteine), subtype F (alpha 2 antiplasmin, factor derived from pigment epithelium), member 1 2. 85 Hs.162200 urotensin 2 H oja 2 of 1 7 Page 3 of 17 FIGURE 3 Table 1 Expression of the Differential Gene in the Gradients of Chemokinesis VS Medium SDF-1 2.50 Hs.3005 Transcription factor AP-4 (activation enhancer-protein 4 of link) 2.50 Hs.293334 ESTs 2.49 Hs.66578 receptor 2 of the hormone of corticotropin release 2. 46 Hs.286233 autoantigenic sperm protein 17 2.38 Hs.99971 zinc finger protein 272 2.34 Hs.24322 ATPase, H + transport, lysosomal (vacuolar proton pump) 9kD 2.34 Hs.154762 HIV-1 rev binding protein 2.32 Hs.84152 cystathionine beta-synthase 2.31 Hs.96 protein 1 induced bybol-12-myristate-13-acetate 2.31 Hs.247043 receptor of tumor necrosis factor type 1 of the aminopeptidase regulator 2.30 Hs.55481 finger protein of zinc 165 2.29 Hs.8074 inhibitor 3 of angiogenesis of the specific brain 2. 29 Hs.103978 serinetreonine kinase 22B (associated spermiogenesis) 2. 27 Hs.306618 Homo sapiens cDNA FLJ11930 fis, clone HEMBB1000441 2. 26 Hs.194669 zeste enhancer (Drosophila) homologue 1 2.26 Hs.16488 calreticulin 2.25 Hs.305979 Homo sapiens clone FLB3024 PRO0756 mRNA, complete cds Sheet 4 of 17 Page 5 of 17 Sheet 6 of 17 FIGURE 3 Table 1 Expression of the Differential Gene in Chemokinesis Gradients VS Medium SDF-1 1.91 Hs.24322 ATPase, H + transport, lysosomal (vacuolar proton pump) 9kD 1.90 Hs.25732 range of initiation factor 4 of eukaryotic translation, 3 1.90 Hs.288771 protein DKFZP586A0522 1.89 Hs. 80919 2 DNA binding inhibitor, helix-loop-helical protein dominant negative 1.89 Hs.42244 Homo sapiens mRNA; CDNA DKFZp564A023 (from clone DKFZp564A023) 1.89 Hs.82101 domain of homology type of pleckstrin, family A, member 1 1.89 Hs.279582 Sara protein GTP binding 1.84 Hs.306639 Homo sapiens cDNA FLJ12624 fis, clone NT2RM4001754 1.83 Hs.55075 product of KIAA0410 gene 1.83 Hs.142023 T cell activation, increased late expression 1. 83 Hs.180919 DNA binding inhibitor 2 DNA, helix-loop-helix protein negative dominant Sheet 7 of 17 Sheet 8 of 17 FIGURE 3 Table 1 Expression of the Differential Gene in the Gradients of Chemokinesis VS Medium SDF-1 REGULATED DESCENSION IN THE QU IMIOCINESIS COMPARED TO THE MEDIUM GRADIENTS SDF-1 -8.38 Hs.12142 WD repetition domain 13 -8.19 Hs .279623 selenoprotein X, 1 -6.29 Hs.180577 granulin -6.24 Hs.41 cell adhesion molecule 8 related to carcinoembryonic antigen -6.08 Hs.99863 elastase 2, neurotrophin -5.87 Hs.99960 membrane tension domains 4, subfamily A , member 3 (specific hematopoietic cell) -5.22 Hs.286124 CD24 antigen (antigen from group 4 of small cell lung carcinoma) -5.14 Hs.29417 Zhangfel factor of HCF-binding transcript -4.87 Hs.25817 BTB domain (POZ) containing 2 -4.87 Hs.193716 receptor 1 component complement (which includes the Knops blood group system) -4.75 Hs.10306 group 7 natural killer cell sequence -4.56 Hs.26319 protein KIAA0833 -4.54 Hs.300772 tropomyosin 2 (beta) Sheet 9 of 17 Sheet 10 of 17 Sheet 11 of 17 FIGURE 3 Table 1 Expression of the Differential Gene in the Gradients of Chemokinesis VS Medium SDF-1 Leaf 12 of 17 -2.40 Hs.7252 protein KIAA1224 -2.40 Hs.12229 response 2 of previous inducible growth TGFB -2.37 Hs.72964 makorin, ring finger protein, 3 -2.36 Hs.272205 Hypothetical protein FLJ10034 -2.34 Hs.10082 channel activated by small conductance calcium intermediary potassium, subfamily N, member 4 -2.32 Hs.300711 annexin A5 - 2.32 Hs.914 human mRNA for histocompatibility class II antigen alpha chain SB -2.31 Hs.278503 regulated in glioma -2.30 Hs.75703 small inducible cytokine A4 (homologous to Mip-1 b mouse) -2.26 Hs.75811 N-acylesphingosine amidohydrolase (ceramidase acid) -2.25 Hs.81256 S100 calcium binding protein A4 (calcium protein, calvasculin, metastasin, murine placental homolog) -2.25 Hs.3066 K granzyme (protease serine, granzyme 3, triptase II) -2.24 Hs71746 Hypothetical protein FLJ11583 -2.23 Hs.155191 villin 2 (ezrin) -2.23 Hs.159263 Collagen, type VI, alpha 2 -2.23 Hs.21497 Homo sapiens, clone IMAGE: 3629896 mRNA, partial cds -222 Hs. 9973 tensin -2.20 Hs.119274 RAS protein activator p21 (GTPase activation protein) 3 (lns (1, 3,4,5) P4-binding protein) -2.18 Hs.15984 homologue pp21 Sheet 13 of 17 Sheet 14 of 17 FIGURE 3 Table 1 Expression of the Differential Gene in the Gradients of Chemokinesis VS Medium SDF-1 Leaf 15 of 17 Page 16 of 17 1.72 Hs, 182982 golgin-67 -1.72 Hs.132807 Homo sapiens (clone 3.8-1) MHC class I Fragment mRNA -1.71 Hs.193163 integrator 1 link • 1.71 Hs.115460 calicin -1.71 Hs.193163 link integrator 1 • 1.70 Hs.75450 delta sleep induction peptide, immunoreactor 1. 70 Hs.1432 protein kinase C substrate 80K-H 1.70 Hs.6150 factor p 114 for guanine specific nucleotide exchange Rho Sheet 17 of 17 Figure 4 depicts Table 2, which indicates genes that are differentially regulated in Fugetaxis gradients against chemotaxis of SDF-1. The positive values are overregulated in Fugetaxis; the negative values are overregulated in chemotaxis; p < 0.05.
FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 REGULATED INCREMENT IN QUIMIOTAXIS COMPARED TO GRADIENTS QUIMIOTAXIS SDF-1 Sheet 1 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 REGULATED INCREMENT IN CHEMOTAXIS COMPARED TO GRADIENTS QUIMIOTAXIS SDF-1 11.00 Hs.75184 chitinase 3; type 1 (glycoprotein-39 cartilage) 8.12 Hs.79658 casein kinase 1, epsilon 6.17 Hs.7358 hypothetical protein FLJ13110 5.66 Hs.89535 bactericidalpermeability-protein that increases 5.61 Hs.100000 S100 calcium binding protein A8 (calgranulin A) . 57 Hs.182740 S11 ribosomal protein 5.15 Hs.78913 chemokine (C-X3-C) receptor 1 5.07 Hs.81665 Homolog of feline sarcoma oncogene Hardy-Zuckerman team v 5.02 Hs.247989 gene of (V4-30.2) variable region of human immunoglobulin heavy chain, partial cds 4.93 Hs.146409 cell division cycle 42 (GTP-binding protein, 25kD) 4.71 Hs.3076 MHC class II transactivator 4.70 gb: AF262973.1 / DEF = murine Homo cell receptor immunoglobulin type 3DL1 sapiens (KIR3DL1) mRNA, KIR3DL1 * 00701 allele, cds. Complete 4.57 gb: NM_000961.1 / DEF = prostaglandin 12 Homo sapiens (prostacyclin) synthase (PTGIS), mRNA. 444 Hs.2962 S100 calcium binding protein P Sheet 1 of 38 3.64 Hs.1 80884 carboxypeptidase B1 (tissue) 3.64 Hs.314762 partial IGKV gene homo sapiens for the variable chain region kappa immunoglobulin, clone 30 3.62 Hs.80620 factor of guanine nucleotide exchange for Rap 1; GEF regulated by M-Ras 3.58 Hs.44278 protein FLJ 12538 hypothetical sim ilar to protein RAB-related ras 3.57 Hs.29417 HCF-factor Zhangfel of transcription binding 3.56 Hs. 1 05859 protein FLJ 1 0260 hypothetical 3.55 Hs. 1 54495 acetylcholinesterase (blood group YT) 3.54 Hs.287269 hormone somatomammotropin chorionic-type 1 3.51 Hs.2582 defensin, alpha 4, corticostatin 3.45 Hs.98658 inhibitor germination by benzimidazoles 1 (homologous yeast) 3.42 Hs.226014 Human DNA sequence of clone 240B8 on chromosome 6p1 1.2-q 1 2. Contains part 3 of a gene for a similar novel protein to T-STAR, Etoile, Sam68, SLM 1 and p62 Tyrosine Phosphoprotein. Contains ESTs, STSs, GSSs and marker D6S 1695 genomic 3.42 Hs.33102 transcription factor AP-2 beta (activator enhancer-protein binding 2 beta) 3.34 Hs.18894 / len = 982 3.29 Hs.287539 protein FLJ 12662 hypothetical 3.28 Hs .1 14316 Alkyltransferase 8C (alpha2, 3Galbeta 1, 4GlcNAcalpha 2, 8-sialyltransferase) H oja 2 of 38 3.27 Hs.323511 Homo sapiens cDNA: FLJ23176 fis, clone LNG10452 3. 24 Hs.89839 EphA1 3.23 Hs.298469 angiotensin I converting enzyme (peptidyl-dipeptidase A) 1 3.18 Hs.314452 fibrousheathin ll 3.17 Hs.85181 v-raf-1 homolog 1 viral oncogene from murine leukemia 3.16 Hs.273621 Homo sapiens CDNA: FLJ21350 fis, clone COL02751 Page 3 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Chemotaxis Gradients SDF-1 3.11 Hs.88411 Lymphocyte antigen 117 3.10 Hs.2056 UDP family glycosyltransferase 1, polypeptide A9 3.10 Hs.233634 hypothetical protein FLJ14220 3.09 Hs.284277 antibody MO30 Mu immunoglobulin chain of Homo sapiens (IgM) mRNA, cds. Full 3.09 gb: BC006196.1 / DEF = Homo sapiens, superfamily of the necrosis factor receptor, member 9, clone MGC: 2172, mRNA, complete cds 3.07 Hs.97084 Lymphocyte antigen 94 (mouse) homologous (NK-receptor activator; NK -p46) 3.05 Hs.172740 microtubule associated protein, RPEB family, member 3 3.00 Hs.73838 Homo sapiens (clone Z146) retinal mRNA, 3 term and repeat region 2.99 Hs.75294 corticotropin releasing hormone 2.99 Hs.949 neutrophil factor 2 cytosolic (65 kD, chronic granulomatous disease, autosomal 2) 2.99 Hs.278984 calcium binding protein 2.99 Hs.73793 vascular endothelial growth factor 2.97 Hs.37169 internally potassium rectification channel, subfamily J, member 3. 2.97 gb: NM_030876.1 / DEF-Olfactory receptor Homo sapiens, family 5, subfamily V member 1 (OR5V1), mRNA. Page 4 of 38 Page 5 of 38 2.72 Hs.274127 CLST 11240 protein 2.71 Hs.70823 KIAA1077 protein 2.69 Hs.75260 transportable mitogen 2 2.68 Hs.302022 PR domain containing 16 2.67 Hs.621 lectin, galactoside link, soluble, 3 (galectin 3) 2. 65 Hs.287872 Hypothetical FLJ14106 protein 2.65 Hs.123062 human mRNA for T cell receptor, IGRA24 clone 2. 62 Hs83484 SRY (Y-region that determines sex) -box 4 2.62 Hs.307138 Human DNA sequence of clone RP3-508D13 on chromosome 6. Contains a pseudogene protein of heat attack pseudogen, ESTs, STSs and GSSs 2.62 Hs.151449 gene product KIAA0535 2.60 Hs.1310 CD1B antigen, polypeptide b 2.59 Hs.6580 Homo sapiens cDNA: FLJ23227 fis, clone CAE00645, highly similar to AF052138 clone mRNA sequence 23718 Homo sapiens 2.59 gb: NM_030788.1 / DEF = Homo sapiens DC-specific transmembrane protein (LOC81501), mRNA. 2.59 Hs.132942 GTPase regulator associated with focal kinase pp125 (FAK) adhesion; protein KIAA0621 2.58 Hs.169401 apolipoprotein E 2.56 Hs.19520 domain FXYD containing regulator 2 ion transport 2.56 Hs.97403 protein KIAA0944 Sheet 6 of 38 Sheet 7 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 Sheet 8 of 38 Page 8 of 38 2.10 Hs.210859 Hypothetical protein FLJ11016 2.09 Hs.127614 protein phosphatase 1, regulatory (inhibitor) subunit 3 (glycogen and sarcoplasmic hairnet binding subunit hairnet, skeletal muscle) 2.08 Hs.82422 G protein sealed (Actin filament), gelsolin type 2. 08 Hs.32168 protein KIAA0442 2.08 Hs.78305 RAB2, family of oncogen member RAS 2.07 Hs.306667 Homo sapiens cDNA FLJ14076 fis, clone HEMBB1001925 2.07 Hs.326198 transcription factor 4 2.06 Hs.60708 calsequestrin 1 (fast pull, skeletal muscle ) 2. 06 Hs.1870 phenylalanine hydroxylase 2.05 Hs.217493 annexin A2 2.05 Hs.33084 family 2 of solute carrier (assisted glucose transporter) member 5 2.05 Hs.80758 asparlil-tRNA synthetase 2.04 Hs.73291 hypothetical protein FLJ10881 2.03 Hs.270549 protein HZFwl 2.00 Hs.5831 tissue inhibitor of metalloproteinase 1 (erythroid potentiation activity, collagenase inhibitor) 2.00 Hs.142023 T cell activation, increased late expression 2. 00 Hs.272789 Hypothetical protein FLJ20217 2.00 Hs.306711 Homo sapiens cDNA: FLJ21215 fis, clone COL00526 2. 00 Hs.323409 Homo sapiens cDNA FLJ14113 fis, clone AMMA1001715 Sheet 9 of 38 1.99 Hs.41143 phosphoinositide-phospholipase C specifies-beta 1 1. 99 Hs.46752 nitric oxide synthase 1 (neuronal) Sheet 10 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxys SDF-1 1.98 Hs.278850 protocaderin beta 1 1.97 Hs.13245 gene product KIAA0455 1.97 Hs. 14070 hypothetical protein FLJ14166 1.96 Hs.269926 Homo sapiens cDNA. FLJ21441 fis, clone COL04422 1. 96 Hs.26776 receptor type 3 neurotrophic tyrosine kinase 1.96 Hs.159003 channel 6 transient receptor potential 1.96 Hs.74614 hermetic binding protein 1 (occluded zone 1) 1.95 Hs.272351 Human DNA sequence of clone RP4-746H2 on chromosome 20 It contains a pseudogene similar to RCAR11 prokaryotic (30S protein S11 ribosomal) ESTs STSs and GSSs 1. 94 Hs.5814 suppression of tumorigenicity 7 1.94 Hs.169824 subfamily B of killer cell lectin-like receptor, member 1 1.94 Hs.283683 chromosome 8 structure 4 open reading 1.94 Hs.326780 Homo sapiens clone KM35 light chain variable mRNA region immunoglobulin, partial cds 1.94 Hs.106185 guanine nucleotide dissociation stimulator ral 1. 93 Hs.172471 channel controlled by potassium voltage, aggregator related subfamily, member 1 beta 1.93 Hs.6654 protein KIAA0657 Page 11 of 38 1.92 Hs.158343 Testicles-PTP-BL specific-related protein in Y 1. 92 Hs.35101 proline rich Gla (G-carboxyglytic acid) polypeptide 2 1. 92 Hs.97109 ESTs 1.92 Hs.106552 Caspr2 cell recognition molecule 1.91 Hs.153445 human mRNA for partial cds of unknown product 1. 91 Hs.12079 calsintenin-2 1.91 Hs.69547 basic myelin protein 1.90 Hs.129914 dwarf-related transcription factor 1 (acute myeloid leukemia 1; onnugen arnM) 1.90 Hs.143212 cystatin F (leukocistatin) 1.90 Hs.90291 laminin, beta 2 (laminin S) 1.89 Hs.287719 hypothetical protein FLJ23074 Hs.91448 protein tyrosine phosphatase type MKP-1 1.88 Hs.21858 trinucleotide repeated containing 3 1.88 Hs.77436 pleckstrin 1.88 Hs.295112 gene product KIAA0618 1.87 Hs.76136 thioredoxin 1.87 Hs .247877 Human DNA sequence of clone 263J7 on chromosome 6q14.3-15. It contains an RPL7 (60S Ribosomal Protein L7) pseudogene, a RAB1 (RAB1 family of RAC member oncogene) pseudogene, ESTs, an STS and GSSs 1.87 Hs.7358 hypothetical protein FLJ13110 1.86 Hs.128322 t-complex 11 (a murine homologue tcp ) Leaf 12 of 38 Page 13 of 38 1.81 Hs.274578 Homo sapiens mRNA; CDNA DKFZp434F0723 (from clone DKFZp434F0723) 1.80 Hs.198281 Pyruvate kinase, muscle Sheet 14 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 180 Hs.158345 testicle-transcript Y 2 of specific testicle 1. 80 Hs.180919 DNA binding inhibitor 2, protein helix-loop-negative helix dominant 1.80 Hs.10247 activated leukocyte cell adhesion molecule 1. 80 Hs.132781 class I receptor cytokine 1.80 Hs.54481 low density lipoprotein receptor-related protein 8, apolipoprotein receptor 1.79 Hs.48778 niban protein 1.79 Hs.9329 chromosome 20 structure 1 open reading 1.79 Hs.272798 protein FLJ20413 Hypothetical 1.79 Hs.100194 arachidonate activation protein 5-lipoxygenase 1. 78 Hs.270010 protein KIAA0508 1.78 Hs.6088 a domain 11 of disintegrin and metalloproteinase 1. 7Í Hs.183075 ATPase, Ca ++ transport, cardiac muscle, pull 1 fast 1.7Í Hs.272375 protein WNT16 1.7Í Hs.288983 protein hypothetical FLJ21477 1.78 Hs.92254 protein hypothetical FLJ20163 1.71 Hs.306531 Homo sapiens caspase-10c mRNA, cds. Complete Sheet 15 of 38 1.78 Hs.76722 CCAAT Enhancer Binding Protein (CEBP), delta 1. 77 Hs.76901 for related disulfide isomerase protein 1.77 Hs.254105 enolase 1, (alpha) 1.77 Hs.311 phosphoribosyl pyrophosphate amidotransferase 1.77 Hs.31869 / len = 680 1.77 Hs.30299 IGF-II mRNA-binding protein 2.77 Hs. 18705 protein KIAA1233 1.77 Hs.121084 eppin-3 1.76 intersectin 1 (protein domain SH3) 1.76 Hs.169081 gene 6 variant ets (TEL oncogene) 1.76 Hs.4975 channel controlled by potassium voltage, subfamily type KQT, member 2 1.76 Hs .170076 variable load, chromosome Y 1.75 Hs.135305 olfactory receptor, family 10, subfamily H. member 3 1. 75 Hs.287388 histamine H4 receptor 1.75 Hs.165 peptide receptor 1 type glucagon 1.74 Hs.306235 protein hypothetical FLJ13954 1.73 Hs.102865 interleukin 1 receptor-type2 1.73 Hs.49500 protein KIAA0746 1.73 Hs.56175 H. sapiens gene PAC 106H8, similar to Dinamin 1.73 Hs.224829 ESTs 1.73 Hs.287445 hypothetical protein FLJ11726 1.72 Hs.287608 hypothetical protein FLJ13892 Sheet 16 of 38 1.72 Hs.3628 protein kinase kinase kinase 4 activated by mitogen 1.72 Hs.97174 rectification channel internally potassium, subfamily K, member 4 1.72 Hs.283330 hypothetical protein PRO1843 1.72 Hs.274509 constant 2 cellular receptor gamma T 1.71 Hs.105115 absent in melanoma 2 1.71 Hs.121576 Homo sapiens cDNA FLJ20153 fis, clone COL08656, highly similar to AJ001381 Incomplete Homo sapiens cDNA for a mutated allele 1.71 Hs.183556 family 1 of solute carrier (neutral amino acid carrier), member 5 1.71 Hs.79732 fibulin 1 1.71 Hs.62954 ferritin, heavy polypeptide 1.70 Hs.88474 prostaglandin-endoperoxide synthase 1 (prostaglandin GH synthase and cyclooxygenase) 1.70 Hs.112259 T cell receptor gamma locus 1.70 Hs.11 carcinoembryonic antigen-related cell adhesion molecule 3 Page 17 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Chemotaxis Gradients SDF-1 REGULATED INCREMENT IN COMPARED CHEMOTAXIS TO GRADIENTES FUGETAXIS SDF-1 -21.01 Hs.323342 protein complex 23 related to actin, subunit 4 (20 kD) -14.05 Hs.78409 Collagen, type XVlll, alpha 1 -10.49 Hs.15075 Hypothetical protein DKFZp434E2216 -10.32 Hs.305960 hemoglobin, gamma A -10.19 Hs.85752 MDS026 protein from uncharacterized haematopoietic stemprogenitor cells -9.17 Hs.76415 inter-alpha (globulin) H4 inhibitor (Kallikrein-sensitive glycoprotein plasma) -8.59 Hs.740 PTK2 protein tyrosine kinase 2 -7.79 Hs.29222 zinc finger protein 76 (expressed in testicles) -7.50 Hs.73931 major histocompatibility complex, class II, beta DQ 1 -7.30 Hs.82979 mitogen-activation protein kinase kinase kinase kinase 2 -6.96 Hs.46907 HEMK homolog 7kb -6.76 Hs.289031 protein hypothetical FLJ11848 -6.59 Hs. 79410 family 4 of solute carrier, anion exchanger, member 2 (membrane protein band erythrocyte membrane protein -tipol) -6.29 Hs.109441 hypothetical protein FLJ20707 Sheet 18 of 38 -6.02 Hs.14142 nudix (X-linked portion of diphosphate of nucleoside) -type motive 2 -5.92 Hs.110457 candidate 1 for Wolf-Hirschhorn syndrome -5.76 Hs.247981 type a Stat2 -5.62 Hs.301636 factor 6 for peroxisomal biogenesis -5.43 Hs.283404 organic cation transporter -5.30 Hs. 300496 mitochondrial solute carrier -5.28 Hs.79019 baculoviral IAP repeated-containing 1 -5.28 Hs.6343 protein KIAA1464 -5.27 Hs.121073 hypothetical protein FLJ10466 -5.25 Hs.280666 Homo sapiens chromosome 19, cosmid R32184 -5.25 Hs.79340 prote ína B1 osteosarcoma of response PTH -5.12 Hs.279862 inhibitor binding protein p21 cdk -5.07 gb: NM_030882.1 / DEF = Homo sapiens apolipoprotein L, 2 (APOL2), mRNA. -4.97 Hs.76289 biliverdin reductase B (Flavin reductase (NADPH)) -4.96 Hs.36972 CD7 antigen (p41) -4.95 Hs.21970 guanine nucleotide binding protein (Protein G), gamma 3, link -4.82 Hs.197335 plasma glutamate carboxypeptidase -4.73 Hs.5378 spondin 1, (f-spondin) extracellular matrix protein -4.67 Hs.250821 hypothetical MGC4054 protein -4.65 Hs.93597 kinase 5 cyclin-dependent, regulatory subunit 1 (P35) -4.63 Hs.22370 Homo sapiens mRNA; CDNA DKFZp56400122 (from clone DKFZp564O0122) Page 19 of 38 Page 20 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 -3.95 Hs.80598 Transcription Elongation Factor A (SU), 2 -3.93 Hs .315478 Homo sapiens, Similar to 1 pericentriolar material, clone MGC: 8458, mRNA, cds. full -3.93 Hs.272814 hypothetical DKFZp434E1723 protein -3.92 Hs.277401 bromodomain adjacent to zinc finger domain, 2A -3.92 Hs.287652 Homo sapiens cDNA: FLJ21258 fis, clone COL01408 -3.88 Hs.41693 aDnJ (Hsp40) homologous, subfamily B, member 4 -3.86 Hs.19554 chromosome 1 structure 2 fragment open -3.84 Hs.112434 Mapping the gene human novel to chromosome 13 -3.80 Hs.4854 inhibitor 2C cyclin-dependent kinase (p18, CDK4 inhibitors) -3.77 Hs.182577 inositol polyphosphate-5-phosphatase, 75kD -3.77 Hs.181223 protein PRO2801 hypothetical -3.77 Hs.256549 protein 2 nucleotide link (type E.coli MinD) -3.73 gb: U41742.1 / DEF = Nucleophosmin human-retinoic acid receptor, NPM-RAR protein of alpha mRNA fusion, long form, cds. Complete -3.65 Thyroid hormone receptor, alpha (avian erythroblastic viral leukemia (v-erb-a) homolog oncogene -3.64 Hs.30250 homolog oncogene fibrosarcoma (avian) musculoaponeurotic v-maf -3.64 Hs.44865 Factor-1 link enhancer lymphoid Leaf 21 of 38 Sheet 22 of 38 Sheet 23 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Chemotaxis Gradients SDF-1 Leaf 24 of 38 -2.95 Hs.168625 associated protein of androgen-induced prostate proliferative disconnection -2.93 Hs.81424 ubiquitin-tipol (sentrin) -2.91 Hs.104916 hypothetical FLJ21940 protein -2.90 Hs.99918 carboxyl ester lipase (lipase bile) stimulated by salt) -2.90 Hs.83347 angio-associated, migratory cell protein -2.89 Hs.86178 phosphoprotein 9 phase -2.84 Hs.251410 chromosome Homo sapiens 19, cosmid R31180 -2.84 Hs.278741 UDP glycosyltransferase 1 family, polypeptide A8 -2.82 Hs.288617 Hypothetical FLJ22621 protein -2.82 Hs.2558 gamma-carboxyglutamate bone (gla) protein (osteocalcin) -2.81 Hs.170307 RalGPSIA guanine nucleotide exchange factor Ral -2.81 Hs.241558 ariadne (Drosophila) homolog 2 -2.80 Hs.272317 Homo sapiens mRNA; CDNA DKFZp434O0213 (from clone DKFZp434O0213); partial cds -2.80 Hs.293334 ESTs -2.78 Hs.3080 protein kinase 7 activated by mitogen -2.78 Hs.94037 Hypothetical protein FLJ23053 -2.78 KIAA0280 protein -2.77 Hs.12328 KIAA1005 protein -2.76 Hs.237825 signal recognition particle 72kD -276 Hs.272792 Hypothetical protein FLJ20307 Sheet 25 of 38 -2.73 Hs.1 32753 protein 2 only box F -2.71 Hs .7451 9 primase, polypeptide 2A (58kD) -2.71 Hs. 1 80338 superfamily of the necrosis factor receptor, member 1 2 (membrane protein of translocation chain association) -2.68 Hs.285005 Tom22 important mitochondrial receptor -2.68 Hs .172052 serinetreonin q uinase 1 8 -2.68 Hs.180903 hypothetical protein 384D8_6 -2.67 Hs .9071 progesterone membrane binding protein -2.64 Hs.1 8443 aldeh gone dehydrogenase 8 family , member A1 -2.63 Hs.1741 85 ectonucleotide pyrophosphatase phosphodiesterase 2 (autotaxin) -2.62 Hs.17883 phosphatase 1 G protein (formerly 2C), magnesium-dependent, gamma isoform -2.62 Hs.1 70482 myosin, light 5 regulatory polypeptide -2.61 Hs.1 80338 superfamily of tumor necrosis factor receptor, member 1 2 (Transplacement chain-membrane protein association) -2.61 Hs .1 53293 protein KIAA0701 -2.60 Hs.238272 1, 4,5-triphosphate receptor inositol, type 2 -2.59 Hs.58362 Hypothetical protein FLJ 12681 -2.59 Chromosome 1 9 Homo sapiens, cosmid R28784, complete sequence. -2.58 Hs.7581 3 polycystic kidney disease 1 (dominant autotosomal) -2.57 Hs.23964 sin3-associated polypeptide, 1 8kD H oja 26 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 -2.54 Hs.283675 protein NPD009 -2.54 Hs.183887 protein hypothetical FLJ22104 -2.53 Hs.80741 carboxylase propionyl Coenzyme A, alpha polypeptide -2.52 Hs.80828 keratin 1 (epidermolytic hyperkeratosis) -2.52 Hs.147587 Homo sapiens mRNA; CDNA DKFZp547F134 (from clone DKFZp547F134) -2.52 Hs.287444 hypothetical protein FLJ11722 -2.51 Hs.17409 protein 1 rich in cysteine (intestinal) -2.50 Hs.325520 Homo sapiens IMM mRNA for hLAT1-3TM, cds. complete -2.49 Hs.301011 protein KIAA0876 -2.48 Hs.16193 Homo sapiens mRNA; CDNA DKFZp586B211 (from clone DKFZp586B211) -2.48 Hs.32942 phosphoinositide-3-kinase, catalytic, gamma polypeptide -2.48 Hs.23240 Homo sapiens cDNA FLJ13496 fis, clACE PLACE1004471, weakly similar to PROTEIN 83 DE FING CINC -2.47 Hs.23796 odz (odd Ozten-m, Drosophila) homologue 1 -2.46 Hs.180338 superfamily of the necrosis factor receptor, member 12 (trans-protein chain-association membrane protein) -2.46 Hs.175941 B-cell protein receptor BAP29 associated -2.44 Hs.57856 PFTAIRE protein kinase 1 Page 28 of 38 Page 29 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 -2.18 Hs.5541 ATPase, Ca + + transport, ubiquitous -2.17 Hs.8173 protein Hypothetical FLJ10803 -2.16 Hs.16079 Hypothetical protein FLJ10233 -2.14 Hs.180338 superfamily of the necrosis factor receptor, member 12 (transplacement chain-membrane association protein) -2.13 Hs.82129 anhydrase carbonic III, muscle or specific -2.13 Hs.143131 glycoprotein A33 (transmembrane) -2.13 Hs.111244 hypothetical protein -2.12 Hs.168640 ankylosis, progressive homologue (mouse) -2.11 Hs.293495 ESTs, Weakly similar to ENTRY OF ALERT OF CONTAMINATION OF SUBFAMILY SEQUENCE J ALU1_HUMANO ALU SUBFAMILIA J H-Sapiens -2.11 Hs.5997 Hypothetical protein FLJ13078 -2.11 Hs.7995 / len = 469 -2.10 Hs.519 oxidoreductase containing WW domain -2.10 Hs.2815 domain POU, class 6, transcription factor 1 -2.09 Hs.278985 hypothetical protein -2.09 Hs.89474 ADP- ribosylation factor -6.0.0 Hs.301114 zinc finger protein 79 (pT7) Sheet 30 of 38 Sheet 32 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 Sheet 33 of 38 Sheet 34 of 38 Sheet 35 of 38 FIGURE 4 Table 2 Expression of the Differential Gene in Fugetaxis VS Gradients Chemotaxis SDF-1 Leaf 36 of 38 Leaf 37 of 38 -1.72 Hs.279819 protein APR-1 -1.71 Hs.295446 ESTs, Moderately similar to 810024C cytochrome oxidase I H. sapiens -1.71 Hs.30696 transcription-type factor (helix-loop-helix basic) -1.71 Hs.31834 Homo sapiens sequence mRNA clone 25129 • 1.71 Hs.14928 protein hypothetical FLJ12903 1.71 Hs.236940 / len = 570 1.71 Hs.48433 endocrine regulator -1.71 Hs.283912 Homo sapiens PAC clone RP4-771P4 of 7q11 .21q11.23 1.71 Hs.129445 Hypothetical protein FLJ12496 -1.70 Hs.49526 f-box and protein 4 leucine-rich repeat -1.70 Hs.70359 KIAA0136 protein • 1.70 Hs.75790 glycan phosphatidylinositol, class C • 1.70 Hs.83790 product of gene KIAA0305 -1.70 Hs.18885 protein CGI-116 -1.70 Hs.232068 transcription factor 8 (expression of interleukin 2 repressed) Sheet 38 of 38 Figure 5 depicts Table 3, which indicates genes that are differentially regulated in the gradients of Chemokinesis against Chemotaxis of SDF-1. Positive values are overregulated in chemotaxis; negative values are deregulated in chemotaxis; p £ 0.05.
FIGURE 5 Table 3 Differential Gene Expression in Chemokinesis VS Chemotaxis Gradients SDF-1 REGULATED INCREMENT IN CHEMOTAXIS COMPARED TO GRADIENTS CHEMOQUINESIS SDF-1 80.37 Hs.99120 DEADH (Asp-Glu-Ala-Asfis) polypeptide box, Y chromosome 51. 46 Hs.80358 SMC (mouse) homologue, Y chromosome 36.38 Hs.180911 S4 ribosomal protein, Y bond 24.62 Hs.155103 eukaryotic translation initiation factor 1A, Y chromosome 21.08 Hs.193145 ubiquitin specific protease 9, Y chromosome (fat aspects) Drosophila related) 16.17 Hs.155397 Homo sapiens mRNA; CDNA DKFZp564K143 (from clone DKFZp564K143) 12.68 Hs.177605 subfamily C of murine cell lecithin receptor type, member 2 11.67 Hs.75658 phosphorylase, glycogen; brain 1031 Hs 155103 factor 1A initiation of eukaryotic translation, Y chromosome 931 Hs 301636 factor 6 of peroxisomal biogenesis 9.28 Hs.99120 DEADH (Asp-Glu-Ala-Asfis) polypeptide box, Y chromosome 8. 95 FK506-binding protein 8 (38kD) 8.15 Hs.37427 4.1 band of erythrocyte membrane protein (eliptocytosis 1, RH-linked) 7.92 Hs.278599 nuclear receptor subfamily 6, group A, member 1 Page 1 of 24 6.60 Hs. 25817 BTB (POZ) domain containing 2 6.45 Hs.79410 family 4 of solute carrier, anion exchanger, member 2 (band 3 of erythrocyte membrane protein-tipol) 6.13 Hs.56336 protein kinase, link Y 6.10 Hs.5541 ATPase, Ca ++ transport, ubiquitous 6.04 Hs. 80577 granulin 5.26 Hs.73931 major histocompatibility complex, class II, DQ beta 1 5.01 Hs.10306 natural killer cell group sequence 7.63 Hs.272108 ESTs 4.28 Hs.121073 hypothetical protein FLJ10466 4.27 Hs .79340 protein B1 response osteosarcoma PTH 4.19 Hs.89560 iduronidase, alpha-L- 3.91 Hs.272438 discs, (Drosophila) homolog 3 long (neuroendocrine-dig) 3. 90 Hs.104555 Neuropeptide FF- precursor amide peptide 3.79 Hs.285753 protein type SCG10 3.75 Hs.99877 Janus kinase 3 (a protein tyrosine kinase, leukocyte) 3. 56 Hs.187378 Hypothetical protein FLJ11278 354 Hs 58362 Hypothetical protein FLJ12681 340 Hs 98614 protein 1 linkage 1 ribosome (homolog dog 180kD) 3. 38 Hs.12142 domain 13 repeated WD 335 Hs 202672 endothelial differentiation, G protein-coupled receptor sfingolipid, 3.35 Hs.41 carcinoembryonic antigen - related cell adhesion molecule 8 Sheet 2 of 24 3.31 Hs.76415 inter-alpha (globulin) inhibitor H4 (Kallikrein-sensitive glycoprotein plasma) 3.21 Hs.326035 growth previous response 1 3.18 Hs.193324 ESTs 3.17 Hs.279651 melanoma inhibitory activity 3.13 Hs.194662 calponin 3, acidic 3.13 Hs.167380 protein BLu 3.10 Hs.181353 UDP- Gal: betaGlcNAc beta 1, 3-galactosyltransferase, polypeptide 2 3.10 Hs.110964 protein hypothetical FLJ23471 310 Hs.147472 chain 2 intermediate dynein 309 Hs.7724 protein KIAA0963 3.03 gb: U52696.1 / DEF = Human adrenal Creb-rp homolog (Creb) -rp), cds. complete, and tenascin-X (XB), partial cds, mRNA. 3.02 Hs.134729 FXYD domain-containing ion transport regulator 7 2.99 Hs.17409 protein 1 enriched for cysteine (intestinal) 2.95 Hs.3066 granzyme K (serine protease, granzyme 3; Tryptase II) 2.90 Hs.92381 nudix (nucleoside diphosphate binding portion X) -type motif 4 2.90 Hs.242407 Protein G-coupled receptor, family C, group 5, member B 2.86 Hs.76289 biliverdin reductase B (flavin reductase ( NADPH)) 2.84 Hs.7258 hypothetical FLJ22021 protein 2.84 gb: NM_030931.1 / DEF = ESP13.2 protein secretory epididymal Homo sapiens (ESP13.2), mRNA. 2.82 Hs.233789 ESTs 2.81 Hs.64096 product of gene KIAA0427 Page 3 of 24 FIGURE 5 Table 3 Expression of the Differential Gene in Quimokinesis VS Gradients Chemotaxis SDF-1 Sheet 4 of 24 Page 5 of 24 2.19 Human DNA sequence of clone RP5-1174N9 on chromosome 1p34.1-35.3. Contains the gene for a novel protein with IBR domain, a (pseudo 2.18 Hs.94970 protein KIAA0306 2.18 Hs.322422 Homo sapiens cDNA FLJ11676 fis, clone HEM8A1004752, highly similar to Homo sapiens mRNA for LAK-4D 2.17 Hs.195464 filamin A, alpha (binding protein-280 actin) 2. 16 gb: NM_030753.1 / DEF = Homo sapiens type aptero family of MMTV integration site, member 3 (WNT3), mRNA. 2.15 Hs.195432 family aldehyde dehydrogenase 2 (mitochondrial) 2. 15 Hs 1724 receptor interleukin 2, alpha 2.14 I Hs 21497 Homo sapiens, clone IMAGE: 3629896, mRNA, partial cds 2.09 Hs.36972 CD7 antigen (p41) 2.08 Hs.11590 cathepsin F 2.08 Hs.57749 gene 2 expressed from synoptic core; protein KIAA1011 2.07 Hs.103382 escramblase 3 phospholipid 2.06 Hs.77858 box 2 homeotic mesenchyme (homeotic box specific growth arrest) 2.06 Hs.64239 Human DNA sequence of clone RP5-1174N9 on chromosome 1p34.1-35.3. Contains the gene for a novel protein with IBR domain, a (pseudo Sheet 6 of 24 2 06 Hs.211584 polypeptide elevated neurofilament (68kD) 2.05 Hs.278295 cholinergic receptor, nicotinic, polypeptide epsilon 2. 05 Hs.11809 Ig related molecule 1L-1 R-single 2.04 Hs.3838 inducible serum kinase 2.03 aquaporin 3 2.02 Hs.307091 ARTS protein Homo sapiens (PNUTL2) mRNA, cds. full; nuclear gene for mitochondrial product 1.99 Hs.71746 hypothetical protein FLJ11583 Sheet 7 of 24 FIGURE 5 Table 3 Expression of the Differential Gene in Quimokinesis VS Gradients Chemotaxis SDF-1 1.98 Hs.58362 / Ien = 594 1.97 Hs.195464 Filamine A, alpha (actin-binding protein-280) 1. 96 Hs.81256 S100 calcium-binding protein A4 (calcium protein, calvasculin, metastasin, homologous murine placental) 1. 96 Hs.79019 baculoviral IAP repeat - contains 1 1.95 Hs.78781 B vascular endothelial growth factor B 1.94 Hs.7019 induced signal-proliferation-associated gene 1.94 Hs.68877 cytochrome b-245, alpha polypeptide 1.94 Hs.195464 filamin A , alpha (actin-binding protein-280) 1. 92 Hs.1103 transformation growth factor, beta 1 1.92 Hs.150540 Homo sapiens, clone IMAGE: 3954961, mRNA, partial cds 1.90 Hs.155191 villin 2 (ezrin) 1.90 Hs.301417 nucleoprotein AHNAK (desmoyocin) 1.90 Hs.112049 link factor 1.88 Hs.73956 oxidoreductase 2 NAD (P) H menadione, dioxin-inducible 1.88 Hs.202687 potassium voltage - controlled channel, Shal-related subfamily, member 2 Sheet 8 of 24 Page 9 of 24 Sheet 10 of 24 FIGURE 5 Table 3 Expression of the Differential Gene in Chemokinesis VS Chemotaxis Gradients SDF-1 REGULATED DESCENT IN CHEMOTAXIS COMPARED TO GRADIENT CHEMOQUINESIS SDF-1 -17.85 Hs.223014 antisense inhibitor -12.91 Hs.76364 inflammatory factor-1 alloherto -5.81 Hs.82985 collagen, type V, alpha 2 -5.69 Hs.7358 hypothetical FLJ13110 protein -5.42 Hs.212587 Homo sapiens mRNA; CDNA DKFZp566M043 (from clone DKFZp566M043) -5.06 Hs.51120 antimicrobial peptide cathelicidin -4.80 Hs.173464 FK506-binding protein 8 (38kD) -4.69 Hs.134503 PR domain containing 8 -4.54 Hs.119500 ribosomal protein, long P2 - 4.27 Hs.139263 calcium channel, voltage-dependent, alpha subunit 1 F -4.24 Hs.76845 phosphoserine type phosphatase -4.23 Hs.182740 protein S11 ribosomal 4.18 gb: M24668.1 / DEF = Human Ig rearrange chain HV-region mRNA ( CD-JH4), cds. full -3.89 Hs.73793 vascular endothelial growth factor Sheet 11 of 24 -3.89 Hs.75105 emopamil-binding protein (sterol isomerase) -3.86 Hs.274 tyrosine kinase associated with megakaryocyte -3.86 Hs 24322 ATPase, H + transport, lysosomal (vacuolar proton pump) 9kD -3.78 Hs 203269 ESTs, Moderately similar to SUBFAMILY SEQUENCE ALERT ENTRY ALU8_HUMAN ALU SUBFAMILIA SX H-Sapiens -3.76 Hs.3112 sodium channel, no controlled voltage 1, gamma -3.72 Hs 10247 activated leukocyte cell adhesion molecule -3.67 Hs 406 family 6 of solute carrier (neurotransmitter transporter, dopamine), member 3 -3.66 Hs.27184 growth factor, ervl (S. cerevisiae) -type (liver regeneration enhancer) -3.63 Hs.321223 keratin 6B -3.57 Hs.173594 serine (or cysteine) proteinase inhibitor, group F (alpha-2 antiplasmino, factor derived from pigment epithelium), member 1 -3.52 Hs.152251 counterpart 5 curled (Drosophila) -3.50 Hs.69752 desmocolin 1 -3.48 Hs.159581 matrix metalloproteinase 17 (membrane-inserted) -3.48 Hs 105927 stem cell growth factor, type C lectin secreted from lymphocyte Sheet 12 of 24 Page 13 of 24 Sheet 14 of 24 FIGURE 5 Table 3 Expression of the Differential Gene in Quimokinesis VS Gradients Chemotaxis SDF-1 Sheet 15 of 24 Sheet 16 of 24 Leaf 17 of 24 -2.30 Hs.7306 related protein 1 curled secreted -2.30 Hs.288931 Homo sapiens cDNA FLJ13034 fis, clone NT2RP3001232 -2.29 Hs.25732 eukaryotic translation initiation gamma factor 4, 3 -2.29 gb: NM 030975.1 / DEF = Homo sapiens protein 9.9 associated by keratin (KRTAP9.9), mRNA. -2.27 Hs20137 Hypothetical protein DKFZp434P0116 -2.26 Hs.97109 ESTs -2.23 Hs.111611 ribosomal protein L27 -2.20 Hs.78629 ATPase, Na + K + transport, beta 1 polypeptide -2.19 Hs.180919 DNA inhibitor binding 2, dominant negative protein helix-loop-helix -2.16 Hs.4147 membrane protein chain association translocation Sheet 18 of 24 FIGURE 5 Table 3 Expression of the Differential Gene in Quimoquinésis VS Gradients Chemotaxis SDF-1 Leaf 19 of 24 Leaf 20 of 24 -1.88 Hs.24385 Human hbc647 sequence mRNA -1.87 Hs. 65662 product of KIAA0675 gene • 1.87 Hs.283037 protein HSPC039 • 1.86 Hs.152939 Sequence mRNA Homo sapiens clone 24630 -1.86 Hs.89474 factor 6 ADP-ribosylation -1.86 Hs.247904 Human DNA sequence of clone 1060K6 on chromosome 20p12.1-13 Contains a pseudogene-like protein 40S ribosomal S11, ESTs, STSs and GSSs 1.86 Hs.121128 BCR signaling 1 downstream • 1.85 Hs .56043 protein CGI-115 -1.84 | Hs.184050 v-Ki-ras2 Kirsten rat sarcoma 2 homologue viral oncogene • 1.84 Hs.50716 hypothetical SIRP-b2 protein -1.83 Hs. Protein PTPRF interactin, binding protein 1 (liprin beta 1) -1.82 Hs.7910 RING1 and YY1 link protein • 1.81 Hs.25732 factor 4 eukaryotic translation initiation gamma, 3 • 1.81 Hs.159526 homolog (Drosophila) patching -1.81 Hs.92254 hypothetical FLJ20163 protein -1.80 Hs.56966 protein KIAA0906 -1.80 Hs.283729 ESTs Sheet 21 of 24 Sheet 22 of 24 FIGURE 5 Table 3 Expression of the Differential Gene in Quimoquinésis VS Gradients Chemotaxis SDF-1 Sheet 23 of 24 Sheet 24 of 24 Figure 6 describes Table 4, which indicates genes that are differentially regulated in the chemokinesis versus Fugetaxis gradients of SDF-1. The positive values are overregulated in Fugetaxis; the negative values are deregulated in Fugétaxis; p < 0.05.
FIGURE 6 Table 4 Expression of the Differential Gene in Chemokinesis VS Gradients Fugetaxis SDF-1 REGULATED INCREMENT IN FUGETAXIS COMPARED TO GRADIENTS CHEMOQUINESIS SDF-1 39.87 Hs.80358 SMC (mouse) homologue, Y chromosome 35.11 Hs.99120 DEADH (Asp-Glu-Ala -AspHis) polypeptide box, Y chromosome 33.40 Hs.180911 S4 ribosomal protein, Y bond 13.24 Hs.155397 Homo sapiens mRNA; CDNA DKFZpS64K143 (from clone DKFZpS64K143) 12.03 Hs.75184 chitinase 3-ti po 1 (cartilage glycoprotein-39) 10.64 Hs.78913 chemokine (C-X3-C) receptor 1 10.01 Hs.155103 eukaryotic translation initiation factor 1A, chromosome And 8.19 Hs.193145 protease 9 of specific ubiquitin, Y chromosome (fat aspects Drosophila) 8.08 Hs.75184 chitinase 3-ti po 1 (cartilage glycoprotein-39) 7.64 Hs.100000 S100 calcium-binding protein A8 (calgranulin A) 6. 57 Hs.10306 natural killer cell group sequence 7.42 Hs.153837 myeloid nuclear cell differentiation antigen 6. 10 Hs.77436 pleckstrin Sheet 1 of 35 Page 2 of 35 Sheet 3 of 35 Sheet 4 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Quimoquinésis VS Gradients Fugetaxis SDF-1 Sheet 5 of 35 Sheet 6 of 35 2.95 Granulisin 2.94 Hs.75990 haptoglobin 2.94 Hs.21425-hydroxytryptamine (serotonin) receptor 3A 2.91 Hs.75260 inducible mitogen 2 2.87 Hs.181353 UDP-Gal: betaGlcNAc beta 1, 3-galactosyltransferase, polypeptide 2 2.85 Hs.278528 Tryptase, alpha 2.81 Hs.274562 Homo sapiens mRNA; CDNA DKFZp434E2028 (from clone DKFZp434E2028) 2.77 Hs.3195 subfamily C small inducible cytokine, member 1 (Lyfotactin) 2.77 Hs.332045 Homo sapiens cDNA FLJ20161 fis, clone COL09252, highly similar to L33930 Homo sapiens CD24 signal transducer mRNA 2.76 Hs.141496 MAGE-type2 2.75 Hs.122552 G-2 and S-phase expressed 1 2.73 Hs. 164960 BarH-type homeocaja 1 2.71 Hs.129706 gene 4 of box tied 2.70 Hs.77436 pleckstrina 2.70 Hs.119597 stearoyl-CoA desaturase (delta-9-desaturase) 2.70 Hs.130546 protein hypothetical FLJ20449 2.69 Hs.301417 nucleoprotein AHNAK (desmoyocin ) Sheet 7 of 35 2.66 Hs.272278 cholinergic receptor, nicotinic, alpha 9 polypeptide 2. 64 Hs.76722 CCAAT enhancer binding protein (CEBP), delta 2. 64 Hs.75607 C substrate protein kinase rich in myristoyl alanine (MARCKS, 80K-L) 2.64 Hs.307177 Human SH3 domain contains SH3P17 protein MRNA, cds. complete 2.63 Hs.250696 KDEL (Lys-Asp-Glu-Leu) receptor 3 retention of endoplasmic reticulum protein 2.61 Hs.108196 protein HSPC037 2.60 Hs.79006 deoxythymidylate kinase (thymidylate kinase) 2.56 Hs.256986 ESTs, Moderately similar to ATPASA 116 KDA SUBUNIDAD TO ISOFORM 2 H. sapiens VPP2_HUMAN TRANSUBICATION OF VACUOUS PROTON 2.55 Hs.198003 sarcosine dehydrogenase 2.54 Hs.112259 T-cell receptor site 2.52 Hs.52931 Adrenergic receptor, alpha-1A Sheet 8 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Chemokinesis VS Gradients Fugetaxis SDF-1 2.51 Hs.50929 Hypothetical protein FLJ13842 2.50 Hs.123030 pseudogen germline kappa-human immunoglobulin (Chr22.4) variable region (subgroup V kappa II) 2.47 Hs.302046 Homo sapiens mRNA; CDNA DKFZp564C163 (from clone DKFZpS64C163) 2.46 Hs.54517 ficolin (collagenfibrinogen domain contains lecithin) 2 (hucolin) 2.44 Hs.79706 plectin 1, intermediate filament binding protein, 500kD 2.44 Hs.274509 constant 2 gamma T cell receptor 2.44 Hs.192662 hypothetical FLJ10469 protein 2.42 Hs.81182 histamin N-methyltransferase 2.41 Hs.272034 hypothetical protein PR02822 2.39 Hs.112259 G-cell receptor site 2.38 gb: BC006252.1 / DEF = Homo sapiens, clone MGC: 10619, mRNA , cds. Complete 2.38 Hs.307187 H. sapiens mRNA for soluble delta TCR 2.37 Hs.283640 protein cig01 2.37 Hs.196352 factor 4 neutrophil cytosolic (40kD) Sheet 9 of 35 2.37 Hs.143212 cystatin F (leukocistatin) 2.36 Hs.211869 dickkopf (Xenopus laevís) homolog 2 2.34 Hs.105700 protein 4 related to secreted ripple 2.34 Hs.183805 ankidinal, erythrocytic 2.34 Hs.78909 factor 2 response butyrate (factor 2 response EGF) 2.34 Hs.150443 protein KIAA0320 2.33 Hs.171921 domain sema, domain (Ig) immunoglobulin, short, secreted, basic domain, (semaphorin) 3C 2.33 Hs.287372 V1R-type1 2.32 Hs.2014 T cell receptor delta site 2.30 gb: M18728.1 / DEF = non-human cross-reactive mRNA specific, cds. complete 2.30 Hs.287778 Human DNA sequence of clone RP11-318P23 on chromosome 20 Contains a TAR DNA-binding protein pseudogene, ESTs, STSs and GSSs 2.29 Hs.16611 protein D52 tumor-tipol 2.29 Hs.195464 filamin A, alpha ( protein -280 link actin) 2.26 Hs.3066 granzyme K (protease serine, granzyme 3, triptase II) 2. 26 Hs.169824 subfamily B of murine cell lectin receptor, member 1 2.25 Hs.171596 EphA2 Sheet 10 of 35 Sheet 11 of 35 2.16 Hs.183805 ankyrin 1, erythrocytic 2.16 Hs.75909 protein KIAA0182 2.16 Hs.287621 protein hypothetical FLJ14069 2.15 Hs.112360 prominin (rat.on) -type1 2.15 Hs.169266 neuropeptide Y receptor Y1 2.15 Hs.118796 Annexin A6 2.15 Hs.195464 Filamine A alpha (actin-binding protein-280) 2. 14 | Hs.50964 carcinoembryonic antigen-related cell adhesion molecule 1 (bile glycoprotein) 2.13 Hs. 83805 ankyrin 1, erythrocytic 2.10 | Hs.553 family 6 of the solute carrier (neurotransmitter transporter, serotonin), member 4 Sheet 12 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Quimokinesis VS Gradients Fugetaxis SDF-1 Leaf 13 of 35 Page 14 of 35 Page 15 of 35 1.82 Hs.274150 Hypothetical FLJ10351 protein 1.81 Hs.289082 ganglioside activating protein GM2 1.81 Hs.785 integrin, alpha 2b (glycoprotein llb complex platelet llbllla, antigen CD41 B) 1.80 Hs.160483 7.2 band erythrocyte membrane protein (stomatin) 1.80 Hs.50477 RAB27A, family of oncogen member RAS 1.79 Hs.306531 Homo sapiens caspase-10c mRNA, cds. full Sheet 16 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Chemokinesis VS Gradients Fugetaxis SDF-1 Page 17 of 35 1.76 Hs.296348 E2k 1.75 Hs.176663 Fc fragment of IgG, low affinity lllb, receptor for (CD16) 1.75 Hs.93837 phosphatidylinositol transfer protein, membrane-associated 1.75 Hs.81256 S100 calcium binding A4 protein (calcium protein, calvasculin, metastasin, homologous murine placental) 1.75 Hs.11801 regulatory factor 6 interferon 1.75 Hs.214982 laminin, gamma 1 (formerly LAMB2) 1.75 Hs.234799 region of bypass point group 1.75 Hs.97672 protein CTAGE-1 1.74 Hs.195175 apoptosis regulator type CASP8 and FADD 1.73 aquaporin 3 1.73 Hs.10247 activated leukocyte cell adhesion molecule 1. 73 Hs.779103-hydroxy-3-methylglutaryl-Coenzyme A synthase 1 (soluble) 1. 73 Hs.44 pleiotrophin (heparin binding growth factor 8, neurite growth promotion factor 1) 1.73 Hs.195175 regulator apoptosis type CASP8 and FADD 1.72 Hs.80645 interferon regulatory factor 1.72 Hs.77422 proteolipid protein 2 ( Colonic-enriched epithelium) 1. 71 Hs.250696 KDEL (Lys-Asp-Glu-Leu) receptor 3 retention of endoplasmic reticulum protein 1.71 Hs.44077 alpha-parvina 1.70 Hs.1103 transformation of growth factor, beta 1 1.70 Hs.159161 inhibitor of dissociation Rho GDP (GDI) alpha 1.70 Hs.153028 cytochrome b-561 1.70 Hs.279562 myelin transcription factor 1 Sheet 18 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Chemokinesis VS Gradients Fugetaxis SDF-1 Sheet 19 of 35 -3.17 Hs.78518 Natriuretic peptide receptor Bguanylate cyclase B (receptor B atrionatriuretic peptide) -3.17 Hs.158688 gene product KIAA0741 -3 17 Hs.20137 hypothetical protein DKFZp434P0116 -3.15 Hs.155049 hypothetical protein FLJ11282 -3.15 Hs.120769 Homo sapiens cDNA FLJ20463 fis, clone KAT06143 -3.13 Hs.173594 serine (or cysteine) proteinase inhibitor, group F (alpha-2 antiplásmino, epithelium pigment-derived factor), member 1 -3.12 Hs.7426 protein K1AA0841 -3.11 Hs.61712 pyruvate dehydrogenase kinase, isoenzyme 1 -3.11 Hs.110796 protein SAR1 -3.11 Hs.105478 phosphoribosilformilglicinamidina synthase (FGAR amidotransferasa) -3.10 Hs.14286 flavin containing monooxygenase 5 -3.08 Hs.61289 synaptojanin 2 -3.06 Hs.23796 odz (odd Ozten-m, Drosophila) homolog 1 - 3.04 Hs.249216 family H2B histone, member J -3.03 Hs.6179 polypeptide box 17 DEADH (Asp-Glu-Ala-AspHis) (72kD) -3.03 Hs.104916 protein hypothetical FLJ21940 -3.03 Hs.184523 protein KIAA0965 -3.03 Hs. 175038 HSPC056 Protein Leaf 20 of 35 Sheet 21 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Chemokinesis VS Gradients Fugetaxis SDF-1 -2.82 Hs.20019 hemochromatosis -2.81 Human DNA sequence of clone RP5-1163J1 on chromosome 22q13.2- 13.33 Contains the 3 'part of a gene for a novel protein containing domain type KIAA0279 - TIPOEGF- (similar to Celsrl mouse, MEGF2 mouse), a novel gene for a protein similar to C. elegans B0035. -2.81 Hs.221040 HBS1 (S. cerevisiae) -type -2.80 Hs.292998 ESTs -2.79 Hs.38783 SKI type -2.79 Hs.168625 protein associated with proliferative disconnection of prostate induced by androgen -2.78 Hs.174185 ectonucleotide pyrophosphatase phosphodiesterase 2 ( autotaxin) -2.77 Hs.1460 glucagon -2.77 Hs.23585 KIAA1078 protein -2.75 Hs.5241 fatty acid binding protein 1, liver -2.75 Hs.82527 sialyltransferase 8 (alpha-N-acetylneuraminate: alpha-2, 8-sialitransferase, GD3 synthase) A -2.75 Hs.31476 Homo sapiens cDNA FLJ13872 fis, clone THYRO1001322 Page 22 of 35 -2.72 Hs.18858 phospholipase A2, group IVC (cytosolic, calcium independent) -2.69 protein KIAA1117 -2.69 Hs.26471 Homo sapiens clone HQ0692 -2.69 Hs.239114 mannosidase, alpha, class 1A, member 2 -2.68 Hs.226213 cytochrome P450, 51 (lanosterol 14-alpha-demethylase) -2.68 Hs.262869 type plasminogen -2.68 gb: BC006356.1 / DEF = Homo sapiens, NCX protein, clone MGC: 12870, mRNA, cds. complete -2.68 Hs.106823 H. sapiens gene of PAC 42616, similar to syntaxin 7 -2.68 Hs.294014 ESTs -2.65 Hs.75574 M12 mitochondrial ribosomal protein -2.65 Hs.168640 anachillosis, progressive (mouse) homologous -2.64 Hs.241493 natural killer tumor recognition sequence -2.62 Hs.100014 glutamate receptor, ionotrophic, AMPA 3 -2.62 Hs.2864 anterior endosome antigen 1, 162kD -2.62 Hs.79368 epithelial membrane protein 1 -2.60 Hs.13980 tetracyclopeptide repeat gene ubiquitously transcribed, X chromosome -2.60 Hs.28777 family H2A histone, member L -2.60 Hs.274131 gene 1 of critical region of Down syndrome - type2 Page 23 of 35 -2.58 Hs.170307 Guanine nucleotide exchange Ral factor RalGPS IA -2.58 Hs.94037 Hypothetical protein FLJ23053 -2.58 Hs.295923 Seventh in absence (Drosophila) homolog 1 -2.57 Hs.46821 Hypothetical protein FLJ20086 -2.56 Hs.144563 tryptophan hydroxylase (tryptophan 5-monooxygenase) -2.56 Hs.35091 Hypothetical protein FLJ 1 0775 -2.55 Hs.288931 Homo sapiens cDNA FLJ 1 3034 fis, clone NT2RP3001232 -2.55 Hs.1 71 545 H IV-1 Rev binding protein -2.53 Hs.59594 len = 529 -2.53 Hs.1 94669 zeste enhancer (Drosophila) homologue 1 -2.53 Hs.151 01 0 ESTs -2.52 Hs.6700 / len = 604 -2.51 Hs.207805 Homo sapiens mRNA; CDNA DKFZp5641 066 (of clone DKFZp5641 066) -2.49 Hs.1 65662 KIAA0675 product of gene -2.49 Hs.1 83291 protein 268 zinc finger -2.49 Hs.73742 ribosomal protein, long, PO -2.49 Hs.12533 Homo sapiens clone 23705 mRNA sequence -2.49 Hs.271 926 serologically defined colon cancer antigen 16 -2.48 Hs.74624 protein tyrosine phosphatase, receptor type, N polypeptide 2 -2.48 Hs.222306 hypothetical MGC3329 protein -2.47 Hs.966 coilin -2.47 Hs.1 58205 nuclear factor 1 basic leucine zipper (J EM-1) -2.45 Hs.271 699 polymerase (targeted DNA) iota -2.44 Hs.51 31 hypothetical FLJ20654 protein -2.43 Hs.237849 ESTs -2.42 Hs.32942 phosphoinositide-3-kinase, catalytic, gamma polypeptide H oja 24 d e 35 FIGURE 6 Table 4 Expression of the Differential Gene in Quimokinesis VS Gradients Fugetaxis SDF-1 Page 25 of 35 -2.34 Hs.278064 Homo sapiens cDNA: FLJ23327 fis, clone HEP12630, highly similar to HSZNF37 Homo sapiens ZNF37A mRNA for zinc finger protein -2.33 Hs.5022 printed in Prader-Willi syndrome -2.32 Hs.78946 cullin 3 -2.32 Hs.23240 Homo sapiens cDNA FLJ13496 fis, clone PLACE1004471, weakly similar to PROTEIN 83 DE DEDO de CINC -2.31 Hs.223241 factor 1 delta lengthening of eukaryotic translation (guanine nucleotide exchange protein) -2.30 Hs. 194148 v-yes-1 Yamaguchi viral sarcoma oncogene homolog 1 -2.30 gb: S82471.1 / DEF = associated box Homo sapiens Kruppel containing the product of gene SSX3 (SSX3) mRNA, cds. full -2.30 Hs.86434 hypothetical FLJ21816 protein -228 Hs.175941 BAP29 protein associated by the B cell receptor -2.26 Hs.96264 Thalassemiamental alpha-linked retardation syndrome X (RAD54 (S. cerevisiae) homolog) -2.26 Hs.75231 family 16 of solute carrier (monocarboxylic acid transporters), member 1 -2.26 Hs.69559 protein KIAA1096 -2.25 Hs.23964 associated polypeptide sin3, 18kD -2.25 Hs.94376 proprotein convertase subtilisincexin type 5 -2.25 Hs.62187 phosphatidylinositol glycan, class K -2.25 Hs.272534 Homo sapiens mRNA; CDNA DKFZp564J062 (from clone DKFZp564J062) -2.24 Hs.74861 cofactor 4 transcription polymerase 11 activated RNA -2.23 Hs.306602 Homo sapiens cDNA FLJ11514 fis, clone HEMBA1002229 Page 26 of 35 Page 27 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Quimoquinésis VS Gradients Fugetaxis SDF-1 Page 28 of 35 -2.01 Hs.300741 sorcin -2.01 Hs.91165 hypothetical protein -2.00 Hs.24485 sulfate proteoglican 6 chondroitin (hammock) -2.00 Hs.311 phosphoribosyl pyrophosphate amidotransferase -2.00 | Hs.296290 Homo sapiens cDNA FLJ13357 fis, clone PLACE1000061, weakly similar to the human protein ribosomal L37a Sequence mRNA -1.99 | Hs.251577 hemoglobin, alpha 1 -1.99 Hs.99847 peroxisome biogenesis factor 1 • 1.99 Hs.7194 protein CGl-74 • 1.98 Hs.39328 / len = 463 1.98 Hs.96063 insulin receptor substrate 1 • 1.98 Hs.93391 protein FLJ10539 hypothetical -1.98 | Hs.48950 novel gene-3 protein of heptacellular carcinoma -1.97 | Hs.13225 U DP-GaI: betaGlcNAc beta 1,4-galactosyltransferase, polypeptide 4 -1.96 | Hs.234757 Lipocortin Human (LIP) 2 pseudogene mRNA, full cds-type region -1.96 | Hs302114 Human DNA sequence of clone RP5-843L14 on chromosome 20. Contains ESTs, STSs and GSSs. It contains a novel gene and the 5 part of a gene for a novel protein similar to the linked 4 ribosomal protein X (RPS4X) Sheet 29 of 35 1.96 Hs.283753 cell cycle protein progression 8 • 1.95 I Hs.184050 homologue shrink viral of sarcoma 2 rat vK¡- ras2 Kirsten • 1.95 Hs.2074 zinc finger protein, X-bond -1.95 Hs.135202 promoter c-myc-binding protein -1.94 Hs.3530 TLS-serine associated -protein 2 arginine - 1.94 Hs.97681 DNA (cytosine-5 -) - methyItransferase 2 -1.94 Hs.12835 A kinase (PRKA) anchor protein 7 • 1.94 | Hs.1592 CDC16 (cell division cycle 16, S. cerevisiae, homologous) • 1.94 I Hs.325520 Homo sapiens IMAA mRNA for hLAT1-3TM, cds. complete -1.94 | Hs.20447 protein kinase related to S. cerevisiae STE20, effector for Cdc42Hs • 1.93 Hs.155995 protein KIAA0643 -1.91 Hs.4310 eukaryotic translation initiation factor-1A -1.91 Hs.20952 Homo sapiens clone 24411 Sequence mRNA • 191 Hs.16951 protein DKFZP586P2219 • 1.90 Hs.158205 nuclear leucine zipper factor 1 basic (JEM-1) -1.90 Hs.14968 gene 1 of denoma pleiomórfica -1.90 Hs.33363 protein DKFZP434N093 Sheet 30 of 35 Sheet 31 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Quimoquinésis VS Gradients Fugetaxis SDF-1 Sheet 32 of 35 Sheet 33 from 35 -1.75 Hs.180324 factor 2 YY1-associated • 1.75 Hs.240112 protein KIAA0276 -1.75 Hs.325667 TMTSP for the transmembrane molecule with thrombospondin module 1.74 Hs.83715 B antigen Sjogren syndrome (La autoantigen) • 1.74 Hs.6241 phosphoinositide-3-kinase, regulatory subunit, polypeptide 1 (p85 alpha) -1.74 Hs.127416 sinaptojanin 1 • 1.74 Hs.236642 3-hydroxylsobutyryl-Coenzyme A hydrolase 1.74 Hs.301800 Homo sapiens cDNA FLJ11568 fis, clone HEMBA1003278 • 1.74 Hs.247782 Human DNA sequence of clone 581F12 on chromosome Xq21. Contains EIF3 P35 Subunit of Eukaryotic Translation Initiation Factor and pseudogenes of Ribosomal Protein L22 60S. Contains ESTs • 1.74 Hs.30057 yeast-like transporter MRS2 -1.73 Hs.79078 MAD2 (yeast, poor homologous, mitotic arrest) -type1 -1.73 Hs.180919 2-DNA binding inhibitor, helix-loop protein-dominant negative helix 1.73 Hs.154740 protein G interactin TBP • 1.73 Hs.247309 succinate-ligase CoA, GDP formation, beta subunit -1.72 Hs.180895 Nuclearly directed protein to the putative brain • 1.72 Hs.84560 hypothetical FLJ11795 protein -1.72 Hs.249495 nuclear heterogeneous nuclear A1 ribonucleoprotein 1. 71 Hs.75140 Low density lipoprotin-related protein-associated protein 1 (alpha-2-receptor macroglobulin-associated protein-1) .71 Hs.285848 KIAA 1454 protein Sheet 34 of 35 FIGURE 6 Table 4 Expression of the Differential Gene in Quimokinesis VS Fugetaxis gradients SDF-1 Sheet 35 of 35 Figure 7 describes Table 5, which indicates genes that are differentially regulated in gradients of Medium against Chemotaxis of SDF-1. Positive values are overregulated in chemotaxis; the negative values are deregulated in chemotaxis; p < 0.05.
FIGURE 7 Table 5 Expression of the Differential Gene in Medium VS Gradients Chemotaxis SDF-1 78.70 Hs.80358 SMC (mouse) homolog, Chromosome Y 71.90 Hs.99120 DEADH (Asp-Glu-Ala-AspHis) polypeptide box, Chromosome Y 54.36 Hs.180911 ribosomal protein S4, Y-linked 29.71 Hs.193145 protease 9 specific ubiquitin, Y chromosome (related Drosophila fat aspects) 22.20 Hs.155103 eukaryotic translation initiation factor 1A, Y chromosome 18.91 Hs.155397 Homo sapiens mRNA; CDNA DKFZp584K143 (from clone DKFZp564K143) 16.39 Hs.73931 major histocompatibility complex, class 11, DQ beta 1 14.73 Hs.99120 DEADH (Asp-Glu-Ala-AspHis) polypeptide box, Chromosome Y 13.49 Hs.177605 C subfamily of murine cell lectin-like receptor, member 2 12.83 Hs.73931 major histocompatibility complex, class 11, DQ beta 1 10.71 Hs.155103 factor 1a of eukaryotic translation initiation 1 A, Y chromosome Leaf 1 of 13 Leaf 2 of 13 Leaf 3 of 13 2.85 Hs.46332 receptor 6 coupled to protein G 2.71 gb: NM 030773.1 / DEF = Homo sapiens beta tubulin 1, class VI (TUBB1), mRNA. 2.71 Hs.21486 signal transducer and transcription activator 1, 91 kD 2.64 Hs.103978 serinatreonin kinase 22B (associated spermiogenesis) 2.59 Hs.164960 BarH-box 1 homeotic type 2.58 Hs.82503 H. sapiens mRNA for 3UTR of unknown protein 2. 57 Hs.319088 Hypothetical protein FLJ10375 2.55 Hs.184915 zinc finger protein, Y-linked 2.49 Hs.37040 growth factor alpha polypeptide 2.44 Hs.23965 family 22 solute carrier (organic anion transporter), member 6 2.41 Hs.306618 Homo sapiens cDNA FLJ11930 fis, clone HEMBB1000441 Sheet 4 of 13 FIGURE 7 Table 5 Expression of the Differential Gene in Medium VS Gradients Chemotaxis SDF-1 Sheet 5 of 13 2.06 Hs.18586 product of gene KIAA0451 2.03 Hs.193606 clone RP5-1093017 PAC Homo sapiens of 7q11.23-q21 1. 99 Hs.146025 hypothetical FLJ23594 protein 1.96 Hs.33862 ESTs 1.96 Hs.274402 heat shock 70kD protein 1B 1.94 Hs.75887 coatomer protein complex, alpha subunit 1.92 Hs.197805 SRY (sex determination Y region) -box 30 1. 91 Hs.1521 Mu-binding protein 2 immunoglobulin 1.90 Hs.15087 product of KIAA0250 gene 1.88 Hs.167927 autoantigen 1 cell islet (69kD) 1.86 Hs.39733 postsynaptic CRIPT protein 1.85 Hs.76722 protein (CEBP) of improved CCAAT binding, delta 1. 84 Hs.265018 Hypothetical protein FLJ20635 1.81 Hs.239737 C-terminal link protein 1.80 Hs.33787 vinexm beta (molecular 1 adapter containing SH3) 1.80 Hs.279582 Sara protein GTP binding 1.80 Hs.324728 SMA5 1.79 Hs.44766 pigmentosa 2 retinitis (recessive X bond) 1.77 Hs.288940 five-interval transmembrane M83 protein 1. 74 Hs.247043 receptor of tumor necrosis factor type 1 that strips the regulator aminopeptidase 1.70 Hs.273099 Homo sapiens cDNA FLJ13712 fis, clone PLACE2000394 Page 6 of 13 FIGURE 7 Table 5 Expression of the Differential Gene in Medium VS Gradients Chemotaxis SDF-1 2.40 Hs.272268 Human DNA sequence of clone RP1-18C9 on chromosome 20 Contains part of a novel gene similar to acetyl-Coenzyme A synthetase, a novel gene (site D20S101) similar to Gamma-qlutamiltranspeptidase (Contains CCA trinucleotide repeat), a simile gene 2.39 Hs.2014 T cell receptor delta site 2.35 Hs.293205 ESTs, Weakly similar to BC39498 1 H. sapiens 2. 34 Hs.55481 zinc finger protein 165 2.31 Hs.122764 BRCA1-associated protein 2.31 Hs.299567 Receptor 44 coupled to G protein 2.28 Hs.3838 serum-inducible kinase 2.25 Hs.79019 baculoviral IAP repeat containing 1 2.25 gb: NM_030895. 1 / DEF = protein FLJ14129 (FLJ14129) hypothetical Homo sapiens, mRNA. 2.20 Hs.36972 CD7 antigen (p41) 2.20 Hs.306797 Homo sapiens cDNA: FLJ21648 fls, clone COL08469 2. 19 Hs.8077 Homo sapiens mRNA; CDNA DKFZp547E184 (from clone DKFZp547E184) 2.16 Hs.753 receptor 1 of the formyl peptide Sheet 7 of 13 Sheet 8 of 13 FIGURE 7 Table 5 Expression of the Differential Gene in Medium VS Gradients Chemotaxis SDF-1 REGULATED DESCENT IN CHEMOTHERAPY COMPARED IN MIDDLE OF GRADIENTS SDF-1 Sheet 9 of 13 Sheet 10 of 13 Sheet 11 of 13 FIGURE 7 Table 5 Expression of the Differential Gene in Medium VS Gradients Chemotaxis SDF-1 -2.48 Hs.19520 regulator 2 of ion transport containing the domain FXYD -2.47 Hs.77643 protein 1B of link FK506 (12.6kD) -2.45 Hs.193716 receptor 1 of the complement component (3b4b), including blood group system Knops -2.45 Hs.296355 Homo sapiens cDNA: FLJ23138 fls, clone LNG08913 -2.45 Hs.19131 transcription factor Dp-2. (E2F dimerization coadyutor 2) -2.45 Hs.274463 defensin, alpha 1, sequence related to myeloid -2.43 Hs.112049 link factor 1 SET -2.43 Hs.283664 beta-hydroxylase aspartate -2.42 Hs.121576 Homo sapiens cDNA FLJ20153 fis, clone COL08656, Highly similar to AJ001381 Homo sapiens incomplete cDNA for a mutant allele -2.37 Hs.287437 Homo sapiens cDNA FLJ11662 fis, clone HEMBA1004629 -2.35 Hs.21858 trinucleotide repeat containing 3 -2.35 Hs.23796 odz (odd Ozten-m, Drosophilia) homologue 1 -2.33 Hs.282344 Homo sapiens cDNA FLJ13387 fis, clACE PLACE1001136 -2.29 Hs.83623 sub-family 1 of the nuclear receiver, group 1, member 3 -2.29 Hs.31432 cardiac ankyrin repeat protein • 2.29 Hs.93758 Family histone H4, member H -2.28 Hs.3781 similar to repeat protein enriched for murine leucine -2.24 Hs.41716 endothelial cell specific molecule 1 Sheet 12 of 13 -2.24 Hs.307353 clone CIT987SK-44M2 of chromosome 16 BAC Homo sapiens -2.22 Hs.292863 ESTs -2.19 Hs.106552 Caspr2 molecule of cellular recognition -2.16 Hs.247910 clone N88K of donor N isolated Homo sapiens, variable chain mRNA region light kappa immunoglobulin, partial cds -2.14 Hs.93597 cyclin kinase 5 dependent, regulatory subunit 1 (P35) -2.07 Hs.283683 open reading structure 4 chromosome 8 -2.01 Hs.289056 ESTs, Highly similar to kininogen L 1312232A, NIW H. sapiens elevated -1.99 Hs.248190 UDP-N-acetyl-alpha-D-galactosamine: N-acetylgalactosaminyltransferase 4 polypeptide (GalNAc-T4) -1.95 Hs.324730 glutathione S-transferase MI -1.95 Hs.3628 protein kinase kinase kinase kinase 4 activated by mitogen • 1.94 Hs.181107 annexin A13 -1.94 Hs.66392 intersectin 1 (SH3 domain protein) -1.94 Hs.3781 leucine-rich repeat protein similar to murine -1.90 Hs.1265 dehydrogenase E1 chain keto acid branched, beta polypeptide (acrid syrup urine disease) -1.89 Hs.957 putative opioid receptor, neuromedin K (neurokinin B) receptor type -1.86 Hs.283330 hypothetical protein PR01843 -1.82 Hs.249727 protein hypothetical FLJ11798 -1.81 Hs.82685 CD47 antigen (antigen related to rH, signal transdutcor associated with integrin) -1.79 Hs.184860 protein CGI-203 -1.74 Hs.212587 Homo sapiens mRNA; CDNA DKFZp566M043 (from clone DKFZp566M043) Sheet 13 of 13 Figure 8 describes Table 6, which indicates genes that are differentially regulated in gradients of Medium vs. Fugetaxis of SDF-1. Positive values are overregulated in chemotaxis; the negative values are deregulated in chemotaxis; p < 0.05.
FIGURE 8 Table 6 Differential Gene Expression in Medium VS Gradients Fugetaxis SDF-1 REGULATED INCREMENT IN COMPARATIVE CHEMOTAXIS IN GRADIENT MEDIUMS SDF-1 45.94 Hs.80358 Homologous SMC (mouse) Chromosome Y 42.44 Hs.180911 Ribosomal S4 protein, Y link 28.32 Hs.99120 DEADH box polypeptide (Asp-Glu-Ala-AspHis), Y Chromosome 13.76 Hs.155397 Homo sapiens mRNA; CDNA DKFZp564K143 (from clone DKFZp564K143) 10.45 Hs.193145 protease 9 specific for ubiquitin, Chromosome Y (related Drosophila fat aspects) 10.07 Hs.155103 eukaryotic translation initiation factor 1a, Chromosome Y 8.93 Hs.78913 chemokine receptor 1 (C -X3-C) 8.52 Hs.2014 T cell receptor delta site 7.9 Hs.100000 S100 calcium binding protein A8 (calgranulin A) 7. 3 Hs.99120 DEADH box polypeptide (Asp-Glu-Ala-AspHis), Chromosome Y 6.58 Hs.3195 sub-family C of small inducible cytosine, member 1 (Lyfotactin) Page 1 of 43 6.36 Hs.73931 Main Histocompatibility Complex, Class II, DQ Beta 1 6.02 Hs.75184 Chitinase Type 3 1 (Glycoprotein-39 Cartilage) 5.57 Hs.76536 Transduction (Beta) -type 1 5.25 Hs.194366 transthyretin (prealbumin amyloidosis type 1) 5.1 Hs.19413 calcium binding protein A12 S100 (calgranulin C) . 07 Hs.251419 DNA sequence Homo sapiens of PAC 845024 on chromosome 1p36.1-36.2. It contains a gene for a protein HNRNP C1 LIKE of Nuclear Ribonucleoprotein and four genes similar to Melanoma Preferentially Expressed Antigen PRAME and counts KIAA0014. Conta 5.01 Hs.156110 kappa immunoglobulin constant 4.97 gb: AF262973.1 / DEF = 3DL1 receptor (KIR3DL1) mRNA, allele KIR3DL1 * 00701 cds. complete 4.9 Hs.326737 Homo sapiens clone MGC: 4655 mRNA cds. full 4. 84 Hs.50929 hypothetical FLJ13842 protein 4.72 Hs.57975 calsequestrin 2 (cardiac muscle) 4.54 Hs.7358 hypothetical FLJ13110 protein 4.51 Hs.177605 family C of murine cell lectin receptor-like, member 2 4.44 Hs.79691 protein domain L 4.33 Hs.37142 efrin-A5 4.31 Hs.198396 ATP binding cassette, sub-family A (ABC1), member 4 Page 2 of 43 Page 3 of 43 Sheet 4 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 3.53 Hs.169910 product of KIAA0173 gene 3.52 Hs.250502 anhydrase carbonic acid 3.5 Hs.2352 adenylate cyclase 2 (brain) 3.5 gb: M32577. 1 / DEF = Human MHC HLA-DQ beta mRNA, cds. Full 3.48 Hs.269926 Homo sapiens cDNA: FLJ21441 fis, clone COL04422 3. 47 Hs.155103 eukaryotic translation initiation factor 1A, Chromosome Y 3.45 Hs.171814 parathymosin 3.42 Hs.203846 member 3 of the TEA domain family 3.42 Hs.21425-hydroxytryptamine (serotonin) receptor 3A 3.38 Hs.14642 structure 3 reading open chromosome 16 3. 38 Hs.76722 protein (CEBP) of the CCAAT enhancer link, delta 3. 36 Hs.64311 a domain 17 of metalloproteinase and disintegrin (tumor necrosis factor, alpha, conversion enzyme) 3.35 Hs.177961 clone CIT987SK-A-388D4 of Human Cromosome of 16 BAC 3.35 Hs.153985 family 7 of the solute carrier ( cationic amino acid transporter, system y +), member 2 Page 5 of 43 Page 6 of 43 Hs.58014 G protein coupled receptor, family C, group 5, member C 2. 99 Hs.118695 Channel potassium voltage controller, subfamily G. member 1 2. 99 Hs.247741 protocaderin alfa 2 2.98 Hs.13040 Receptor 86 coupled protein G 2.98 Hs.69319 CA11 2.98 Hs.146409 cycle 42 cell division (GTP binding protein, 25kD) 2. 97 Hs.79876 spheroid sulfatase (microsomal), arylsulfatase C, isosima S 2. 96 Hs.325722 variable 3D-15 immunoglobulin kappa 2.93 Hs.284277 antibody MO30 (IgM) mRNA chain mu immunoglobulin Homo sapiens, cds. full 2.87 gb: AF349720.1 DEF = beta (TRO) magfinin Homo sapiens, cds. Complete 2.82 Hs.79706 plectin 1, intermediate filament binding protein, 500kD 2.79 Hs.694 interleukin 3 (colony stimulation factor, multiple) 2. 79 Hs.131361 pyruvate dehydrogenase (lipoamide) alpha 2 2.74 Hs.265848 myomegalin similar to rat 2.72 Hs.2742303-phosphoadenosin 5-phosphosulfate synthase 2 2.72 Hs.306643 Homo sapiens cDNA FLJ13302-fis, clone OVARC1001357 2. 71 Hs.153837 Myeloid nuclear cell differentiation antigen 2. 71 Hs.621 lectin, galactoside link, soluble, 3 (galectin 3) 2. 69 Hs.227751 lectin, galactoside link, soluble, 1 (galectin 1) 2. 69 Hs.84152 cystathionine beta-synthase 2.69 Hs.48778 niban protein 2.68 Hs.158315 accessory protein 18 receptor interleukin 2. 67 Hs.8074 inhibitor 3 of brain-specific angiogenesis 2. 66 Hs.1915 folate hydrolase (specific cell membrane antigen) 1 Page 7 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Sheet 8 of 43 Page 9 of 43 Page 10 of 43 Sheet 11 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Page 12 of 43 2.12 Hs.821 biglican 2.11 Hs.288771 protein DKFZP586A0522 2.1 Hs.225641 Hypothetical protein FLJ13Í71 2.1 Hs.75825 genome pleomorphic adenoma-tipol 2.09 Hs.241570 tumor necrosis factor (TNF superfamily, member 2) 2.08 Hs.727 inhibin, beta A (activin A, activin AB alpha polypeptide) 2.08 Hs.164371 protein hypothetical FLJ12439 2.08 Hs.94210 homolog 1 (Drosophila) absent from eyes 2.08 Hs.306455 Homo sapiens mRNA; DKFZp434K1126 cDNA (from the clone DKFZp434K1126) 2.07 Hs.69049 tocopherol (alpha) transfer protein (ataxia (similar to Friedreich) with vitamin E deficiency) 2.07 Hs.181353 UDP-Gai: betaGlcNAc beta 1, 3-qalactosiItransferase, polypeptide 2 2.06 Hs.171811 adenylate kinase 2 2.05 Hs.84298 CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated) 2.04 gb: K03226.1 / DEF = Human preprourokinase mRNA, cds. full 2. 04 Hs.858 Vi viral oncogene homologous viral avian reticuloendotheliosis v-rel (nuclear factor enhancer light kappa gene in 3 B cells) 2.03 Hs.41707 protein 327kD heat attack Sheet 13 of 43 Sheet 14 of 43 1.99 Hs.6580 Homo sapiens cDNA: FLJ23227 fis, clone CAE00645, Highly similar to AF052138 Homo sapiens clone 23718 Sequence mRNA 1.98 Hs.35101 Polypeptide 2 of proline-rich Gla (G-carboxyglutamic acid) 1.98 Hs.41270 Procollagen-lysine, 2-oxogyutarate 5-dioxygenase (lysine hydroxylase) 2 1.97 Hs.13046 thioredoxin reductase 1 1.97 Hs.2667 metallothionein 1 H 1.96 Hs.1481 histidine decarboxylase 1.96 Hs.100194 activation protein of 5-lipoxygenase arachidonate 1.95 Hs.41143 phospholipase C-beta 1 specific for inositide 1.94 Hs.166072 annexin A2 pseudogen 2 Sheet 15 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Sheet 16 of 43 Sheet 17 of 43 Sheet 18 of 43 1.77 Hs.195175 CASP8 and FADD-type of apoptosis regulator 1. 77 Hs.248183 olfactory receptor, family 1, subfamily G. member 1 1.77 Hs.273294 hypothetical protein FLJ20069 1.77 KIAA0674 protein 1.76 Growth disruption and DNA-damaged-inducible 34 1.76 Hs.11091 5 Interleukin 22 receptor 1.76 Hs.102471 KIAA0680 Gene product 1.75 Hs.75825 genome pleiomorphic adenoma-tipol Sheet 19 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Sheet 20 of 43 Sheet 21 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 REGULATED DESCENSION IN FUGETAXIS COMPARED TO GRADIENTS MEDIUM SDF-1 Sheet 22 of 43 Leaf 23 of 43 -4.97 Hs.79340 PTH-respective osteosarcoma protein B1 -4.96 Hs.36977 hemoglobin, delta -4.92 Hs.278483 Histone family H4, member E -4.9 Hs.97176 hypothetical protein FLJ13906 similar to finger protein RING - 4.9 Hs.2399 metalloproteinase 14 matrix (membrane-inserted) -4.85 Hs.78146 molecule (CD31 antigen) plateletendothelial cell adhesion -4.8 Hs.12820 SnRNP defective homologue 1 assembly -4.8 Hs.129903 polymerase (DNA-administered), lambda -4.8 Hs.7943 protein RPB5-mediated -4.78 Hs. 326457 ESTs -4.75 Hs.325530 KIAA1067 protein -4.64 Hs.197335 plasma glutamate carboxypeptidase -4.56 Hs.6092 f-box and repeat protein rich in leucine -4.55 Hs.159241 polycystic kidney disease 2 type -4.47 Hs. 99987 transverse complementation rodent repair deficiency - excision repair, group 2 complementation (xeroderma pigmentosum D) -4.45 Hs.155204 protein 174 zinc finger -4.42 Hs.11135 major histocompatibility complex, class 11, alpha DN -4.39 Hs.20017 chromosome 22 structure 4 open reading Sheet 24 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 -4.39 gb: M24668.1 / DEF = Human Ig redisposed H chain, V-region mRNA (CD-JH4), cds. complete -4.31 Hs.156115 voltage controlled channel potassium, KQT subfamily type, member 1 -4.28 Hs.97574 component Rrp41 exosome -4.28 Hs.168737 ESTs, Highly similar to REGULATORY SUBUNITY KDA - PROTEINPHOSPHATASE 2A, 65 DE SERINETREONINE 2AAB_HUMANA, BETA ISOFORM H. sapiens -4.22 Hs.300772 tropomyosin 2 (beta) -4.2 Hs.283404 organic cation transporter -4.19 Hs.103839 band 4.1 erythrocyte membrane protein-type 3 -4.18 Hs.101874 double mouse minute 4, human homologue of; p53 link protein -4.13 Hs.33818 Protein RecQ-type 5 -4.1 Hs.121102 vanina 2 -4.09 Hs.22370 Homo sapiens mRNA; CDNA DKFZp56400122 (clone DKFZp56400122) -4.06 Hs.36 alpha limfotoxin (TNF superfamily, member 1) -4.06 M10098 Human 18S rRNA sequence, length 1969 bases, middle object bases 647-1292 -4.05 Hs.79386 leiomodin 1 (smooth muscle ) Sheet 25 of 43 -4.02 Hs.110796 protein SAR1 -4.01 Hs.14286 flavin containing monooxygenase 5 -3.99 Hs.272108 ESTs -3.98 Hs.112751 protein KIAA0892 -3.98 Hs.47822 guanine exchange Rho (GEF) factor 11 -3.96 Hs.90443 dehydrogenase NADH (ubiquinone) Fe-S protein 8 (23kD) (NADH-Coenzyme Q reductase) -3.94 Hs.7426 protein KIAA0841 -3.9 Hs.154085 protein 1 zipper leucine -3.89 gb: NM_030925.1 IDEF = Homo sapiens hypothetical FLJ12577 protein (FLJ12577), mRNA. -3.86 Hs.102867 3 high-affinity sodium-dependent affinity dicarboxylate transporter -3.81 gb: BC006441.1 / DEF = Homo sapiens, Similar to the RRN3 factor of polymerase I RNA, clone MGC: 13169, mRNA, cds. full -3.81 Hs.9857 carbonyl reductase -3.79 Hs.119498 interactor 6 of the thyroid hormone receptor -3.77 Hs.194148 v-yes-1 homologue 1 viral oncogene of the Yamaguchi sarcoma -3.76 Hs.277401 Bromodomain adjacent to zinc finger domain, 2A -3.75 Hs.78921 kinase anchor protein 1 (PRKA) -3.74 Hs.278064 Homo sapiens cDNA: FLJ23327 fis, clone HEP12630, highly similar to HSZNF37 Homo sapiens ZNF37A mRNA for zinc finger protein -3.73 gb: NM_030930.1 / DEF = Homo sapiens unc93 (C.elegans) homolog B (UNC93B), mRNA. Sheet 26 of 43 -3.7 Hs.1 265 dehydrogenase E 1 of branched chain keto acid, beta polypeptide (maple syrup-urine disease) -3.69 Hs.48269 q related to vaccinia -3.66 Hs.1 68670 protein famesylated peroxisomal -3.66 Hs.1 55597 component D of the complement (adipsin) -3.66 Hs.291 972 ESTs, Moderately similar to SC14_H U MAN SEC 14- LI KE PROTE INH. sapiens -3.64 Hs.1 3405 gephyrin -3.64 Hs. 701 9 gene 1 associated with the proliferation of signal induction -3.61 Hs.285005 Tom22 receptor I mitochondrial carrier -3.61 Hs .21 0546 interleukin-21 receptor -3.58 KIAA1 1 1 7 protein -3.57 Hs.44865 limfoid enhancer-1 binding factor -3.57 Hs.23585 KIAA1 protein 078 -3.56 Hs.14846 Homo sapiens mRNA; CDNA DKFZp564D016 (from clone DKFZp564D016) -3.56 Hs.47344 advillina -3.55 Hs.296821 gene region (FSH D) human facioscapulohumeral muscular dystrophy, tandem D4Z4 repeat unit -3.53 Hs.2558 gamma-carboxyglutamate bone (gla) protein (osteocalcin) -3.52 Hs.80741 propionyl Coenzyme A carboxylase, alpha polypeptide -3.47 Hs.59544 transverse complementation rodent repair deficiency- excision repair, complementing group 1 (includes overlapping antisense sequence) -3.47 Hs.248007 actin (ACTBP9) beta-cytoplasmic human pseudogene -3.47 Hs.300496 carrier of mitochondrial solution -3.45 Hs.1 1 1 244 hypothetical protein -3.44 Hs.193716 receptor 1 (3b4b) of complement component, including Blood group system -3.37 Hs.79064 deoxyhipusinsynthase H oja 27 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 -3.36 Hs.5378 Spondine 1, (spondine-f) extracellular matrix protein -3.35 Hs.94229 hypothetical protein FLJ11939 -3.33 gb: M24669.1 / DEF = lg Human rearrangement of H chain, V region mRNA (C-D-JH6), cds. complete -3.33 Hs.16193 Homo sapiens mRNA: cDNA DKFZp586B211 (clone DKFZp586B211) -3.32 Hs.5353 caspase 10, cysteine protease-related apoptosis -3.32 Hs.117242 Meningioma expressed antigen 6 (rich proline-rusked loop) -3.3 Hs.5378 spondine 1, (spondin-f) extracellular matrix protein -3.29 Hs 203269 ESTs, Moderately similar to ENTRY OF CONTAMINATION ALERT OF SEQUENCE OF SUBFAMILY ALU8_HUMAN ALU SUBFAMILY SX H-Sapiens -3.27 Hs.184523 protein KIAA0965 -3.27 Homo sapiens chromosome 19, cosmic R28784, complete sequence. -3.26 Hs.21542 protein KIAA1035 -3.26 Hs.83765 dihydrofolate reductase -3.25 Hs.283860 Homo sapiens partial mRNA for MOZCBP chimeric transcription type II Leaf 28 of 43 -3.24 Hs.168625 Protein associated with prostatic proliferative disruption induced by androgen -3.24 Hs.9846 protein KIAA1040 -3.23 Hs.104916 protein hypothetical FLJ21940 -3.22 gb: BC006222.1 / DEF = Homo sapiens, clone MGC: 10279, mRNA, cds. complete -3.21 Hs.73980 troponin T1, skeletal, slow -3.2 Hs.85195 factor 1 of myeloid leukemia -3.19 Hs.288697 hypothetical protein MGC11349 -3.17 Hs.26899 product of gene KIAA0285 -3.17 Hs.262869 type plasminogen -3.16 Hs. 226581 COX15 (yeast) homologue, cytochrome oxidase assembly protein c -3.16 Hs.4854 inhibitor or cyclin-dependent kinase 2C (p18, CDK4 inhibitors) -3.15 Hs.184938 Mapping of the novel human gene to chromosome 13 -3.15 Hs.44697 ATPase, Class V, type 10C -3.14 Hs.25155 neuroepithelial cell transformation gene -3.13 Hs.267263 hypothetical protein -3.13 Hs.21361 protein KIAA1023 -3.11 Hs.180686 ligase E3A protein ubiquitin (protein associated by the E6 virus of the human papilloma, Angelman syndrome) -3.11 Hs.86178-phase M of phosphoprotein 9 -3.1 Sheet 29 of 43 -3.08 Hs.31324 protein 155 of zinc finger (pHk-96) -3.06 Hs.61712 pyruvate of kinase dehydrogenase, isoenzyme 1 -3.06 Hs.103839 band 4.1 erythrocyte membrane protein-type 3 -3.06 Hs.292998 ESTs -3.05 Hs.31476 Homo sapiens cDNA FLJ13872 nS, clone THYRO1001322 -3.04 Hs.26471 Homo sapiens clone HQ0692 -3.04 Hs.100602 MAD (mothers against decapentaplégico, Drosofila) homologue 7 -3.03 Hs.79993 factor 7 of peroxisomal biogenesis -3.03 Hs.82919 culilna 2 -3.02 Hs.1975 hypothetical protein FLJ21007 - 3.01 Hs.118738 product of gene KIAA0800 -3 Hs.222306 protein hypothetical MGC3329 -2.99 Hs.100090 tetraspan 3 -2.98 Hs.18889 Protein DKFZP434M183 -2.98 Hs.20019 hemochromatosis -2.96 Hs.21811 protein hypothetical FLJ10374 -2.92 Hs.308332 ESTs , Highly similar to protein A42735 ribosomal L10, H. sapiens cytosolic -2.9 Hs.9003 protein hypothetical FLJ13868 -2.89 Hs.234265 protein DKFZP586G011 -2.88 Hs.26468 precursor protein binding amyloid precursor beta (A4), family A, member 2 (type-X11) -2.85 Hs.68398 period (Drosophila) homologue 1 -2.84 Hs.153639 hypothetical SBBI03 protein -2.84 Hs.184019 Homo sapiens clone 23551 mRNA sequence -2.82 Hs.5378 spondine 1, (spondin-f) protein extracellular matrix -2.8 Hs.159900 receptor 15 coupled to Protein G Page 30 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Page 31 of 43 -2.66 Hs.142245 HERV-H LTR-association 3 -2.65 Hs.283032 Hypothetical protein PRO2015 -2.63 Hs.182595 dynein, axonemal, light polypeptide -2.63 Hs.9071 progesterone membrane binding protein -2.62 gb: U31110.1 / DEF = Human protein trp-1 and protein trp-1 alternatively spliced and (trp-1) mRNA not spliced, cds. full -2.62 Hs.168670 Farnesylated peroxisomal protein -2.6 Hs.66191 Homo sapiens clone 24675 mRNA sequence -2.6 Hs.9196 Hypothetical protein -2.58 Hs.184376 sinaptosomal-associated protein, 23kD -2.58 Hs.27610 retinoic acid- and protein (58kD) inducible interferon -2.57 Hs.77868 ORF -2.57 Hs.77152 deficient maintenance of minichromosome (S. cerevisiae) 7 -2.56 Hs.115537 putative dipeptidase -2.54 Hs.2006 glutathione S-transferase M3 (brain) -2.54 Hs.7854 type transporter regulated by zinciron -2.53 Hs.19561 dehydrogenase NADH (ubiquinone) subcomplex alpha 1, 7 (14.5kD, B14.5a) -2.53 Hs.72980 Protein P3 -2.53 Hs.262023 Homo sapiens mRNA cDNA DKFZp564N1623 (from clone DKFZp564N1623); cds. complete -2.53 Hs.79368 epithelial membrane protein 1 sheet 32 of 43 -2.52 Hs.23964 sin3-associated polypeptide, 18kD -2.51 Hs.966 coilin -2.5 Hs.158982,4-dienoyl CoA reductase 2, peroxisomal -2.49 Hs. 118722 fucosyltransferase 8 (alpha (1,6) fucosyltransferase -2.49 Hs.278503 regulated in glioma -2.48 Hs.180338 superfamily of the necrosis factor receptor, member 12 (transposition of membrane protein chain association) -2.48 Hs.46465 T-cell, immune regulator 1 -2.48 Hs.100915 factor 16 peroxisomal biogenesis -2.48 Hs.119274 RAS protein activator p21 (GTPase activation protein) 3 (lns (1, 3,4,5) P4 -protein binding) -2.46 Hs.50748 chromosome 21 structure 18 open reading -2.46 Hs.25155 gene 1 transforming neuroepithelial cell -2.46 Hs.108779 protein DKFZP586E1519 -2.46 Hs.82527 sialyltransferase 8 (alpha-N-acetyl) lneuraminate: alpha-2, 8-sialitransferase, GD3 synthase) A -2.46 Hs.99491 RAS guanil release protein 2 (calcium and DAG-regulated) -2.44 Hs.143131 glycoprotein A33 (transmembrane) -2.42 gb: BC006332.1 / DEF = Homo sapiens, clathrin, light polypeptide (Lcb), clone MGC: 12930, mRNA, cds. Complete -2.4 Hs.7594 family 2 soluble carrier (assisted glucose transporter), member 3 -2.4 Hs.15984 pp21 homologous -2.39 Hs.78056 cathepsin L -2.39 Hs.152981 CDP-diacylglycerol synthase (phosphatidate cytidyl-transferase) 1 -2.39 Hs.271699 polymerase (direct DNA) particle Sheet 33 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Sheet 34 of 43 -2.28 Hs.285737 Homo sapiens cDNA: FLJ20895 fis, clone ADKA03483 -2.27 Hs.183291 protein 268 zinc finger -2.27 Hs.82919 culilna 2 -2.26 Hs.5881 ELL gene (leukemia gene 11-19 lysine-enriched) -2.26 Hs.294014 ESTs -2.25 Hs.62187 glycan phosphatidylinositol, class K -2.24 Hs.1117 tripeptidyl peptidase 11 -2.22 Hs.153299 DOM-3 (C. elegans) homologue Z -2.22 Hs.250619 protein MDS019 phorboline type -2.22 Hs.301114 zinc finger protein 79 (pT7) -2.22 Hs. 300741 sorcin -2.21 Hs.295923 seven in the absence of (Drosophila) homologue 1 -2.21 Hs.17775 executor of cell death associated with p75NTR; ovarian granulosa cell protein (13kD) -2.2 Hs.174185 ectonucleotide pyrophosphatase phosphodiesterase 2 (autotaxin) -2.19 Hs.2864 previous endosome antigen 1, 162kD -2.19 Hs.321567 complexin 2 -2.19 Hs.31432 cardiac ankyrin repeat protein -2.18 Hs.77508 glutamate dehydrogenase 1 -2.18 Hs.293495 ESTs, weakly similar to ALU1_HUMAN ALU SUBFAMILIY J SECUENCE CONTAMINATED WARNING ENTRY H.sapiens -2.17 Hs.48924 KIAA0512 gene product; ALEX2 Sheet 35 of 43 Sheet 36 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 Leaf 37 of 43 -2.01 Hs.16193 Homo sapiens mRNA cDNA DKFZp586B211 (from clone DKFZp586B211) -2 Hs.7158 protein DKFZP566H073 Hs.5002 copper chaperone for superoxide dismutase Hs.279777 hypothetical protein 1.99 Hs.264330 N-acylesphingosine amidohydrolase type ( ceramidase acid) -1.97 Hs.9456 SWISNF related, associated matrix, chromatin actin-dependent regulator, subfamily a, member 5 -1.97 Hs302114 Human DNA sequence of clone RP5-843L14 on chromosome 20. Contains ESTs, STSs and GSSs. It contains a novel gene and part 5 of a gene for a new protein similar to the X bond of ribosomal protein 4 (RPS4X) -1.97 Hs.102 aminomethyltransferase (glycine cleavage system of T protein) • 1.97 Hs.75790 phosphatidylinositol glycan, class C -1.96 Hs.180338 superfamily of tumor necrosis factor receptor, member 12 (transplacement of chain association membrane protein) -1.95 Hs.81687 non-metastatic cells 3, protein expressed in • 1.93 Hs.31659 thyroid hormone receptor associated protein, 95-kD subunit • 1.92 Hs.99847 peroxisome biogenesis factor 1 -1.92 Hs.46736 hypothetical protein FLJ23476 • 1.91 Hs.8198 zinc finger protein 204 Sheet 38 of 43 Page 39 of 43 -1.85 Hs.7194 protein CGI-74 -1.84 Hs.322478 protein KIAA0117 • 1.84 Hs.12835 A kinase (PRKA) anchoring protein 7 • 1.83 Hs.43803 receptor 2 of Ig type associated by leukocyte -1.83 Hs .66708 membrane-associated protein 3 per vesicle (celubrevine) -1.82 Hs.249495 heterogeneous nuclear ribonucleoprotein A1 Sheet 40 of 43 FIGURE 8 Table 6 Expression of the Differential Gene in Medium VS Gradients Fugetaxis SDF-1 -1.82 Hs.266933 domain hect and RLD 2 -1.81 Hs.119000 actinin, alpha 1 1.81 Hs.177486 beta amyloid precursor protein (A4) (nexin-ll protease, Alzheimer's disease) • 1.8 Hs.300684 protein of the peptide receptor component related to the calcitonin gene -1.8 Hs.18490 protein FLJ20452 hypothetical 1.8 Hs.279785 putative secreted protein -1.8 Hs.17409 protein 1 enriched for cysteine (intestinal) -1.79 Hs.301201 Homo sapiens cDNA FLJ14152 fis, clone MAMMA1003089 -1.79 Hs.16803 LUC7 type (S. cerevisiae) • 1.79 Hs. 265561 CD2-associated protein • 1.79 Hs.30696 transcription factor-type 5 (helix-loop-basic helix) • 1.79 Hs.46907 / len = 607 -1.78 Hs.240112 protein KIAA0276 • 1.78 Hs.41072 serine (or cysteine) proteinase inhibitor, clade B (ovalbumin), member 6 Sheet 41 from 43 -1.77 Hs.330056 Hypothetical protein FLJ22795 -1.77 Hs.5353 caspase 10, cistern protease-related apoptosis -1.77 Hs.75061 macrophage mylanine-rich alanine-rich C kinase substrate • 1.77 gb.NM_031214.1 / DEF = Homo sapiens hypothetical protein (AF311304), mRNA. -1.76 Hs.286848 protein KIAA1454 -1.76 Hs.75692 asparagine synthetase -1.75 Hs.36972 CD7 antigen (p41) -1.75 Hs.83958 improved type transducin separation 4, homolog of Drosophila E (sp1) -1.75 Hs.64310 receptor interleukin 11, alpha -1.74 Hs.198726 receptor 1 of the vasaoactive intestinal peptide -1.74 Hs.194329 protein hypothetical FLJ21174 -1.74 Hs.102456 motor neuron protein survival that interacts with protein 1 -1.74 Hs.26471 Homo sapiens clone HQ0692 1.74 Homo sapiens Chromosome 16 BAC clone CIT987SK-A-67A1, complete sequence. • 1.74 Hs.158195 heat shock transcription factor -1.74 Hs.306173 phosphatidylinositol glycan, class C, pseudogene 1 -1.73 Hs.9452 KIAA0770 protein -1.73 Hs.31834 Homo sapiens clone 25129 mRNA sequence -1.73 Hs.77602 protein 2 DNA link (48kD) specific damage Sheet 42 of 43 Sheet 43 of 43 Figure 9 describes Table 7, which indicates extracellular matrix / adhesion of actin / cytoskeleton, proteins related to the activation and migration of T-cell differentially regulated under different gradient conditions of SDF-1.
FIGURE 9 Table 7 Chemotaxis against Fugetaxis: Transcriptional changes downstream Actin / Cistoscepletal Increased in Increased Chemotaxis in Fugetaxis NDE / Increased Adhesion in Increased Chemotaxis in Fugetaxis Activation of Increased T Cell in Increased Chemotaxis in Fugetaxis Related Migration Increased in Increased Chemotaxis in Fugetaxis Figures 10A to 10P describe the migration of human neutrophils in a continuous linear gradient (0.12 nm, 120 nM or 1.2 mM) of L-8 in microfabricated devices. Cell migration at uniform concentrations or continuous gradients of IL-8 (screened with the assistance of MetaMorph software) is described in Figures 10E to 10H. The normalized cell concentration through the migration channel (measured by MetaMorph software) is described in Figures 101 through L. The distribution of the angles of the motion vector for all cells for all time points is described in the Figures 10M to P. A ') Without chemokine; B ') Position in channel (um); C) Initial position and tracks; D ') Normalized cell concentration; E ') Frequency angle. Figures 11A and 11B describe batches of average velocities (11A) and average quadratic displacement (11B) for cells tracked over time in videos of cells migrating in the absence of I L-8 or defined as continuous linear gradients of chemokine in peak concentrations of 12nM, 120nM and 1.2 mM. A) Mean neutrophil migration rates; B) Average speed (um / min); C) Mean square displacement of neutrophil migration; D) Mean square displacement; E) Time. Figure 12 describes the effect of SB225002 on a directional migration of neutrophils into and out of IL-8. A) Effect of SB225002 on Fugetaxis induced by IL-8; B) Normalized angle frequency radio (30 to 150 degrees); C) Effect of SB225002 on chemotaxis induced by I L-8; D) Normalized angle frequency radio (210 to 330 degrees); E) Values P- 'Figure 13 describes effects of chemokine signal transduction pathway inhibitors on directional human neutrophil migration in defined continuous gradients of IL-8. A) Intracellular signal transduction inhibitors in bi-directional neutrophil migration; B) Normalized angle radiofrequency; C) Toxin pertussis I L-8 120 nM; D) Wortmanin I L-8 120 nM; E) Wortmanin I L-8 1.2 μM; F) Experimental conditions; G) Chemotaxis; H) Fugetaxis. Figures 14A to 141 describe intravital microscopic quantification of rat neutrophil migration in response to continuous diffusive gradients of the IL-8 ortholog, CINC-1. The continuous diffusive gradients are mathematically modeled and described in Figures 14A, 14B and 14C. A simple photomicrograph derived from the first frame of the time-lapse video is described in 14D (Video 5), 14E (Video 6) and 14F (Video 7). Figures 14G, 14H and 141 describe traces of cells normalized to an original and again use the same color code as in Figure 14 for directional and random cell movement. A) Capillary wall distance (μm); B) [CINC-1] predicted (nM). Figure 15 describes quantitative parameters defined by measuring the directional inclination and orientation of the cell movement of the cells traced in neutrophil videos that migrate in the absence of I L-8 (No. IL-8), a constant concentration of chemokine (120 nM I L-8 without gradient), and three continuous linear gradient conditions with peak concentrations of IL-8, 12nM, 120nM and 1.2 mM within the microfabricated devices (Table 8). r in or in FIGURE 15 TABLE 8 r or in Figure 16 describes quantified motility parameters for rat neutrophils that migrate in response to continuous diffusive CIN-1 gradients in vivo (Table 9).
FIGURE 16 TABLE 9 r 00 DETAILED DESCRIPTION OF THE INVENTION The invention is theorized in part in the discovery that cells exposed to a gradient undergo changes in gene expression associated with the presence of the gradient and movement through the gradient. It has unexpectedly developed that exposure of cells to an agent gradient causes differential gene expression in cells thus exposed as compared to cells exposed to a uniform agent concentration (i.e., no gradient). As a result, the profiles of genetic expression during or after exposure to gradients are significantly different from those observed during or after exposure to concentrations of uniform agents. In addition, gene expression profiles are dependent on the structure of the gradient. That is, if the gradient is oriented so that the cell is attracted to a source of the agent (an attractant gradient or a chemoattractant), the gene expression profile will be different than if the gradient is oriented in such a way that the cell is repels from the source of the agent (a gradient or fugactic agent). The gene expression profiles for cells exposed to a fugectic gradient are clearly different from those seen in the chemotactic gradients. As an example, when a cell is exposed to a SDF-1 gradient (CXCL12), it begins to differentially express genes involved in chemokine signal transduction depending on whether it migrates to or out of the source of the agent.
Definitions: As used in accordance with the terms appearing herein, the following definitions are provided: An "agent" is a disseminable substance that can alter expression in a migratory cell, either alone or in combination with other agents. Preferably, the agent is an attractant or repellent of a migratory cell. An "agent concentration gradient" is a gradually increased concentration of an agent, wherein the location of the highest agent concentration is at the source of the agent. A "continuous gradient" is a continuous, physiologically relevant agent concentration over a fixed distance. A "cap gradient" comprises agent concentrations that descend or rise abruptly to another concentration of the agent. A "source of the agent" is the point at which the concentration of an agent is highest. Since a cell migrates to the source, it moves toward the highest agent concentration, and since it migrates out of the source, it moves toward the lowest agent concentration.
A "ligand" is a molecule, such as a protein, lipid or cation, capable of binding to another molecule for which it has an affinity, such as a receptor. A ligand is therefore a member of the binding interaction or association. "Chemotactic migration" or "chemotaxis" is the movement of a migratory cell towards a source of the agent (ie, towards a higher concentration of the agent). "Fugectic migration" or "fugetaxis" is the movement of a migratory cell out of a source of the agent (that is, toward a lower concentration of the agent). "Chemokinetic migration" or "chemokinesis" is the random movement of the irrespective cells of a gradient. A "cytosine" is the genetic term for all proteins or extracellular peptides that mediate cell-to-cell communication, often with the effect of altering the activation state of cells. A "chemokine" is a cytosine with a conserved cysteine motif and which can serve as an attractant. A "signaling molecule" is a molecule involved in the transduction of a signal cascade from one compartment of the cell to another (e.g., in the case of cell movement, a signaling molecule can be involved in the transduction of a signal cascade from the cell membrane to the actin cytoskeleton). A "cytoskeleton-related molecule" is a component of the cytoskeleton, which is a system of protein filaments (eg, actin filaments, integrins, microtubules and intermediate filaments) in the cytoplasm of a eukaryotic cell that gives shape and capacity of cell movement. A "cell cycle molecule" is a molecule involved to regulate, initiate or paralyze the reproductive cycle of a cell, which is the cycle by which a cell doubles its contents and divides them into two. A "molecule related to the extracellular matrix" is a molecule that is a component of the extracellular matrix, which is a network of structural elements, such as polysaccharides and proteins, secreted by the cells. A "molecule related to immune response" is a molecule involved in the generation, propagation or termination of an immune response, which is a response by a cell immune to an antigen. An "immune cell" is a cell of hematopoietic origin that is involved in the specific recognition of antigens. Immune cells include, but are not limited to T cells, B cells, NK cells, dendritic cells, monocytes and macrophages. "Primary cells" are cells obtained directly from living tissue or damaged tissue. An "inflammatory cell" is a cell that contributes to an immune response, including, but not limited to, neutrophils, basophils, eosinophils, and mast cells. The definitions and additional descriptions appear in the following context. Other aspects of the invention are described in, or are obvious from, the following description and are within the scope of the invention.
Methods of the Invention The methods of the invention can be used to determine the differences between cells undergoing chemotaxis against those undergoing fugetaxis, or difference between cells undergoing chemotaxis or fugetaxis against those undergoing chemokinesis (ie, random movement). In some cases, the profiles of gene expression of cells that undergo chemokinesis are considered "antecedents" and thus are deduced from the profiles of chemiotactic or fugectic gene expression. These expression differences also identify mediators of chemotaxis and fugetaxis and provide novel targets that can be affected to modulate directed cell movement. In some cases, these newly identified targets can be delivered to cells directly. Alternatively, newly identified targets can be upregulated or deregulated in ways that are independent of current exposure to a chemotactic or fugactic gradient. These include the introduction of nucleic acids into cells (e.g., antisense or gene therapy) and exposure of cells to compounds that modulate newly identified targets (e.g., agonists or antagonists). Yet another unexpected finding of the invention is the observation that the cells are capable of detecting not only differences in the concentration of the agent, but also differences in the concentration of the agent along its length. Previous work is related to concentration gradients and cells compared in different concentrations. The invention is based in part on the finding that the cells respond to changes in concentration, but may also be able to detect their position in a gradient based on the difference in agent concentration along the length of the cell . That is, a cell can detect its position in a gradient, and therefore modulate its expression profile, detecting that its opposite ends are exposed to different concentrations of agent. In one aspect, the invention provides a method for identifying a nucleic acid expressed in a manner dependent on the concentration of the agent. The method comprises determining a first expression profile of the nucleic acid of a first cell in a first position in a gradient of concentration of the agent, determining a second expression profile of the nucleic acid of a second cell in a second position in the concentration gradient of the agent, and determining a difference between the first and second expression profiles of the nucleic acid, wherein the first position in the concentration gradient of the agent corresponds to a first concentration of the agent, and the second position in the gradient of the concentration of the agent corresponds to a second concentration of the agent. In some embodiments, at least the second cell has migrated through the concentration gradient of the agent. Therefore, the invention provides a method for identifying a nucleic acid expressed in a concentration dependent manner, comprising determining a first expression profile of the nucleic acid of a first cell in a first position in a concentration gradient of the agent, determining a second expression profile of the nucleic acid of a second cell that has migrated through the concentration gradient of the agent, and determining a difference between the first and second expression profiles of the nucleic acid. In another embodiment, the second cell is placed in the gradient, so that a gradient exists along the length (or diameter) of the cell. In other words, the concentration of the agent at one end of the cell (for example, the front end of the cell) is different from the concentration of the agent at the opposite end of the cell (e.g., the trailing end of the cell). In this way, the method can be performed by placing a cell within a preformed concentration gradient, or allowing the cell to move through the concentration gradient, depending on the application and desired information. The chemotactic, fugactic or chemokinetic response can be measured as described herein, or according to the transmigration assays described in greater detail in US Pat. No. 6,448,054 B1 and US Pat. No. 5,514,555, entitled: "Assays and therapeutic methods based on lymphocyte chemoattractans ", issued May 7, 1996, to Springer, TA, et al.). Other suitable methods will be known to one of ordinary skill in the art and can be employed using only routine experimentation. The concentration gradients of the agent can be established using a source of the agent. The source of the agent is the location in a gradient that has the highest concentration of the agent, and is generally the location to which the agent is supplied to establish the gradient. The agent can be continuously supplied or the source can be supersaturated with the agent which is not necessary for the replenishment of the agent during the course of screening. In the preferred embodiments, the gradient is established and remains constant throughout the screening process. That is, the concentration differential between the agent source and the end of the gradient is constant, as is the concentration differential between the different locations in the gradient. In some embodiments, the first concentration of the agent is a zero concentration of the agent, and the second concentration of the agent is a zero concentration of the agent, while in other embodiments, the first concentration of the agent is greater than the second concentration of the agent. The cells could migrate through the gradient, and in these modalities, one or both cells will migrate through the concentration gradient of the agent. The migration can be fugactic migration, or chemotactic migration. The gradient can be either a step gradient or a continuous gradient, although a continuous gradient is preferred in some embodiments. In yet another embodiment, there may be a second overlapping gradient on the first gradient. In one important embodiment, the first cell has undergone chemotaxis and the second cell has undergone fugetaxis, and the expression profiles of these cells are compared. The expression profile of the nucleic acid can be an RNA profile (preferably an mRNA), or it can be a protein profile. Depending on which expression product is analyzed, the method of analysis and quantification of the expression product will differ. If the nucleic acid expression product is likewise a nucleic acid, such as an RNA (e.g., mRNA), then it can be quantified using a number of methods including, but not limited to Norhtern analysis, polymerase chain reaction transcriptase- Reverse (RT-PCR), subtractive hybridization, differential deployment, representational difference analysis and microdata analysis of cDNA. In some embodiments, the nucleic acids are harvested from the cells and analyzed without the need for in vitro amplification. The differentially expressed molecule can be identified in a number of ways. If the expression product is a nucleic acid (i.e., an mRNA), then it can be identified using techniques such as subtractive hybridization (including subtractive substraction hybridization), differential display, representational difference analysis, or microdisposition analysis (e.g. Affymetrix microchip analysis). These techniques have been reported in the literature, and in this way someone with ordinary experience will be familiar with them. (See for example, Methods Enzymol 303: 349-380, 1999; Ying and Lin in Biotechniques 26: 966-8, 1999; Zhao et al., J. Biotechnol 73: 35-41, 1999; and Blumberg and Belmonte in Methods Mol Biol. 97: 555-574, 1999). The sequences isolated in this screening process can then be sequenced and compared to the non-redundant EST and GenBank databases using BLAST algorithm. Another important technique to identify differentially expressed transcripts involving DNA microchip technology and hybridization of cDNA microdevice. This technique is capable of analyzing hundreds, if not thousands, of coding sequences at the same time. Customized, standardized DNA microchips are now commercially available from manufacturers such as Affymetrix and InCyte. These methods have evolved to the extent that high total throughput screening for different sequences can be easily achieved. (Von Stein, et al., Nucleic Acids Res 25: 2598-602, 1997. Carulli, et al., J Cell Biochem Suppl 30-31: 286-96, 1998). One of the greatest advantages of DNA microchip technology is that no RNA amplification is required. If the expression product of the nucleic acid is a protein, then it can be identified using, for example, separation of gel electrophoresis followed by Coomassie Blue staining. In this latter approach, differences between the experimental cell and a control can be revealed by the presence or absence of stained protein bands. The additional separation, sequencing and cloning of these "difference bands" would then require, all of which are within the domain of the ordinary technician. Other methods can be used in a similar manner to identify and / or quantify nucleic acid expression products that are proteins, and these include, but are not limited to, immunohistochemistry. Western analysis, and fluorescence activated cytometry.
The agent that is used to establish a gradient is not intended to be limiting. Any agent that induces a change in the gene expression profile would be adequate. The agent can be a ligand, resulting in a gradient of ligand concentration. Accordingly, the ligand can also be a receptor. In some embodiments, the agent is a molecule that induces chemotaxis or fugetaxis. The agent can be a cytosine (including a chemokine). For an additional description of a cytosine, see Human Cytokines: Hanbook for Basic & Clinical Research (Aggrawal, et al., Eds., Blackwell Scientific, Boston, Mass. 1991) (which is incorporated herein by reference in its entirety for all purposes). Examples of cytokines include PAF, N-formylated peptides, C5a, LTB4 and LXA4, chemokines: CXC, IL-8, GCP-2, GRO, GROa, GROß, GRO ?, ENA-78, NAP-2, IP-10, MIG, I-TAC, SDF-1a, BCA-1, PF4, Bolequin, MIP-1a, MIP-1β, RANTES, HCC-1, MCP-1, MCP-2, MCP-3, MCP-4, MCP- 5 (mouse), Leukotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin-3 (TSC), MDC, TARC, SLC (Exodus-2, 6CKine), MIP-3a (LARC, Exodus-1), ELC (MIP-3β), 1-309, DC-CK1 (PARC, AMAC-1), TECK, CTAK, MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), MIP-1? (mouse), C-10 (mouse), Lymphotactin C and Franchisin CX3C (Neurotactin). Cytosine can be a member of the Cys-X-Cys family of chemokines (for example, chemokines that bind to the CXCR-4 receptor). Preferred cytokines of the invention include SDF-1a, SDF-1β, met-SDF-1β, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL -10, IL-12, IL-15, IL-18, TNF, IFN-a, lFN-β, IFN- ?, factor that stimulates the granulocyte-macrophage colony (GM-CSF), a factor that stimulates the colony of granulocyte (G-CSF), a factor that stimulates macrophage colony (M-CSF), TGF-β, ligand FLT-3, VEGF, DMDA, endothelin, and CD40 ligand. This first list does not mean that it is exhaustive and someone with ordinary experience will be able to identify other cytokines that can be used in the methods of the invention. In certain embodiments, cytosine is a cytisine with chemoattractant and / or chemokinetic properties. The agent can be a chemokine. Chemokines, or chemoattractant cytokines, are a family of small proteins with a conserved cysteine motif. These small proteins have been implicated in a wide range of disease states, such as acute and chronic inflammatory processes, angiogenesis, leukocyte migration, regulation of cell proliferation and maturation, hematopoiesis, viral replication, and other immunoregulatory functions. Chemokines are expressed by a number of different cells and have different cellular but overlapping targets. Chemokines have been classified into four subgroups, depending on the nature of the space of two highly conserved cysteine amino acids that are located near the amino terminus of the polypeptide. The first subgroup of chemokine is referred to as "CXX", the second subgroup is referred to as "CC", the third subgroup of chemokine is referred to as "CX3C"; the fourth subgroup of chemokine is referred to as "C". Within these subgroups, chemokines are further divided into related families that are based on amino acid sequence homology. The CXC chemokine families include the IP-10 and MIG family; GROa, GROß and the GROß family; the family of interleukin-8 (IL-8); and the family of PF4. The CC chemokine families include the monocyte chemoattractant protein family (MCP); the family including the macrophage inhibitor protein 1a (MIP-1a), macrophage inhibitor protein 1β (MIP-1β), and regulated in the activation of expressed normal T cell (RANTES). Stromal cell-derived factor 1a (SF-1a) and stromal cell-derived factor 1ß (SDF-1ß) represents a chemokine family that is approximately equal related by amino acid sequence homology to the CXC and CC chemokine subgroups . The CX3C chemokine family includes fractalkine; the chemokine C family includes lymphotactin. In general, CXC chemokines are joined by members of the CXCR class of receptors: CC chemokines are joined by the CCR class of receptors; CX3R chemokines are joined by the CX3CR class of receptors; and C-chemokines are joined by the CR class of receptors. Most chemokine receptors are transmembrane extension molecules that belong to the family of G-protein coupled receptors. Many of these receptors couple proteins that bind to the guanine nucleotide to transmit cellular signals. Chemokines and receptor expression are upregulated during inflammatory responses and cellular activation. Chemokines, when bound to their respective receptors, have been shown to be involved in a number of physiological conditions. For example, chemokines of the CXE group, such as interlucin-8, can stimulate angiogenesis, while platelet factor-4, growth-related oncogene-β (GRO-β) and interferon-induced protein-10? (IP-10) inhibit endothelial cell proliferation and angiogenesis. Interleukin-8 stimulates endothelial cell proliferation and chemotaxis in vitro, and appears to be a primary inducer of macrophage-induced angiogenesis. It was demonstrated that the activities of these chemokines are dependent on the NH2-terminal amino acid sequence (Streiter et al., J. Biol. Chem., 270: 27348-27357). SDF-1, another CXC chemokine, is active in the recruitment and mobilization of hematopoietic cells from the spinal cord, as well as the attraction of monocytes and lymphocytes. The agent can be any molecule, either naturally occurring or synthetically produced. The agent can be isolated from a biological sample such as a biological fluid. Biological fluids include, but are not limited to synovial fluid, cerebral spinal fluid, fallopian tube fluid, seminal fluid, ocular fluid, pericardial fluid, pleural fluid, inflammatory exudate, and dropsy fluid. The agent can also be presented in a tumor cell culture supernatant, tumor cell eluate and / or tumor cell lysate. In preferred embodiments, the agent is a molecule that induces chemotaxis or fugetaxis. In another embodiment, the agent is a fugactactic agent in a high concentration and a chemotactic agent in a lower concentration. The cells that are used in the methods of the invention are not limited to any cell type, provided that it has migratory capacity. An example of a migratory cell is a hematopoietic cell, such as neutrophils, basophils, eosinophils, monocytes, macrophages, dendritic cells, T cells and the like. In some modalities, the cell with migratory capacity is a neural cell. In additional modalities, the cell with migratory capacity is an epithelial cell. In still further modalities, the cell with migratory capacity is a mesenchymal cell. In some modalities, the cell with migratory capacity is an embryonic stem cell. In certain modalities, the cell with migratory capacity is a germ cell. In important embodiments, the cells are mammalian cells, such as human cells. In important modalities, the cells are primary human T cells. In other embodiments, the cells are neural cells, such as neurons capable of undergoing chemotaxis or fugetaxis for example in response to a neurotransmitter. Cells that express chemokine receptors include migratory cells, such as lymphocytes, granulocytes, and antigen presenting cells (APCs) that are believed to participate in immune responses or that can release other factors to mediate other cellular processes in vivo. The presence of a chemokine gradient serves to attract migratory cells that express the chemokine receptors. For example, migrating cells can be attracted by a chemokine gradient to a particular site of inflammation, at whose location, they play a role in further modifying the immune response. The "immune cells" as used herein are cells of hematopoietic origin, which are involved in the specific recognition of antigens. Immune cells include antigen presenting cells (APCs), such as dendritic cells or macrophages, B cells, T cells, etc. The "mature T cell" cells as used herein include T cells of the CD4 + 0 CD8hi CD69 + TCR +, CD4 + CD8 + CD69 + TCR +, CD4 + CD45 + RA +, CD4 + CD3 + RO + and / or CD8 + CD3 + RO phenotypes. +. Fugetaxis may play a role in the migration of T cells from the thymus during development. Cells of "hematopoietic origin" include, but are not limited to pluripotent stem cells, multipotent progenitor cells and / or progenitor cells committed to specific hematopoietic lineages. Progenitor cells committed to specific hematopoietic lineages may be of T cell lineage, B cell lineage, dendritic cell lineage, Langerhans cell lineage and / or lymphoid tissue-specific macrophage cell lineage. Hematopoietic cells can be derived from a tissue such as spinal cord, peripheral blood (including mobilized peripheral blood), umbilical cord blood, placental blood, fetal liver, embryonic cells (including embryonic stem cells), cells derived from aortal-gonadal-mesonephros , and soft lymphoid tissue. The soft lymphoid tissue includes the thymus, spleen, liver, lymph node, skin, tonsil and Peyer patches. In other embodiments, cells of "hematopoietic origin" can be derived from in vitro cultures of any of the above cells, and in particular, in in vitro cultures of progenitor cells. Cells of neural origin, including neurons and glia, and / or cells from both central and peripheral nervous tissue expressing RR / B (see U.S. Patent No. 5,863,744, entitled "Neural cell protein marker RR / B and DNA encoding. same, "issued on January 26, 1999, to Avraham, et al.). The work in Xenopus indicates that neurons and growth cones respond to netrines. Neurons are expected to respond by either chemotaxis or fugetaxis in the presence of neurotransmitters. Cells of epithelial origin include cells of a tissue that covers and coats the free surfaces of the body. Such epithelial tissue includes skin cells and sensitive organs as well as specialized cells that line blood vessels, the gastrointestinal tract, air passages, kidney ducts and endocrine organs. Cells of mesenchymal origin include cells that express typical fibroblast markers such as collagen, vimentin and fibronectin. The cells involved in angiogenesis are cells that are involved in the formation of blood vessels and include cells of epithelial origin and cells of mesenchymal origin. An embryonic stem cell is a cell that can give rise to cells of all lineages; it also has the capacity to self-renew. A germ cell is a specialized cell to produce monosomatic gametes. This is an additional differentiated cell from a stem cell that can still result in more differentiated germline cells. The cell can be a eukaryotic cell or a prokaryotic cell. In some embodiments, the cells used in the screening assays are adult cells. Preferably, they are human cells. These can be primary cells (for example directly harvested cells), or they can be secondary cells (including cells of a cell line). The invention in one aspect identifies differential expression products that are up-regulated or sub-regulated during chemokinesis (ie random movement), when compared to cells in medium alone. The identification of these products can be exploited in cases where it is desired to inhibit or facilitate cell movement. The over-regulated products during chemokinesis include the signaling of PTK2 molecules (focal adhesion kinase) (over-regulated by a value of 6.88) and the regulator of G protein signaling 10 (over-regulated by a value of 2.53) . Sub-regulated products during chemokinesis include the signaling of phospholipase C beta 3 molecules (sub-regulated by a value of 2.54), the RAS protein activator p21 (GAP) 3 (sub-regulated by a value of 2.20), RASguanil (calcium / DAG) release protein 2 (sub-regulated by a value of 2.16), protein G-coupled receptor kinase 6 (sub-regulated by a value of 2.15), Rho-specific GEF (p 114) ) (de-regulated by a value of 1.70) and substrate 80K-H protein kinase C (de-regulated by a value of 1.70). The knowledge of these products at a minimum allows the identification of products that are differentially regulated in a specific way in response to either chemotaxis or fugetaxis (that is, it is possible to distinguish between those products that are impacted by the directional movement determined instead of simply random movement). The data provided in the following tables are generally presented as expression levels of a particular gene product relative to the level of that gene product when the cell from which it is derived is placed in a medium alone or chemokinesis is allowed to occur. Knowledge of these products also leads to methods to inhibit or stimulate the movement of cells, depending on the desired effect. It is possible that many of these products are required in chemotaxis and fugetaxis and thus provide another objective to avoid or stimulate these directional migrations. In this way, these specific products of "chemokinesis" can be thought of as "household products" of cell movement in general (that is, they are required for movement, regardless of whether the movement is directional or not). The agents that stimulate these products include agonists and nucleic acids that encode the products, but are not limited as much. Agents that inhibit these products include antagonists, antibodies, and antisense nucleic acids, but are not so limited. In another aspect, the invention provides a method for identifying a compound that can modulate cell migration in one or more concentration gradients of one or more agents comprising contacting a migrating cell in a concentration gradient of agent with a test compound. , determining the expression profile of the nucleic acid in the cell and identifying a change in the expression of a gene expression product. The cell movement can be chemotaxis or fugetaxis and therefore, the gene expression product can be a specific genetic product of chemotaxis or fugetaxis. A test compound is any compound that is thought to potentially modulate chemotaxis or fugetaxis. The invention further provides methods for modulating chemotaxis and fugetaxis. As used in this, modular means affect or change, and includes stimulation or inhibition. To modulate chemotaxis or fugetaxis, the cells are in contact or exposed to agents that are differential expression products as identified according to the invention, or that impact the differential expression products. The differential expression products identified according to the invention are thus previously unrecognized, additional objectives that can be manipulated to modulate chemotaxis or fugetaxis. The ability to modulate chemotaxis and fugetaxis is important for manipulating bodily processes, such as but not limited to immune responses, chemical migration, and neural outcome (e.g., in response to neurotransmitters). In some cases, it would be desirable to inhibit an immune response that is occurring or is likely to occur in a subject. Examples include subjects having asthma, allergy, autoimmune diseases, such as rheumatoid arthritis, infections that are deleterious due to the immune response that is formed in response (eg, RSV infection, particularly in children), inflammatory conditions, host disease against graft (GVHD), and the like. In other cases, it would be desirable to promote or stimulate an immune response in which a subject is likely to benefit from such a response. These subjects include those who have or are likely to develop infections (eg, bacterial infections, viral infections, fungal infections, parasitic infections), and those who have or are likely to develop cancer to raise the immune survival of cancer cells. Other subjects include those who are diagnosed as having an impaired immune response, particularly when the defect falls on the inability of immune cells to respond to chemotactic factors. Accordingly, in one embodiment, a cell undergoing or probably experiencing fugetaxis is contacted or exposed to an agent that inhibits a specific gene expression product of fugetaxis in an amount effective to inhibit fugetaxis. The agent that inhibits fugetaxis can act at the level of the nucleic acid or protein. Fugetaxis-specific gene expression products are those that are up-regulated in response to fugetaxis when compared to their level when cells move randomly (ie chemokinesis) or when cells are produced by chemotaxis. Since these products are over-regulated in response to fugetaxis, the fugetaxis can be inhibited by blocking the activity of these products using a number of methods known in the art, including but not limited to antisense and antibody methods. The products can also be oriented to modulate chemotaxis, as someone with ordinary experience will understand. The signaling molecules may be, but are not limited to, the cell division cycle 42, annexin A3, guanine nucleotide exchange factor Rap1, adenylate cyclase 1, JAK binding protein, and alpha inhibitor of Rho GDP dissociation. In another embodiment, the signaling molecule is the cell division cycle 42 (cdc42), ribosomal protein S6 kinase, protein 2 associated with BAI1, GTPase regulator associated with FAK, protein kinase C-beta, phosphoinositide specific phospholipase C-beta 1, nitric oxide synthase 1, phosphatidylinositol-4-phosphate 5-kinase, and MAP kinase, kinase kinase kinase 4. The molecules related to the extracellular matrix can be, but are not limited to Chitinase 1 similar to 3 (glycoprotein 39 cartilage), cell adhesion molecule related to carcinoembryonic antigen 6, matrix metalloproteinase 8 (neutrophil collagenase), protein 1 associated with cytoplasmic integrin domain, ficolin (containing the collagen fibrinogen domain) 1, antigen 1 similar to epithelial V, factor of vascular endothelial growth (VEGF), fibulin 1, cell adhesion molecule 3 related to carcinoembryonic antigen, and lysosomal associated membrane protein 1. Cytoskeletal-related molecules can be, but are not limited to, Ankyrin 1 (erythrocytic), A12 protein to antisense and antibody methods. The products can also be oriented to modulate chemotaxis, as someone with ordinary experience will understand. The signaling molecules may be, but are not limited to, the cell division cycle 42, annexin A3, guanine nucleotide exchange factor Rap1, adenylate cyclase 1, JAK binding protein, and alpha inhibitor of Rho GDP dissociation. In another embodiment, the signaling molecule is the 42 cell division cycle (cdc42), ribosomal protein S6 kinase, protein 2 associated with BAI1, GTPase regulator associated with FAK, protein kinase C-beta, phosphoinositide specific phospholipase C-beta 1, nitric oxide synthase 1, phosphatidylinositol-4-phosphate 5-kinase, and MAP kinase, kinase kinase kinase 4. The molecules related to the extracellular matrix can be, but are not limited to Chitinase 1 similar to 3 (glycoprotein 39 cartilage), cell adhesion molecule related to carcinoembryonic antigen 6, matrix metalloproteinase 8 (neutrophil collagenase), protein 1 associated with cytoplasmic integrin domain, ficolin (containing the collagen fibrinogen domain) 1, antigen 1 similar to epithelial V, factor of vascular endothelial growth (VEGF), fibulin 1, cell adhesion molecule 3 related to carcinoembryonic antigen, and associated membrane protein 1 gives lysosomal. Cytoskeletal related molecules can be from the T cell receptor, gamma site of the T cell receptor. The specific gene expression product of fugetaxis can also be chemokine receptor 1 (C-X3-C). The invention further provides a method for inhibiting cellular chemotaxis. The method involves contacting a cell that is experiencing or is likely to undergo chemotaxis with an agent that inhibits a specific gene expression product of chemotaxis in an amount effective to inhibit chemotaxis. The agent that inhibits chemotaxis can act at the level of nucleic acid or protein. Chemotaxis-specific gene expression products are those that are up-regulated in response to chemotaxis when compared to their level in chemokinesis or fugetaxis. Since these products are upregulated in response to chemotaxis, chemotaxis can be inhibited by blocking the activity of these products using a number of methods known in the art, including but not limited to antisense and antibody methods. The signaling molecules can be, but are not limited to, kinase 6 of the G protein-coupled receptor, vaccine-related kinase 1, tyrosine kinase 2 of the PTK2 protein, STAM-like protein containing 2 SH3 domains and ITAM, gene 1 associated with signal-induced proliferation, CD47 antigen (Rh-related antigen, integrin-associated signal transducer), and protein tyrosine phosphatase (without type 12 receptor). The signaling molecule can also be selected from the group consisting of PTK2 (focal adhesion kinase), MAP kinase kinase kinase kinase 2, guanine nucleotide binding protein, PT phosphatase (receptor), cdc binding protein kinase beta -42, Ral GEF (RalGPSIA), MAP kinase 7, autotaxin, inositol, 1, 4,5-triphosphate receptor, phosphoinositide-3-kinase gamma, PT phosphatase (without receptor), p21 RAS protein activator (GAP), 2 guanyl RAS release protein, and 20kDa subunit of the Arp23 complex. Cell-related molecules between cells can be, but are not limited to, spondin-1 (spondin-f, extracellular matrix protein), collagen of type XVII I (alpha 1), adhesion molecule of CD31, tetraspan 3, glycoprotein A33 , and angio migratory cell protein associated. The molecules related to cytoskeleton can be, but are not limited to the protein complex 23 related to actin (subunit 4, 20kD), tropomyocin 2 (beta), actin-dependent regulator associated with chromatin-related SWISNF matrix (subfamily A, member 5), beta espretin (noeritrócitical), myocin (light 5 polypeptides, regulatory), keratin 1, placofilin 4, and sealed protein (actin filament, muscle Z line, alpha 2). Cell cycle molecules can be, but are not limited to, protein 1 that activates the FGF receptor, musculoaponeurotic fibrosarcoma v-maf (avian) oncogene homolog, cyclin-dependent kinase (similar to CDC2) 10, early growth response 2 inducible with TGFB, retinoic acid alpha receptor, subunit 10 of anaphase-promoting complex, RAS p21 protein activator (GTPase-activating protein, 3-lns-1, 3,4,6-P4 binding protein), cycle 27 of cell division, programmed cell death 2, c-myc binding protein, protein kinase kinase kinase 1 activated with mitogen, receptor III beta TGF (betaglycan, 300 kDa) and G1 for transition 1 of phase S. Molecules related to immune response may, but are not limited to, major histocompatibility complex of class II DQ beta I, spinal cord stromal cell antigen 2, Burkitt lymphoma receptor 1 (GTP binding protein); CXCR5), CD7 antigen (p41), Stat2 type a, cellular immune regulator 1, and interleukin 21 receptor. Contacting the cells with the inhibitory or stimulating agents of the invention can occur in vivo. And as mentioned above, the subject that receives the agent will vary depending on the type of agent being administered. Thus, in an embodiment wherein the method is intended to inhibit chemotaxis, the subject is one that has or is at risk of having an immune response to normal. The normal immune response may be an inflammatory response or an autoimmune response, but it is not so limited. Autoimmune disease is a class of diseases wherein the subject's own antibodies react with the host tissue or where the immune effector T cells are self-reactive to endogenous peptides and cause tissue destruction. Autoimmune diseases include, but are not limited to, rheumatoid arthritis, Crohn's disease, multiple sclerosis, lupus erythematosus (SLE), autoimmune encephalomyelitis, severe myiastemia (MG), Hashimoto's thyroiditis, Goodpasture's syndrome, fenfigo (for example, pemphigus) vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis, pernicious anemia, idiopathic Adison's disease, autoimmune associated infertility, glomerulonephritis (eg, growing glomerulonephritis, proliferative glomerulonephritis), pemphigoid bolus, Sjögren's syndrome, insulin resistance, insulin-dependent diabetes mellitus, uveitis, rheumatic fever, Guillain-Barre syndrome, psoriasis, and autoimmune hepatitis. According to yet another aspect of the invention, a method for promoting fugetaxis is provided. The method involves contacting a cell with an agent without chemokine that promotes fugetaxis in an effective amount to promote fugetaxis. In one embodiment, contact occurs in vivo in a subject who has a disorder characterized by abnormal fugetaxis. As used herein, an agent without chemokine is an agent without a chemokine such as those reported above. The agent without chemokine is preferably one of the downstream targets of fugetaxis identified according to the invention, or is an agonist thereof. The invention further provides a method for promoting chemotaxis. The method involves contacting a cell with an agent without chemokine that promotes chemotaxis in an amount effective to promote chemotaxis. In one embodiment, contact occurs in vivo in a subject who has a disorder characterized by a lack of chemotaxis. The agent without chemokine is preferably one of the downstream targets of fugetaxis identified according to the invention, or is an agonist thereof. As stated above, in some cases, modulation occurs by the administration of nucleic acids (e.g., in antisense therapy), or by proteins or polypeptides (e.g., antibody therapy). In some embodiments, the nucleic acids or proteins / peptides are isolated. In still further embodiments, the nucleic acids or proteins / peptides are substantially pure. As used herein, with respect to nucleic acids, the term "isolated" means: (i) amplified in vitro by, for example, polymerase chain reaction (PCR); (ii) produced recombinantly by cloning; (iii) purified, as by splitting and gene separation; or (iv) synthesized by, for example, chemical synthesis. An isolated nucleic acid is one which is easily manipulated by recombinant DNA techniques well known in the art. Thus, a nucleotide sequence contained in a vector wherein the 5 'and 3' restriction sites are known or for which the sequences of the polymerase chain reaction (PCR) primer have been described is considered isolated but a nucleic acid sequence existing in its native state in its natural host no. An isolated nucleic acid can be substantially purified, but it does not need to be. For example, a nucleic acid that is isolated within an expression or cloning vector is not pure in that it can comprise only a minute percentage of the material in the cell in which it resides. Such a nucleic acid is isolated, however, when the term is used herein because it is easily manipulated by standard techniques known to those of ordinary skill in the art. As used herein, with respect to proteins / peptides, the term "isolated" means separated from its native environment in a sufficiently pure form so that it can be manipulated or used for any of the purposes of the invention. Thus, isolated means sufficiently pure to be used (i) to increase and / or isolate antibodies, (i) as a reagent in an assay, or (iii) for sequencing, etc. The term "substantially pure" means that the nucleic acid or protein / peptide is essentially free of other substances with which it can be found in nature or in in vitro systems, to a practical degree and appropriate for its intended use. Substantially pure polypeptides can be produced by techniques well known in the art. As an example, because an isolated protein can be mixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the protein can comprise only a small percentage by weight of the preparation. The protein is isolated, however, in that it has been separated from many of the substances with which it can be associated in active systems, that is, isolated from certain other proteins. According to another aspect, the invention provides compositions and methods that are related to attracting or repelling immune cells to or from a material surface. These aspects of the invention involve coating or loading surfaces of the material alternately with the chemotactic inhibitory agents, the chemotactic stimulating agents, the fugactic-inhibiting agents, or the fugactic-stimulating agents provided herein. "Material Surfaces" as used herein, include, but are not limited to prosthetic orthopedic and dental implants, artificial valves and organic implantable tissue such as a stent, allogeneic and / or xenogeneic tissue, organ and / or vasculature. Implantable prosthetic devices have been used in the surgical repair or replacement of internal tissue for many years. Orthopedic implants include a wide variety of devices, each suitable for the particular medical needs carried out. Examples of such devices are hip replacement devices, knee replacement devices, shoulder replacement devices, and nails, braces and plates used to hold fractured bones. Some contemporary orthopedic and dental implants use high performance metals such as chrome-cobalt alloy and titanium to achieve high strength. These materials are easily manufactured in the normal complex shapes of these devices using mature metal working techniques including casting and machining. The surface of the material is coated with an amount of an effective agent to repel or attract cells (e.g., immune cells), depending on the desired therapeutic effect. In important modalities, the surface of the material is part of an implant. In particular embodiments, in addition to a fugactic agent, the surface of the material can also be coated with a cell growth potentializing agent, an anti-infective agent, and / or an anti-inflammatory agent. A cell growth potentizing agent as used herein is an agent which stimulates the growth of a cell and includes growth factors such as PDGF, EGF, FGF, TGF, NGF, CNTF, and GDNF. An anti-infective agent as used herein, is an agent that reduces the activity of or eliminates a microorganism and includes: Aztreonam; Chlorhexidine gluconate; Imidurea; Licetamine; Nibroxane; Sodium pirazmonam; Propionic acid; Sodium pyrithione; Sanguinario Chloride; Tigemonam Dicoline; Acedapsona; Sodium Acetosulfone; Alamecin; Alexidina; Amdinocillin; Amdinocilin Pivoxil; Amicycline; Amifloxacin; Amifloxacin mesylate; Amikacin; Amikacin sulfate; Aminosalicylic acid; Sodium Aminosalicylate; amoxicillin; Amphotomy; Ampicillin; Sodium Ampicillin; Sodium apaccilin; Apramycin; Aspartocin; Astromycin Sulfate; Avilamycin; Avoparcin; Azithromycin; Aziocillin; Sodium aziocillin; Bacampicillin hydrochloride; Bacitracin; Methylene Bacitracin Disilicylate; Zinc Bacitracin; Bambermicins; Benzoilpas de Calcio; Berithromycin; Betamycin Sulfate; Biapenem; Biniramycin; Bifenamine hydrochloride; Bispiritiona Magsulfex; Butikacina; Butyrosine sulfate; Capreomycin Sulfate; Carbadox; Disodium Carbenicillin; Indane Sodium Carbenicillin; Sodium phenyl carbenicillin; Potassium Carbenicillin; Sodium Carumonam; Cefaclor; Cefadroxil; Cefamandole; Cefamandole Hydrochloride; Sodium cefamandole; Cefaparol; Cefatrizine; Sodium cefazaflur; cefazolin; Sodium cefazolin; Cefbuperazone; Cefdinir; Cefepime; Cefepime hydrochloride; Cefetecol; Cefixime; Cefmenoxim hydrochloride; Cefmetazole; Sodium cefmetazole; Monosodium cefonicide; Sodium Cefonicide; Sodium Cefoperazone; Ceforanide; Sodium cefotaxime; Cefotetan; Cefotetan Disodium; Cefotiam hydrochloride; Cefoxitin; Sodium cefoxitin; Cefpimizole; Sodium Cefpimizole; Cefpyramide; Sodium cefpyramide; Cefpiroma Sulfate; Proxetil cefpodoxime; Cefprozil; Cefroxadine: Sodium cefsulodin; Ceftazidime; Ceftibuten; Sodium Ceftizoxime; Sodium Ceftriaxone; Cefuroxime; Axetil cefuroxime; Pivoxetil cefuroxime; Sodium cefuroxime; Sodium cefacetril; Cephalexin; Cefalexin hydrochloride; Cephaloglycine; Cefaloridine; Sodium cephalothin; Sodium cefaparin; Cephradine; Cetocycline hydrochloride; Cetofenicol; Chloramphenicol; Chloramphenicol Palmitate; Chloramphenicol Pantothenate Complex; Sodium Chloramphenicol Succinate; Chlorhexidine Phosphanylate; Chloroxylenol; Chlortetracycline bisulfate; Chlortetracycline hydrochloride; Cinoxacin; Ciprofloxacin; Ciprofloxacin hydrochloride; Cirolemycin; Clarithromycin; Clinafloxacin hydrochloride; Clindamycin; Clindamycin hydrochloride; Hydrochloride-Palmitate Clindamycin; Clindamycin Phosphate; Clofazimine; Cloxacillin benzathine; Sodium Cloxacillin; Cloxiquin; Sodium Colistimethate; Colistin sulfate; Coumermycin; Sodium coumermycin; Cyclacillin; Cycloserine; Dalfopristin; Dapsone; Daptomycin; Demeclocycline; Demeclocycline hydrochloride; Demecycline; Denofungin; Diaveridine; Dicloxacillin; Sodium dicloxacillin; Dihydrostreptomycin Sulfate; Dipyrithione; Dirithromycin; Doxycycline; Calcium Doxycycline; Doxycycline Fosfatex; Doxycycline histalate; Sodium Droxacin; Enoxacin; Epicillin; Epitetracycline hydrochloride; Erythromycin; Acrylate of Erythromycin; Erythromycin estolate; Erythromycin Etiisuccinate; Glycetate of Erythromycin; Erythromycin Lactobionate; Erythromycin Propinate; Erythromycin stearate; Ethambutol Hydrochloride; Ethionamide; Fleroxacin; Floxacillin; Fludalanin; Flumequine; Fosfomycin; Fosfomycin tromethamine; Fumoxycillin; Furazolium Chloride; Furazolium tartrate; Sodium Fusidate; Fusidic acid; Gentamicin Sulfate; Gloximonam; Gramicidin; Haloprogin; Hetacillin; Potassium hetacycline; Hexedine; Ibafloxacin; Imipenem; Isoconazole; Isepamycin; Isoniazid; Josamycin; Kanamycin Sulfate; Kitasamycin; Levofuraltadone; Potassium Levopropylillin; Lexithromycin; Lincomycin; Lincomycin Hydrochloride; Lomefloxacin; Lomefloxacin hydrochloride; Lomefloxacin mesylate; Loracarbef; Mafenide; Mecycycline; Meclocycline sulfosalicylate; Potassium Megalomycin Phosphate; Mequidox; Meropenem; Metacycline; Metacycline hydrochloride; Metenamine; Metenamine Hipurate; Metenamine mandelate; Sodium methicillin; Methioprim; Metronidazole hydrochloride; Metronidazole phosphate; Mezlocillin; Sodium mezlocillin; Minocycline; Minocycline hydrochloride; Mirincamycin hydrochloride; Monensin; Sodium Monensin; Sodium Nafcillin; Nalidixato de Sodio; Nalidixic acid; Natamycin; Nebramycin; Neomycin palmitate; Neomycin sulfate; A Neomycin Decylenate; Netilmicin sulfate; Neutramycin; Nifuradene; Nifuraldezona; Nifuratel; Nifuratrone; Nifurdazil; Nifurimide; Nifurpirinol; Nifurquinazol; Nifurtiazole; Nitrocycline; nitrofurantoin; Nitromide; Norfloxacin; Sodium Novobiocin; Ofloxacin; Ormetroprima; Sodium Oxacillin; Oximonam; Sodium Oximonam; Oxolinic acid; Oxytetracycline; Calcium Oxytetracycline; Oxytetracycline hydrochloride; Paldimycin; Parachlorophenol; Paulomycin; Pefloxacin; Mesylate of Pefloxacin; Penamecillin; Benzathine of Penicillin G; Potassium Penicillin G; Penicillin G Procaine; Sodium Penicillin G; Penicillin V; Benzathine of Penicillin V; Hydraminine of Penicillin V; Potassium penicillin V; Sodium Pentizidone; Phenyl Aminosalicylate; Sodium piperacillin; Sodium pirbenicillin; Sodium pyridicillin; Pirlimycin hydrochloride; Hydrochloride Pivampicillin; Pivampicillin Pamoate; Probenate from Pivampicillin; Polymyxin B Sulfate; Porphyromycin; Propikacin; Pyrazinamide; Zinc pyrithione; Quindecamine Acetate; Quinupristin; Racefenicol; Ramoplanin; Ranimycin; Relomycin; Repromycin; Rifabutin; Rifamethane; Rifamexyl; Rifamide; Rifampin; Rifapentine; Rifaximin; Rolitetracycline; Nitrate Rolitetracycline; Rosaramycin; Rosaramycin butyrate; Rosaramycin propionate; Phosphate of Sodium Rosaramycin; Rosaramycin stearate; Rosoxacin; Roxarsone; Roxithromycin; Sancycline; Sodium Sanfetrinem; Sarmoxycillin; Sarpicillin; Scopafungin; Sisomycin; Sisomycin Sulfate; Esparfloxacin; Spectinomycin Hydrochloride; Spiramycin; Estalimicin hydrochloride; Estefimicina; Streptomycin Sulfate; Streptonicozid; Sulfabenz; Sulfabenzamide; Sulfacetamide; Sodium sulfacetamide; Sulfacitin; Sulfadiazine; Sodium sulfadiazine; Sulfadoxine; Sulfalene; Sulfamerazine; Sulfameter; Sulfamethazine; Sulfametizole; Sulfamethoxazole; Sulfamonomethoxine; Sulfamoxol; Zinc sulfanilate; Sulfanitran; Sulfasalazine; Sulfasomizol; Sulfatiazole; Sulfazamet; Sulfisoxazole; Acetyl sulfisoxazole; Sulfisoxazole diolamine; Sulfomixin; Sulopenem; Sultamicillin; Sodium suncillin; Talampicillin Hydrochloride; Teicoplanin; Temafloxacin Hydrochloride; Temocillin; Tetracycline; Tetracycline hydrochloride; Tetracycline Phosphate Complex; Tetroxoprim; Tianfenicol; Potassium diphencillin; Ticarcilin Sodium Crosyl; Disodium Ticarcillin; Monosodic ticarcillin; Ticlatone; Thiodonium Chloride; Tobramycin; Tobramycin Sulfate; Tosufloxacin; Trimethoprim; Trimethoprim Sulfate; Trisulfapyrimidines; Troleandomycin; Trospectomycin Sulfate; Tirothricin; Vancomycin; Vancomycin Hydrochloride; Virginiamicin; Zorbamycin; Difloxacin hydrochloride; Lauryl isoquinolinium bromide; Moxalactam Disodium; Ornidazole; Pentisomycin; And Sarafloxacin Hydrochloride. An anti-inflammatory agent is an agent that jointly reduces or inhibits an inflammatory response in vivo and includes Alclofenac; Alclometasone dipropionate; Algestone acetonide; Alpha Amylase; Amcinafal; Amcinafide; Sodium Amfenac; Amiprilose hydrochloride; Anakinra; Anirolac; Anitrazafen; Apazon; Disodium Balsalazide; Bendazac; Benoxaprofen; Hydrochloride and Benzydamine; Bromelains; Broperamol; Budesonide; Caprofen; Cycloprofen; Cintazona; Cliprofen; Clobetasol propionate; Clobetasone Butyrate; Clopirac; Cloticasone propionate; Cormetasone acetate; Cortodoxona; Deflazacort; Desonida; Deoxymethasone; Dexamethasone dipropionate; Potassium Diclofenac; Sodium Diclofenac; Diflorasone Diacetate; Sodium diflumidone; Dif I a isa I; Difftalone difluprednate; Dimethyl sulfoxide; Drocinonide; Endrisone Enlimomab; Sodium Enolicam; Epirizol; Etodolac; Ethofenamate Felbinac; Fenamol; Fenbufen; Fenclofenac; Fenclorac; Fendosal Fenpipaione; Fentiaza; Flazalona; Fluazacort; Flufenic acid Flumizol; Flunisolide Acetate; Flunixin; Flunixin Meglumine Fluocortin Butyl; Fluorometholone acetate; Fluquazone Flurbiprofen; Fluretofen; Fluticasone propionate Furaprofen; Furobufeno; Halcinonide; Halobetasol Propionate Halopredone Acetate; Ibufenaco; Ibuprofen; Aluminum Ibuprofen; Ibuprofen Piconol; llonidap; Indomethacin; Sodium Indomethacin; Indoprofen; Indoxol; Intrazole; Isoflupredone acetate; Isoxepac; Isoxicam; Ketroprofen; Lofemizol hydrochloride; Lornoxicam; Loteprednol etabonate; Sodium Meclofenamate; Meclofenamic acid; Meclorisone dibutyrate; Mefenamic acid; Mesalamine; Meseclazone; Methylprednisolone suleptanate; Morniflumate; Nabumetone; Naproxen; Sodium Naproxen; Naproxol; Nimazone; Olsalazine sodium; Orgotein; Orpanoxin; Oxaprozin; Oxyphenbutazone; Paraniline hydrochloride; Pentosan Sodium Polysulfate; Sodium Penbutazone glycerate; Pirfenidone; Piroxicam; Piroxicam cinnamate; Olamina from Piroxicam; Pirprofen; Prenazate; Prifelone; Prodolic acid; Proquazone; Proxazole; Proxazole Citrate; Rimexolone; Romazarit; Salcolex; Salnacedina; Saisalate; Sanguinary Chloride; Seclazone; Sermetacin; Sudoxicam; Sulindac; Suprofen; Talmetacin; Talniflumate; Talosalate; Tebufelone; Tenidap; Sodium Tenidap; Tenoxicam; Tesicam; Tesimida; Tetridamine; Tiopinac; Tixocortol pivalate; Tolmetin; Sodium tolmetin; Triclonide; Triflumidate; Zidomethacin; Sodium zomepirac. According to one aspect of the invention, there is provided a method for inhibiting the migration of immune cells to a specific site in a subject. The method involves locally administering to a specific site in a subject in need of such treatment an agent that promotes fugetaxis in an amount effective to inhibit the migration of immune cells to the specific site in a subject. In an important embodiment, the invention provides a method for inhibiting migration of immune cells to a site of inflammation in the subject. "Inflammation" as used herein, is a localized protective response produced by a foreign antigen (not own) and / or by a damage or destruction of the or of the tissues, which serves to destroy, dilute or sequester the foreign antigen, the damaged agent, and / or the damaged tissue. Inflammation occurs when tissues are damaged by viruses, bacteria, trauma, chemicals, heat, cold or any other dangerous stimulus. In such cases, the classic weapons of the immune system interface (T cells, B cells, macrophages) with cells and soluble products that mediate inflammatory responses (neutrophils, eosinophils, basophils, quinine and coagulation systems, and complete cascade ). A typical inflammatory response is characterized by (i) leukocyte migration at the site of antigen location (damage); (ii) specific and non-specific recognition of "foreign" and other antigens (necrotic / damaged tissue) mediated by B and T lymphocytes, macrophages and the alternative complementary pathway; (iii) the amplification of the inflammatory response with the recruitment of specific and non-specific effector cells by complementary components, lymphokines and monoquinas, quinines, arachidonic acid metabolites and mast cell / basophilic products; and (iv) participation of macrophage, neutrophil and lymphocyte in the destruction of the antigen with final removal of antigen particles (damaged tissue) by phagocytosis. According to yet another aspect of the invention, a method for improving an immune response in a subject having a condition involving a specific site is provided. The method involves locally administering to a specific site in a subject in need of such treatment an agent that inhibits fugetaxis or stimulates chemotaxis in an amount effective to inhibit specific cell-site-specific fu- geneatic activity at a specific site in the subject. In some modalities, the specific site is a site of a pathogenic infection. The efficient recruitment of immune cells to help eliminate the infection is therefore beneficial. In certain modalities, the specific site is a site that contains the germ cell. In this case, the recruitment of immune cells to these specific sites will help eliminate unwanted germ cells, and / or implanted and non-implanted embryos. In the additional embodiments, co-administration of contraceptive agents other than anti-phyletic agents is also provided. In the additional modalities, the specific site is an area that immediately surrounds a tumor. Since most of the known tumors escape immune recognition, it is beneficial to improve the migration of immune cells to the tumor site. In additional embodiments, co-administration of anti-cancer agents other than anti-fugactic agents is also provided. Anti-non-antigenated anti-cancer agents include: Acivicin; Aclarubicin; Acodazole hydrochloride; Acronine; Adozelesin; Aldesleucin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlina; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene hydrochloride; Bisnafida dimesylate; Bizelesina; Bleomycin sulfate; Brequinar de sodio; Bupirimine; Busulfan; Cactinomycin; Calusterona; Caracemide; Carbetimero; Carboplatin; Carmustine; Carubicin hydrochloride; Carzelesin; Cedefingol; Chlorambucil; Cirolemycin; Cisplatin; Cladribine; Crisantol mesylate; Cyclophosphamide; Cytarabine; Dacarbazine; Dactinomycin; Daunorubicin hydrochloride; Decitabine; Dexormaplatin; Dezaguanine; Dezaguanine mesylate; Diaziquona; Docetaxel; Doxorubicin; Doxorubicin hydrochloride; Droloxifene; Droloxifene Citrate; Dromostanolone propionate; Duazomycin; Edatrexate; Hydrochloride Eflornithine; Elsamitrucin; Enloplatin; Enpromato; Epipropidin; Epirubicin hydrochloride; Erbulozole; Esorubicin Hydrochloride; Estramustine; Sodium Estramustine Phosphate; Etanidazole; Etoposide; Etoposide Phosphate; Etoprine; Fadrozole hydrochloride; Fazarabina; Fenretidine; Floxuridine; Phosphate Fludarabine; Fluorouracil; Flurocitabine; Fosquidone; Sodium Phosphomycin; Gencitabine; Gencitabine hydrochloride; Hydroxyurea; Idarubicin hydrochloride; Ifosfamide; Ilmofosin; Interferon Alfa-2a; Interferon Alfa-2b; Interferon Alfa-n1; Interferon Alfa-n3; Interferon Beta-I a; Interferon Gamma-I b; Iproplatin; Irinotecan hydrochloride; Lanreotide Acetate; Letrozole; Acetate Leuprolide; Liarozole Hydrochloride; Sodium Lometrexol; Lomustine; Losoxantrone hydrochloride; Masoprocol; Maytansina; Mechloretamine hydrochloride; Megestrol Acetate; Melengestrol Acetate; Melphalan; Menogaril; Mercaptopurine; Methotrexate; Sodium methotrexate; Metoprine; Meturedepa; Mitindomide; Mitocarcin; Mitochromin; Mitogiline; Mitomalcin; Mitomycin; Mitosper; Mitotano; Mitoxantrone hydrochloride; Mycophenolic acid; Nocodazole; Nogalamycin; Ormaplatin; Oxisuran; Paclitaxel; Pegaspargasa; Peliomycin; Pentamustine; Peplomycin Sulfate; Perfosfamide; Pipobroman; Piposulfan; Piroxantrone Hydrochloride; Piicamycin; Plommethane; Podofilox; Sodium Porfimer; Porphyromycin; Prednimustine; Procarbazine hydrochloride; Puromycin; Puromycin hydrochloride; Pyrazofurin; Riboprine; Rogletimida; Safingol; Safingol Hydrochloride; Semustine; Simtrazeno; Sodium esparfosato; Esparsomycin; Spirogermanium hydrochloride; Spiromustine; Spiroplatin; Streptonigrin; Estroptozocine; Sulofenur; Talisomycin; Taxotero; Tecogalán de Sodio; Tegafur; Teloxantrone Hydrochloride; Temoporfin; Teniposide; Teroxirone; Testoiactone; Tiamiprin; Thioguanine; Tiotepa; Thiazofurine; Tirapazamine; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine phosphate; Trimetrexate; Trimetrexate gluconate; Triptorelin; Tubulozole hydrochloride; Uraciio mustard; Uredepa; Vapreotide; Verteporfin; Vinblastine Sulfate; Vincristine Sulfate; Vindesina; Vindesine Sulfate; Vinepidine sulfate; Vinglicinato-Sulfato; Vinleurosine sulfate; Vinorelbine tartrate; and Vinrosidine Sulfate. In some embodiments, the fugetaxis stimulation, fugetaxis inhibition, chemotaxis stimulation or chemotaxis inhibition agents of the invention are administered substantially simultaneously with other therapeutic agents. By "substantially simultaneously" it is meant that the agents are administered to the subject fairly closely in time, so that the other therapeutic agents can exert a potentiating effect on the inhibition of migration or stimulating activity of the fugactic or chemotactic agent. The fugactic or chemotactic agent can be administered before, at the same time, and / or after the administration of the other therapeutic agent. The methods provided herein can in some cases be carried out by the administration of antisense molecules to block the transcription or translation of the nucleic acid expression products. As used herein, the term "antisense oligonucleotide" or "antisense" describes an oligonucleotide that is an oligoribonucleotide, oligodeoxyribonucleotide, modified oligoribonucleotide or modified oligodeoxyribonucleotide which hybridizes under physiological conditions to DNA comprising a particular gene or a transcription of mRNA of that gene and, therefore, inhibits transcription of that gene and / or translation of that mRNA. Antisense molecules are designed to interfere with the transcription or translation of a target gene until hybridization with the target gene or transcript. Those skilled in the art will recognize that the exact length of the antisense oligonucleotide and its degree of complementarity with its objective will depend on the specific objective selected, including the target sequence and the particular bases comprising that sequence. It is preferred that the antisense oligonucleotide be constructed and arranged so as to selectively bind to the target under physiological conditions, i.e., hybridize substantially more to the target sequence than to any other sequence in the target cell under physiological conditions. Based on the identification of molecules that are over-regulated in fugetaxis or chemotaxis (see Tables herein), one skilled in the art can easily choose and synthesize any of a number of appropriate antisense molecules for use in accordance with the present invention. invention. To be selective and potent enough for inhibition, such antisense oligonucleotides must comprise at least about 10 and, more preferably, at least about 15 consecutive bases that are complementary to the target, although in certain cases the modified oligonucleotides as short as 7 bases in length have have been used successfully as antisense oligonucleotides. See Wagner et al., Nat. Med. 1 (11): 1116-1118, 1995. More preferably, the antisense oligonucleotides comprise a complementary sequence of 20-30 bases. Although oligonucleotides, which are antisense to any region of the gene or mRNA transcripts, can be chosen, in the preferred embodiments, the antisense oligonucleotides correspond to upstream 5 'or N-terminal sites such as translation initiation, transcription initiation or promoter sites. In addition, the 3'-untranslated regions can be directed by the antisense oligonucleotides. To target mRNA splice sites has also been used in the art, but may be less preferred, if splicing of alternative mRNA occurs. In addition, the antisense is directed, preferably to sites where the secondary mRNA structure is not expected (see for example, Sainio et al., Cell Mol. Neurobiol., 14 (5): 439-457, 1994) and where the proteins are not expected to join. In a set of embodiments, the antisense oligonucleotides of the invention may be composed of "native" deoxyribonucleotides, ribonucleotides, or any combination thereof. That is, the 5 'end of a native nucleotide and the 3' end of another native nucleotide can be covalently linked, as in natural systems, through a phosphodiester internucleoside linkage. These oligonucleotides can be prepared by recognized methods of the art that can be carried out manually or by an automated synthesizer. These can be produced recombinantly by vectors. In preferred embodiments, however, the antisense oligonucleotides of the invention may also include "modified" oligonucleotides. That is, the oligonucleotides can be modified in a number of ways that do not prevent them from hybridizing to their target, but which improve their stability or target or which otherwise improve their therapeutic effectiveness. The term "modified oligonucleotide" as used herein describes an oligonucleotide, wherein (1) at least two of its nucleotides are covalently linked through a synthetic internucleoside link (i.e., a different connection of a phosphodiester linkage). between the 5 'end of one nucleotide and the 3' end of the other nucleotide) and / or (2) a chemical group not normally associated with the nucleic acid molecules has been covalently linked to the oligonucleotide. The preferred synthetic internucleoside linkages are phosphorothioates, alkyl phosphonates, phosphorodithioates, phosphate esters, alkylphosphonothioates, phosphoramidates, carbamates, carbonates, phosphate triesters, acetamides, carboxymethyl esters and peptides. The term "modified oligonucleotide" also encompasses oligonucleotides with a covalently modified base and / or sugar. For example, modified oligonucleotides include oligonucleotides having major structure sugars that are covalently attached to organic groups of low molecular weight other than a hydroxyl group at the 3 'position and different from a phosphate group at the 5' position. Thus, the modified oligonucleotides can include a 2'-O-alkylated ribose group. In addition, the modified oligonucleotides can include sugars such as arabinose instead of ribose. The present invention, thus, contemplates pharmaceutical preparations containing modified antisense molecules together with pharmaceutically acceptable carriers. The antisense oligonucleotides can be administered as part of a pharmaceutical composition. In this latter modality, it is preferable that a slow intravenous administration be used. Such a pharmaceutical composition may include the antisense oligonucleotides in combination with any physiologically and / or pharmaceutically acceptable carriers that are known in the art. The compositions must be sterile and must contain a therapeutically effective amount of the antisense oligonucleotides in a unit of weight or volume suitable for administration to a patient. The compositions, as described above, are administered in effective amounts. The effective amount will depend on the mode of administration, the particular condition being treated and the desired result. This will also depend, as discussed above, on the stage of the condition, the age and physical condition of the subject, the nature of the concurrent therapy, if any, and similar factors well known to the physician. For therapeutic applications, it is that amount sufficient to achieve a medically desirable result. In some cases, it is a local (site-specific) reduction of inflammation. In other cases, its inhibition of tumor growth and / or metastasis. In still other embodiments, the effective amount is that amount sufficient to stimulate an immune response that leads to the inhibition of an infection, or cancer. Generally, the doses of the active compounds of the present invention would be from about 0.01 mg / kg per day to 1000 mg / kg per day. It is expected that doses ranging from 50-500 mg / kg will be adequate. A variety of administration routes are available. The methods of the invention, generally speaking, can be practiced using any mode of administration that is medically acceptable, meaning any mode that produces effective levels of the active compounds without causing clinically unacceptable adverse effects. Such modes of administration include oral, rectal, topical, nasal, interdermal or parenteral routes. The term "parenteral" includes subcutaneous, intravenous, intramuscular or infusion. Intravenous or intramuscular routes are not particularly suitable for long-term therapy and prophylaxis. These could, however, be preferred in emergency situations. Oral administration will be preferred for prophylactic treatment because of the patient's convenience as well as the dosing schedule. When the peptides are used therapeutically, in certain embodiments a desirable route of administration is by pulmonary aerosol. Techniques for preparing aerosol delivery systems containing peptides are well known in the art. Generally, such systems must utilize components that do not significantly impair the biological properties of the antibodies, such as the binding capacity of paratope (see, for example, Sciarra and Cutie, "Aerosols," in Remington's Pharmaceutical Sciences, 18th edition, 1990, pp 1694-1712; incorporated for reference). Those of ordinary skill in the art can easily determine the various parameters and conditions for producing antibody or peptide aerosols without resorting to undue experimentation. Compositions suitable for oral administration may be presented as discrete units, such as capsules, tablets, lozenges, each containing a predetermined amount of the active agent. Other compositions include suspensions in aqueous or non-aqueous liquids such as a syrup, elixir or an emulsion. Preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic / aqueous solutions, emulsions or suspensions, including saline and regulated media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer or fixed oils. Intravenous vehicles include fluid and nutrient replenisher, electrolyte replenishers (such as those based on Ringer's dextrose) and the like. The preservatives and other additives may also be presented such as, for example, antimicrobials, anti-oxidants, chelating agents and inert gases and the like. Lower doses will result from other forms of administration, such as intravenous administration. In the event that a response in a subject is insufficient to the initial dose applied, higher doses (or effectively higher doses for a different route of delivery more located) can be used to the extent that the tolerance of the patient allows. Multiple doses per day are contemplated to achieve appropriate systemic levels of the compounds. Agents that may be combined, optionally, with a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" as used herein means one or more compatible solid or liquid fillers, diluents or encapsulating substances that are suitable for administration to a human being. The term "carrier" denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions are also capable of co-mingling with the molecules of the present invention, and with each other, in such a manner that there is no interaction that would substantially impair the desired pharmaceutical efficacy. The invention in other aspects includes pharmaceutical compositions of the agents. When administered, the pharmaceutical preparations of the invention are applied in pharmaceutically acceptable amounts and in pharmaceutically acceptable compositions. Such preparations may routinely contain salt, regulating agents, preservatives, compatible carriers and optionally other therapeutic agents. When used in medicine, the salts should be pharmaceutically acceptable, but pharmaceutically unacceptable salts can conveniently be used to prepare pharmaceutically acceptable salts thereof and not be excluded from the scope of the invention. Such pharmacologically and pharmaceutically acceptable salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, maleic, acetic, salicylic, citric, formic, malonic, succinic and the like. Also, the pharmaceutically acceptable salts can be prepared as alkali metal or alkaline earth metal salts, such as sodium, potassium or calcium salts. Various techniques can be employed to introduce nucleic acids of the invention, (e.g., antisense nucleic acids) into cells, depending on whether the nucleic acids are introduced in vitro or in vivo in a host. Such techniques include transfection of CaP04-nucleic acid precipitates, transfection of nucleic acids associated with DEAE, transfection with a retrovirus including the nucleic acid of interest, liposome-mediated transfection and the like. For certain uses, it is preferred to route the nucleic acid to the particular cells. In such cases, a vehicle used to deliver a nucleic acid of the invention within a cell (e.g., a retrovirus, or another virus: a liposome) may have a target molecule attached thereto. For example, a molecule such as an antibody specific for a surface membrane protein in the target cell or a ligand for a receptor in the target cell can be found in, or incorporated into, the nucleic acid delivery vehicle. For example, when liposomes are used to deliver the nucleic acids of the invention, proteins that bind to a surface membrane protein associated with endocytosis can be incorporated into the liposome formulation to direct and / or facilitate incorporation. Such proteins include capsid proteins or fragments thereof for a particular cell type, antibodies for proteins that undergo internalization in cyclization, proteins that direct intracellular localization and improve intracellular half-life, and the like. Polymeric delivery systems have also been successfully used to deliver nucleic acids into cells, as is known to those skilled in the art. Such systems still allow oral delivery of the nucleic acids. Other delivery systems may include delivery systems of time release, delayed release or sustained release (collectively referred to herein as controlled release). Such systems can avoid repeated administrations of the fugactic agent, increasing the convenience to the subject and the doctor. Many types of delivery delivery systems are available and known to those of ordinary skill in the art. These include polymer-based systems such as poly (lactide-glycolide), copolyoxalates, polycarprolactones, polyesteramides, polyorthoesters, polyhydroxybutyric acid and polyanhydrides. Microcapsules of the above drug-containing polymers are described in, for example, US Pat. No. 5,075,109. The delivery systems also include non-polymeric systems which are: lipids including sterols such as cholesterol, cholesterol esters and fatty acids or neutral fats such as mono-di and triglycerides; hydrogel release systems; silastic systems; peptide-based systems; waxy coatings; compressed tablets using conventional binders and excipients; partially combined implants; and similar. Specific examples include, but are not limited to: (a) erosional systems wherein the anti-inflammatory agent is contained in a form within a matrix such as those described in U.S. Patent Nos. 4,452,775, 4,667,014, 4,748,034 and 5,239,660 and (b) diffusional systems wherein an active component permeates at a controlled rate of a polymer as described in US Pat. Nos. 3,832,253 and 3,854,480. A preferred delivery system of the invention is a colloidal dispersion system. Colloidal dispersion systems include lipid-based systems that include oil-in-water emulsions, micelles, mixed micelles, and liposomes. A preferred colloidal system of the invention is a liposome. Liposomes are artificial membrane vessels that are useful as a delivery vector in vivo or in vitro. It has been shown that large unilamellar vessels (LUV), which vary in size from 0.2-4.0 μm, can encapsulate large macromolecules, RNA, DNA, and intact virions can be encapsulated within the aqueous interior and delivered to cells in a biologically active form (Fraley , et al., Trends Biochem. Scí., (1981) 6:77). For a liposome to be an efficient gene transfer vector, one or more of the following characteristics must be presented: (1) encapsulation of the gene of interest at high efficiency with retention of biological activity; (2) preferential and substantial binding to a target cell compared to non-target cells; (3) supply of the aqueous contents of the vesicle to the cytoplasm of the target cell at high efficiency; and (4) exact and effective expression of genetic information. The liposomes can be targeted to a particular tissue by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid or protein. Liposomes are commercially available from Gibco BRL, for example, as LIPOFECTIN ™ and LIPOFECTACE ™, which are formed from cationic lipids such as N- [1- (2,3-dioleyloxy) -propyl] -N, N, N chloride -trimethylammonium (DOTMA) and dimethyl dioctadecylammonium bromide (DDAB). Methods for making iiposomes are well known in the art and have been described in many publications. Liposomes have also been reviewed by Gregoriadis, G. in Trends in Biotechnology, (1985) 3: 235-241. In an important embodiment, the preferred vehicle is a microparticle or biocompatible implant that is suitable for implantation in the mammalian receptor. Exemplary bioerodible implants that are useful in accordance with this method are described in PCT International Application No. PCT / US / 03307 (Publication No. WO 95/24929, entitled "Polymeric Gene Delivery System"). PCT / US / 0307 preferably discloses a biodegradable, biocompatible polymer matrix containing an exogenous gene under the control of an appropriate promoter. The polymer matrix is used to achieve sustained release of the exogenous gene in the patient. In accordance with the present invention, the fugectic agents described herein are encapsulated or dispersed within the biocompatible, preferably biodegradable polymer matrix described in PCT / US / 03307. The polymer matrix is preferably in the form of a microparticle such as a microsphere (where an agent is dispersed through a solid polymer matrix) or a microcapsule (wherein an agent is stored in the core of a polymeric shell). Other forms of the polymer matrix containing an agent include films, coatings, gels, implants and stent. The size and composition of the polymeric matrix device is selected to result in favorable release kinetics in the tissue within the matrix being introduced. The size of the polymer matrix is further selected according to a delivery method, which will be used. Preferably, when an aerosol route is used, the polymeric matrix and the fugactic agent is encompassed in a surfactant vehicle. The polymeric matrix composition can be selected to have favorable degradation rates and also be formed of a material that is bioadhesive, to further increase the transfer efficiencies. The matrix composition can also be selected not to degrade, but rather, to release the diffusion over an extended period of time. In another important embodiment, the delivery system is a biocompatible microsphere that is suitable for local site-specific delivery. Such microspheres are described in Chickering et al., Biotech. And Bioeng., (1996) 52: 96-101 and Mathiowitz et al., Nature (1997) 386: 410-414. Both biodegradable and non-biodegradable polymer matrices can be used to deliver the agents of the invention to the subject. Biodegradable matrices are preferred. Such polymers may be natural or synthetic polymers. Synthetic polymers are preferred. The polymer is selected based on the period of time over which the release is desired, generally in the order of a few hours to a year or more. Normally, the release for a period ranging from a few hours to three to twelve months is more desirable. The polymer optionally is in the form of a hydrogen which can absorb up to about 90% of its weight in water and in addition, optionally cross-linked with multivalent ions or other polymers. In general, fugactic agents are delivered using a bioerodible implant in the manner of diffusion, or more preferably, by degradation of the polymer matrix. Exemplary synthetic polymers that can be used to form the biodegradable delivery system include: polyamides, polycarbonates, polyalkylene, polyalkylene glycols, polyalkylene oxides, polyalkylene terephthalates, polyvinyl alcohols, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinyl pyrrolidone, polyglycolides, polysiloxanes, polyurethanes and copolymers thereof, alkylcellulose, hydroxyalkylcellulose, cellulose ethers, cellulose esters, nitrocelluloses, acrylic polymers and methacrylic esters, methylcellulose, ethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, hydroxybutylmethylcellulose, cellulose acetate, cellulose propionate, acetate-butyrate of cellulose, cellulose acetate phthalate, carboxyethylcellulose, cellulose triacetate, sodium salt of cellulose sulfate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate) , poly (methacrylate) hexyl), poii (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly ( octadecyl acrylate), polyethylene, polypropylene, poly (ethylene glycol), poly (ethylene oxide), poly (ethylene terephthalate), poly (vinyl alcohol), polyvinyl acetate, polyvinyl chloride, polystyrene, polyvinyl pyrrolidone and lactic acid polymers and glycolic acid, poly (ortho) ester polyanhydrides, poly (bútic acid), poly (valeric acid), and poly (lactide-cocaprolactone), and natural polymers such as alginate and other polysaccharides including dextran and cellulose, collagen, chemical derivatives of ios (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations and other modifications routinely made by those skilled in the art), albumin and other hydrophilic proteins, zein and other prol amines and hydrophobic proteins, copolymers and mixtures thereof. In general, these materials are degraded either by enzymatic hydrolysis or exposure to water in vivo, by surface or volume of erosion. Examples of non-biodegradable polymers include ethylene vinyl acetate, (poly (meth) acrylic acid), polyamides, copolymers and mixtures thereof. Bioadhesive polymers of particular interest include bioerodible hydrogels described by H.S. Sawhney, C.P. Pathak and J.A. Hubell in Macromolecules, (1993) 26: 581-587, the teachings of which are incorporated herein, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly (methyl methacrylates), poly (ethyl methacrylates), poly (butyl methacrylate), poly (isobutyl methacrylate), poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methacrylate) methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate) and poly (octadecyl acrylate). In addition, important embodiments of the invention include pump-based hardware delivery systems, some of which are adapted for implantation. Such implantable pumps include controlled release microchips. A preferred controlled release microchip is described in Santini, JT Jr., et al., Nature, 1999, 397: 335-338, the contents of which are expressly incorporated herein by reference.
The use of a long-term sustained release implant may be particularly suitable for the treatment of chronic conditions. Long-term release, as used herein, means that the implant is constructed and disposed at therapeutic levels of delivery of the active ingredient for at least 30 days, and preferably 60 days. Long-term sustained release implants are well known to those of ordinary skill in the art and include some of the delivery systems described above. In certain embodiments, the agents of the invention are delivered directly to the site in which there is inflammation, for example, the joints in the case of a subject with rheumatoid arthritis, the blood vessels of an atherosclerotic organ, etc. For example, this may be achieved by attaching an agent (nucleic acid or polypeptide) to the surface of a balloon catheter; inserting the catheter into the subject until the portion of the balloon is located at the site of inflammation, eg, an atherosclerotic vessel, and inflating the balloon to bring the surface of the balloon into contact with the vessel wall at the site of occlusion . In this manner, the compositions can be directed locally to particular inflammatory sites to modulate immune cell migration at these sites. In another example, local administration involves an implantable pump to the site in need of such treatment. Preferred pumps are as described above. In a further example, when treatment of an abscess is involved, the fugactic agent can be delivered topically, for example in an ointment / dermal formulation. Optionally, the agents are supplied in combination with other therapeutic agents (e.g., anti-inflammatory agents, immunosuppressive agents, etc.). The invention will be understood more broadly for reference to the following examples. These examples, however, are merely intended to illustrate the embodiments of the invention and are not construed to limit the scope of the invention.
EXAMPLE 1 Example 1 describes experiments and findings demonstrating that bidirectional migratory response of T cells to specific gradients of chemokine are associated with differential changes in the expression of genes encoding proteins involved in the signal transduction path SDF-1 / CXCR4 .
Methods Primary human or murine T cells were exposed to specific gradients of SDF-1 to induce chemotaxis or fugetaxis in vitro and in vivo. The Zigmund / Hirsch chamber and microfabricated devices as well as a murine model of allergic peritonitis were used to establish SDF-1 gradients defined in vitro and live, respectively. Purified T cells were generated from these systems and RNA amplified was examined using genomic array technology (Affymetrix). These results were validated by RT-PCR and Northern blot. The experiments were performed on T cells that had not been exposed to SDF-1 or that had been exposed to chemokine in the absence of a gradient. Cell Cultures: CD4 + CD45 + RA cells were obtained from peripheral blood leukocyte layer samples from healthy donors. Transpoxide assays: Trans-well assays were performed using 0.4 μm pore size filters (23 mm diameter, with polycarbonate membrane: Corning Inc., New York). 10 x 106 cells suspended in 0.5% IMDM containing FBS were added to the upper chamber of the trans-well. To create positive, negative, uniform and absent gradients, either 0.5% of medium FBS IMDM alone or medium plus SDF-1a. Total RNA Extraction: Total RNA was extracted from all samples using Gibco TRIzol protocol (GIBC-BRL, Life Technologies, Rockville, MD) with 1 ml of Trizol per 10-20 x 10 6 cells. The total RNA was brought to a concentration of 1 μg / μl and 5-10 μg was used in the Affymetrix microchips. CRNA preparation and Microchip Hybridization Conditions: cRNA probes were prepared according to the GeneChip Technical Expression Analysis Manual and as previously described (Warrington et al., 2000). Briefly, 5-10 μg of total RNA was used to synthesize double-stranded cDNA using SuperScript Choice System (GIBCO-BRL) and a T7- (dT) -24 primer (Geneset Oligos, La Jolla, CA). The cDNA was purified by extraction of phenol / chloroform / isoamyl alcohol with Phase Immobilization Gel (5Prime 3Prime, Boulder, CO) and concentrated by EtOH precipitation. In vitro transcription yielded biotin-labeled cRNA using a BioDisposited Elevated Production Transcript Labeling Device (Affymetrix) according to the manufacturer's instructions. The cRNA was linearly amplified with T7 polymerase, the biotinylated cRNA was cleaned with RNeasy Mini (Qiagen) and 20 μg of labeled cRNA was fragmented (Warrington et al., 2000). The fragmented cRNA was hybridized to the microarray for 16 hours at 45 ° C with a constant rotation of 60 rpm in a 640 GeneChip Hybridization Furnace (Affymetrix). After washing, the arrays were stained with streptavidin-phycoerythrin (Molecular Probes, Eugene, OR) and amplified by biotinylated anti-streptavidin (Vector Laboratories, Burlingame, CA) using the Fluidica GeneChip 400 Station (Affymetrix) and screened in the scanner. GeneArray (Affymetrix). The intensity for each feature of the arrangement was captured with Affymetrix GeneChip Software v5.0, according to standard Affymetrix procedures (Warrington et al.2000). Statistical Analysis of Expression Data: To allow comparison between experiments, Affymetrix image files (.cel) were loaded into the Expression Data Analysis System v4.0 of the Rosette Resolver and normalized according to the Resolver error model (See Waring et al., 2001, Lock et al., 2002 for description). Q-PCR Verification of Genetic Targets: Total RNA from primary T cells was isolated, purified and quantified as described above. QRT-PCR was performed using the Brilliant Stage QRT-PCR equipment (Stratagene, La Jolla, CA) containing 1 SYBR Green (1: 30,000, Molecular Probes), forward and reverse primers (50 nM each); Invitrogen) and sample RNA (the amount was variable, depending on the abundance of transcription).
Results The microchips of the same combination were combined using software based on the Rosette Resolver error model, as described in the Methods. The combined experiments were then compared with each other in different combinations to direct different sub-components of the hypothesis. M / M - basal conditions; CM - chemokinesis: CT - positive gradient SDF-1 chemotaxis; and FT - negative gradient SDF-1 fugetaxis. The gene expression profile for T cells that underwent chemotaxis differs from the profile generated for T cells that underwent fugetaxis in response to SDF-1 gradients in several significant respective. Cluster analysis of gene expression showed that molecules encoding genes known to be involved in SDF-1 signal transduction were expressed significantly and differentially (p <0.05 for 1.7 to 21-fold changes in RNA expression) when The cells that had experienced fugetaxis or chemotaxis were compared. Of particular observation, these differentially expressed genes encoded members of the G protein-coupled receptor kinase, cellular tyrosine kinase, PI-3 kinase and Rho GTPase cascades as well as the cyclic nucleotide metabolic pathway. The gene expression profile for the control T cells were exposed to SDF-1 in the absence of a also deferred gradient of the profiles generated from the cells corresponding to chemokine gradients. The data is presented in Tables 1-6 (Figures 3-8). Signaling molecules that are up-regulated in a uniform gradient of the SDF-1 (chemokinetic) gradient of SDF-1 include PTK2 (+ 6.88) and the Regulator of protein signaling 10 (+ 2.53). The signaling molecules are sub-regulated in a uniform gradient of the SDF-1 (chemokinetic) gradient of SDF-1 including Phospholipase C, beta 3 (-2.54), RAS protein activator p21 (GAP) 3 (-2.20), Ras guanil release protein 2 (calcium / DAG) (-2.16), protein G-coupled receptor kinase 6 (-2.15), Rho-specific GEF (p114) (-1.70), 80K-H substrate kinase C of protein (-1.70). Signaling molecules that are up-regulated in the presence of a directional gradient (chemotactic or fugactic) against neutral include transform the growth factor, beta 1 (1.92 Chemiokinetic versus Chimatactic: 1.70 Fumetic Chemo) and the nucleotide binding protein of Guanine (1.74 Chemokinetic versus Chemotherapy: 1.78 Chemokinetic vs. Fugactic). Signaling molecules that are sub-regulated in the presence of a directional gradient (chemotactic and fugactic) versus neutral (chemokinetic) include inflammatory factor 1 allograft (12.9 Chemokinetic versus Chemotactic; -11.9 Chemokinetic versus Fugactic), phosphoserine similar to phosphothase (-4.24 Chemokineetics versus chemotactic; 5-76 Chemokinetics versus Fugetica) downstream signaling of BCR 1 (-1.86 Chemokineetics versus Chemotactics; -2.14 Chemokinetics versus Fugectatic) rat sarcoma 2 viral oncogene v-Kit-ras2 Kirsten (-1.84 Chemokinetic versus Chemotactic; -1.95 Chemokinetic versus Fugactic). Signaling molecules differentially expressed between a positive (chemotactic) and negative (fugectic) gradient of SDF-1. Signaling molecules that are most highly expressed in a chemotactic gradient of SDF-1 (against the fugectic gradient) include PTK2 (focal adhesion kinase) (8.59), MAP kinase kinase kinase kinase 2 (7.30), nucleotide binding protein of guanine (4.95), phosphatase receptor PT (4.20), beta kinase binding protein CDC42 (3.23), Ral GEF (RalGPSIA) (2.81), MAP kinase 7 (2.78), Autotaxin (2.63), Inositol receptor 1, 4,5-triphosphate (2.60) Phosphoinositide-3-kinase, gamma (2.48), PT phosphatase, non-receptor (2.02), Ras p21 protein activator (GAP) (1.98), guanyl release protein 2 Ras (1.98) and 2 kDa subunit of Arp23 complex (1.95). Signaling molecules that are most highly expressed in a fugactic gradient of SDF-1 (against a chemotactic gradient) include cycle 42 of cell division (4.93), protein kinase S6 Ribosomal (2.91), protein 2 associated with BAII (2.84) ), GPTase regulator associated with FAK (2.59), Protein kinase C, beta 1 (2.16), phosphoinositide specific phospholipase C-beta (1.99), nitric oxide synthase I (1.99), phosphotidylinositol-4-phosphate 5-kinase (1.82) and MAP kinase kinase kinase kinase 4 (1.72).
Conclusions This work represents the mechanism of T cell migration in vitro and in vivo in response to SDF-1 gradient and shows that the regulation of gene expression associated with the signal transduction pathway for chemotaxis is different from that associated with fugetaxis. This work forms the basis for identifying potential molecular targets for specific therapeutic agents that could selectively block or ameliorate the chemotactic or fugectic responses of T cells to SDF-1 gradients in vivo.
EXAMPLE 2 Example 2 describes experiments and findings demonstrating a new aspect of neutrophil migration in response to chemokine, interleukin-8, primarily bi-directional movement. Specifically, the use of non-peptide antagonists of the IL-8 receptor, CXCR2, and known inhibitors of chemokine signal transduction reveal that neutrophils can make a directional decision to move up and down a gradient of I L-8 and that this The decision is dependent on the slope of the gradient, the absolute concentration of the chemokine at which the neutrophil is exposed to, and the occupancy level of the CXCR2 receptor. In addition, the directional decision of neutrophils to migrate below a gradient was also found to be differentially sensitive to inhibitors of signal transduction when compared to migration above the gradient.
Methods Primary human T cells are exposed to specific gradients of I L-8 to induce chemotaxis or fugetaxis in vitro and in vivo in microfabricated devices. The intravital microscope and digital image analysis were used to examine bidirectional movement of neutrophils in response to IL-8. Isolation of Neutrophils: Human whole blood was obtained from healthy volunteers by pricking the vein in tubes containing sodium heparin (Becton Dickinson, San Jose, CA). The whole blood was centrifuged for 4 minutes at 2400 rpm and the plasma was removed The resulting granule was resuspended in an Iscove Modified Dulbecco's Medium (IMDM; Cellgro MediaTech, Rendon, VA) with 0.5% (w / v) fetal calf serum (FCS; Cellgro MediaTech), 25 ml suspension stratified on 10 ml of Lymphocyte Separation Medium (ICN, Irvine, CA) and centrifuged for 40 minutes at 1600 rpm at 22 ° C. The supernatant was aspirated, the resulting granule was resuspended in IMDM with 0.5% (w / v) FCS and 2% (w / v) dextran (Sigma-Aldrich, St. Louis, MO), and red blood cells ( RBC) were allowed to settle for 30 minutes at room temperature. The supernatant was transferred in a clean tube and centrifuged for 5 minutes at 200 rpm. The supernatant was aspirated, the granule was mixed with cold ddH20 for hypotonic lysis of the remaining RBCs, and transferred to IMDM with 0.5% (w / v) FCS. Isolated neutrophils were washed and resuspended in IMDM with 0.5% (w / v) FCS, determined to be 95% pure, and 99% viable by exclusion of Trypan Blue. The Manufacture and Preparation of the Microfluidic Linear Gradient Generator: The microfluidic linear gradient generator was manufactured in poly (dimethylsiloxane) (PDMS; Sylgard 184, Dow Corning, NY) using rapid prototyping and flexible lithography as previously described. Briefly, a high-resolution printer was used to generate a transparency mask from a computer-aided design image file. The mask was used in contact photolithography with SU-8 photoresistor (Microlithography Chemical Co. Newton MA) to generate a positive relief of photoresistor stamped on a silicon wafer. The replicas with increased channels were manufactured by curing PDMS prepolymer against the stamped wafer. The input and output ports for media and cell suspension were punched out of the cured PDMS replica using a shaped syringe needle. The PDMS replica and the glass substrate were placed in an oxygen plasma generator (150 mTorr, 100 W) for 1 minute. Immediately after the plasma treatment, the PDMS and glass replica were placed against each other and joined irreversibly. The polyethylene tube (Becton Dickenson) was inserted into the inlet and outlet ports to make the fluidic connections. The tube was connected to a PHD 2000 syringe pump (Harvard Apparatus, Holliston, MA) to complete the configuration. Hemostats were used to control the flow during cell loading. Characterization of the Linear Gradient Generator: Verification of the gradient formations in the microfluidic device was carried out using phosphate-buffered saline solutions (PBS); Cellgro MediaTech) and fluorescein isothiocyanate (FITC; Sigma-Aldrich) as previously described. Verification of the gradient formations in the microfluidic device was carried out using solutions of Dulbecco's phosphate saline solution (DPBS, Cellgro MediaTech) and fluorescein isocyanate (FITC; Sigma-Aldrich) as previously described. Briefly, PBS and PBS with 100 μM FITC were introduced into the device. Fluorescent micrographs were taken from stable gradients at various stable flow rates (0.1, 1, 10, 10 mm / s). The graphs of the fluorescent intensity profile through the migration channel demonstrate the generation of linear temporal and spatially stable gradients; profiles at low flow rates are smooth and continuous, while step gradients of increased flow velocity yields as fluid flow become more laminar. (Li Jeon et al, Nat Biotech 2002, Li Jeon et al, Langmuir 2000). Microfluidic Migration and Microscopy Trial Time Lapse: Neutrophils (1 x 103 cells) were uniformly placed through the migration channel and allowed to migrate under a linear gradient of human lterleucine-8 (72a., PeproTech, Rocky Hill, NJ) in IMDM with 0.5% (w / v) of FCS flowing at 0.1 mm / seconds. The migration was observed in a Nikon Eclipse TE2000-S microscope (Nikon, Japan) through a 10X Plan-Fluor (Nikon) objective. Bright field images were taken every 30 seconds using a Hamamatsu digital camera C4742-95 (Hamamatsu, Japan), controlled by IPLab 3.6.1 (Scanalytics, Fairfax, VA). The cell movement was always observed at the reference point along the migration channel. The gradients were also calibrated at this reference point. The migration was quantified for all cells across the gradient. The Construction of Digital Videos for Analysis Quantitative: Digital videos made from time-lapse video microscopy file stacks or S-VHS videotapes were made using a combination of IPLab 3.6.1, Photoshop 6.0 (Adobe Systems, San Jose, CA) and Apple QuickTime Pro 5 ( Apple Computer, Cupertino, CA). Migration tracking was carried out using MetaMorph 4.5 (Universal Imaging, Downington, PA), the object tracking application, which generated Cartesian coordinate data tables for each cell traced. Mathematical Analysis of Cell Migration in Linear Gradient Generator: The angular correlation function, or cosine correlation function, was calculated for each experiment. For experiments without a gradient, the correlation function decayed exponentially with the increased time interval, although the function decayed much slower, potentially with a power law, for experiments with a gradient; in all cases, the angle correlation over time was increased as absolute [I L-8] increased. The fact that the angular choice correlates with time allowed us to compare the angular frequency distributions as a directional migration index. The cell movement within the linear gradient generator was characterized based on a predisposed random walking model (Moghe et al, J Immun Methods 1995, Tan et al, J. Biomed Mater Res 2000), thus the movement between the tracked positions in successive frames of a video can be considered as a vector, with an associated length and angle. The MetaMorph tracking data was analyzed in Excel (Microsoft, USA) and MATLAB 13 (Mathworks, Inc.) to determine average quadratic shifts, motility coefficients, angular frequencies and correlations, random path lengths, and migration speeds . Cell motility was characterized as follows: For each cell, the quadratic displacement R2 (t) was calculated in time interval t, < R2 (t) > = < (x (t0 + t) - x (t0)) 2 + (y (t0 + t) - y (t0)) 2 > , where t0 is the time at the origin. The origin was changed throughout the data set and the offsets were averaged for overlapping time intervals. A global average was made on all the cells in the set to calculate the average quadratic displacement. The cellular movement is mathematically molded into a random, biased, correlated gear, this can be written as < R2 (t) > = 2S2P [t - P (1 - e -t / p)], where S and P are measurements of the movement index and persistence time respectively. When the time interval t is much larger than the persistence time P, the average quadratic displacement becomes linearly proportional to t, analogous to Brownian diffusion. < R2 (t) > = 2S2Pt = 4 μt where μ is the motility coefficient. The decline and intercept of at least the quadratic region adapted to the linear section of the average quadratic shift gives an estimate of μ and P, respectively. Additionally, a "persistence index" (Pl) of the movement or total free trajectory, was calculated as the total displacement of the cell divided by the total distance traveled along the route. Pl is an indicator of rotation behavior, with 1 indicating movement in a straight line and 0 indicating no real displacement. The directional tilt of cell motility was quantified as follows: For each cell, angle frequency histograms show the distribution of angles associated with each displacement vector between successive time intervals of migration. The union of these histograms can be varied to reduce the stochastic noise associated with a random walk. The x-axes of these histograms are folded around a point to create a circular histogram that presents the angular frequencies at 360 °. The angular correlation function (or cosine correlation function) was calculated as: G (t) = «p (t) .f (t + t) >; = < C0S [f (t - = - t)] > , Where f (t) is the angle that the displacement vector makes with respect to the direction of the gradient. The weakening of this function with increased time interval indicates the correlation between the successive angles of rotation and is a measure of the persistent direction or memory of the cells. The directional inclination is quantified with respect to the established gradient, the "average chemo-tropic index" (MCI) was calculated, which is defined as the actual path length crossed by a cell with respect to the direction of the established gradient divided by the total distance traveled and is a measure of the accuracy of orientation. Cl = The index of each cell was calculated and then averaged over the entire population. The average chemotropic index will be 1 if the cells move directly over the gradient, 0 if there is no preferred orientation, and -1 for migration directly below the gradient. Signaling Pathway Inhibitors: The cells were treated with pertussis toxin (100 ng / ml, 30 minutes at 37 ° C), wortmanin (1 μM, 10 μM, 20 minutes at 37 ° C 8-Br-cAMP (1 mM 15 minutes at room temperature), 8-Br-cGMP (1 mM, 15 minutes at room temperature) (Sigma-Aldrich), or the non-peptide antagonist CXCR2, SB225002 (1 pM, 100 pM, 1 nM, or 1 μM during 15 minutes at 37 ° C, Calbiochem, CA) Immediately after the treatment, cells were seeded into the migration channel of the microfluidic device and allowed to migrate as described above Intravital Microscopy: Sprague Dawley rats were purchased (200-300 g) from Harlan-Olac (Bicester, UK) Male rats were prepared for intravital microscopy Briefly, after sedation with Hipnorm im (mixture of fentanyl-fluanisone, 0.1 ml, Janssen-Cilag, High Wycombe, UK), animals were anesthetized with sodium pentobarbitone iv (30 mg / kg loading dose followed by 30 mg / kg / h; Rhone Mérieux, Harlow, U.K.). Animals were kept at 37 ° C in a heated microscope stage constructed as usual. After the abdominal incision of the midline, the mesentery next to the terminal ileum was carefully placed on a glass window in the microscope stage and nailed into position. The mesentery was kept warm and moist by continuous application of balanced salt solution Tyrode (Sigma Aldrich). The small mesenteric post-capillary veins (15-40 μm in diameter) were seen in a straight fixed stage microscope (Axioskop FS, Carl Zeiss, Welwyn Garden City, U.K) adapted with water immersion objectives. The images were captured with a digital camera (C5810-01, Hamamatsu Photonics UK, Enfield, UK) to be viewed on a monitor (PVM-1453 MD, Sony, UK, Weybridge, UK) and stored by the video recorder (AG-MD830E, Panasonic UK, Bracknell, UK). Since the intravital microscope resolution does not allow definitive distinctions to be made between different subpopulations of leukocytes, all responses were quantified in terms of leukocyte behavior. Therefore, circulating leukocytes were defined as those cells that move slower than flowing erythrocytes, and the circulating flow was quantified as the number of circulating cells moving beyond a fixed point in the venular wall per minute, averaged for 4-5 minutes. The tight adherent leukocytes were defined as those that remain stationary by at least 30 within 100 μm of the segment of a small vein. Extravasated leukocytes were defined as those in the perivenular tissue adjacent to, but remaining within a distance of 150 μm of a 100 μm length of the vessel segment under study. After the baseline readings of performance, adhesion and transmigration were taken; CINC-1 in the final concentrations of 10"9, M, 10 ~ 8 M or 1 O" 7 M (Peprotech) was applied topically to the mesenteric tissue in the superfusion regulator. The leukocyte responses within the chosen vessels were quantified for up to 180 minutes, during which the topical application of CINC-1 was maintained. In each animal, the multiple vessel segments of the appropriate vessels were quantified. The migratory cell videos were built for quantitative and mathematical analysis as described above; At the end of certain in vivo experiments, the mesentery was stained with acridine orange (Sigma Aldrich), nuclear stain, scanned with 488 nm laser line generated from argon laser, and observed by confocal microscope (LSM5 PASCAL, Axioskop 11 FS, Carl Zeiss) to verify that the migratory cells were neutrophils. Mathematical Modeling of Continuous Gradients in vivo: The chemokine concentration profile in the steady state mesentery was predicted using a novel in vivo model based on classical diffusion equations applied in a spherical model of the post-capillary vein girl, and the assumption that the chemokine receptor-dependent transport by endothelial cells is the main mechanism to generate the closeness of the gradient of the small post-capillary veins. The steady-state solution was calculated for the concentration gradient around a sphere in a homogeneous medium, with the two boundary conditions: 1) the distant concentration of the sphere is constant, and 2) the chemokine flow through the surface of the sphere is also constant. Other mechanisms of chemokine transport outside of tissue were considered less significant due to the low lipid solubility of CINC-1 and IL 8 and the presence of tight intercellular junctions between endothelial cells in the absence of vasoactive signals (Middleton et al, Cell 1997). In this way, the steady-state concentration C at distance r from the capillary wall was calculated as: F0a2 C (r) = Co-D (r + a) 'where, C0 is the chemokine concentration in the perfusion solution (either 10 or 100 nM), in the vessel radius (12.5 μm), F0 the rate of chemokine incorporation, and D the diffusion coefficient. The rate of chemokine uptake by endothelial cells was estimated in the range of 1,000 to 10,000 molecules / cells / minute by comparison with endocytosis rates for other proteins (Schwartz, Annu Rev Immunol, 1990). A value of 0.6 x 10"7 cm2 / s for diffusion coefficient of CINC-1 (MW 7,800) in the mesentery was interpolated from the albumin diffusion coefficient (MW 66,000) experimentally determined in similar tissues (Parameswaran et al. ., Microcirculation 1999).
Results To examine whether neutrophils were capable of continuous gradients of bi-directional migration of IL-8, variable steepening in microfabricated devices were established as previously described (Li Jeon, N., et al., (2002) Nat Biotechnol, 20 (8): 826-30). Previous work with microfabricated devices demonstrated solid chemotaxis of primary human neutrophils in gradients of recombinant human L-8 between 0 and 50 nM and 0 and 100 nM (Li Jeon, N., et al., (2002) Nat Biotechnol. (8): 826-30). Since it had previously been shown that the T cell undergoes fugetaxis at high concentrations of chemokine, SDF-1, gradients from 0 to 12 nM, 0 to 120 nM, 0 to 1.2 μM and 0 to 2.4 μM for IL-8 are they examined additionally. Each gradient was initially calibrated and characterized as shown in Figures 10A to D and as previously described (Li Jeon, N., et al., (2002) Nat Biotechnol, 20 (8): 826-30). The differential concentration of chemokine across the migration channel ranges from 0.0267 nM per miera to 5.34 nM per miera or the equivalent of a difference in chemokine concentration of 0.267 nM or 50.34 nM through the length of a neutrophil of 10. long mieras. Neutrophils were also exposed to control conditions not including chemokine or uniform concentrations of I L-8 of 12 nM, 120 nM or 1.2 μM in the migration channel. Human neutrophils were loaded into the device and their migration was tracked and quantified using MetaMorph software together with the MatLab software, respectively (Figures 10E to H). The initial and final density of the cells through the migration channel was plotted for each of the conditions and the angular frequency of all the directional movements determined for each cell using MetaMorph (Figures 101 to L). Cells exposed to non-chemokine or chemokine at a uniform concentration through the migration channel experienced chemokinesis characterized by angular frequencies in all directions. In contrast, cells placed on gradients between 0 and 12 nM and 0 and 120 nM demonstrated predominantly chemotaxis with predominant angular frequencies occurring towards the peak concentration of chemokine in the gradient (Figures 10M to P). Surprisingly, when the cells were exposed to the chemokine gradient of migratory behaviors 0 to 1.2 μM they were more complex. Cells in the lower third of the chemotaxed gradient toward higher levels of chemokine while cells originating in the upper third of the gradient experienced fugetaxis down the gradient and outside the peak concentration of chemokine: Cells initially start in a position in the central third of the gradient that experienced chemokinesis. The cell density through the migration channel before and after the neutrophil migration reflects a re-distribution of cells randomly arranged to the central third of this gradient (Figure 10L). In addition, the angular frequency distribution for this gradient reflects a predominant movement outside the chemokine in this gradient (Figure 10P): Cells exposed to the pronounced chemokine gradient studied, (0 to 2.4 μM) underwent chemokinesis despite its position within of the gradient (data not shown). In this manner, solid bidirectional neutrophil migration within a pronounced and temporally and spatially stable gradient of I L-8 was observed. In addition, the videos of cells that migrate in gradients of I L-8 were analyzed using MetaMorph software and each position of each cell in each frame was defined by its Cartesian coordinates within that frame. It was therefore possible to examine quantitative parameters that describe each migratory path of cells. A random gait mode was used to quantify cell migration, and previously defined parameters of average velocity, Random motility coefficient and persistence time to measure how "diffusive" or "ballistic" cell migration is and the average chemotropic index to measure the directionality of the movement towards or outside of a chemokine were used. Mean velocity and mean square displacement for cells that migrate in the absence of a chemokine or within gradients where chemotaxis (0 to 12 nM and 0 to 120 nM) or fugetaxis (0 to 1.2 μM) is predominantly seen (Figure 11A and 11B). The average speed measurement demonstrates that the cells undergo chemotaxis in the gradient from 0 to 120 nM or fugetaxis in the gradient from 0 to 1.2 μM migrates at similar rates. The average quadratic shift reflects the directional inclination of the random gait of the cells. Producing chemotaxis and fugetaxis in the cells demonstrates an exponentially increased directional tilt when migrating in gradients 0 to 120 nM and 0 to 1.2 μM, respectively. The gradient of the linear section of the average quadratic displacement graph for cells migrating in each experimental and control condition defines the random motility coefficient for cell migration (Figure 15, Table 8). Random motility coefficients are significantly higher in cell direction migration in the gradient from 0 to 120 nM and 0 to 1.2 μM in the presence of a uniform concentration of I L-8 of 120 nM where chemokinesis predominates. The interceptor y of the linear segment of the average quadratic displacement graph indicates the persistence time which is a measure of how the "ballistic" cell movement is (Figure 15, Table 8). The persistence time for cells that migrate in linear gradients of variable steepness are greater than those for cells presented without chemokine or a uniform concentration of chemokine. The persistence times for cell movement in the gradient of I L-8 where chemotaxis (21.5 minutes) or fugetaxis (10.9 minutes) are predominantly seen to be higher than those seen for cells undergoing chemokinesis in the absence of a gradient (0 minutes) or a uniform concentration of I L-8 (4.5 minutes). Chemotaxis and fugetaxis increase or decrease a "ballistic" movement of the gradient approach of IL-8, while cell movement in the absence of a chemokine gradient is more "diffusive". The analysis of cell displacement within a random migration model of cell migration does not measure the directionality of movement towards or away from a chemokine. In addition, by treating all cells equally within a gradient it assumes that all cells behave in the same way in the same gradient. Since it had been identified that the cells can migrate above or below a gradient in a manner that is dependent on their precise position within the gradient, the measurement of the average chemotropic index (MCI) was used to define the directionality of movement on ( positive values) or below (negative values) of a gradient and cell movement was analyzed three arbitrary sectors of each gradient (Figure 15, Table 8). Cells exposed for uniform concentrations of chemokine at 120 nM or without chemokine and MCI values of -0.02 +/- 0.01 and 0.00 +/- 0.02 respectively. Cells that underwent chemotaxis in gradients between 0 and 12 nM and 0 and 120 nM showed MCI of + 0.32 and +0.39 respectively. In contrast, cells exposed to the pronounced gradient from 0 to 1.2 μM demonstrated a negative MCI of -0.13 which supports the view that the predominant movement of cells in the gradient was outside the peak concentration of IL-8. Cells that migrate in the steepest gradient from 0 to 2.4 μM exhibit chemokinesis. To further analyze the effect of the gradient steepness influence and the absolute concentration of the chemokine gradient each gradient was divided into three equal segments and the cell populations, and starting the movement in each segment were analyzed separately. Cells that migrate in all sectors of positive MCI that reveal the gradient from 0 to 12 nM and 0 to 120 nM from +0.21 to +0.44. While the cells that migrate in the lower segment of the 0 to 1.2 μM gradient had an average sectional MCI of +0.2, the cells in the middle third and upper third of the gradient had negative MCIs of -0.14 and -0.22 respectively. These quantitative data that examine both the steepness and direction of the random walk confirm the finding of bi-directional neutrophil migration. In addition, these quantitative data confirm that the directional decision of a cell to move above or below a gradient is determined by the steepness of the gradient and the absolute concentration of the chemokine that is exposed within the gradient. Since receptor occupancy is known to play a role in the conformation and directional decision gradient sensation in the context of eukaryotic chemotaxis cells, it was postulated that chemokine receptor occupancy by a chemokine could also play a critical role in the decision of a cell to move up or down a chemokine gradient. Thus, an SB25002, the receptor-specific non-receptor antagonist (L-8, CXCR2, was used to examine this hypothesis.) Neutrophils were pre-treated with SB225002 at concentrations between 1 pM and 1 μM and then exposed from gradients 0 to 1.2. μM of I L-8 in microfabricated devices as described above The cell migration videos were analyzed using MetaMorph software and MathLab to generate normalized angular frequencies determined for cell migration in each of the three gradient sectors. a normalized angular frequency of 1.0 whereas the inhibition of angular frequencies fugectic or chemotactic results in a normalized frequency of <; 1.0 and the increase of any directional response results in a value greater than 1. This analysis allows to quantify precisely the effect of a given concentration of the inhibitor on the directional decision of the cell to move up or down a gradient. The lower concentrations of SB225002 (1 pM and 100 pM) lead to significant inhibition (p = 0.0037 and 0.0210) of fugetaxis whereas chemotaxis was actually increased under these conditions (Figure 12). Gradually increasing concentrations of SB225002 eventually inhibited both fugetaxis and chemotaxis. These data indicate that receptor occupancy plays a significant role in determining the directional decision of a cell to move up or down a pronounced I L-8 gradient. In addition, although I L-8 binds both CXCR2 and CXCR1 on the cell surface, bidirectional human neutrophil signaling was evidently critically dependent on CXCR2. It has previously been shown that the signaling path for chemotaxis is different from that for chemo-rejection or fugetaxis. It is known that T-cell fugetaxis in response to SDF-1 in standard transmigration assays was differentially more sensitive for inhibition by the cyclic 8-Br-cAMP agonist intracytoplasmic than chemotaxis. In addition, it has been shown that T cell chemotaxis was differentially more sensitive for inhibition by the tyrosine kinase inhibitor, genistein, then it was fugetaxis. The study of neutrophil migration in microfabricated devices allows us to precisely examine the effects of these inhibitors on quantitative parameters of cell migration including the directional tilt of cells in the context of precisely defined and stable chemokine gradients. Primary human neutrophils were pre-treated with known inhibitors of the chemokine signal transduction pathway including pertussi toxin, wortmanin, genistein, 8-Br-cAMP and 8-Br-cGMP and then exposed to gradients of I L-8 where the chemotaxis or fugetaxis were seen. The effect of the inhibitor on the directional migration towards or away from chemokine was quantified by determining the directional motility index of the cells that migrate in the context of these gradients. The vector angles of movement corresponding to the movement above the gradient (30 to 150 degrees - see Figure 13) were defined as the vector angles of chemotactic and measured movement corresponding to the movement below the gradient (210 to 330 degrees - see the figure) were defined as fugactic. The directional choice of cells to move up or down a chemokine gradient were therefore compared in the presence and absence of an inhibitor. Active movement with the selective inhibition of directional sensation manifests as an inverse relationship in the distribution of angular frequencies between the fugactic and chemotactic sectors; if the fugetaxis is inhibited (< 1) the chemotaxis will be increased above normal (> 1). The reversal of the directional sensation manifests as a decrease in the angular frequency distributions in both sectors to zero. In this way, neutrophil chemotaxis and fugetaxis were shown to be significantly inhibited by pertussis toxin (p = 0.007 and p = 0.003 respectively), 8-Br-cAMP also selectively inhibited fugetaxis (p = 4.6 x 10"6) while the same concentration of this intracytoplasmic nucleotide agonist increased chemotaxis (p = 0.0008) .Pre-treatment of wortmanin cells before placement in the gradient from 0 to 120 nM or 0 to 1.2 μM generated more complex results than expected. Wortmanin significantly inhibited chemotaxis (p = 0.0020) and increased fugetaxis (p = < 0.0001) in the gradient from 0 to 120 nM and in contrast to this chemotaxis increasing significantly (p <; 0.0001) and inhibiting fugetaxis (p < 0.0001) in the gradient context of 0 to 1.2 μM of IL-8. In addition, the differential sensitivities of chemotaxis and neutrophil fugetaxis to wormanin and 8-Br-cAMP were demonstrated. Both PI3K and cAMP have been shown to play a significant role in the gradient sensation and directional decision making in eukaryotic cells including Dictiostelio, neutrophils, neurons and T cells. It was also shown that levels of ctracitoplasmic cAMP differentially inhibit fugetaxis or chemorepulsion that is consistent with previous findings in eukaryotic neurons and T cells. The wortmanin system inhibited the predominant direction of movement observed under control conditions in the gradient and increased the opposite directional decision that was not predominantly seen previously under control conditions. The distribution of PI3K and PTEN to the end of cell conduction or tracking is thought to play a critical role in directional decision making in the context of eukaryotic cell chemotaxis. Chemotaxis is deregulated in the context of wortmanin in the surface gradient from 0 to 120 nM as expected, but fugetaxis is surprisingly increased. When fugetaxis is inhibited by the wortmanin in the chemotaxis of the pronounced I L-8 gradient it is increased. These data support previous work that indicates an independent PI3K path that governs the directional decision of neutrophils and indicates that the conduction and tracking ends may be interchangeable and that the location of PI3K and, or a second protein or proteins such as PTEN may determine the directional decision in the absence of the PI3K activity. Having demonstrated the solid bidirectional migration of neutrophils to the I L-8 gradient defined in vitro, this observation was confirmed in vivo. The neutrophil migration responses to the ortholog of IL-8, the cytosine-induced neutrophil chemoattractant-1 (CINC-1), was evaluated in a rat model. CINC-1 and I L-8 are known and potent chemoattrants for murine neutrophils and signal migration through CXCR2. Rat CINC-1, unlike rat I L-8 has been cloned and is commercially available. Diffusive chemokine gradients were established in tissues adjacent to small veins in the mesentery that had been externalized in anesthetized animals. The diffusive gradients with peak concentrations adjacent to the point of superfusion and declining towards the small vein as a result of adsorption of chemokine by matrix proteins, the binding of the chemokine to the receptor and the internalization of the chemokine / receptor complexes and the representation of chemokine on the luminal surface of endothelial cells. Chemokine gradients can be mathematically modeled in this context on the basis of the predictable absorption and diffusion rates of chemokine through the tissue (Figures 14A to C). It is important to note that this gradient model predicts that the gradient shape between the source of the chemokine superfusion and the vessel wall is the same shape for all peak chemokine concentrations. The steepness of the gradient at any fixed point between the supercombined chemokine and the vessel wall will therefore remain constant while the absolute concentration of the chemokine seen at that point varies. The in vivo model therefore confirms its use for determining the effect of gradient steepening and the absolute concentration of the directional decision of cells in vivo. Two types of experiments were established in this model. First, the mesenteric tissue adjacent to a small vein was supercombed with the chemokine in a fixed concentration of 1 nM, 10 nM or 100 nM for 90 minutes. Neutrophil migration was subsequently recorded by lapse video microscopy and positively identified migrating neutrophils such as by subsequent acridine orange staining (Figure 14D). Under these conditions, the trans-endothelial migration of neutrophil peak from blood to peak concentrations of chemokine of 10 nM. Concentrations of 1 nM lead to minimal neutrophil adhesion to the luminal surface of the small vein and transmigration and concentrations of 100 nM leads to the accumulation of neutrophils around the vessel without transmigration towards the peak concentrations of CINC-1 (data not shown). In the second set of experiments the application of a chemokine gradient with a peak concentration of 10 nM (Figures 14E and Video 6) or 100 nM for 45 minutes was sequentially replaced by a gradient with a peak concentration of 100 nM (Figure 14F and Video 7) or 10 nM to replace a potentially chemotactic gradient with a fugactic gradient. Cell migration is scanned as previously described using the MetaMorph software (Figure 141). Cells were observed undergoing chemotaxis out of the mesenteric small vein towards peak concentrations of chemokine of 10 nM in adjacent tissues as previously described (Figure 12H). However, in contrast, when a gradient with a peak concentration at the superfusion point of 100 nM replaced the previous lower concentration of chemokine, the neutrophils were observed to migrate back to the mesenteric small vein (Figure 141). Directional movement above or below a gradient was quantified as previously described for cells that migrate in defined gradients in vitro. Cell velocities and random motility coefficients of neutrophils migrating under these in vivo gradient conditions toward or away from the chemokine peak concentrations of 10 nM and 100 nM varied between 7.70 and 7.87 μm per minute and 64.57 to 135.11 μm2 / minute (Figure 16, Table 9). These velocities and the random motility coefficients were not significantly different from those seen for cell migration in similar steep gradients and the absolute chemokine concentration in vitro and ranged between 2.0 and 5.1 microns / minutes and between 504.11 and 831.33 μm2 / minutes . Interestingly, the persistent times for cells that migrate in vivo were significantly lower in vivo (2.31 to 5.25 minutes) than those seen in vitro (11.1 to 14.6 minutes) in gradients with peak concentrations of 10 nM and 100 nM and 12 nM and 120 nM, respectively, and may reflect the complexity of the surface on which cells migrate in vivo when compared to in vitro fixation. The finally quantitative measurement of bi-directional inclination of cells in gradients in vivo, including the average chemotropic index, indicated that the cells migrate predominantly towards a diffusive gradient of CINC-1 with a peak concentration of 10 nM with MCI of + -0.32 + / - 0.06 while the cells move away when this gradient was replaced with a gradient with a peak concentration of 100 nM of CINC-1 with an MCI of -0.35 +/- 0.12.
Conclusions The in vitro and in vivo analyzes presented above rigorously demonstrated the ability of neutrophils to move up or down a chemokine gradient. In contrast to the current paradigm, which argues that neutrophils allow only tissues to enter as a result of positive chemotactic agents, these findings indicate the existence of neutrophil chemorepellant that actively exclude neutrophils from healthy uninfected tissues. Finally, these findings increase the possibility for the design of a new class of anti-inflammatory agents that actively repel neutrophils from specific anatomical sites.
Equivalents The above written specification is considered sufficient to enable one skilled in the art to practice the invention. The present invention is not limited in scope by the examples provided, since the examples are intended as a simple illustration of one aspect of the invention and other functionally equivalent embodiments are within the scope of the invention. Various modifications of the invention in addition to those shown and described herein will be apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. The advantages and objects of the invention are not necessarily encompassed by each embodiment of the invention.

Claims (121)

  1. CLAIMS 1. A method for identifying a nucleic acid expressed in a concentration-dependent manner, characterized in that it comprises: determining a first expression profile of nucleic acid in a first cell in a first position in a gradient of agent concentration, determining a second nucleic acid expression profile of a second cell in a second position in the concentration gradient of the agent, determining a difference between the first and second nucleic acid expression profiles, wherein the first position in the concentration gradient of the agent corresponds at a first concentration of the agent, and the second position in the concentration gradient of the agent corresponds to a second concentration of the agent, and at least the second cell has migrated through the concentration gradient of the agent.
  2. 2. The method according to claim 1, characterized in that the expression profile of the nucleic acid is an expression profile of mRNA.
  3. 3. The method according to claim 1, characterized in that the concentration gradient of the agent is a concentration gradient of the ligand.
  4. 4. The method of claim 1, characterized in that the concentration gradient of the agent is a chemokine concentration gradient.
  5. 5. The method according to claim 4, characterized in that the chemokine concentration gradient is selected from the group consisting of SDF-1a, SDF-1β, 1P-10, MIG, GROa, GROß, GRO ?, IL -8, PF4, MCP, MIP-1a, MIP-1β, MIP-1? (mouse), MCP-2, MCP-3, MCP-4, MCP-5 (mouse), RANTES, fractalkine, lymphotactin, CXC, IL-8, GPC-2, ENA-78, NAP-2, IP-10 , MIG, l-TAC, SDF-1a, BCA-1, PF4, Bolequina, HCC-1, Leucotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin-3 (TSC) ), MDC, TARC, SLC (Exodus-2, 6CKina), MIP-3a (LARC, Exodus-1), ELC (MIP-3ß), I-309 DC-CK1 (PARC, AMA-1), TECK, CTAK , MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), C-10 (mouse), lymphotactin C, and CX3C Fractelquine (Neurotactin) and ITAC concentration gradients.
  6. 6. The method according to claim 3, characterized in that the concentration gradient of the agent is a cytosine concentration gradient.
  7. The method according to claim 6, characterized in that the cytosine concentration gradient is selected from the group consisting of PAF, N-formylated peptides, C5a, LTB4 and LXA4, CXC, I L-8, GCP-2, GRO, GROa, GROß, GRO ?, ENA-78, NAP-2, IP-10, MIG, l-TAC, SDF-1a, BCA-1, PF4, Bolequina, MIP-1a, MIP-1β, RANTE
  8. S, HCC -1, MCP-1, MCP-2, MCP-3, MCP-4, MCP-5 (mouse), Leukotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin- 3 (TSC), MDC, TARC, SLC (Exodus-2, 6CKina), MIP-3a (LARC, Exodus-1), ELC (MIP-3ß), 1-309 DC-CK1 (PARC, AMAC-1), TECK, CTAK, MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), MIP-1? (mouse), C-10 (mouse), lymphotactin C, and CX3C Fractelquine (Neurotactin), SDF-1a, SDF-1β, IL-1, IL-2, IL-3, IL-4, IL-5, IL -6, IL-7, IL-10, IL-12, IL-15, IL-18, TNF, IFN-a, IFN-β, lFN-, factor that stimulates the granulocyte-macrophage colony (GM-CSF ), a factor that stimulates the granulocyte colony (G-CSF), a factor that stimulates the macrophage colony (M-CSF), TF-β, FLT-3 ligand, VEGF, DMDA, endothelin and CD40 ligand concentration gradients. The method according to claim 1, characterized in that the first concentration of the agent is a zero concentration of the agent, and the second concentration of the agent is a non-zero concentration of the agent.
  9. 9. The method according to claim 1, characterized in that the first concentration of the agent is greater than the second concentration of the agent.
  10. The method according to claim 1, characterized in that the first cell has migrated through the concentration gradient of the agent.
  11. 11. The method according to claim 1 or 10, characterized in that the migration through the concentration gradient of the agent is fugactactic migration.
  12. 12. The method according to claim 1 or 10, characterized in that the migration through the concentration gradient is chemotactic migration.
  13. The method according to claim 1, characterized in that the expression profile of nucleic acid is determined by Northern analysis.
  14. The method according to claim 1, characterized in that the expression profile of nucleic acid is determined by the polymerase chain reaction (PCR) analysis.
  15. The method according to claim 1, characterized in that the expression profile of nucleic acid is determined by microchip analysis of the nucleic acid.
  16. 16. The method according to claim 1, characterized in that the gradient is a step gradient.
  17. 17. The method according to claim 1, characterized in that the gradient is a continuous gradient.
  18. 18. The method according to claim 1, characterized in that the gradient comprises a second co-existing gradient with the first gradient.
  19. 19. The method according to claim 1, characterized in that the first and second cells are adult cells.
  20. 20. The method according to claim 1, characterized in that the first and second cells are human cells.
  21. 21. The method according to claim 1, characterized in that the first and second cells are primary cells.
  22. 22. The method according to claim 1, characterized in that the first and second cells are hematopoietic cells.
  23. 23. The method according to claim 1, characterized in that the first and second cells are T lymphocytes.
  24. The method according to claim 1, characterized in that the first and second cells are neural cells.
  25. 25. A method for identifying a compound that can modulate cell migration in one or more concentration gradients of the agent, characterized in that it comprises: contacting a migrating cell in a concentration gradient of the agent with a test compound; determining the expression profile of nucleic acid in the cell; and identify a change in the expression of a gene expression product.
  26. 26. The method according to claim 25, characterized in that the cell migration is a migration of chemotaxis and the gene expression product is a product of specific chemotactic gene expression.
  27. 27. The method according to claim 25, characterized in that the cell migration is a fugetáxica migration and the gene expression product is a product of specific chemotactic gene expression.
  28. 28. The method according to claim 25, characterized in that the expression profile of nucleic acid is an expression profile of mRNA.
  29. 29. The method according to claim 25, characterized in that the concentration gradient of the agent is a gradient of ligand concentration.
  30. 30. The method according to claim 25, characterized in that the concentration gradient of the agent is a chemokine concentration gradient.
  31. 31. The method according to claim 30, characterized in that the chemokine concentration gradient is selected from the group consisting of SDF-1a, SDF-1β, IP-10, MIG, GROa, GROß, GRO ?, IL-8. , PF4, MCP, MIP-1a, MIP-1β, MIP-1? (mouse), MCP-2, MCP-3, MCP-4, MCP-5 (mouse), RANTES, fractalkine, lymphotactin, CXC, IL-8, GPC-2, ENA-78, NAP-2, IP-10 , MIG, l-TAC, SDF-1a, BCA-1, PF4, Bolequina, HCC-1, Leucotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin-3 (TSC) ), MDC, TARC, SLC (Exodus-2, 6CKina), MIP-3a (LARC, Exodus-1), ELC (MIP-3ß), I-309 DC-CK1 (PARC, AMA-1), TECK, CTAK , MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), C-10 (mouse), lymphotactin C, and CX3C Fractelquine (Neurotactin) and ITAC concentration gradients.
  32. 32. The method according to claim 25, characterized in that the concentration gradient is a cytosine concentration gradient.
  33. 33. The method according to claim 32, characterized in that the cytosine concentration gradient is selected from the group consisting of PAF, N-formylated peptides, C5a, LTB4 and LXA4, CXC, IL-8, GCP-2. , GRO, GROa, GROß, GRO ?, ENA-78, NAP-2, IP-10, MIG, l-TAC, SDF-1a, BCA-1, PF4, Bolequina, MIP-1a, MIP-1β, RANTES, HCC-1, MCP-1, MCP-2, MCP-3, MCP-4, MCP-5 (mouse), Leukotactin-1 (HCC-2, MIP-5), Eotaxin, Eotaxin-2 (MPIF2), Eotaxin -3 (TSC), MDC, TARC, SLC (Exodus-2, 6CKina), MIP-3a (LARC, Exodus-1), ELC (MIP-3ß), I-309 DC-CK1 (PARC, AMAC-1) , TECK, CTAK, MPIF1 (MIP-3), MIP-5 (HCC-2), HCC-4 (NCC-4), MIP-1? (mouse), C-10 (mouse), lymphotactin C, and CX3C Fractelquine (Neurotactin), SDF-1a, SDF-1β, met-SDF-1β, IL-1, IL-2, IL-3, IL-4 , IL-5, IL-6, IL-7, IL-10, IL-12, IL-15, IL-18, TNF, IFN-a, IFN-β, IFN-α, factor that stimulates the granulocyte colony -Macrophage (GM-CSF), a factor that stimulates the granulocyte colony (G-CSF), a factor that stimulates the macrophage colony (M-CSF), TF-β, ligand FLT-3, VEGF, DMDA, endothelin and gradients of concentration of CD40 ligand.
  34. 34. The method according to claim 25, characterized in that the expression profile of the nucleic acid is determined in two different concentrations of the agent.
  35. 35. The method according to claim 34, characterized in that the two different concentrations of the agent are a zero concentration of the agent and a non-zero concentration of the agent.
  36. 36. The method according to claim 35, characterized in that the cell in a zero concentration of the gradient has migrated through a gradient.
  37. 37. The method according to claim 25, characterized in that the expression profile of nucleic acid is determined by Northern analysis.
  38. 38. The method according to claim 25, characterized in that the expression profile of the nucleic acid is determined by polymerase chain reaction (PCR) analysis.
  39. 39. The method according to claim 25, characterized in that the expression profile of nucleic acid is determined by nucleic acid microchip analysis.
  40. 40. The method according to claim 25, characterized in that the gradient is a step gradient.
  41. 41. The method according to claim 25, characterized in that the gradient is a continuous gradient.
  42. 42. The method according to claim 25, characterized in that the gradient comprises a second gradient co-existing with the first gradient.
  43. 43. The method according to claim 25, characterized in that the cell is an adult cell.
  44. 44. The method according to claim 25, characterized in that the cell is a human cell.
  45. 45. The method according to claim 25, characterized in that the cell is a primary cell.
  46. 46. The method according to claim 25, characterized in that the cell is a hematopoietic cell.
  47. 47. The method according to claim 25, characterized in that the cell is a T lymphocyte.
  48. The method according to claim 25, characterized in that the cell is a neural cell.
  49. 49. A method for inhibiting fugetaxis, characterized in that it comprises contacting a cell experiencing or probably experiencing fugetaxis with an agent that inhibits a specific gene expression product of fugetaxis in an amount effective to inhibit fugetaxis.
  50. 50. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is a nucleic acid or a peptide.
  51. 51. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is a signaling molecule.
  52. 52. The method according to claim 49, characterized in that the signaling molecule is selected from the group consisting of the cell division cycle 42, annexin A3, guanine nucleotide exchange factor Rap1, adenylate cyclase 1, JAK binding protein and dissociation alpha inhibitor Rho GDP.
  53. 53. The method according to claim 52, characterized in that the signaling molecule is selected from the group consisting of the cell division cycle 42 (cdc42), ribosomal protein S6 kinase, protein 2 associated with BAI1, GTPase regulator associated with FAK, C-beta 1 protein kinase, phosphoinositide specific phospholipase C-beta, nitric oxide synthase 1, phosphatidylinositol-4-phosphate 5-kinase and MAP kinase kinase kinase 4.
  54. 54. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is a molecule related to the extracellular matrix.
  55. 55. The method according to claim 54, characterized in that the molecule related to the extracellular matrix is selected from the group consisting of chitinase 3-similar to 1 (cartilage glycoprotein-39), cell adhesion molecule 6 related to carcinoembryonic antigen. , matrix metalloproteinase 8 (neutrophil collagenase), protein 1 associated with the cytoplasmic domain of integrin ficoiin (domain containing collagen fibrinogen) 1, antigen 1 similar to epithelial V, vascular endothelial growth factor (VEGF), fibulin 1, molecule 3 of cell adhesion related to caricinoembryonic antigen and lysosomal associated membrane protein 1.
  56. 56. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is a cytoskeleton-related molecule.
  57. 57. The method according to claim 56, characterized in that the cytoskeletal-related molecule is selected from the group consisting of ankyrin 1 (erythrocytic), calcium binding protein S12 (calgranulin C), plectin 1 (protein binding intermediate filament, 500kD), RPEB3 protein associated with microtubule, protein similar to microtubule-associated protein 1A (MILP), sealed protein (actin filament, similar to gelsolin), and ankyrin 2 (neuronal).
  58. 58. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is a cell cycle molecule.
  59. 59. The method according to claim 58, characterized in that the cell cycle molecule is selected from the group consisting of feline sarcoma viral oncogene v-kit Ardí-Zuckerman 4 homolog, lipocalin 2 (24p3 oncongene), lectin (galectin) 3 of galactoside binding), RAB31 (RAS member oncogene family), neutralized homolog 2 (Drosophila) (mitogenic response phosphoprotein), RABD9 (family of the RAS member oncogene, psudógeno 1), and factor 8 of differentiation of increase.
  60. 60. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is a molecule related to the immune response.
  61. 61. The method according to claim 61, characterized in that the molecule related to the immune response is selected from the group consisting of the major histocompatibility complex (class II, DR alpha), calcium binding protein A8 S100 (calgranulin A) , small inducible cytosine subfamily A (Cys-Cys), eukaryotic translation initiation factor 5A, small inducible cytosine subfamily B (Cys-X-Cys) (member 6, granulocyte chemoattractant protein 2), Fc fragment of the protein of IgG binding, CD24 antigen (antigen from clustering 4 of small cell lung carcinoma), MHC class II transactivator, T cell receptor (alpha chain), T cell activation (increased final expression), protein tyrosine phosphatase similar to MKP-1, 2 gamma constant of the T cell receptor, gamma site of the T cell receptor, cytochrome P450 (subfamily IVF, polypeptide 3, omega hydroxylase of leukotriene B4).
  62. 62. The method according to claim 49, characterized in that the cell is an immune cell.
  63. 63. The method according to claim 49, characterized in that the contact occurs in vivo in a subject who has or is at risk of having an abnormal immune response.
  64. 64. The method according to claim 49, characterized in that, the cell is a neural cell.
  65. 65. The method according to claim 49, characterized in that the specific gene expression product of fugetaxis is chemokine receptor 1 (C-X3-C).
  66. 66. A method for inhibiting cellular chemotaxis, characterized in that it comprises: contacting a cell undergoing or likely undergoing chemotaxis with an agent that inhibits a specific gene expression product of chemotaxis in an amount effective to inhibit chemotaxis.
  67. 67. The method according to claim 66, characterized in that the gene expression product specific for chemotaxis is a nucleic acid or peptide.
  68. 68. The method according to claim 66, characterized in that the cell is an immune cell.
  69. 69. The method according to claim 66, characterized in that the contact occurs in vivo in a subject who has or is at risk of having an abnormal immune response.
  70. 70. The method according to claim 66, characterized in that the abnormal immune response is an inflammatory response.
  71. 71. The method according to claim 66, characterized in that the abnormal immune response is an autoimmune response.
  72. 72. The method according to claim 71, characterized in that the autoimmune response is selected from the group consisting of rheumatoid arthritis, Crohn's disease, multiple sclerosis, systemic lupus erythematosus (SLE), autoimmune encephalomyelitis, myasthenia gravis (MG), thyroiditis of Hashimoto, Goodspasture syndrome, fenfigus (eg, fenfigus vulgaris), Grave's disease, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, scleroderma with anti-collagen antibodies, mixed connective tissue disease, polymyositis, pernicious anemia, Addison's idiopathic disease , associated autoimmune infertility, glomerulonephritis (eg, growing glomerulonephritis, proliferative glomerulonephritis), bolus pemphigoid, Sjögren's syndrome, insulin resistance, and autoimmune diabetes mellitus.
  73. 73. The method according to claim 66, characterized in that the abnormal tissue response is a host versus graft response.
  74. 74. The method according to claim 66, characterized in that the specific gene expression product of chemotaxis is a signaling molecule.
  75. 75. The method according to claim 74, characterized in that the signaling molecule is selected from the group consisting of the kinase 6 of the G-protein coupled receptor., kinase 1 related to vaccine, tyrosine kinase 2 of PTK2 protein, SH3 and ITAM domains containing the STAM-like protein, gene 1 associated with signal-induced proliferation, CD47 antigen (Rh-related antigen, signal transducer associated with integrin) and tyrosine phosphatase protein (type 12 without receptor).
  76. 76. The method according to claim 74, characterized in that the signaling molecule is selected from the group consisting of PTK2 (focal adhesion of the kinase), MAP kinase kinase kinase kinase 2, guanine nucleotide binding protein, PT phosphatase (receptor), beta kinase of cdc42 binding protein, Ral GEF (RalGPSIA), MAP kinase 7, autotaxin, inositol, 1, 4,5-triphosphate receptor, phosphoinositide-3-kinase gamma, PT phosphatase (without receptor) , RAS p21 protein activator (GAP), RAS p21 protein activator (GAP), guanyl RAS release protein 2 and 20kDa subunit of the Arp23 complex.
  77. 77. The method according to claim 66, characterized in that the specific gene expression product of chemotaxis is a molecule related to the extracellular matrix.
  78. 78. The method according to claim 77, characterized in that the molecule related to the extracellular matrix is selected from the group consisting of spondine 1 (f-spondine, extracellular matrix protein), collagen of type XVlll (alpha 1), molecule of CD31 adhesion, tetraspan 3, glycoprotein A33, and angi-associated migratory cell protein.
  79. 79. The method according to claim 66, characterized in that the specific gene expression product of chemotaxis is a molecule related to the cytoskeleton.
  80. 80. The method according to claim 79, characterized in that the cytoskeleton-related molecule is selected from the group consisting of the protein complex 23 related to actin (subunit 4, 20 kD), tropomyosin 2 (beta), actin-dependent regulator associated with chromatin SWISNF related matrix (subfamily a, member 5), beta spectrin (non-erythrocytic 1), light myosin polypeptide (regulator), keratin 1, placofilin 4, and sealing protein (actin filament, Z line of muscle, alpha 2).
  81. 81. The method according to claim 66, characterized in that the specific gene expression product of chemotaxis is a cell cycle molecule.
  82. 82. The method according to claim 81, characterized in that the cell cycle molecule is selected from the group consisting of protein 1 that activates the FGF receptor, fibrosarcoma musculoaponeurotic oncogene v-maf (avian) homolog, cyclin-dependent kinase (as CDC2) 10, inducible early growth response of TGFB, retinoic acid alpha receptor, subunit 10 of anaphase-promoting complex, RAS p21 protein activator (GTPase-activating protein, 3-lns-1 binding protein, 3,4,5, -P4), cell division cycle 27, programmed cell death 2, c-myc binding protein, protein kinase kinase kinase 1 activated with mitogen, receptor III TGF beta (betaglycan, 300 kDa) and Gl to transition 1 of phase S.
  83. 83. The method according to claim 66, characterized in that the specific gene expression product of chemotaxis is a molecule related to the immune response.
  84. 84. The method according to claim 83, characterized in that the molecule related to the immune response is selected from the group consisting of class II DQ beta 1 of highest histocompatibility complex, spinal cord stromal cell antigen 2, receptor 1 of Burkitt lymphoma (GTP binding protein, CXCR5), CD7 antigen (p41), Stat2 a type, T cell immune regulator 1, and interleukin receptor 21.
  85. 85. The method according to claim 66, characterized in that the cell is a neural cell.
  86. 86. A method for promoting cellular fugetaxis, characterized in that it comprises: contacting a cell with an agent without chemokine that promotes fugetaxis in an amount effective to promote fugetaxis.
  87. 87. The method according to claim 86, characterized in that contact occurs in vivo in a subject having a disorder characterized by lack of fugetaxis.
  88. 88. The method according to claim 86, characterized in that the cell is a hematopoietic cell.
  89. 89. The method according to claim 88, characterized in that the hematopoietic cell is a T lymphocyte.
  90. The method according to claim 86, characterized in that the cell is a neural cell.
  91. 91. A method for promoting cellular chemotaxis, characterized in that it comprises contacting a cell with an agent without chemokine that promotes chemotaxis in an amount effective to promote chemotaxis.
  92. 92. The method according to claim 91, characterized in that the contact occurs in vivo in a subject having a disorder characterized by a lack of chemotaxis.
  93. 93. The method according to claim 91, characterized in that the cell is a hematopoietic cell.
  94. 94. The method according to claim 93, characterized in that the hematopoietic cell is a T lymphocyte.
  95. The method according to claim 91, characterized in that the cell is a neural cell.
  96. 96. The method according to claim 62 or 68, characterized in that the immune cell is one selected from the group consisting of T cells, B cells, NK cells, dendritic cells, monocytes and macrophages.
  97. 97. The method according to claim 96, characterized in that the immune cell is an inflammatory cell selected from the group consisting of neutrophils, basophils, eosinophils and mast cells.
  98. 98. The method according to claim 88 and 93, characterized in that the hematopoietic cell is an immune cell, selected from the group consisting of T cells, B cells, NK cells, dendritic cells, monocytes and macrophages.
  99. 99. The method according to claim 98, characterized in that the immune cell is an inflammatory cell selected from the group consisting of neutrophils, basophils, eosinophils and mast cells.
  100. 100. A method for producing neutrophil chemotaxis, characterized in that it comprises contacting a neutrophil with I L-8 in an effective amount to promote chemotaxis.
  101. 101. The method according to claim 100, characterized in that the neutrophil is in contact with a low concentration of IL-8.
  102. 102. The method according to claim 101, characterized in that the low concentration of I L-8 is between about 10 ng / ml to about 500 ng / ml.
  103. 103. The method of compliance with the claim 100, characterized in that contact occurs in vivo in a subject having a disorder characterized by a lack of neutrophil chemotaxis.
  104. 104. The method according to claim 103, characterized in that the disorder is selected from the group consisting of bacterial infections and granulomatous diseases.
  105. 105. The method according to claim 103, characterized in that i L-8 is provided to the subject on a surface of material coated with IL-8.
  106. 106. The method according to claim 105, characterized in that the surface of the material is implanted within the subject.
  107. 107. The method according to claim 103, characterized in that I L-8 is provided to the subject in a controlled release formulation.
  108. 108. A method for promoting neutrophil fugetaxis, characterized in that it comprises contacting a neutrophil with I L-8 in an amount effective to promote fugetaxis.
  109. 109. The method of compliance with the claim 108, characterized in that the neutrophil is in contact with a high concentration of IL-8.
  110. 110. The method of compliance with the claim 109, characterized in that the concentration of I L-8 is between about 1 microgram / ml to about 10 microgram / ml.
  111. 111. The method according to claim 108, characterized in that contact occurs in vivo in a subject having a disorder characterized by lack of neutrophil fugetaxis.
  112. 112. The method according to claim 111, characterized in that the disorder is selected from the group consisting of inflammatory or immune mediated diseases, rejection of a transplanted organ or tissue, rheumatoid arthritis, autoimmune diseases and asthma.
  113. 113. The method according to claim 108, characterized in that I L-8 is provided to the subject on a material surface coated with IL-8.
  114. 114. The method according to claim 113, characterized in that the surface of the material is implanted within the subject.
  115. 115. The method according to claim 108, characterized in that I L-8 is provided to the subject in a controlled release formulation.
  116. 116. A method for inhibiting I L-8 that induces neutrophil chemotaxis, characterized in that it comprises contacting a neutrophil with wortmanin in an amount effective to inhibit chemotaxis and optionally induce neutrophil fugetaxis.
  117. 117. A method for inhibiting I L-8 induced by neutrophil fugetaxis, characterized in that it comprises contacting a neutrophil with LY294002 in an amount effective to inhibit fugetaxis and optionally induce chemotaxis by the neutrophil.
  118. 118. The method according to claim 4 or 30, characterized in that the chemokine concentration gradient is a concentration gradient of SDF-1.
  119. 119. The method according to claim 4 or 30, characterized in that the chemokine concentration gradient is a concentration gradient of IL-8.
  120. 120. The method according to claim 6 or 32, characterized in that the cytosine concentration gradient is a concentration gradient of SDF-1.
  121. 121. The method according to claim 6 or 32, characterized in that the cytosine concentration gradient is a concentration gradient of IL-8.
MXPA/A/2005/006086A 2002-12-06 2005-06-06 Methods and compositions relating to gradient exposed cells MXPA05006086A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/431,424 2002-12-06
US60/438,848 2003-01-09
US60/445,049 2003-02-05

Publications (1)

Publication Number Publication Date
MXPA05006086A true MXPA05006086A (en) 2006-10-17

Family

ID=

Similar Documents

Publication Publication Date Title
EP1169348B1 (en) Purposeful movement of human migratory cells away from an agent source
Du et al. Experimental study on 1, 25 (OH) 2D3 amelioration of oral lichen planus through regulating NF‐κB signaling pathway
US20070134657A1 (en) Methods and compositions relating to gradient exposed cells
US8133857B2 (en) NOTCH inhibition in the treatment of atherosclerosis
Sakai et al. 1-Alpha, 25-dihydroxy vitamin D 3 inhibits osteoclastogenesis through IFN-beta-dependent NFATc1 suppression
WO2002085285A2 (en) Methods and reagents for regulating bone and cartilage formation
Carinci et al. Calcium sulfate: Analysis of MG63 osteoblast‐like cell response by means of a microarray technology
Somebang et al. CCR2 deficiency alters activation of microglia subsets in traumatic brain injury
JP2010521135A (en) Cell chemical repellent
De Rossi et al. Cementocytes express receptor activator of the nuclear factor kappa-B ligand in response to endodontic infection in mice
Oldani et al. In vitro and bioinformatics mechanistic-based approach for cadmium carcinogenicity understanding
Sollazzo et al. Genetic effect of anatase on osteoblast‐like cells
Sollazzo et al. Genetic effect of zirconium oxide coating on osteoblast‐like cells
Wang et al. Enhanced osteogenic differentiation of rat bone marrow mesenchymal stem cells on titanium substrates by inhibiting Notch3
Martinez et al. Gene profiles during root canal treatment in experimental rat periapical lesions
MXPA05006086A (en) Methods and compositions relating to gradient exposed cells
Cardemil et al. Jaw Bone Samples From Bisphosphonate‐Treated Patients: A Pilot Cohort Study
KR20120013693A (en) Markers for diagnosing angiogenesis-related diseases and use thereof
Carinci et al. P‐15 cell‐binding domain derived from collagen: Analysis of MG63 osteoblastic‐cell response by means of a microarray technology
Barkana et al. Gene profile in periodontal ligament cells and clones with enamel matrix proteins derivative
Hadziabdic et al. Quantitative analysis of CCL5 and ep300 in periapical inflammatory lesions.
Zheng et al. Integrated analysis of lncRNA–mRNA networks associated with an SLA titanium surface reveals the potential role of HIF1A-AS1 in bone remodeling
Kersey et al. Inorganic ions activate lineage-specific gene regulatory networks
EP4056205A1 (en) Micro-rna composition functionalized for dental implants
Shirazi et al. Evaluation of nanoscale versus hybrid micro/nano surface topographies for endosseous implants