WO2009039542A2 - Methods of screening for compounds having anti- inflammatory activity and/or prevent / treat vascular leak - Google Patents

Methods of screening for compounds having anti- inflammatory activity and/or prevent / treat vascular leak Download PDF

Info

Publication number
WO2009039542A2
WO2009039542A2 PCT/AT2008/000337 AT2008000337W WO2009039542A2 WO 2009039542 A2 WO2009039542 A2 WO 2009039542A2 AT 2008000337 W AT2008000337 W AT 2008000337W WO 2009039542 A2 WO2009039542 A2 WO 2009039542A2
Authority
WO
WIPO (PCT)
Prior art keywords
screening
compounds
thrombin
proteins
antibodies
Prior art date
Application number
PCT/AT2008/000337
Other languages
French (fr)
Other versions
WO2009039542A3 (en
WO2009039542A9 (en
Inventor
Peter Petzelbauer
Rainer Henning
Sonja Reingruber
Waltraud Pasteiner
Marion GRÖGER
Original Assignee
Fibrex Medical Research & Development Gmbh
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
Priority claimed from US11/860,488 external-priority patent/US20090081192A1/en
Priority claimed from AT19392007A external-priority patent/AT506121A1/en
Application filed by Fibrex Medical Research & Development Gmbh filed Critical Fibrex Medical Research & Development Gmbh
Priority to EP08799926A priority Critical patent/EP2193370A2/en
Priority to JP2010525162A priority patent/JP2010540892A/en
Publication of WO2009039542A2 publication Critical patent/WO2009039542A2/en
Publication of WO2009039542A9 publication Critical patent/WO2009039542A9/en
Publication of WO2009039542A3 publication Critical patent/WO2009039542A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5064Endothelial cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/42Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving phosphatase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/48Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase
    • C12Q1/485Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving transferase involving kinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5082Supracellular entities, e.g. tissue, organisms
    • G01N33/5088Supracellular entities, e.g. tissue, organisms of vertebrates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • G01N33/54313Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals the carrier being characterised by its particulate form
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/582Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with fluorescent label
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/585Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances with a particulate label, e.g. coloured latex
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/06Gastro-intestinal diseases
    • G01N2800/065Bowel diseases, e.g. Crohn, ulcerative colitis, IBS
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/06Gastro-intestinal diseases
    • G01N2800/067Pancreatitis or colitis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/08Hepato-biliairy disorders other than hepatitis
    • G01N2800/085Liver diseases, e.g. portal hypertension, fibrosis, cirrhosis, bilirubin
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/12Pulmonary diseases
    • G01N2800/122Chronic or obstructive airway disorders, e.g. asthma COPD
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/12Pulmonary diseases
    • G01N2800/125Adult respiratory distress syndrome
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/16Ophthalmology
    • G01N2800/164Retinal disorders, e.g. retinopathy
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/20Dermatological disorders
    • G01N2800/202Dermatitis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/20Dermatological disorders
    • G01N2800/205Scaling palpular diseases, e.g. psoriasis, pytiriasis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/22Haematology
    • G01N2800/224Haemostasis or coagulation
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/24Immunology or allergic disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/24Immunology or allergic disorders
    • G01N2800/245Transplantation related diseases, e.g. graft versus host disease
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/26Infectious diseases, e.g. generalised sepsis
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/324Coronary artery diseases, e.g. angina pectoris, myocardial infarction
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/32Cardiovascular disorders
    • G01N2800/328Vasculitis, i.e. inflammation of blood vessels
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/34Genitourinary disorders
    • G01N2800/347Renal failures; Glomerular diseases; Tubulointerstitial diseases, e.g. nephritic syndrome, glomerulonephritis; Renovascular diseases, e.g. renal artery occlusion, nephropathy

Definitions

  • the present invention relates to the field of drug screening. More specifically, the present invention relates to methods for screening, identification and characterization of proteins, peptides, peptidomimetics, antibodies and small molecules, which bind to vascular endothelial (VE)-cadherin and influence certain signalling processes that are mediated by VE-cadherin. It also relates to the use of these compounds to prevent the opening of endothelial adherens junctions between endothelial cells and certain morphological changes of endothelial cells as a consequence of these events. Compounds with these characteristics are useful for the treatment of all diseases, where inflammatory responses, vascular leak and endothelial dysfunction play a role. They can also prevent formation of new blood capillaries and are therefore useful for the treatment of cancer.
  • VE vascular endothelial
  • the endothelial layer which seamlessly covers the inside of all blood vessels, has a very important barrier function, preventing blood constituents such as blood borne substances, cells and serum from entering the underlying tissue.
  • the barrier function is tightly regulated through a number of homo- and heterotopic interactions between molecules on neighbouring endothelial cells as well as similar interaction with molecules on circulating blood cells. The breakdown of this barrier function leads to severe physiological consequences and injury to the underlying tissue.
  • Endothelial dysfunction can be manifested in a number of ways, for example as an imbalance between the release of relaxant and contractile factors, the release of anti- and pro-coagulant mediators, or as a loss in barrier function (Rubanyi, Journal of Cardiovascular Pharmacology 22, Sl- 14.1993; McQuaid & Keenan Experimental Physiology 82, 369-376 (1997)).
  • Such dysfunction has been associated with numerous pathological conditions, including hypercholesterolemia, hypertension, vascular disease associated with diabetes mellitus, atherosclerosis, septic shock and the adult respiratory distress syndrome (Sinclair, Braude, Haslam & Evans, Chest. 106:535-539 (1994); Davies, Fulton & Hagen, Br J Surg. 82:1598- 610 (1995).
  • endothelial barrier function One of the principal abnormalities associated with acute inflammatory disease is the loss of endothelial barrier function. Structural and functional integrity of the endothelium is required for maintenance of barrier function and if either of these is compromised, solutes and excess plasma fluid leak through the monolayer, resulting in tissue oedema and migration of inflammatory cells. Many agents increase monolayer permeability by triggering endothelial cell shape changes such as contraction or retraction, leading to the formation of intercellular gaps (Lum & Malik, Am. J. Physiol. 267: L223-L241 (1994). These agents include e.g thrombin, bradykinin and vascular endothelial growth factor (VEGF).
  • VEGF vascular endothelial growth factor
  • Endothelial cell contraction resembles the regulation of actin-myosin interaction in smooth muscle cells, but occurs over a longer time scale and is more properly described as a contracture.
  • the mechanism of this contraction is thought to involve increases in intracellular Ca2+ concentrations, activation of myosin light chain kinase, phosphorylation of myosin light chain and reorganization of F-actin microfilaments. Retraction is a more passive process, is independent of myosin light chain kinase and involves protein kinase C (PKC)-stimulated phosphorylation of actin-linking proteins critical for maintaining cell-cell and cell-matrix interactions (Lum & Malik, Am. J. Physiol. 267: L223-L241 (1994).
  • PKC protein kinase C
  • Hyperpermeability of the blood vessel wall permits leakage of excess fluids and protein into the interstitial space.
  • This acute inflammatory event is frequently allied with tissue ischemia and acute organ dysfunction.
  • Thrombin formed at sites of activated endothelial cells (EC) initiates this microvessel barrier dysfunction due to the formation of large paracellular holes between adjacent EC (Carbajal et al, Am J Physiol Cell Physiol 279: C195-C204, 2000).
  • This process features changes in EC shape due to myosin light chain phosphorylation (MLCP) that initiates the development of F-actin-dependent cytoskeletal contractile tension ( Garcia et al, J Cell Physiol. 1995; 163:510-522 Lum & Malik, Am J Physiol Heart Circ Physiol. 273(5): H2442 - H2451. (1997).
  • MLCP myosin light chain phosphorylation
  • the signalling mechanism of this contractile process involves the proteolytic cleavage and activation of the thrombin receptor.
  • This receptor is coupled to heterotrimeric G proteins of the Gq family that stimulate phospholipase CB, release D-wr ⁇ -inositol 1,4,5-trisphosphate, mobilizing Ca ions from intracellular stores.
  • the subsequent rise in intracellular Ca ion concentration activates Ca ion-calmodulin-dependent MLC kinase, which phosphorylates serine- 19 and threonine- 18 of MLC (Goeckeler & Wysolmerski, J. Cell Biol. 1995; 130:613- 627.).
  • MLCP initiates myosin Mg ion-ATPase activity, causing the binding of myosin to F-actin and subsequent actomyosin stress fiber formation (Ridley & Hall, Cell, 70, 389-399 (1992).
  • the phosphorylation of MLC converts the soluble folded 1OS form of non-muscle myosin II to the insoluble unfolded 6S form.
  • This process is characterized by reorganization of myosin from a diffuse intracellular cloud to punctate spots and ribbons associated with large bundles of F-actin (Verkhovsky et al, The Journal of Cell Biology, 131, 989-1002 (1995).
  • the final consequence is a persistent shape change of endothelial cells and a disruption of the barrier function.
  • Thrombin-induced endothelial hyperpermeability may also be mediated by changes in cell- cell adhesion (Dejana J. Clin. Invest. 98: 1949-1953 (1996).
  • Endothelial cell-cell adhesion is determined primarily by the function of vascular endothelial (VE) cadherin (cadherin 5), a Ca-dependent cell-cell adhesion molecule that forms adherens junctions.
  • Cadherin 5 function is regulated from the cytoplasmic side through association with the accessory proteins beta-catenin, plakoglobin (g-catenin), and pi 20 that are linked, in turn, to alpha-catenin (homologous to vinculin) and the F-actin cytoskeleton.
  • VE-cadherin has emerged as an adhesion molecule that plays fundamental roles in microvascular permeability and in the morphogenic and proliferative events associated with angiogenesis (Vincent et al, Am J Physiol Cell Physiol, 286(5): C987 - C997 (2004). Like other cadherins, VE-cadherin mediates calcium-dependent, homophilic adhesion and functions as a plasma membrane attachment site for the cytoskeleton. However, VE-cadherin is integrated into signaling pathways and cellular systems uniquely important to the vascular endothelium.
  • VE-cadherin represents a cadherin that is both prototypical of the cadherin family and yet unique in function and physiological relevance.
  • Evidence is accumulating that the VE-cadherin-mediated cell-cell adhesion is controlled by a dynamic balance between phosphorylation and dephosphorylation of the junctional proteins including cadherins and catenins.
  • VE-cadherin monomers in adherens junctions is indispensable for a correct signalling activity of VE-cadherin, since cell bearing a chimeric mutant (IL2-VE) containing a full-length VE-cadherin cytoplasmic tail is unable to cause a correct signalling despite its ability to bind to beta-catenin and pi 20 (Lampugnani et al, MoI. Biol, of the Cell, 13, 1 175-1 189 (2002).
  • Rho GTPases are a family of small GTPases with profound actions on the actin cytoskeleton of cells. With respect to the functioning of the vascular system they are involved in the regulation of cell shape, cell contraction, cell motility and cell adhesion.
  • the three most prominent family members of the Rho GTPases are RhoA, Rac and cdc42. Activation of RhoA induces the formation of f-actin stress fibres in the cell, while Rac and cdc42 affect the actin cytoskeleton by inducing membrane ruffles and microspikes, respectively (Hall, Science,279:509-5 l4 ⁇ 99S).
  • RhoA has a prominent stimulatory effect on actin-myosin interaction by its ability to stabilize the phosphorylated state of MLC (Katoh et al., Am. J. Physiol. Cell. Physiol. 280, C1669-C1679 (2001). This occurs by activation of Rho kinase that in its turn inhibits the phosphatase PPlM that hydrolyses phosphorylated MLC.
  • Rho kinase inhibits the actin-severing action of cofilin and thus stabilizes f-actin fibres (Toshima et al., MoI. Biol, of the Cell. 12, 1131-1 145 (2001). Furthermore, Rho kinase can also be involved in anchoring the actin cytoskeleton to proteins in the plasma membrane and thus may potentially act on the interaction between junctional proteins and the actin cytoskeleton (Fukata et al. . Cell Biol 145:347-361 (1999).
  • RhoA can activate RhoA via G ⁇ l2/13 and a so-called guanine nucleotide exchange factor (GEF) (Seasholtz et al; MoI: Pharmacol. 55, 949-956 (1999).
  • GEF guanine nucleotide exchange factor
  • the GEF exchanges RhoA- bound GDP for GTP, by which RhoA becomes active.
  • RhoA is translocated to the membrane, where it binds by its lipophilic geranyl-geranyl-anchor.
  • RhoA can be activated by a number of vasoactive agents, including lysophosphatidic acid, thrombin and endothelin.
  • the membrane bound RhoA is dissociated from the membrane by the action of a guanine dissociation inhibitor (GDI) or after the action of a GTPase-activating protein (GAP).
  • GDIs guanine dissociation inhibitors
  • GAP GTPase-activating protein
  • RhoA inhibits the activity of RhoA by retarding the dissociation of GDP and detaching active RhoA from the plasma membrane.
  • Thrombin directly activates RhoA in human endothelial cells and induces translocation of RhoA to the plasma membrane.
  • the related GTPase Rac was not activated.
  • Specific inhibition of RhoA by C3 transferase from Clostridium botulinum reduced the thrombin-induced increase in endothelial MLC phosphorylation and permeability, but did not affect the transient histamine-dependent increase in permeability (van Nieuw Amerongen et al. Circ Res. 1998;83: 1 1 15-11231 (1998).
  • the effect of RhoA appears to be mediated via Rho kinase, because the specific Rho kinase inhibitor Y27632 similarly reduced thrombin-induced endothelial permeability.
  • RhoA have antagonistic effects on endothelial barrier function.
  • Acute hypoxia inhibits Racl and activates RhoA in normal adult pulmonary artery endothelial cells (PAECs), which leads to a breakdown of barrier function (Wojciak-Stothard and Ridley, Vascul Pharmacol. ,39: 187-99 (2002).
  • PAECs from piglets with chronic hypoxia induced pulmonary hypertension have a stable abnormal phenotype with a sustained reduction in Racl and an increase in RhoA activitity. These activities correlate with changes in the endothelial cytoskeleton, adherens junctions and permeability.
  • RhoA Activation of Racl as well as inhibition of RhoA restored the abnormal phenotype and permeability to normal (Wojciak-Stothard et al., Am. J. Physiol, Lung Cell MoI. Physiol. 290, Ll 173-Ll 182 (2006).
  • Focal adhesion kinase is composed of a central catalytic domain flanked by large N- and C- terminal domains.
  • the N-terminal region contains the FERM homology that can bind integrins and growth factor receptors.
  • the non-catalytic domain in the C-terminal also referred to as FRNK(FAK-related non-kinase), carries the FATsequence which not only directs FAK to adhesion complexes for signalling, but also provides binding sites for other docking molecules to interact with the cytoplasmic To date, at least five tyrosine residues have been identified in FAK.
  • FAK activation and focal adhesion reorganization actively contribute to the opening of endothelial cell-cell junctions by providing a mechanical basis for endothelial cells to contract or change shape.
  • the focal complex serves as a point of convergence for multiple scaffold proteins or signalling molecules to be integrated, which in turn affect the barrier function.
  • potential signalling events downstream from FAK include the myosin light chain phosphorylation- triggered actin-myosin contraction and Rho-dependent stress fiber formation which are characteristic features of paracellular permeability (Wu, J Physiol 569. , 359-366 (2005). It is therefore desirable to inhibit FAK phosphorylation in order to promote endothelial integrity.
  • Endothelial dysfunction and leakiness of the endothelial barrier is an important component of a range of inflammatory diseases.
  • the inflammatory response is characterized by an extravasation of blood constituents such as plasma proteins and of blood serum leading to severe interstitial tissue edema.
  • neutrophils which are the primary agents of the inflammatory response, are able to emigrate from the blood stream into the underlying tissue.
  • ARDS adult respiratory distress syndrome
  • ALI acute lung injury
  • glomerulonephritis acute and chronic allograft rejection
  • inflammatory skin diseases rheumatoid arthritis
  • asthma atherosclerosis
  • SLE systemic lupus erythematosus
  • connective tissue diseases vasculitis
  • ischemia-reperfusion injury in limb replantation myocardial infarction
  • crush injury shock, stroke and organ transplantation.
  • the present invention is directed to methods for screening, identification, characterization and use of proteins, peptides, peptidomimetics, antibodies and small molecules that modulate interactions and signalling events mediated by agents that cause endothelial hyperpermeability.
  • the agents identified by these screening methods exert their effect by binding to and modulating the conformation and/or phosphorylation status of vascular endothelial (VE)-cadherin expressed in the adherens junctions of endothelial cell layers. More specifically these agents promote endothelial integrity by stabilizing the clustering of VE-cadherin at intercellular junctions. Given the importance of disruption of the endothelial barrier function for a broad range of diseases, these agents have broad applicability as therapeutic and/or prophylactic medicinal products.
  • VE vascular endothelial
  • the present invention provides a method of screening for proteins, peptides, peptidomimetics, antibodies or small organic molecules that increase the activity of Racl by virtue of their binding to the extracellular portion of this protein, the method comprising the steps of:
  • the present invention provides a method of screening for proteins, peptides, peptidomimetics, antibodies or small organic molecules that increase the activity of Racl by virtue of their binding to the extracellular portion of this protein, the method comprising the steps of:
  • the present invention provides a method for screening of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent the activation of RhoA and consequentially the change in the cytoskeletal structure of the endothelial cells, the method comprising the steps of:
  • a contacting a confluent layer of cultured endothelial cells with thrombin in the presence of at least one of the test compounds b. lysing the endothelial cells with a lysation buffer c. measuring the RhoA activity with a specific assay
  • the present invention provides a method for screening of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent the phosphorylation of focal adhesion kinase, the method comprising the steps of:
  • a contacting a confluent layer of cultured endothelial cells with thrombin in the presence of at least one of the test compounds b. lysing the endothelial cells with a lysation buffer c. measuring the phosphorylation of focal adhesion kinase with a specific assay
  • the present invention provides a method for screening of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent vascular leak in a warm-blooded animal undergoing systemic inflammatory response, the method comprising the following steps:
  • a Initiation of a systemic inflammatory response by applying an appropriate dose of bacterial lipopolysaccharide (LPS) b. Exposing the animal to at least one of the test compounds c. injecting the animal with an appropriate amount of fluorescence labelled micro-beads of appropriate size d. sacrificing the animal after an appropriate time period e. excising and homogenizing an organ or tissue of the animal f. measuring the amount of fluorescence in the homogenate
  • LPS bacterial lipopolysaccharide
  • these assays are performed in the sequence described above, which constitutes a screening tree to selectively identify compounds with the specific physiological activities claimed by the current invention.
  • the useful method for measuring the permeability of a confluent layer of endothelial cells is based on a dual chamber method, where endothelial cells are grown on a porous membrane separating the chambers.
  • the cells preferentially human umbilical vein endothelial cells (HUVEC) are incubated with a test compound alone or together with a stimulant of endothelial permeability for 30 to 120 min.
  • a stimulant of endothelial permeability for 30 to 120 min.
  • thrombin is used as such stimulant.
  • the permeability can be measure with a permeating dye, whose absorption can be measured colorimetrically. Evans Blue can be used preferentially as such dye, either as such or bound to albumin.
  • the useful methods of analysing the activation status of Racl and RhoA are based on the principle of the so-called pull down assay.
  • the GTP-bound active state of the respective protein in a cell lysate is bound to an immobilized binding partner and detected with a monoclonal antibody (MAb) specifically directed against the protein in question.
  • MAb monoclonal antibody
  • the amount of the GTP-bound active state can subsequently be quantified through suitable detection methods, including but not limited to Western blotting or luminescence detection.
  • the cells are incubated with the test compound under suitable conditions for various periods of time up to 30 min and lysed afterwards.
  • the lysate is added to a suitably immobilized p21- binding domain
  • PBD p21 -activated protein kinase
  • the cells are incubated under suitable conditions with a suitable amount of thrombin with and without the test compound for various periods of time up to 10 min and lysed afterwards.
  • the lysate is added to a suitably immobilized rhotekin-binding domain (RBD) and the signal measured with an appropriate detection method.
  • RBD rhotekin-binding domain
  • the cells are incubated under suitable conditions with a suitable amount of thrombin with and without the test compound for various periods of time up to 60 min and lysed afterwards, lysates were subjected to SDS-PAGE and western blot analysis with site-specific antibodies directed against FAK phosphorylated tyrosine residues.
  • the animals receive injections of amounts of gram-negative lipopolysaccharide (LPS) suitable to achieve a systemic inflammatory response.
  • LPS gram-negative lipopolysaccharide
  • test substance is injected intravenously, followed by the injection of a suitable amount of fluorescent microbeads. Subsequently the animals are sacrificed, and organs (lung, kidney, spleen, heart, brain) are excised and cut into thin slices suitable for microscopic analysis and fixated with paraformaldehyde. The numbers of extravasated microspheres are counted using a fluorescent microscope.
  • the organs are homogenized and the amount of microbeads trapped in the tissues are measured using a suitable fluorescence detection device.
  • the compounds identified with the screening methods according to the present invention are useful for development of drugs for the prevention and/or treatment of diseases which are caused by an inflammatory reaction and/or endothelial disruption and vascular leak. Therefore, according to another embodiment of the current invention, the compounds of the present invention are administered for treatment and/or prevention of, but not restricted to, septic shock, wound associated sepsis, post-ischemic reperfusion injury, such as after myocardial infarction/reperfusion or organ transplantation), frost-bite injury or shock, acute inflammation mediated lung injury, such as respiratory distress syndrome, acute pancreatitis, liver cirrhosis, uveitis, asthma, traumatic brain injury, nephritis, atopic dermatitis, psoriasis, inflammatory bowel disease, macula degeneration of the eye, diabetic retinopathy, neovascular glaucoma, retinal vein occlusion and tumour progression.
  • septic shock wound associated sepsis
  • the compounds of the present invention may be given orally or parenterally and maybe be formulated into suitable pharmaceutical formulation with pharmaceutically acceptable excipients or carriers.
  • the present invention therefore also relates to a pharmaceutical composition containing an active ingredient identified by the method of screening according to the present invention and further comprising pharmaceutically acceptable excipients or carriers.
  • compositions are those which are approved by a regulatory agency of the Federal or State governments or listed in the U.S. Pharmacopeia or any other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of animal, vegetable and synthetic origin, e.g. peanut oil, soybean oil., mineral oil and the like.
  • Aqueous carriers nay contain for instance also contain dextrose or glycerol.
  • Suitable excipients may include, but are not restricted to, starch, glucose, lactose, sucrose, gelatin, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, glycerol, propylene glycol, ethanol and the like.
  • the composition may also include wetting and/or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsions, tablets, capsules, powders, or slow release formulations.
  • Such compositions will contain a therapeutically effective amount of the compound together with a suitable amount of carrier so as to provide a form for proper administration to the subject to be treated and suitable for the form of treatment.
  • Fig. 1 Compounds identified through the claimed method of screening.
  • Fig. 2 Permeability of a confluent HUVEC layer stimulated with thrombin after incubation for 45 min.
  • Fig. 3 Activation of Racl with compound IA.
  • This gel is the result of a pull-down assay as described in Example 1.
  • Lane 1 medium control
  • lane 2 HUVEC activation with thrombin for 1 min
  • lane 3 treatment with compound IA alone for 1 min
  • lane 4 HUVEC with thrombin and compound IA for 1 min
  • lane 5 thrombin for 5 min
  • lane 6 compound IA for 5 min
  • lane 7 thrombin and compound IA for 5 min.
  • Beta-actin was used to control for total protein content.
  • Fig. 4 Inhibition of thrombin induced activation of RhoA by compound IA. This gel is the result of a pull-down assay as described in Example 1. Lane 1 : medium control, lane 2:
  • HUVEC activation with thrombin for 1 min lane 3: treatment with compound IA alone for 1 min; lane 4: HUVEC with thrombin and compound IA for 1 min, lane 5: thrombin for 5 min; lane 6: compound IA for 5 min, lane 7: thrombin and compound IA for 5 min.
  • Beta-actin was used to control for total protein content.
  • Fig. 5 Activation of Racl with compound IB.
  • This gel is the result of a pull-down assay as described in Example 1.
  • Lane 1 medium control
  • lane 2 HUVEC activation with thrombin for 1 min
  • lane 3 treatment with compound IB alone for 1 min
  • lane 4 HUVEC with thrombin and compound IB for 1 min
  • lane 5 thrombin for 5 min
  • lane 6 compound IB for 5 min
  • lane 7 thrombin and compound IB for 5 min.
  • Fig. 6 Inhibition of thrombin induced activation of RhoA by compound IB at time points 1, 5 and 10 min after stimulation with thrombin.
  • Fig. 9 Inhibition of thrombin induced phosphorylation of FAK by compound IA. This graphs shows the time-course of inhibition of phosphorylation of FAK induced by thrombin.
  • Fig. 10 Inhibition of LPS induced vascular leak by compound IA.
  • These are fluorescent images of lung slices from rats, in which vascular leak was induced by LPS treatment.
  • Slice a) is from a control animal
  • slice b) is from an animal treated with compound IA
  • HUVECs are grown to confluence on 12mm transwell plates, polycarbonate membrane 3 ⁇ m pore size (Corning), under standard conditions. 2h before the start of the experiment the standard growth medium is exchanged in the upper and lower chamber by growth medium w/o phenol red. To induce permeability 5U/ml thrombin (Calbiochem) or 5U/ml plus the test compound are added to the upper well. HUVEC are incubated for 45 min. At the end of incubation time the medium of the upper chamber is replaced by growth medium w/o phenolred containing 4% BSA and 0,67mg/ml Evans Blue. The diffusion of Evans Blue into the lower chamber is allowed for 10 min, afterwards the upper chamber is removed and the optical density of the medium recovered from the lower chamber measured at wavelength of 620nm.
  • HUVECs are grown to confluence under standard conditions. Before induction of Racl activity HUVECs were starved for 4h by using IMDM (Gibco) without growth factor and serum supplements. Racl activity is induced by adding 50 ⁇ g/ml of test compound into starvation medium for 1, 5 and 10 min. Active Racl was isolated using Racl / Cdc42 Assay Reagent from Upstate according to manufactures instructions. Isolates were separated on a 15% polyacrylamid gel and blotted on Nitrocellulose-Membranes (Bio-Rad). Racl was dedected by using Anti-Racl clone23A8, anti-mouse from Upstate (1 :250).
  • Example 3 A compound screening method for identification of substances inhibiting thrombin induced RhoA activation
  • HUVEC are grown to confluence under standard conditions. Before induction of Rho activity HUVEC were starved for 4h by using IMDM (Gibco) without growth factor and serum supplements. After the starvation period 5 U/ml Thrombin (Calbiochem) or 5U thrombin plus 50 ⁇ g/ml of test compound are added to the starvation medium for 1, 5 and 10 min. Active RhoA was isolated using Rho Assay Reagent from Upstate according to manufactures instructions. Isolates were separated on a 15% polyacrylamid gel and blotted on Nitrocellulose-Membrane (Bio-Rad). RhoA was dedected by using Anti-Rho (-A, -B, -C), clone55 from Upstate (1 :500).
  • HUVEC HUVEC were incubated with FX06 (50 ⁇ g/ml), Thrombin (lU/ml, Sigma Aldrich) and Thrombin/ test compound for indicated time points. After washing with ice cold PBS (GIBCO), cells were scrapped in Tris-lysis buffer (plus 1% Triton X (Bio-Rad), NP40 (Sigma Aldrich) and proteinase and phosphatase inhibitory cocktails (Sigma Aldrich)) from culture flasks and lysed for 20 min on ice. Lysates were heavily vortexed every 5 min.
  • lysis lysates were centrifuged (15.000rpm/10min/4°C) and supernatants were added to 50 ⁇ l sepahrose beads (Sigma Aldrich) preincubated with 1 ⁇ g total FAK antibody (BD Transduction Laboratories). Beads were agitated on the wheel for 2h at 4°C, followed by 3 times washing with ice cold PBS, the addition of 2x sample buffer and incubation at 95°C for 5 min. The sample buffer was then removed from the beads and applied to western blotting.
  • HRP-labeled goat anti-mouse Ab (1 :25 000; Bio-Rad) in TBST was used and bound Abs were visualized by chemi luminescence (ECL-system, Amersham Corp., Arlington Heights, IL) and recorded on film.
  • Rats Male Him OFA/SPF rats (Institute for Biomedical Research, Medical School Vienna) with a body weight of 260-32Og are housed at the Institute for Biomedical Research, Medical School Vienna. All experiments were approved by Amt der Wiener Austin, MA58. Rats are anaesthetised with 100 mg/ kg sodium thiopentone (Sandoz). The trachea is cannulated to facilitate respiration. The right jugular vein is cannulated for the administration of drugs. To measure the Mean Arterial Blood Pressure (MAP) a catheter is placed into the right carotid artery. After surgery the animals are randomized in treatment groups. All rats receive a fluid replacement (600 ⁇ l 0,9% saline as an i.v.
  • MAP Mean Arterial Blood Pressure
  • the endotoxic shock is induced by a bolus injection of 12 mg/kg LPS (E.coli serotype 0.127:B8; Sigma). 60min after LPS administration the animals receive a bolus injections of 3 mg/kg of test compound or saline. 5h 50 min after the LPS administration the rats receive an bolus injection of fluorescenct microspheres; 125 x 10 beads /kg body weight (Fluo Spheres Polystyrene Microspheres; 1 ⁇ m yellow-green fluorescent (505/515) Invitrogen Molecular Probes).
  • vascular leakage of the lung is assessed by measurement of the fluorescence per g of tissue.
  • the lung tissue was digested with ethanolic KOH and the fluorescent microspheres are recovered by sedimetation as recommended by the "Manual for using Fluorescent Microspheres to measure organ perfusion" Fluorescent Microsphere Resource Center; University of Washington. Fluorescence is measured using a Spectra Max Gemini S Fluorometer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Analytical Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Toxicology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Investigating Or Analysing Materials By The Use Of Chemical Reactions (AREA)

Abstract

Methods of screening for modulators of Racl, RhoA or focal adhesion kinase (FAK) in endothelial cells are disclosed. The effect is determined by- measuring endothelial permeability, Racl or RhoA activity and phosphorylation of FAK. Vascular permeability may be stimulated by thrombin. Method of screening for compounds that prevent vascular leak in a warm-blooded animal undergoing systematic inflammatory response triggered by LPS, using fluorescence labelled micro-beads. A method of screening comprising a combination of all said methods is also disclosed. Compounds and pharmaceutical preparations being identified by said methods.

Description

METHODS OF SCREENING FOR COMPOUNDS HAVING ANTIINFLAMMATORY ACTIVITY
FIELD OF THE INVENTION
The present invention relates to the field of drug screening. More specifically, the present invention relates to methods for screening, identification and characterization of proteins, peptides, peptidomimetics, antibodies and small molecules, which bind to vascular endothelial (VE)-cadherin and influence certain signalling processes that are mediated by VE-cadherin. It also relates to the use of these compounds to prevent the opening of endothelial adherens junctions between endothelial cells and certain morphological changes of endothelial cells as a consequence of these events. Compounds with these characteristics are useful for the treatment of all diseases, where inflammatory responses, vascular leak and endothelial dysfunction play a role. They can also prevent formation of new blood capillaries and are therefore useful for the treatment of cancer.
BACKGROUND OF THE INVENTION
The endothelial layer which seamlessly covers the inside of all blood vessels, has a very important barrier function, preventing blood constituents such as blood borne substances, cells and serum from entering the underlying tissue. The barrier function is tightly regulated through a number of homo- and heterotopic interactions between molecules on neighbouring endothelial cells as well as similar interaction with molecules on circulating blood cells. The breakdown of this barrier function leads to severe physiological consequences and injury to the underlying tissue. It is involved in the pathogenesis of inflammatory diseases, edema formation as well as angiogenesis, for instance, but not limited to ischemia reperfusion injury caused by for instance myocardial infarction or organ transplatation, systemic inflammatory response syndrome (SIRS) as a sequel of trauma/resuscitation and septicaemia, macula degeneration in the eye and in cancer progression. It is therefore desirable to identify compounds which are able to maintain the integrity of the endothelial adherens junction. These compounds can be used to treat or prevent these disease processes. ENDOTHELIAL BARRIER DYSFUNCTION
Endothelial dysfunction can be manifested in a number of ways, for example as an imbalance between the release of relaxant and contractile factors, the release of anti- and pro-coagulant mediators, or as a loss in barrier function (Rubanyi, Journal of Cardiovascular Pharmacology 22, Sl- 14.1993; McQuaid & Keenan Experimental Physiology 82, 369-376 (1997)). Such dysfunction has been associated with numerous pathological conditions, including hypercholesterolemia, hypertension, vascular disease associated with diabetes mellitus, atherosclerosis, septic shock and the adult respiratory distress syndrome (Sinclair, Braude, Haslam & Evans, Chest. 106:535-539 (1994); Davies, Fulton & Hagen, Br J Surg. 82:1598- 610 (1995).
One of the principal abnormalities associated with acute inflammatory disease is the loss of endothelial barrier function. Structural and functional integrity of the endothelium is required for maintenance of barrier function and if either of these is compromised, solutes and excess plasma fluid leak through the monolayer, resulting in tissue oedema and migration of inflammatory cells. Many agents increase monolayer permeability by triggering endothelial cell shape changes such as contraction or retraction, leading to the formation of intercellular gaps (Lum & Malik, Am. J. Physiol. 267: L223-L241 (1994). These agents include e.g thrombin, bradykinin and vascular endothelial growth factor (VEGF). Endothelial cell contraction resembles the regulation of actin-myosin interaction in smooth muscle cells, but occurs over a longer time scale and is more properly described as a contracture. The mechanism of this contraction is thought to involve increases in intracellular Ca2+ concentrations, activation of myosin light chain kinase, phosphorylation of myosin light chain and reorganization of F-actin microfilaments. Retraction is a more passive process, is independent of myosin light chain kinase and involves protein kinase C (PKC)-stimulated phosphorylation of actin-linking proteins critical for maintaining cell-cell and cell-matrix interactions (Lum & Malik, Am. J. Physiol. 267: L223-L241 (1994).
Hyperpermeability of the blood vessel wall permits leakage of excess fluids and protein into the interstitial space. This acute inflammatory event is frequently allied with tissue ischemia and acute organ dysfunction. Thrombin formed at sites of activated endothelial cells (EC) initiates this microvessel barrier dysfunction due to the formation of large paracellular holes between adjacent EC (Carbajal et al, Am J Physiol Cell Physiol 279: C195-C204, 2000). This process features changes in EC shape due to myosin light chain phosphorylation (MLCP) that initiates the development of F-actin-dependent cytoskeletal contractile tension ( Garcia et al, J Cell Physiol. 1995; 163:510-522 Lum & Malik, Am J Physiol Heart Circ Physiol. 273(5): H2442 - H2451. (1997).
The signalling mechanism of this contractile process involves the proteolytic cleavage and activation of the thrombin receptor. This receptor is coupled to heterotrimeric G proteins of the Gq family that stimulate phospholipase CB, release D-wrμø-inositol 1,4,5-trisphosphate, mobilizing Ca ions from intracellular stores. The subsequent rise in intracellular Ca ion concentration activates Ca ion-calmodulin-dependent MLC kinase, which phosphorylates serine- 19 and threonine- 18 of MLC (Goeckeler & Wysolmerski, J. Cell Biol. 1995; 130:613- 627.). MLCP initiates myosin Mg ion-ATPase activity, causing the binding of myosin to F-actin and subsequent actomyosin stress fiber formation (Ridley & Hall, Cell, 70, 389-399 (1992). The phosphorylation of MLC converts the soluble folded 1OS form of non-muscle myosin II to the insoluble unfolded 6S form. This process is characterized by reorganization of myosin from a diffuse intracellular cloud to punctate spots and ribbons associated with large bundles of F-actin (Verkhovsky et al, The Journal of Cell Biology, 131, 989-1002 (1995). The final consequence is a persistent shape change of endothelial cells and a disruption of the barrier function.
VASCULAR ENDOTHELIAL (VE)-CADHERIN
Thrombin-induced endothelial hyperpermeability may also be mediated by changes in cell- cell adhesion (Dejana J. Clin. Invest. 98: 1949-1953 (1996). Endothelial cell-cell adhesion is determined primarily by the function of vascular endothelial (VE) cadherin (cadherin 5), a Ca-dependent cell-cell adhesion molecule that forms adherens junctions. Cadherin 5 function is regulated from the cytoplasmic side through association with the accessory proteins beta-catenin, plakoglobin (g-catenin), and pi 20 that are linked, in turn, to alpha-catenin (homologous to vinculin) and the F-actin cytoskeleton.
VE-cadherin has emerged as an adhesion molecule that plays fundamental roles in microvascular permeability and in the morphogenic and proliferative events associated with angiogenesis (Vincent et al, Am J Physiol Cell Physiol, 286(5): C987 - C997 (2004). Like other cadherins, VE-cadherin mediates calcium-dependent, homophilic adhesion and functions as a plasma membrane attachment site for the cytoskeleton. However, VE-cadherin is integrated into signaling pathways and cellular systems uniquely important to the vascular endothelium. Recent advances in endothelial cell biology and physiology reveal properties of VE-cadherin that may be unique among members of the cadherin family of adhesion molecules. For these reasons, VE-cadherin represents a cadherin that is both prototypical of the cadherin family and yet unique in function and physiological relevance. Evidence is accumulating that the VE-cadherin-mediated cell-cell adhesion is controlled by a dynamic balance between phosphorylation and dephosphorylation of the junctional proteins including cadherins and catenins. Increased tyrosine phosphorylation of beta-catenin resulted in a dissociation of the catenin from cadherin and from the cytoskeleton, leading to a weak adherens junction (AJ) . Similarly, tyrosine phosphorylation of VE-cadherin and beta-catenin occurred in loose AJ and was notably reduced in tightly confluent monolayers (Tinsley et al., J Biol Chem, 274, 24930-24934 (1999).
In addition the correct clustering of VE-cadherin monomers in adherens junctions is indispensable for a correct signalling activity of VE-cadherin, since cell bearing a chimeric mutant (IL2-VE) containing a full-length VE-cadherin cytoplasmic tail is unable to cause a correct signalling despite its ability to bind to beta-catenin and pi 20 (Lampugnani et al, MoI. Biol, of the Cell, 13, 1 175-1 189 (2002).
Rho- AND Rac-GTPases AND VASCULAR PERMEABILITY
Rho GTPases are a family of small GTPases with profound actions on the actin cytoskeleton of cells. With respect to the functioning of the vascular system they are involved in the regulation of cell shape, cell contraction, cell motility and cell adhesion. The three most prominent family members of the Rho GTPases are RhoA, Rac and cdc42. Activation of RhoA induces the formation of f-actin stress fibres in the cell, while Rac and cdc42 affect the actin cytoskeleton by inducing membrane ruffles and microspikes, respectively (Hall, Science,279:509-5 l4Λ99S). While Rac and cdc42 can affect MLCK activity to a limited extent via activation of protein PAK ( Goeckeler et al. J. Biol. Chem., 275, 24, 18366-18374 (2000), RhoA has a prominent stimulatory effect on actin-myosin interaction by its ability to stabilize the phosphorylated state of MLC (Katoh et al., Am. J. Physiol. Cell. Physiol. 280, C1669-C1679 (2001). This occurs by activation of Rho kinase that in its turn inhibits the phosphatase PPlM that hydrolyses phosphorylated MLC. In addition, Rho kinase inhibits the actin-severing action of cofilin and thus stabilizes f-actin fibres (Toshima et al., MoI. Biol, of the Cell. 12, 1131-1 145 (2001). Furthermore, Rho kinase can also be involved in anchoring the actin cytoskeleton to proteins in the plasma membrane and thus may potentially act on the interaction between junctional proteins and the actin cytoskeleton (Fukata et al. . Cell Biol 145:347-361 (1999).
Thrombin can activate RhoA via Gαl2/13 and a so-called guanine nucleotide exchange factor (GEF) (Seasholtz et al; MoI: Pharmacol. 55, 949-956 (1999). The GEF exchanges RhoA- bound GDP for GTP, by which RhoA becomes active. By this activation RhoA is translocated to the membrane, where it binds by its lipophilic geranyl-geranyl-anchor.
RhoA can be activated by a number of vasoactive agents, including lysophosphatidic acid, thrombin and endothelin. The membrane bound RhoA is dissociated from the membrane by the action of a guanine dissociation inhibitor (GDI) or after the action of a GTPase-activating protein (GAP). The guanine dissociation inhibitors (GDIs) are regulatory proteins that bind to the carboxyl terminus of RhoA.
GDIs inhibit the activity of RhoA by retarding the dissociation of GDP and detaching active RhoA from the plasma membrane. Thrombin directly activates RhoA in human endothelial cells and induces translocation of RhoA to the plasma membrane. Under the same conditions the related GTPase Rac was not activated. Specific inhibition of RhoA by C3 transferase from Clostridium botulinum reduced the thrombin-induced increase in endothelial MLC phosphorylation and permeability, but did not affect the transient histamine-dependent increase in permeability (van Nieuw Amerongen et al. Circ Res. 1998;83: 1 1 15-11231 (1998). The effect of RhoA appears to be mediated via Rho kinase, because the specific Rho kinase inhibitor Y27632 similarly reduced thrombin-induced endothelial permeability.
Racl and RhoA have antagonistic effects on endothelial barrier function. Acute hypoxia inhibits Racl and activates RhoA in normal adult pulmonary artery endothelial cells (PAECs), which leads to a breakdown of barrier function (Wojciak-Stothard and Ridley, Vascul Pharmacol. ,39: 187-99 (2002). PAECs from piglets with chronic hypoxia induced pulmonary hypertension have a stable abnormal phenotype with a sustained reduction in Racl and an increase in RhoA activitity. These activities correlate with changes in the endothelial cytoskeleton, adherens junctions and permeability. Activation of Racl as well as inhibition of RhoA restored the abnormal phenotype and permeability to normal (Wojciak-Stothard et al., Am. J. Physiol, Lung Cell MoI. Physiol. 290, Ll 173-Ll 182 (2006).
It is therefore desirable to screen for substances that restore the physiologic balance of Racl and RhoA activity to a level that is observed in endothelial cells in normal and stable conditions. Preferably this effect is caused by a stabilization of the clustering of VE-cadherin in the adherens junction.
FOCAL ADHESION KINASE AND ITS ROLE IN ENDOTHELIAL PERMEABILITY
Focal adhesion kinase is composed of a central catalytic domain flanked by large N- and C- terminal domains. The N-terminal region contains the FERM homology that can bind integrins and growth factor receptors. The non-catalytic domain in the C-terminal, also referred to as FRNK(FAK-related non-kinase), carries the FATsequence which not only directs FAK to adhesion complexes for signalling, but also provides binding sites for other docking molecules to interact with the cytoplasmic To date, at least five tyrosine residues have been identified in FAK.
Phosphorylation of these tyrosine residues directly correlates with the kinase activity. This is supported by a reciprocal relationship between FAK activity and monolayer permeability. Several models have been proposed to explain the effect of focal adhesion formation on the barrier structure. The increased adhesion of endothelial cells to the extracellular matrix may help to stabilize monolayers against detachment due to the lateral contractile forces produced by inflammatory mediators. Thus, focal adhesion activation may occur in parallel with cell contraction to compensate for the diminished cell-cell binding during inflammatory stimulation. Another hypothesis proposes that FAK activation and focal adhesion reorganization actively contribute to the opening of endothelial cell-cell junctions by providing a mechanical basis for endothelial cells to contract or change shape. The last, but not the least, possibility is that the focal complex serves as a point of convergence for multiple scaffold proteins or signalling molecules to be integrated, which in turn affect the barrier function.
In addition to the well-characterized activation of MAPK, PDK, and eNOS , potential signalling events downstream from FAK include the myosin light chain phosphorylation- triggered actin-myosin contraction and Rho-dependent stress fiber formation which are characteristic features of paracellular permeability (Wu, J Physiol 569. , 359-366 (2005). It is therefore desirable to inhibit FAK phosphorylation in order to promote endothelial integrity.
INFLAMMATION AND ENDOTHELIAL DYSFUNCTION: PATHOLOGICAL AND THERAPEUTIC CONSEQUENCES
Endothelial dysfunction and leakiness of the endothelial barrier is an important component of a range of inflammatory diseases. The inflammatory response is characterized by an extravasation of blood constituents such as plasma proteins and of blood serum leading to severe interstitial tissue edema.
In addition, neutrophils, which are the primary agents of the inflammatory response, are able to emigrate from the blood stream into the underlying tissue.
Together, these effects of endothelial layer leakiness cause substantial damage to healthy organs and tissues. They have been implicated in organ damage of a number of diseases including, but not limited to adult respiratory distress syndrome (ARDS), acute lung injury (ALI), glomerulonephritis, acute and chronic allograft rejection, inflammatory skin diseases, rheumatoid arthritis, asthma, atherosclerosis, systemic lupus erythematosus (SLE), connective tissue diseases, vasculitis, as well as ischemia-reperfusion injury in limb replantation, myocardial infarction, crush injury, shock, stroke and organ transplantation. It is also a prerequisite for new blood vessel formation by proliferation of endothelial and therefore can result in disease where such angiogenesis has been shown to play a pathogenetic role, including but not limited to wet age-related macula degeneration and cancer progression and metastasis.
BRIEF DESCRIPTION OF THE INVENTION
The present invention is directed to methods for screening, identification, characterization and use of proteins, peptides, peptidomimetics, antibodies and small molecules that modulate interactions and signalling events mediated by agents that cause endothelial hyperpermeability. The agents identified by these screening methods exert their effect by binding to and modulating the conformation and/or phosphorylation status of vascular endothelial (VE)-cadherin expressed in the adherens junctions of endothelial cell layers. More specifically these agents promote endothelial integrity by stabilizing the clustering of VE-cadherin at intercellular junctions. Given the importance of disruption of the endothelial barrier function for a broad range of diseases, these agents have broad applicability as therapeutic and/or prophylactic medicinal products.
According to one aspect, the present invention provides a method of screening for proteins, peptides, peptidomimetics, antibodies or small organic molecules that increase the activity of Racl by virtue of their binding to the extracellular portion of this protein, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells grown on porous membranes with at least one of the test compounds alone or together with a stimulant of vascular permeability b. Measuring the endothelial permeability with a suitable agent, which can be colorimetrically detected
According to another embodiment, the present invention provides a method of screening for proteins, peptides, peptidomimetics, antibodies or small organic molecules that increase the activity of Racl by virtue of their binding to the extracellular portion of this protein, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells with at least one of the test compounds b. lysing the endothelial cells with a lysation buffer c. measuring the amount of Racl activity with a specific assay
In another embodiment, the present invention provides a method for screening of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent the activation of RhoA and consequentially the change in the cytoskeletal structure of the endothelial cells, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells with thrombin in the presence of at least one of the test compounds b. lysing the endothelial cells with a lysation buffer c. measuring the RhoA activity with a specific assay
In another embodiment, the present invention provides a method for screening of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent the phosphorylation of focal adhesion kinase, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells with thrombin in the presence of at least one of the test compounds b. lysing the endothelial cells with a lysation buffer c. measuring the phosphorylation of focal adhesion kinase with a specific assay
In another embodiment, the present invention provides a method for screening of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent vascular leak in a warm-blooded animal undergoing systemic inflammatory response, the method comprising the following steps:
a. Initiation of a systemic inflammatory response by applying an appropriate dose of bacterial lipopolysaccharide (LPS) b. Exposing the animal to at least one of the test compounds c. injecting the animal with an appropriate amount of fluorescence labelled micro-beads of appropriate size d. sacrificing the animal after an appropriate time period e. excising and homogenizing an organ or tissue of the animal f. measuring the amount of fluorescence in the homogenate
BEST METHOD OF CARRYING OUT THE INVENTION
Preferentially, these assays are performed in the sequence described above, which constitutes a screening tree to selectively identify compounds with the specific physiological activities claimed by the current invention.
The useful method for measuring the permeability of a confluent layer of endothelial cells is based on a dual chamber method, where endothelial cells are grown on a porous membrane separating the chambers. The cells, preferentially human umbilical vein endothelial cells (HUVEC) are incubated with a test compound alone or together with a stimulant of endothelial permeability for 30 to 120 min. Preferentially thrombin is used as such stimulant. The permeability can be measure with a permeating dye, whose absorption can be measured colorimetrically. Evans Blue can be used preferentially as such dye, either as such or bound to albumin.
The useful methods of analysing the activation status of Racl and RhoA are based on the principle of the so-called pull down assay. In this format, the GTP-bound active state of the respective protein in a cell lysate is bound to an immobilized binding partner and detected with a monoclonal antibody (MAb) specifically directed against the protein in question. The amount of the GTP-bound active state can subsequently be quantified through suitable detection methods, including but not limited to Western blotting or luminescence detection.
For measuring the activation of Racl in human umbilical vein endothelial cells (HUVECs), the cells are incubated with the test compound under suitable conditions for various periods of time up to 30 min and lysed afterwards. The lysate is added to a suitably immobilized p21- binding domain
(PBD) of p21 -activated protein kinase (PAK) and the amount of activated Racl is quantified with a Rac 1 -specific MAb.
For measuring the inhibition of thrombin induced activation of RhoA in HUVECs, the cells are incubated under suitable conditions with a suitable amount of thrombin with and without the test compound for various periods of time up to 10 min and lysed afterwards. The lysate is added to a suitably immobilized rhotekin-binding domain (RBD) and the signal measured with an appropriate detection method.
For measuring the inhibition of thrombin meditated FAK phosphorylation in HUVECs, the cells are incubated under suitable conditions with a suitable amount of thrombin with and without the test compound for various periods of time up to 60 min and lysed afterwards, lysates were subjected to SDS-PAGE and western blot analysis with site-specific antibodies directed against FAK phosphorylated tyrosine residues. For measuring the inhibition of LPS-induced vascular leak in rodents, the animals receive injections of amounts of gram-negative lipopolysaccharide (LPS) suitable to achieve a systemic inflammatory response. After various periods of time between 0 and 4 hours, the test substance is injected intravenously, followed by the injection of a suitable amount of fluorescent microbeads. Subsequently the animals are sacrificed, and organs (lung, kidney, spleen, heart, brain) are excised and cut into thin slices suitable for microscopic analysis and fixated with paraformaldehyde. The numbers of extravasated microspheres are counted using a fluorescent microscope.
Alternatively, the organs are homogenized and the amount of microbeads trapped in the tissues are measured using a suitable fluorescence detection device.
The compounds identified with the screening methods according to the present invention are useful for development of drugs for the prevention and/or treatment of diseases which are caused by an inflammatory reaction and/or endothelial disruption and vascular leak. Therefore, according to another embodiment of the current invention, the compounds of the present invention are administered for treatment and/or prevention of, but not restricted to, septic shock, wound associated sepsis, post-ischemic reperfusion injury, such as after myocardial infarction/reperfusion or organ transplantation), frost-bite injury or shock, acute inflammation mediated lung injury, such as respiratory distress syndrome, acute pancreatitis, liver cirrhosis, uveitis, asthma, traumatic brain injury, nephritis, atopic dermatitis, psoriasis, inflammatory bowel disease, macula degeneration of the eye, diabetic retinopathy, neovascular glaucoma, retinal vein occlusion and tumour progression.
In order to achieve their therapeutic effects in these diseases, the compounds of the present invention may be given orally or parenterally and maybe be formulated into suitable pharmaceutical formulation with pharmaceutically acceptable excipients or carriers. The present invention therefore also relates to a pharmaceutical composition containing an active ingredient identified by the method of screening according to the present invention and further comprising pharmaceutically acceptable excipients or carriers.
Pharmaceutically acceptable excipients are those which are approved by a regulatory agency of the Federal or State governments or listed in the U.S. Pharmacopeia or any other generally recognized pharmacopeia for use in animals, and more particularly in humans. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of animal, vegetable and synthetic origin, e.g. peanut oil, soybean oil., mineral oil and the like. Aqueous carriers nay contain for instance also contain dextrose or glycerol.
Suitable excipients may include, but are not restricted to, starch, glucose, lactose, sucrose, gelatin, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, glycerol, propylene glycol, ethanol and the like. The composition may also include wetting and/or emulsifying agents, or pH buffering agents.
The compositions can take the form of solutions, suspensions, emulsions, tablets, capsules, powders, or slow release formulations. Such compositions will contain a therapeutically effective amount of the compound together with a suitable amount of carrier so as to provide a form for proper administration to the subject to be treated and suitable for the form of treatment.
BRIEF DESCRIPTION OF THE FIGURES
Fig. 1 Compounds identified through the claimed method of screening.
Fig. 2 Permeability of a confluent HUVEC layer stimulated with thrombin after incubation for 45 min.
Fig. 3 Activation of Racl with compound IA. This gel is the result of a pull-down assay as described in Example 1. Lane 1 : medium control, lane 2: HUVEC activation with thrombin for 1 min, lane 3: treatment with compound IA alone for 1 min; lane 4: HUVEC with thrombin and compound IA for 1 min, lane 5: thrombin for 5 min; lane 6: compound IA for 5 min, lane 7: thrombin and compound IA for 5 min. Beta-actin was used to control for total protein content.
Fig. 4 Inhibition of thrombin induced activation of RhoA by compound IA. This gel is the result of a pull-down assay as described in Example 1. Lane 1 : medium control, lane 2:
HUVEC activation with thrombin for 1 min, lane 3: treatment with compound IA alone for 1 min; lane 4: HUVEC with thrombin and compound IA for 1 min, lane 5: thrombin for 5 min; lane 6: compound IA for 5 min, lane 7: thrombin and compound IA for 5 min. Beta-actin was used to control for total protein content.
Fig. 5 Activation of Racl with compound IB. This gel is the result of a pull-down assay as described in Example 1. Lane 1 : medium control, lane 2: HUVEC activation with thrombin for 1 min, lane 3: treatment with compound IB alone for 1 min; lane 4: HUVEC with thrombin and compound IB for 1 min, lane 5: thrombin for 5 min; lane 6: compound IB for 5 min, lane 7: thrombin and compound IB for 5 min.
Fig. 6 Inhibition of thrombin induced activation of RhoA by compound IB at time points 1, 5 and 10 min after stimulation with thrombin.
F'g- 7 Quantification and time dependency of activation of Racl by compound IB
Fig. 8 Quantification and time dependence of inhibition of thrombin induced RhoA activation by compound 1 B
Fig. 9 Inhibition of thrombin induced phosphorylation of FAK by compound IA. This graphs shows the time-course of inhibition of phosphorylation of FAK induced by thrombin.
Fig. 10 Inhibition of LPS induced vascular leak by compound IA. These are fluorescent images of lung slices from rats, in which vascular leak was induced by LPS treatment. Slice a) is from a control animal, slice b) is from an animal treated with compound IA
Example 1
A compound screening method for identification of substances reducing Thrombin- induced permeability of Endothelial Monolayers, through stabilization of VE-cadherin junctions
HUVECs are grown to confluence on 12mm transwell plates, polycarbonate membrane 3μm pore size (Corning), under standard conditions. 2h before the start of the experiment the standard growth medium is exchanged in the upper and lower chamber by growth medium w/o phenol red. To induce permeability 5U/ml thrombin (Calbiochem) or 5U/ml plus the test compound are added to the upper well. HUVEC are incubated for 45 min. At the end of incubation time the medium of the upper chamber is replaced by growth medium w/o phenolred containing 4% BSA and 0,67mg/ml Evans Blue. The diffusion of Evans Blue into the lower chamber is allowed for 10 min, afterwards the upper chamber is removed and the optical density of the medium recovered from the lower chamber measured at wavelength of 620nm.
Relative optical density at 620 nm Control peptide 45 min 1
Compound IB, 45 min 0,99 +/-0.15 thrombin 45 min 9,5 +/- 1,25* thrombin + compound IB 45 min 5,3 +/-1,6 #
*denotes p<0.05 compared to control
# denotes p<0.05 between thrombin and thrombin + compound IB
Example 2
A compound screening method for identification of substances activating Racl through stabilization of VE-cadherin junctions
HUVECs are grown to confluence under standard conditions. Before induction of Racl activity HUVECs were starved for 4h by using IMDM (Gibco) without growth factor and serum supplements. Racl activity is induced by adding 50μg/ml of test compound into starvation medium for 1, 5 and 10 min. Active Racl was isolated using Racl / Cdc42 Assay Reagent from Upstate according to manufactures instructions. Isolates were separated on a 15% polyacrylamid gel and blotted on Nitrocellulose-Membranes (Bio-Rad). Racl was dedected by using Anti-Racl clone23A8, anti-mouse from Upstate (1 :250).
Relative values compared to unstimulated control
Control peptide 1 min 1
Control peptide 5 min 1
Control peptide 10 min 1
Compound IB, 1 min 2+/-0.2 *
Compound IB, 5 min 2+/-0.1 *
Compound IB, 10 min 1+/-0.1 thrombin 1 min 0.5+/-0.2 * thrombin 5 min 0.5+/-0.2 * thrombin 10 min 1+/-0.1 thrombin + compound IB, 1 min 1+/-0.2 # thrombin + compound IB, 5 min 1+/-0.1 # thrombin + compound IB, 10 min 1+/-0.1
* denotes p<0.05 compared to control
# denotes p<0.05 between thrombin and thrombin + compound IB
Example 3 A compound screening method for identification of substances inhibiting thrombin induced RhoA activation
HUVEC are grown to confluence under standard conditions. Before induction of Rho activity HUVEC were starved for 4h by using IMDM (Gibco) without growth factor and serum supplements. After the starvation period 5 U/ml Thrombin (Calbiochem) or 5U thrombin plus 50μg/ml of test compound are added to the starvation medium for 1, 5 and 10 min. Active RhoA was isolated using Rho Assay Reagent from Upstate according to manufactures instructions. Isolates were separated on a 15% polyacrylamid gel and blotted on Nitrocellulose-Membrane (Bio-Rad). RhoA was dedected by using Anti-Rho (-A, -B, -C), clone55 from Upstate (1 :500).
Relative values compared to unstimulated control
Control peptide 1 min 1
Control peptide 5 min 1
Control peptide 10 min 1
Compound IB, 1 min 1+/-0.2
Compound IB 5 min 1+/-0.1
Compound IB 10 min 1+/-0.1 thrombin 1 min 2.5+/-0.2 * thrombin 5 min 2.5+/-0.2 * thrombin 10 min 1+/-0.2 thrombin + compound IB 1 min 1+/-0.3 # thrombin + compound IB 5 min 1+/-0.1 # thrombin + compound IB 10 min 1+/-0.1
* denotes p<0.05 compared to control
# denotes p<0.05 between thrombin and thrombin + compound IB
Example 4
A compound screening method for identification of substances inhibiting the thrombin induced FAK phosphorylation at autophosphorylation site Tyr397
Immunoprecipitation :
HUVEC were incubated with FX06 (50μg/ml), Thrombin (lU/ml, Sigma Aldrich) and Thrombin/ test compound for indicated time points. After washing with ice cold PBS (GIBCO), cells were scrapped in Tris-lysis buffer (plus 1% Triton X (Bio-Rad), NP40 (Sigma Aldrich) and proteinase and phosphatase inhibitory cocktails (Sigma Aldrich)) from culture flasks and lysed for 20 min on ice. Lysates were heavily vortexed every 5 min. After lysis lysates were centrifuged (15.000rpm/10min/4°C) and supernatants were added to 50μl sepahrose beads (Sigma Aldrich) preincubated with 1 μg total FAK antibody (BD Transduction Laboratories). Beads were agitated on the wheel for 2h at 4°C, followed by 3 times washing with ice cold PBS, the addition of 2x sample buffer and incubation at 95°C for 5 min. The sample buffer was then removed from the beads and applied to western blotting.
Western Blot:
10% polyacrylamide gels were run for separating precipitated proteins. Gels were blotted onto PVDF (Bio-Rad) membranes using the hoefer semi dry blotting system. Membranes were then washed with TBS/0, 5% TWEEN (TBST), blocked with 1% BSA/TBST for Ih at RT and then incubated with the p397 FAK antibody (0,2 μg/ ml; BD Transduction Laboratories) in 1% BSA/TBST over night at 4°C. For detection, a HRP-labeled goat anti-mouse Ab (1 :25 000; Bio-Rad) in TBST was used and bound Abs were visualized by chemi luminescence (ECL-system, Amersham Corp., Arlington Heights, IL) and recorded on film.
Relative values compared to unstimulated control
Control peptide 1 min 1
Control peptide 5 min 1
Control peptide 10 min 1
Compound IA, 1 min 5.5+/-0.2 *
Compound I A, 5 min 2+/-0.1 *
Compound 1 A, 10 min 1+/-0.1 thrombin 1 min 4.5+/-0.2 * thrombin 5 min 4.5+/-0.2 * thrombin 10 min 3.8+/-0.1 * thrombin + compound IA, 1 min 3+/-O.5 * thrombin + compound IA, 5 min 2+/-0.1 *# thrombin + compound IA, 10 min 1.3+/-0.1 *#
* denotes p<0.05 compared to control
# denotes p<0.05 between thrombin and thrombin + FX06 Example 5
A compound screening method for indentification of substances inhibiting the LPS induced vascular leak in rodents
Male Him OFA/SPF rats (Institute for Biomedical Research, Medical School Vienna) with a body weight of 260-32Og are housed at the Institute for Biomedical Research, Medical School Vienna. All experiments were approved by Amt der Wiener Landesregierung, MA58. Rats are anaesthetised with 100 mg/ kg sodium thiopentone (Sandoz). The trachea is cannulated to facilitate respiration. The right jugular vein is cannulated for the administration of drugs. To measure the Mean Arterial Blood Pressure (MAP) a catheter is placed into the right carotid artery. After surgery the animals are randomized in treatment groups. All rats receive a fluid replacement (600 μl 0,9% saline as an i.v. infusion) and are allowed to stabilize for 15 min. Body temperature is controlled with a homeothermic blanket throughout the whole experiment. After the stabilisation period, the endotoxic shock is induced by a bolus injection of 12 mg/kg LPS (E.coli serotype 0.127:B8; Sigma). 60min after LPS administration the animals receive a bolus injections of 3 mg/kg of test compound or saline. 5h 50 min after the LPS administration the rats receive an bolus injection of fluorescenct microspheres; 125 x 10 beads /kg body weight (Fluo Spheres Polystyrene Microspheres; 1 μm yellow-green fluorescent (505/515) Invitrogen Molecular Probes).
6h after LPS administration the animals are sacrificed and the lungs are removed to assess vascular leakage. Vascular leakage of the lung is assessed by measurement of the fluorescence per g of tissue. For these purpose the lung tissue was digested with ethanolic KOH and the fluorescent microspheres are recovered by sedimetation as recommended by the "Manual for using Fluorescent Microspheres to measure organ perfusion" Fluorescent Microsphere Resource Center; University of Washington. Fluorescence is measured using a Spectra Max Gemini S Fluorometer
Relative fluorescence within lungs of LPS-treated animals sham 656+/-210
LPS 3454+/-790 *
LPS + compound IA 2275+A795 *#
* denotes p<0.05 compared to control
# denotes p<0.05 between thrombin and thrombin + compound IA

Claims

Claims
1. A method of screening for compounds out of the group consisting of proteins, peptides, peptidomimetics, antibodies or small organic molecules that increase the activity of the protein Racl by virtue of their binding to the extracellular portion of this protein, the method comprising the steps of: a. contacting a confluent layer of cultured endothelial cells grown on porous membranes with at least one of the test compounds alone or together with a stimulant of vascular permeability, b. measuring the endothelial permeability with a suitable agent, which can be colorimetrically detected.
2. A method of screening for compounds out of the group consisiting of proteins, peptides, peptidomimetics, antibodies and small organic molecules that increase the activity of the protein Racl by virtue of their binding to the extracellular portion of this protein, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells with at least one of test compounds, b. lysing the endothelial cells with a lysation buffer, and c. measuring the amount of Rac 1 activity.
3. A method for screening for compounds out of the group consisiting of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent the activation of RhoA and consequentially the change in the cytoskeletal structure of endothelial cells, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells with thrombin in the presence of at least one of test compounds, b. lysing the endothelial cells with a lysation buffer, and c. measuring the RhoA activity.
4. A method for screening for compounds out of the groups consisting of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent the phosphorylation of focal adhesion kinase, the method comprising the steps of:
a. contacting a confluent layer of cultured endothelial cells with thrombin in the presence of at least one of the test compounds, b. lysing the endothelial cells with a lysation buffer, and c. measuring the phosphorylation of focal adhesion kinase.
5. A method for screening for compounds out of the group consisting of proteins, peptides, peptidomimetics, antibodies and small organic molecules that prevent vascular leak in a warm-blooded animal undergoing systemic inflammatory response, the method comprising the following steps:
a. initiation of a systemic inflammatory response by applying an appropriate dose of bacterial lipopolysaccharide (LPS), b. exposing the animal to at least one of the test compounds, c. injecting the animal with an appropriate amount of fluorescence labelled micro-beads of appropriate size, d. sacrificing the animal after an appropriate time period, e. excising and homogenizing an organ or tissue of the animal, and f. measuring the amount of fluorescence in the homogenate.
6. A method for screening for compounds out of the group consisting of proteins, peptides, peptidomimetics, antibodies and small organic molecules, characterized in that it comprises one or more methods according to claims 1-5.
7. A method for screening for compounds out of the group consisting of proteins, peptides, peptidomimetics, antibodies and small organic molecules according to claim 6, characterized in that it comprises all 5 methods according to claims 1-5.
8. A method for screening for compounds out of the group consisting of proteins, peptides, peptidomimetics, antibodies and small organic molecules according to claim 6, characterized in that it comprises all 5 methods according to claims 1-5, wherein said methods are carried out subsequently beginning with the method according to claim 1 or claim 2, followed by the methods according to claim 2, 3 and 4.
9. Compounds out of the group consisting of proteins, peptides, peptidomimetics, antibodies and small organic molecules identified with the screening methods according to one of the claim 1-8 for development of drugs for the prevention and/or treatment of diseases which are caused by an inflammatory reaction and/or endothelial disruption and vascular leak.
10. Pharmaceutical preparation containing compounds according to claim 9.
11. Pharmaceutical preparation according to claim 10 for treatment and/or prevention of septic shock, wound associated sepsis, post-ischemic reperfusion injury, such as after myocardial infarction/reperfusion or organ transplantation), frost-bite injury or shock, acute inflammation mediated lung injury, such as respiratory distress syndrome, acute pancreatitis, liver cirrhosis, uveitis, asthma, traumatic brain injury, nephritis, atopic dermatitis, psoriasis, inflammatory bowel disease, macula degeneration of the eye, diabetic retinopathy, neovascular glaucoma, retinal vein occlusion and tumour progression.
PCT/AT2008/000337 2007-09-24 2008-09-22 Methods of screening for compounds having anti- inflammatory activity and/or prevent / treat vascular leak WO2009039542A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP08799926A EP2193370A2 (en) 2007-09-24 2008-09-22 Methods of screening for compounds having anti-inflammatory activity
JP2010525162A JP2010540892A (en) 2007-09-24 2008-09-22 Method for screening compound having anti-inflammatory activity

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US11/860,488 US20090081192A1 (en) 2007-09-24 2007-09-24 Methods of screening for compounds having anti-inflammatory activity
US11/860,488 2007-09-24
AT19392007A AT506121A1 (en) 2007-11-28 2007-11-28 Screening for compounds, e.g. proteins or peptides, that increase the activity of the protein Rac1 comprises contacting cultured endothelial cells with a test compound and measuring endothelial permeability
ATA1939/2007 2007-11-28

Publications (3)

Publication Number Publication Date
WO2009039542A2 true WO2009039542A2 (en) 2009-04-02
WO2009039542A9 WO2009039542A9 (en) 2009-05-14
WO2009039542A3 WO2009039542A3 (en) 2009-07-02

Family

ID=40263422

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AT2008/000337 WO2009039542A2 (en) 2007-09-24 2008-09-22 Methods of screening for compounds having anti- inflammatory activity and/or prevent / treat vascular leak

Country Status (3)

Country Link
EP (1) EP2193370A2 (en)
JP (1) JP2010540892A (en)
WO (1) WO2009039542A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009137851A1 (en) * 2008-05-15 2009-11-19 Fibrex Medical Research & Development Gmbh Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
WO2010043972A2 (en) * 2008-10-15 2010-04-22 Fibrex Medical Research & Development Gmbh Pharmaceutical compositions and methods of use for the prevention and treatment of hypoxic injury
KR101127225B1 (en) 2010-01-07 2012-03-29 건국대학교 산학협력단 Biomarker of Urothelial Cancer and the Diagnostic Method of the Same
WO2013004332A1 (en) 2011-07-07 2013-01-10 Merck Patent Gmbh Substituted azaheterocycles for the treatment of cancer

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0405315A2 (en) * 1989-06-30 1991-01-02 American Cyanamid Company Use of a phagocyte-activating agent : LPS or IL-2 for the manufacture of a medicament for treating staphylococcus aureus infection in cows
WO2006000007A1 (en) * 2004-06-25 2006-01-05 Fibrex Medical Research & Development Gesmbh Use of peptides derived from the a alpha or b beta chain of human fibrinogen for the treatment of shock
WO2007070899A2 (en) * 2005-12-23 2007-06-28 Fibrex Medical Research & Development Gmbh Pharmaceutical composition for treating haemorrhagic shock and its consecutive symptoms

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1494699A4 (en) * 2002-04-11 2009-07-22 Childrens Medical Center Methods for inhibiting vascular hyperpermeability
JP2006101755A (en) * 2004-10-05 2006-04-20 Tokyoto Igaku Kenkyu Kiko Allergic mouse

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0405315A2 (en) * 1989-06-30 1991-01-02 American Cyanamid Company Use of a phagocyte-activating agent : LPS or IL-2 for the manufacture of a medicament for treating staphylococcus aureus infection in cows
WO2006000007A1 (en) * 2004-06-25 2006-01-05 Fibrex Medical Research & Development Gesmbh Use of peptides derived from the a alpha or b beta chain of human fibrinogen for the treatment of shock
WO2007070899A2 (en) * 2005-12-23 2007-06-28 Fibrex Medical Research & Development Gmbh Pharmaceutical composition for treating haemorrhagic shock and its consecutive symptoms

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
HINSBERGH VAN V W M ET AL: "Intracellular signalling involved in modulating human endothelial barrier function" JOURNAL OF ANATOMY, CAMBRIDGE UNIVERSITY PRESS, CAMBRIDGE, GB, vol. 200, 1 January 2002 (2002-01-01), pages 549-560, XP002261213 ISSN: 0021-8782 *
HOLINSTAT MICHAEL ET AL: "Suppression of RhoA activity by focal adhesion kinase-induced activation of p190RhoGAP: role in regulation of endothelial permeability." THE JOURNAL OF BIOLOGICAL CHEMISTRY 27 JAN 2006, vol. 281, no. 4, 27 January 2006 (2006-01-27), pages 2296-2305, XP002512837 ISSN: 0021-9258 *
KLINGER ET AL: "Natriuretic peptides differentially attenuate thrombin-induced barrier dysfunction in pulmonary microvascular endothelial cells" EXPERIMENTAL CELL RESEARCH, ACADEMIC PRESS, US, vol. 312, no. 4, 15 February 2006 (2006-02-15), pages 401-410, XP005207531 ISSN: 0014-4827 *
KNEZEVIC NEBOJSA ET AL: "GDI-1 phosphorylation switch at serine 96 induces RhoA activation and increased endothelial permeability." MOLECULAR AND CELLULAR BIOLOGY SEP 2007, vol. 27, no. 18, 1 September 2007 (2007-09-01), pages 6323-6333, XP002512836 ISSN: 0270-7306 *
MARCUS ET AL: "Measurement of Endothelial Permeability" ANNALS OF VASCULAR SURGERY, QUALITY MEDICAL PUBLISHING, ST. LOUIS, MO, US, vol. 12, no. 4, 1 July 1998 (1998-07-01), pages 384-390, XP005940431 ISSN: 0890-5096 *
SAENZ-MORALES ET AL: "Requirements for proximal tubule epithelial cell detachment in response to ischemia: Role of oxidative stress" EXPERIMENTAL CELL RESEARCH, ACADEMIC PRESS, US, vol. 312, no. 19, 15 November 2006 (2006-11-15), pages 3711-3727, XP005716783 ISSN: 0014-4827 *
TORIUMI Y ET AL: "Pioglitazone reduces monocyte adhesion to vascular endothelium under flow by modulating RhoA GTPase and focal adhesion kinase" FEBS LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 553, no. 3, 23 October 2003 (2003-10-23), pages 419-422, XP004468259 ISSN: 0014-5793 *
VAN NIEUW AMERONGEN GEERTEN P ET AL: "GIT1 mediates thrombin signaling in endothelial cells: role in turnover of RhoA-type focal adhesions." CIRCULATION RESEARCH 30 APR 2004, vol. 94, no. 8, 30 April 2004 (2004-04-30), pages 1041-1049, XP002512838 ISSN: 1524-4571 *
WOJCIAK-STOTHARD BEATA ET AL: "Rac and Rho play opposing roles in the regulation of hypoxia/reoxygenation-induced permeability changes in pulmonary artery endothelial cells." AMERICAN JOURNAL OF PHYSIOLOGY. LUNG CELLULAR AND MOLECULAR PHYSIOLOGY APR 2005, vol. 288, no. 4, April 2005 (2005-04), pages L749-L760, XP002512834 ISSN: 1040-0605 *
WOJCIAK-STOTHARD BEATA ET AL: "Rac1 and RhoA as regulators of endothelial phenotype and barrier function in hypoxia-induced neonatal pulmonary hypertension." AMERICAN JOURNAL OF PHYSIOLOGY. LUNG CELLULAR AND MOLECULAR PHYSIOLOGY JUN 2006, vol. 290, no. 6, June 2006 (2006-06), pages L1173-L1182, XP002512835 ISSN: 1040-0605 cited in the application *
WOJCIAK-STOTHARD BEATA ET AL: "Rho GTPases and the regulation of endothelial permeability." VASCULAR PHARMACOLOGY NOV 2002, vol. 39, no. 4-5, November 2002 (2002-11), pages 187-199, XP002512833 ISSN: 1537-1891 cited in the application *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009137851A1 (en) * 2008-05-15 2009-11-19 Fibrex Medical Research & Development Gmbh Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
CN102124026A (en) * 2008-05-15 2011-07-13 菲布雷克斯医疗研究及开发有限责任公司 Peptides and derivatives thereof, the manufacturing thereof as well as their use for preparing a therapeutically and/or preventively active pharmaceutical composition
WO2010043972A2 (en) * 2008-10-15 2010-04-22 Fibrex Medical Research & Development Gmbh Pharmaceutical compositions and methods of use for the prevention and treatment of hypoxic injury
WO2010043444A2 (en) * 2008-10-15 2010-04-22 Fibrex Medical Research & Development Gmbh Pharmaceutical preparation for the treatment and/or prevention of ischemia/reperfusion injury and the sequels thereof
WO2010043444A3 (en) * 2008-10-15 2010-06-24 Fibrex Medical Research & Development Gmbh Bbeta (15-42) and its analogues combined with cyclosporine for treatment of reperfusion injury
WO2010043972A3 (en) * 2008-10-15 2010-06-24 Fibrex Medical Research & Development Gmbh Bbeta (15-42) and its analogues combined with cyclosporine for treatment of reperfusion injury
KR101127225B1 (en) 2010-01-07 2012-03-29 건국대학교 산학협력단 Biomarker of Urothelial Cancer and the Diagnostic Method of the Same
WO2013004332A1 (en) 2011-07-07 2013-01-10 Merck Patent Gmbh Substituted azaheterocycles for the treatment of cancer
US9199962B2 (en) 2011-07-07 2015-12-01 Merck Patent Gmbh Substituted azaheterocycles for the treatment of cancer

Also Published As

Publication number Publication date
JP2010540892A (en) 2010-12-24
EP2193370A2 (en) 2010-06-09
WO2009039542A3 (en) 2009-07-02
WO2009039542A9 (en) 2009-05-14

Similar Documents

Publication Publication Date Title
Chiu et al. Arp2/3-and cofilin-coordinated actin dynamics is required for insulin-mediated GLUT4 translocation to the surface of muscle cells
Dudek et al. Abl tyrosine kinase phosphorylates nonmuscle Myosin light chain kinase to regulate endothelial barrier function
White Pathobiology of troponin elevations: do elevations occur with myocardial ischemia as well as necrosis?
Corsetti et al. Decreased expression of Klotho in cardiac atria biopsy samples from patients at higher risk of atherosclerotic cardiovascular disease
Friedrich et al. Mechano-regulation of the beating heart at the cellular level–mechanosensitive channels in normal and diseased heart
Hus-Citharel et al. Apelin counteracts vasopressin-induced water reabsorption via cross talk between apelin and vasopressin receptor signaling pathways in the rat collecting duct
Aydin et al. Elevated adropin: a candidate diagnostic marker for myocardial infarction in conjunction with troponin-I
Dolai et al. Effects of ethanol metabolites on exocytosis of pancreatic acinar cells in rats
Lockamy et al. Functional and physical competition between phospholamban and its mutants provides insight into the molecular mechanism of gene therapy for heart failure
Bck et al. Insulin and IGF1 receptors in human cardiac microvascular endothelial cells: metabolic, mitogenic and anti-inflammatory effects
Tian et al. Asef controls vascular endothelial permeability and barrier recovery in the lung
Sumida et al. Sphingosine-1-phosphate enhancement of cortical actomyosin organization in cultured human Schlemm's canal endothelial cell monolayers
EP2193370A2 (en) Methods of screening for compounds having anti-inflammatory activity
AU2013279604A1 (en) BAG3 as biochemical serum and tissue marker
Chen et al. Apoptosis of bone marrow mesenchymal stem cells caused by hypoxia/reoxygenation via multiple pathways
Iwata et al. Production of TRPV2-targeting functional antibody ameliorating dilated cardiomyopathy and muscular dystrophy in animal models
Gangopadhyay et al. Intracellular translocation of calmodulin and Ca2+/calmodulin-dependent protein kinase II during the development of hypertrophy in neonatal cardiomyocytes
Peters et al. Identification, localization and interaction of SNARE proteins in atrial cardiac myocytes
US20100267615A1 (en) Methods for treatment and/or prevention of a disease associated with vascular leak
Li et al. Irisin alleviates high glucose-induced hypertrophy in H9c2 cardiomyoblasts by inhibiting endoplasmic reticulum stress
Liu et al. Impact of traumatic muscle crush injury as a cause of cardiomyocyte-specific injury: an experimental study
Turcotte et al. A perinuclear calcium compartment regulates cardiac myocyte hypertrophy
Wette et al. Nuclei isolation methods fail to accurately assess the subcellular localization and behaviour of proteins in skeletal muscle
Khalaf et al. Vascular endothelial ERp72 is involved in the inflammatory response in a rat model of skeletal muscle injury
Kendig et al. Sphingosine-1-phosphate induced contraction of bladder smooth muscle

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08799926

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2008799926

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2010525162

Country of ref document: JP

NENP Non-entry into the national phase in:

Ref country code: DE