AU2005270734B2 - Compositions and methods for angiogenesis-related molecules and treatments - Google Patents
Compositions and methods for angiogenesis-related molecules and treatments Download PDFInfo
- Publication number
- AU2005270734B2 AU2005270734B2 AU2005270734A AU2005270734A AU2005270734B2 AU 2005270734 B2 AU2005270734 B2 AU 2005270734B2 AU 2005270734 A AU2005270734 A AU 2005270734A AU 2005270734 A AU2005270734 A AU 2005270734A AU 2005270734 B2 AU2005270734 B2 AU 2005270734B2
- Authority
- AU
- Australia
- Prior art keywords
- sirna
- cells
- molecule
- isolated
- angiogenesis
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
- 238000000034 method Methods 0.000 title claims description 72
- 230000033115 angiogenesis Effects 0.000 title claims description 58
- 238000011282 treatment Methods 0.000 title claims description 32
- 239000000203 mixture Substances 0.000 title description 6
- 239000004055 small Interfering RNA Substances 0.000 claims description 147
- 108020004459 Small interfering RNA Proteins 0.000 claims description 108
- 108090000623 proteins and genes Proteins 0.000 claims description 87
- 239000013598 vector Substances 0.000 claims description 71
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims description 68
- 230000014509 gene expression Effects 0.000 claims description 58
- 208000035475 disorder Diseases 0.000 claims description 56
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 39
- 206010028980 Neoplasm Diseases 0.000 claims description 29
- 239000002773 nucleotide Substances 0.000 claims description 20
- 125000003729 nucleotide group Chemical group 0.000 claims description 19
- 230000000295 complement effect Effects 0.000 claims description 16
- 238000004519 manufacturing process Methods 0.000 claims description 14
- 108020004999 messenger RNA Proteins 0.000 claims description 14
- 239000003814 drug Substances 0.000 claims description 12
- 238000002560 therapeutic procedure Methods 0.000 claims description 12
- 201000011510 cancer Diseases 0.000 claims description 11
- 208000024172 Cardiovascular disease Diseases 0.000 claims description 8
- 206010012689 Diabetic retinopathy Diseases 0.000 claims description 8
- 201000004681 Psoriasis Diseases 0.000 claims description 8
- 206010003246 arthritis Diseases 0.000 claims description 8
- 230000002207 retinal effect Effects 0.000 claims description 8
- 230000002441 reversible effect Effects 0.000 claims description 8
- 208000005590 Choroidal Neovascularization Diseases 0.000 claims description 7
- 206010060823 Choroidal neovascularisation Diseases 0.000 claims description 7
- 206010055665 Corneal neovascularisation Diseases 0.000 claims description 7
- 208000007135 Retinal Neovascularization Diseases 0.000 claims description 7
- 201000000159 corneal neovascularization Diseases 0.000 claims description 7
- 208000027866 inflammatory disease Diseases 0.000 claims description 7
- 208000002780 macular degeneration Diseases 0.000 claims description 6
- 239000008194 pharmaceutical composition Substances 0.000 claims description 5
- 238000012228 RNA interference-mediated gene silencing Methods 0.000 claims description 3
- 230000009368 gene silencing by RNA Effects 0.000 claims description 3
- 239000008177 pharmaceutical agent Substances 0.000 claims description 3
- 239000003937 drug carrier Substances 0.000 claims description 2
- 230000002757 inflammatory effect Effects 0.000 claims 1
- 210000004027 cell Anatomy 0.000 description 174
- 108090000765 processed proteins & peptides Proteins 0.000 description 87
- 150000007523 nucleic acids Chemical class 0.000 description 81
- 102000004196 processed proteins & peptides Human genes 0.000 description 80
- 102000039446 nucleic acids Human genes 0.000 description 79
- 108020004707 nucleic acids Proteins 0.000 description 79
- 229920001184 polypeptide Polymers 0.000 description 71
- 230000000694 effects Effects 0.000 description 45
- 102000004169 proteins and genes Human genes 0.000 description 40
- 235000018102 proteins Nutrition 0.000 description 36
- 238000003556 assay Methods 0.000 description 29
- 239000012634 fragment Substances 0.000 description 27
- 150000001875 compounds Chemical class 0.000 description 26
- 108010029485 Protein Isoforms Proteins 0.000 description 25
- 102000001708 Protein Isoforms Human genes 0.000 description 25
- 101800001318 Capsid protein VP4 Proteins 0.000 description 24
- 102100027609 Rho-related GTP-binding protein RhoD Human genes 0.000 description 24
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 24
- 102000018898 GTPase-Activating Proteins Human genes 0.000 description 22
- 241000282414 Homo sapiens Species 0.000 description 22
- 108091006094 GTPase-accelerating proteins Proteins 0.000 description 21
- 210000002889 endothelial cell Anatomy 0.000 description 21
- 210000001519 tissue Anatomy 0.000 description 21
- 108020004414 DNA Proteins 0.000 description 20
- 241000699670 Mus sp. Species 0.000 description 20
- 230000006870 function Effects 0.000 description 20
- 210000004881 tumor cell Anatomy 0.000 description 20
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 19
- 241001465754 Metazoa Species 0.000 description 19
- 241000699666 Mus <mouse, genus> Species 0.000 description 15
- 239000000523 sample Substances 0.000 description 15
- 150000001413 amino acids Chemical group 0.000 description 14
- 238000009396 hybridization Methods 0.000 description 14
- 102000011068 Cdc42 Human genes 0.000 description 13
- 108050001278 Cdc42 Proteins 0.000 description 13
- 230000000692 anti-sense effect Effects 0.000 description 13
- 238000002474 experimental method Methods 0.000 description 13
- 230000003612 virological effect Effects 0.000 description 13
- 230000002491 angiogenic effect Effects 0.000 description 12
- 230000015572 biosynthetic process Effects 0.000 description 12
- 201000010099 disease Diseases 0.000 description 12
- 230000008569 process Effects 0.000 description 12
- 229950010131 puromycin Drugs 0.000 description 12
- 241000699660 Mus musculus Species 0.000 description 11
- 238000004458 analytical method Methods 0.000 description 11
- 238000013459 approach Methods 0.000 description 11
- 238000003745 diagnosis Methods 0.000 description 11
- 239000013604 expression vector Substances 0.000 description 11
- 238000004393 prognosis Methods 0.000 description 11
- 230000001225 therapeutic effect Effects 0.000 description 11
- 230000027455 binding Effects 0.000 description 10
- 230000012010 growth Effects 0.000 description 10
- 238000001727 in vivo Methods 0.000 description 10
- 238000013508 migration Methods 0.000 description 10
- 230000035772 mutation Effects 0.000 description 10
- 230000000875 corresponding effect Effects 0.000 description 9
- 230000030279 gene silencing Effects 0.000 description 9
- 230000005012 migration Effects 0.000 description 9
- 238000003752 polymerase chain reaction Methods 0.000 description 9
- 108091033319 polynucleotide Proteins 0.000 description 9
- 102000040430 polynucleotide Human genes 0.000 description 9
- 239000002157 polynucleotide Substances 0.000 description 9
- 239000000047 product Substances 0.000 description 9
- 230000035755 proliferation Effects 0.000 description 9
- 230000001177 retroviral effect Effects 0.000 description 9
- 238000012216 screening Methods 0.000 description 9
- 102000042463 Rho family Human genes 0.000 description 8
- 108091078243 Rho family Proteins 0.000 description 8
- 239000002299 complementary DNA Substances 0.000 description 8
- 238000011534 incubation Methods 0.000 description 8
- 238000003753 real-time PCR Methods 0.000 description 8
- 230000001105 regulatory effect Effects 0.000 description 8
- 238000011830 transgenic mouse model Methods 0.000 description 8
- 210000005166 vasculature Anatomy 0.000 description 8
- 108091034117 Oligonucleotide Proteins 0.000 description 7
- 101150036876 cre gene Proteins 0.000 description 7
- 230000003247 decreasing effect Effects 0.000 description 7
- 238000007877 drug screening Methods 0.000 description 7
- 108020001507 fusion proteins Proteins 0.000 description 7
- 102000037865 fusion proteins Human genes 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 239000002609 medium Substances 0.000 description 7
- 230000004048 modification Effects 0.000 description 7
- 238000012986 modification Methods 0.000 description 7
- 230000002829 reductive effect Effects 0.000 description 7
- 210000003518 stress fiber Anatomy 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- 230000008685 targeting Effects 0.000 description 7
- QTBSBXVTEAMEQO-UHFFFAOYSA-N Acetic acid Chemical compound CC(O)=O QTBSBXVTEAMEQO-UHFFFAOYSA-N 0.000 description 6
- 102000004190 Enzymes Human genes 0.000 description 6
- 108090000790 Enzymes Proteins 0.000 description 6
- 241000282412 Homo Species 0.000 description 6
- 101001106406 Homo sapiens Rho GTPase-activating protein 1 Proteins 0.000 description 6
- OKKJLVBELUTLKV-UHFFFAOYSA-N Methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 6
- 102220492952 Nuclear RNA export factor 1_R82A_mutation Human genes 0.000 description 6
- 102100021433 Rho GTPase-activating protein 1 Human genes 0.000 description 6
- 239000006146 Roswell Park Memorial Institute medium Substances 0.000 description 6
- 101001092180 Saccharomyces cerevisiae (strain ATCC 204508 / S288c) RHO GTPase-activating protein RGD1 Proteins 0.000 description 6
- JLCPHMBAVCMARE-UHFFFAOYSA-N [3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[3-[[3-[[3-[[3-[[3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-[[5-(2-amino-6-oxo-1H-purin-9-yl)-3-hydroxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxyoxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(5-methyl-2,4-dioxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(6-aminopurin-9-yl)oxolan-2-yl]methoxy-hydroxyphosphoryl]oxy-5-(4-amino-2-oxopyrimidin-1-yl)oxolan-2-yl]methyl [5-(6-aminopurin-9-yl)-2-(hydroxymethyl)oxolan-3-yl] hydrogen phosphate Polymers Cc1cn(C2CC(OP(O)(=O)OCC3OC(CC3OP(O)(=O)OCC3OC(CC3O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c3nc(N)[nH]c4=O)C(COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3COP(O)(=O)OC3CC(OC3CO)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3ccc(N)nc3=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cc(C)c(=O)[nH]c3=O)n3cc(C)c(=O)[nH]c3=O)n3ccc(N)nc3=O)n3cc(C)c(=O)[nH]c3=O)n3cnc4c3nc(N)[nH]c4=O)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)n3cnc4c(N)ncnc34)O2)c(=O)[nH]c1=O JLCPHMBAVCMARE-UHFFFAOYSA-N 0.000 description 6
- 229940088598 enzyme Drugs 0.000 description 6
- 238000010195 expression analysis Methods 0.000 description 6
- 238000003197 gene knockdown Methods 0.000 description 6
- 238000010363 gene targeting Methods 0.000 description 6
- 239000003102 growth factor Substances 0.000 description 6
- 208000015181 infectious disease Diseases 0.000 description 6
- 108010082117 matrigel Proteins 0.000 description 6
- 230000001404 mediated effect Effects 0.000 description 6
- 238000010232 migration assay Methods 0.000 description 6
- 238000012546 transfer Methods 0.000 description 6
- 102000007469 Actins Human genes 0.000 description 5
- 108010085238 Actins Proteins 0.000 description 5
- 108091026890 Coding region Proteins 0.000 description 5
- 108020004705 Codon Proteins 0.000 description 5
- 102100024785 Fibroblast growth factor 2 Human genes 0.000 description 5
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 5
- 102000016359 Fibronectins Human genes 0.000 description 5
- 108010067306 Fibronectins Proteins 0.000 description 5
- 108010001441 Phosphopeptides Proteins 0.000 description 5
- 102000005789 Vascular Endothelial Growth Factors Human genes 0.000 description 5
- 108010019530 Vascular Endothelial Growth Factors Proteins 0.000 description 5
- 241000700605 Viruses Species 0.000 description 5
- 235000001014 amino acid Nutrition 0.000 description 5
- 229940024606 amino acid Drugs 0.000 description 5
- 238000010171 animal model Methods 0.000 description 5
- 239000005557 antagonist Substances 0.000 description 5
- 230000008901 benefit Effects 0.000 description 5
- 238000001516 cell proliferation assay Methods 0.000 description 5
- 230000008859 change Effects 0.000 description 5
- 230000002950 deficient Effects 0.000 description 5
- 230000002068 genetic effect Effects 0.000 description 5
- 238000000338 in vitro Methods 0.000 description 5
- 238000003780 insertion Methods 0.000 description 5
- 230000037431 insertion Effects 0.000 description 5
- 239000002245 particle Substances 0.000 description 5
- 238000012360 testing method Methods 0.000 description 5
- 238000001890 transfection Methods 0.000 description 5
- 230000004614 tumor growth Effects 0.000 description 5
- 241000701161 unidentified adenovirus Species 0.000 description 5
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 238000000134 MTT assay Methods 0.000 description 4
- 231100000002 MTT assay Toxicity 0.000 description 4
- 238000005481 NMR spectroscopy Methods 0.000 description 4
- 241000700159 Rattus Species 0.000 description 4
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 4
- 238000002835 absorbance Methods 0.000 description 4
- 239000002253 acid Substances 0.000 description 4
- 210000001124 body fluid Anatomy 0.000 description 4
- 239000010839 body fluid Substances 0.000 description 4
- 238000004113 cell culture Methods 0.000 description 4
- 230000003915 cell function Effects 0.000 description 4
- 230000010261 cell growth Effects 0.000 description 4
- 238000006243 chemical reaction Methods 0.000 description 4
- 239000003153 chemical reaction reagent Substances 0.000 description 4
- 239000013078 crystal Substances 0.000 description 4
- 238000013461 design Methods 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 238000009510 drug design Methods 0.000 description 4
- 239000012737 fresh medium Substances 0.000 description 4
- 238000001415 gene therapy Methods 0.000 description 4
- 238000002744 homologous recombination Methods 0.000 description 4
- 230000006801 homologous recombination Effects 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 230000005764 inhibitory process Effects 0.000 description 4
- 150000002484 inorganic compounds Chemical class 0.000 description 4
- 229910010272 inorganic material Inorganic materials 0.000 description 4
- 230000010354 integration Effects 0.000 description 4
- 238000011813 knockout mouse model Methods 0.000 description 4
- 238000002493 microarray Methods 0.000 description 4
- 210000000287 oocyte Anatomy 0.000 description 4
- 230000037361 pathway Effects 0.000 description 4
- 238000012340 reverse transcriptase PCR Methods 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- DAEPDZWVDSPTHF-UHFFFAOYSA-M sodium pyruvate Chemical compound [Na+].CC(=O)C([O-])=O DAEPDZWVDSPTHF-UHFFFAOYSA-M 0.000 description 4
- UCSJYZPVAKXKNQ-HZYVHMACSA-N streptomycin Chemical compound CN[C@H]1[C@H](O)[C@@H](O)[C@H](CO)O[C@H]1O[C@@H]1[C@](C=O)(O)[C@H](C)O[C@H]1O[C@@H]1[C@@H](NC(N)=N)[C@H](O)[C@@H](NC(N)=N)[C@H](O)[C@H]1O UCSJYZPVAKXKNQ-HZYVHMACSA-N 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- 239000013603 viral vector Substances 0.000 description 4
- JKMHFZQWWAIEOD-UHFFFAOYSA-N 2-[4-(2-hydroxyethyl)piperazin-1-yl]ethanesulfonic acid Chemical compound OCC[NH+]1CCN(CCS([O-])(=O)=O)CC1 JKMHFZQWWAIEOD-UHFFFAOYSA-N 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- 206010006187 Breast cancer Diseases 0.000 description 3
- 241000282472 Canis lupus familiaris Species 0.000 description 3
- 108010035532 Collagen Proteins 0.000 description 3
- 102000008186 Collagen Human genes 0.000 description 3
- 241000282326 Felis catus Species 0.000 description 3
- 102000016285 Guanine Nucleotide Exchange Factors Human genes 0.000 description 3
- 108010067218 Guanine Nucleotide Exchange Factors Proteins 0.000 description 3
- 239000007995 HEPES buffer Substances 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 102000015636 Oligopeptides Human genes 0.000 description 3
- 108010038807 Oligopeptides Proteins 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 3
- 102000004357 Transferases Human genes 0.000 description 3
- 108090000992 Transferases Proteins 0.000 description 3
- 230000002159 abnormal effect Effects 0.000 description 3
- 239000002671 adjuvant Substances 0.000 description 3
- 239000000556 agonist Substances 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 239000011324 bead Substances 0.000 description 3
- 230000033228 biological regulation Effects 0.000 description 3
- 230000003197 catalytic effect Effects 0.000 description 3
- 230000012292 cell migration Effects 0.000 description 3
- 239000003795 chemical substances by application Substances 0.000 description 3
- 238000010367 cloning Methods 0.000 description 3
- 229920001436 collagen Polymers 0.000 description 3
- 239000003184 complementary RNA Substances 0.000 description 3
- 230000009918 complex formation Effects 0.000 description 3
- 230000001351 cycling effect Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 229940079593 drug Drugs 0.000 description 3
- 230000002708 enhancing effect Effects 0.000 description 3
- 210000003527 eukaryotic cell Anatomy 0.000 description 3
- 239000000284 extract Substances 0.000 description 3
- 210000004602 germ cell Anatomy 0.000 description 3
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 3
- 210000004408 hybridoma Anatomy 0.000 description 3
- 230000007062 hydrolysis Effects 0.000 description 3
- 238000006460 hydrolysis reaction Methods 0.000 description 3
- 238000003018 immunoassay Methods 0.000 description 3
- 230000001965 increasing effect Effects 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 238000011835 investigation Methods 0.000 description 3
- 238000002955 isolation Methods 0.000 description 3
- 239000002502 liposome Substances 0.000 description 3
- 210000004962 mammalian cell Anatomy 0.000 description 3
- 239000003550 marker Substances 0.000 description 3
- 239000011159 matrix material Substances 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 230000007246 mechanism Effects 0.000 description 3
- 239000012528 membrane Substances 0.000 description 3
- 210000004379 membrane Anatomy 0.000 description 3
- 230000003278 mimic effect Effects 0.000 description 3
- 210000004940 nucleus Anatomy 0.000 description 3
- 238000011580 nude mouse model Methods 0.000 description 3
- 150000002894 organic compounds Chemical class 0.000 description 3
- 239000013612 plasmid Substances 0.000 description 3
- 229920000642 polymer Polymers 0.000 description 3
- 230000001023 pro-angiogenic effect Effects 0.000 description 3
- 230000000644 propagated effect Effects 0.000 description 3
- 230000004853 protein function Effects 0.000 description 3
- 238000003127 radioimmunoassay Methods 0.000 description 3
- 230000010076 replication Effects 0.000 description 3
- 238000011160 research Methods 0.000 description 3
- 230000004044 response Effects 0.000 description 3
- 230000019491 signal transduction Effects 0.000 description 3
- 229940124597 therapeutic agent Drugs 0.000 description 3
- 230000009261 transgenic effect Effects 0.000 description 3
- 241001430294 unidentified retrovirus Species 0.000 description 3
- FWBHETKCLVMNFS-UHFFFAOYSA-N 4',6-Diamino-2-phenylindol Chemical compound C1=CC(C(=N)N)=CC=C1C1=CC2=CC=C(C(N)=N)C=C2N1 FWBHETKCLVMNFS-UHFFFAOYSA-N 0.000 description 2
- 229920000936 Agarose Polymers 0.000 description 2
- 108020005544 Antisense RNA Proteins 0.000 description 2
- 239000004475 Arginine Substances 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 108090001008 Avidin Proteins 0.000 description 2
- 208000026310 Breast neoplasm Diseases 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 241000282693 Cercopithecidae Species 0.000 description 2
- 108020004635 Complementary DNA Proteins 0.000 description 2
- 102100028473 DNA-directed RNA polymerases I, II, and III subunit RPABC4 Human genes 0.000 description 2
- 102000016911 Deoxyribonucleases Human genes 0.000 description 2
- 108010053770 Deoxyribonucleases Proteins 0.000 description 2
- 239000006144 Dulbecco’s modified Eagle's medium Substances 0.000 description 2
- 239000006145 Eagle's minimal essential medium Substances 0.000 description 2
- 241000283086 Equidae Species 0.000 description 2
- 108700039887 Essential Genes Proteins 0.000 description 2
- 102000010834 Extracellular Matrix Proteins Human genes 0.000 description 2
- 108010037362 Extracellular Matrix Proteins Proteins 0.000 description 2
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 2
- 102000013446 GTP Phosphohydrolases Human genes 0.000 description 2
- 108091006109 GTPases Proteins 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 102000016805 Guanine Nucleotide Dissociation Inhibitors Human genes 0.000 description 2
- 108010092964 Guanine Nucleotide Dissociation Inhibitors Proteins 0.000 description 2
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 2
- 101000723789 Homo sapiens DNA-directed RNA polymerases I, II, and III subunit RPABC4 Proteins 0.000 description 2
- 108060003951 Immunoglobulin Proteins 0.000 description 2
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 2
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 2
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 2
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 2
- 206010027476 Metastases Diseases 0.000 description 2
- 241001467552 Mycobacterium bovis BCG Species 0.000 description 2
- 206010029113 Neovascularisation Diseases 0.000 description 2
- 108091028043 Nucleic acid sequence Proteins 0.000 description 2
- 208000037273 Pathologic Processes Diseases 0.000 description 2
- 241001494479 Pecora Species 0.000 description 2
- 229930182555 Penicillin Natural products 0.000 description 2
- JGSARLDLIJGVTE-MBNYWOFBSA-N Penicillin G Chemical compound N([C@H]1[C@H]2SC([C@@H](N2C1=O)C(O)=O)(C)C)C(=O)CC1=CC=CC=C1 JGSARLDLIJGVTE-MBNYWOFBSA-N 0.000 description 2
- 239000002202 Polyethylene glycol Substances 0.000 description 2
- 102000009572 RNA Polymerase II Human genes 0.000 description 2
- 108010009460 RNA Polymerase II Proteins 0.000 description 2
- 238000011530 RNeasy Mini Kit Methods 0.000 description 2
- -1 Rac Proteins 0.000 description 2
- 206010038997 Retroviral infections Diseases 0.000 description 2
- 229920002684 Sepharose Polymers 0.000 description 2
- 108091081024 Start codon Proteins 0.000 description 2
- 241000700618 Vaccinia virus Species 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 210000000577 adipose tissue Anatomy 0.000 description 2
- 239000011543 agarose gel Substances 0.000 description 2
- 230000003321 amplification Effects 0.000 description 2
- 238000000540 analysis of variance Methods 0.000 description 2
- 230000019552 anatomical structure morphogenesis Effects 0.000 description 2
- 238000000137 annealing Methods 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- ODKSFYDXXFIFQN-UHFFFAOYSA-N arginine Natural products OC(=O)C(N)CCCNC(N)=N ODKSFYDXXFIFQN-UHFFFAOYSA-N 0.000 description 2
- 229960000190 bacillus calmette–guérin vaccine Drugs 0.000 description 2
- 229960002685 biotin Drugs 0.000 description 2
- 235000020958 biotin Nutrition 0.000 description 2
- 239000011616 biotin Substances 0.000 description 2
- 208000024055 brain glioblastoma Diseases 0.000 description 2
- 201000011609 brain glioblastoma multiforme Diseases 0.000 description 2
- 210000000481 breast Anatomy 0.000 description 2
- 238000004422 calculation algorithm Methods 0.000 description 2
- 239000000969 carrier Substances 0.000 description 2
- 238000000423 cell based assay Methods 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 230000003399 chemotactic effect Effects 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 238000012875 competitive assay Methods 0.000 description 2
- 239000003636 conditioned culture medium Substances 0.000 description 2
- 230000008878 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 230000007850 degeneration Effects 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- 239000012895 dilution Substances 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 230000003511 endothelial effect Effects 0.000 description 2
- 230000002255 enzymatic effect Effects 0.000 description 2
- ZMMJGEGLRURXTF-UHFFFAOYSA-N ethidium bromide Chemical compound [Br-].C12=CC(N)=CC=C2C2=CC=C(N)C=C2[N+](CC)=C1C1=CC=CC=C1 ZMMJGEGLRURXTF-UHFFFAOYSA-N 0.000 description 2
- 229960005542 ethidium bromide Drugs 0.000 description 2
- 210000002744 extracellular matrix Anatomy 0.000 description 2
- 239000012909 foetal bovine serum Substances 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 230000004927 fusion Effects 0.000 description 2
- 102000034356 gene-regulatory proteins Human genes 0.000 description 2
- 108091006104 gene-regulatory proteins Proteins 0.000 description 2
- RWSXRVCMGQZWBV-WDSKDSINSA-N glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 2
- 230000013595 glycosylation Effects 0.000 description 2
- 238000006206 glycosylation reaction Methods 0.000 description 2
- PCHJSUWPFVWCPO-UHFFFAOYSA-N gold Chemical compound [Au] PCHJSUWPFVWCPO-UHFFFAOYSA-N 0.000 description 2
- 239000010931 gold Substances 0.000 description 2
- 229910052737 gold Inorganic materials 0.000 description 2
- 102000018358 immunoglobulin Human genes 0.000 description 2
- 238000012405 in silico analysis Methods 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 150000002611 lead compounds Chemical class 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 238000013507 mapping Methods 0.000 description 2
- 239000000463 material Substances 0.000 description 2
- 230000009401 metastasis Effects 0.000 description 2
- 230000001617 migratory effect Effects 0.000 description 2
- 239000013642 negative control Substances 0.000 description 2
- 238000010899 nucleation Methods 0.000 description 2
- 238000003199 nucleic acid amplification method Methods 0.000 description 2
- 230000009054 pathological process Effects 0.000 description 2
- 229940049954 penicillin Drugs 0.000 description 2
- 239000000546 pharmaceutical excipient Substances 0.000 description 2
- 229920001223 polyethylene glycol Polymers 0.000 description 2
- 239000011148 porous material Substances 0.000 description 2
- 230000001323 posttranslational effect Effects 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 238000012545 processing Methods 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- 230000009467 reduction Effects 0.000 description 2
- 108700038288 rhodamine-phalloidin Proteins 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 238000007423 screening assay Methods 0.000 description 2
- 238000012163 sequencing technique Methods 0.000 description 2
- 102000030938 small GTPase Human genes 0.000 description 2
- 108060007624 small GTPase Proteins 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000001509 sodium citrate Substances 0.000 description 2
- 229940054269 sodium pyruvate Drugs 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 230000009870 specific binding Effects 0.000 description 2
- 239000003381 stabilizer Substances 0.000 description 2
- 238000010186 staining Methods 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 238000007619 statistical method Methods 0.000 description 2
- 210000000130 stem cell Anatomy 0.000 description 2
- 230000004936 stimulating effect Effects 0.000 description 2
- 229960005322 streptomycin Drugs 0.000 description 2
- 238000013337 sub-cultivation Methods 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- HRXKRNGNAMMEHJ-UHFFFAOYSA-K trisodium citrate Chemical compound [Na+].[Na+].[Na+].[O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O HRXKRNGNAMMEHJ-UHFFFAOYSA-K 0.000 description 2
- 229940038773 trisodium citrate Drugs 0.000 description 2
- 210000003606 umbilical vein Anatomy 0.000 description 2
- 230000002792 vascular Effects 0.000 description 2
- 238000005406 washing Methods 0.000 description 2
- 238000002424 x-ray crystallography Methods 0.000 description 2
- WZUVPPKBWHMQCE-XJKSGUPXSA-N (+)-haematoxylin Chemical compound C12=CC(O)=C(O)C=C2C[C@]2(O)[C@H]1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-XJKSGUPXSA-N 0.000 description 1
- ASWBNKHCZGQVJV-UHFFFAOYSA-N (3-hexadecanoyloxy-2-hydroxypropyl) 2-(trimethylazaniumyl)ethyl phosphate Chemical compound CCCCCCCCCCCCCCCC(=O)OCC(O)COP([O-])(=O)OCC[N+](C)(C)C ASWBNKHCZGQVJV-UHFFFAOYSA-N 0.000 description 1
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 1
- CXNVOWPRHWWCQR-UHFFFAOYSA-N 4-Chloro-ortho-toluidine Chemical compound CC1=CC(Cl)=CC=C1N CXNVOWPRHWWCQR-UHFFFAOYSA-N 0.000 description 1
- 101150096316 5 gene Proteins 0.000 description 1
- 229920001817 Agar Polymers 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 108700028369 Alleles Proteins 0.000 description 1
- 108020000948 Antisense Oligonucleotides Proteins 0.000 description 1
- 108010002913 Asialoglycoproteins Proteins 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 1
- 241000972773 Aulopiformes Species 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 241000282817 Bovidae Species 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 108090000994 Catalytic RNA Proteins 0.000 description 1
- 102000053642 Catalytic RNA Human genes 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- KRKNYBCHXYNGOX-UHFFFAOYSA-K Citrate Chemical compound [O-]C(=O)CC(O)(CC([O-])=O)C([O-])=O KRKNYBCHXYNGOX-UHFFFAOYSA-K 0.000 description 1
- 108700010070 Codon Usage Proteins 0.000 description 1
- 108020004394 Complementary RNA Proteins 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- 241000186216 Corynebacterium Species 0.000 description 1
- 108010051219 Cre recombinase Proteins 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- FBPFZTCFMRRESA-KVTDHHQDSA-N D-Mannitol Chemical compound OC[C@@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-KVTDHHQDSA-N 0.000 description 1
- FBPFZTCFMRRESA-JGWLITMVSA-N D-glucitol Chemical compound OC[C@H](O)[C@@H](O)[C@H](O)[C@H](O)CO FBPFZTCFMRRESA-JGWLITMVSA-N 0.000 description 1
- WQZGKKKJIJFFOK-QTVWNMPRSA-N D-mannopyranose Chemical compound OC[C@H]1OC(O)[C@@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-QTVWNMPRSA-N 0.000 description 1
- 102000011724 DNA Repair Enzymes Human genes 0.000 description 1
- 108010076525 DNA Repair Enzymes Proteins 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108091027757 Deoxyribozyme Proteins 0.000 description 1
- 241000702421 Dependoparvovirus Species 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 239000004375 Dextrin Substances 0.000 description 1
- 229920001353 Dextrin Polymers 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 108010073385 Fibrin Proteins 0.000 description 1
- 102000009123 Fibrin Human genes 0.000 description 1
- BWGVNKXGVNDBDI-UHFFFAOYSA-N Fibrin monomer Chemical compound CNC(=O)CNC(=O)CN BWGVNKXGVNDBDI-UHFFFAOYSA-N 0.000 description 1
- 108010049003 Fibrinogen Proteins 0.000 description 1
- 102000008946 Fibrinogen Human genes 0.000 description 1
- 108010027920 GTPase-Activating Proteins Proteins 0.000 description 1
- 241000287828 Gallus gallus Species 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- WZUVPPKBWHMQCE-UHFFFAOYSA-N Haematoxylin Natural products C12=CC(O)=C(O)C=C2CC2(O)C1C1=CC=C(O)C(O)=C1OC2 WZUVPPKBWHMQCE-UHFFFAOYSA-N 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 101000703463 Homo sapiens Rho GTPase-activating protein 35 Proteins 0.000 description 1
- 101000666640 Homo sapiens Rho-related GTP-binding protein RhoJ Proteins 0.000 description 1
- 101000666657 Homo sapiens Rho-related GTP-binding protein RhoQ Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000026350 Inborn Genetic disease Diseases 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- ODKSFYDXXFIFQN-BYPYZUCNSA-P L-argininium(2+) Chemical compound NC(=[NH2+])NCCC[C@H]([NH3+])C(O)=O ODKSFYDXXFIFQN-BYPYZUCNSA-P 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- KDXKERNSBIXSRK-YFKPBYRVSA-N L-lysine Chemical compound NCCCC[C@H](N)C(O)=O KDXKERNSBIXSRK-YFKPBYRVSA-N 0.000 description 1
- 108091026898 Leader sequence (mRNA) Proteins 0.000 description 1
- 241000713666 Lentivirus Species 0.000 description 1
- 239000004472 Lysine Substances 0.000 description 1
- KDXKERNSBIXSRK-UHFFFAOYSA-N Lysine Natural products NCCCCC(N)C(O)=O KDXKERNSBIXSRK-UHFFFAOYSA-N 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 229930195725 Mannitol Natural products 0.000 description 1
- 108010085220 Multiprotein Complexes Proteins 0.000 description 1
- 102000007474 Multiprotein Complexes Human genes 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 101100412856 Mus musculus Rhod gene Proteins 0.000 description 1
- 102100023648 N-chimaerin Human genes 0.000 description 1
- 101710140152 N-chimaerin Proteins 0.000 description 1
- 108091061960 Naked DNA Proteins 0.000 description 1
- 238000012408 PCR amplification Methods 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 102000057297 Pepsin A Human genes 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 102100024616 Platelet endothelial cell adhesion molecule Human genes 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 108010076504 Protein Sorting Signals Proteins 0.000 description 1
- 239000012979 RPMI medium Substances 0.000 description 1
- 238000011529 RT qPCR Methods 0.000 description 1
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 1
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 1
- 102100030676 Rho GTPase-activating protein 35 Human genes 0.000 description 1
- 102100038337 Rho-related GTP-binding protein RhoJ Human genes 0.000 description 1
- 102100038339 Rho-related GTP-binding protein RhoQ Human genes 0.000 description 1
- 102000007562 Serum Albumin Human genes 0.000 description 1
- 108010071390 Serum Albumin Proteins 0.000 description 1
- 238000000692 Student's t-test Methods 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L Sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 238000011053 TCID50 method Methods 0.000 description 1
- 239000004098 Tetracycline Substances 0.000 description 1
- 101100242191 Tetraodon nigroviridis rho gene Proteins 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 108700019146 Transgenes Proteins 0.000 description 1
- 238000002441 X-ray diffraction Methods 0.000 description 1
- LWZFANDGMFTDAV-BURFUSLBSA-N [(2r)-2-[(2r,3r,4s)-3,4-dihydroxyoxolan-2-yl]-2-hydroxyethyl] dodecanoate Chemical compound CCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O LWZFANDGMFTDAV-BURFUSLBSA-N 0.000 description 1
- 238000000367 ab initio method Methods 0.000 description 1
- 238000002679 ablation Methods 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 239000013543 active substance Substances 0.000 description 1
- 230000010933 acylation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 230000002411 adverse Effects 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 239000008272 agar Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 239000002168 alkylating agent Substances 0.000 description 1
- 238000003016 alphascreen Methods 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- WNROFYMDJYEPJX-UHFFFAOYSA-K aluminium hydroxide Chemical compound [OH-].[OH-].[OH-].[Al+3] WNROFYMDJYEPJX-UHFFFAOYSA-K 0.000 description 1
- 229910021502 aluminium hydroxide Inorganic materials 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 230000001772 anti-angiogenic effect Effects 0.000 description 1
- 230000001028 anti-proliverative effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 230000000890 antigenic effect Effects 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 239000000074 antisense oligonucleotide Substances 0.000 description 1
- 238000012230 antisense oligonucleotides Methods 0.000 description 1
- 230000006907 apoptotic process Effects 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 229960001230 asparagine Drugs 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 238000011717 athymic nude mouse Methods 0.000 description 1
- 238000011888 autopsy Methods 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 210000003719 b-lymphocyte Anatomy 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000004204 blood vessel Anatomy 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 201000008274 breast adenocarcinoma Diseases 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 210000001043 capillary endothelial cell Anatomy 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 230000033077 cellular process Effects 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- IRPXADUBAQAOKL-UHFFFAOYSA-N chembl1408927 Chemical compound C1=CC=C2C(N=NC3=C4C=CC(=CC4=CC(=C3O)S(O)(=O)=O)S(O)(=O)=O)=CC=C(S(O)(=O)=O)C2=C1 IRPXADUBAQAOKL-UHFFFAOYSA-N 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 210000003711 chorioallantoic membrane Anatomy 0.000 description 1
- 239000003593 chromogenic compound Substances 0.000 description 1
- 210000000349 chromosome Anatomy 0.000 description 1
- 229940121657 clinical drug Drugs 0.000 description 1
- 239000000512 collagen gel Substances 0.000 description 1
- 238000004737 colorimetric analysis Methods 0.000 description 1
- 230000009137 competitive binding Effects 0.000 description 1
- 239000013068 control sample Substances 0.000 description 1
- 208000029078 coronary artery disease Diseases 0.000 description 1
- 230000002596 correlated effect Effects 0.000 description 1
- 230000001054 cortical effect Effects 0.000 description 1
- 238000012258 culturing Methods 0.000 description 1
- 210000004292 cytoskeleton Anatomy 0.000 description 1
- 239000002254 cytotoxic agent Substances 0.000 description 1
- 229940127089 cytotoxic agent Drugs 0.000 description 1
- 231100000599 cytotoxic agent Toxicity 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000001419 dependent effect Effects 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 235000019425 dextrin Nutrition 0.000 description 1
- 238000002050 diffraction method Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 150000002016 disaccharides Chemical class 0.000 description 1
- 238000002224 dissection Methods 0.000 description 1
- 238000009509 drug development Methods 0.000 description 1
- 238000007876 drug discovery Methods 0.000 description 1
- 239000003596 drug target Substances 0.000 description 1
- 238000001493 electron microscopy Methods 0.000 description 1
- 230000013020 embryo development Effects 0.000 description 1
- 231100001129 embryonic lethality Toxicity 0.000 description 1
- 210000001671 embryonic stem cell Anatomy 0.000 description 1
- 230000012202 endocytosis Effects 0.000 description 1
- 210000003038 endothelium Anatomy 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000005284 excitation Effects 0.000 description 1
- 229950003499 fibrin Drugs 0.000 description 1
- 229940012952 fibrinogen Drugs 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 239000012530 fluid Substances 0.000 description 1
- 238000005558 fluorometry Methods 0.000 description 1
- 230000005714 functional activity Effects 0.000 description 1
- 238000010230 functional analysis Methods 0.000 description 1
- 238000002825 functional assay Methods 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 238000003167 genetic complementation Methods 0.000 description 1
- 208000016361 genetic disease Diseases 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 229960003180 glutathione Drugs 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 238000003306 harvesting Methods 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 210000002443 helper t lymphocyte Anatomy 0.000 description 1
- 210000003494 hepatocyte Anatomy 0.000 description 1
- 210000000688 human artificial chromosome Anatomy 0.000 description 1
- 229920001477 hydrophilic polymer Polymers 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 238000010166 immunofluorescence Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 229940072221 immunoglobulins Drugs 0.000 description 1
- 238000001114 immunoprecipitation Methods 0.000 description 1
- 239000003547 immunosorbent Substances 0.000 description 1
- 238000000126 in silico method Methods 0.000 description 1
- 238000000099 in vitro assay Methods 0.000 description 1
- 238000010874 in vitro model Methods 0.000 description 1
- 238000005462 in vivo assay Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 230000006698 induction Effects 0.000 description 1
- 238000001802 infusion Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 102000006495 integrins Human genes 0.000 description 1
- 108010044426 integrins Proteins 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 230000000302 ischemic effect Effects 0.000 description 1
- 238000012917 library technology Methods 0.000 description 1
- 239000003446 ligand Substances 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 210000005265 lung cell Anatomy 0.000 description 1
- 210000004698 lymphocyte Anatomy 0.000 description 1
- 210000003712 lysosome Anatomy 0.000 description 1
- 230000001868 lysosomic effect Effects 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 239000000594 mannitol Substances 0.000 description 1
- 235000010355 mannitol Nutrition 0.000 description 1
- 230000013011 mating Effects 0.000 description 1
- 102000006240 membrane receptors Human genes 0.000 description 1
- 230000008172 membrane trafficking Effects 0.000 description 1
- 230000011987 methylation Effects 0.000 description 1
- 238000007069 methylation reaction Methods 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 210000003632 microfilament Anatomy 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 238000003032 molecular docking Methods 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- 238000012544 monitoring process Methods 0.000 description 1
- 150000002772 monosaccharides Chemical class 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 229930014626 natural product Natural products 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 239000002736 nonionic surfactant Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 102000006271 p21-Activated Kinases Human genes 0.000 description 1
- 108010058266 p21-Activated Kinases Proteins 0.000 description 1
- 239000013610 patient sample Substances 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 238000010647 peptide synthesis reaction Methods 0.000 description 1
- 239000000816 peptidomimetic Substances 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000000704 physical effect Effects 0.000 description 1
- 230000035790 physiological processes and functions Effects 0.000 description 1
- 238000007747 plating Methods 0.000 description 1
- 229920001983 poloxamer Polymers 0.000 description 1
- 229920000729 poly(L-lysine) polymer Polymers 0.000 description 1
- 102000054765 polymorphisms of proteins Human genes 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 239000001267 polyvinylpyrrolidone Substances 0.000 description 1
- 229920000036 polyvinylpyrrolidone Polymers 0.000 description 1
- 235000013855 polyvinylpyrrolidone Nutrition 0.000 description 1
- 238000010149 post-hoc-test Methods 0.000 description 1
- 238000002360 preparation method Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000004952 protein activity Effects 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000012846 protein folding Effects 0.000 description 1
- 230000018883 protein targeting Effects 0.000 description 1
- 230000006337 proteolytic cleavage Effects 0.000 description 1
- 210000001938 protoplast Anatomy 0.000 description 1
- 230000007420 reactivation Effects 0.000 description 1
- 238000012755 real-time RT-PCR analysis Methods 0.000 description 1
- 102000005962 receptors Human genes 0.000 description 1
- 108020003175 receptors Proteins 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000003362 replicative effect Effects 0.000 description 1
- 108091008146 restriction endonucleases Proteins 0.000 description 1
- 230000000717 retained effect Effects 0.000 description 1
- 238000010839 reverse transcription Methods 0.000 description 1
- 108091092562 ribozyme Proteins 0.000 description 1
- 235000019515 salmon Nutrition 0.000 description 1
- 230000003248 secreting effect Effects 0.000 description 1
- 230000028327 secretion Effects 0.000 description 1
- 238000013207 serial dilution Methods 0.000 description 1
- 239000013605 shuttle vector Substances 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 238000002741 site-directed mutagenesis Methods 0.000 description 1
- 150000003384 small molecules Chemical class 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000007790 solid phase Substances 0.000 description 1
- 239000000243 solution Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 235000011067 sorbitan monolaureate Nutrition 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 230000000638 stimulation Effects 0.000 description 1
- 238000012916 structural analysis Methods 0.000 description 1
- 125000002730 succinyl group Chemical group C(CCC(=O)*)(=O)* 0.000 description 1
- 150000005846 sugar alcohols Chemical class 0.000 description 1
- 229910021653 sulphate ion Inorganic materials 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 208000024891 symptom Diseases 0.000 description 1
- 238000003786 synthesis reaction Methods 0.000 description 1
- 238000012353 t test Methods 0.000 description 1
- 229960002180 tetracycline Drugs 0.000 description 1
- 229930101283 tetracycline Natural products 0.000 description 1
- 235000019364 tetracycline Nutrition 0.000 description 1
- 150000003522 tetracyclines Chemical class 0.000 description 1
- 229940126585 therapeutic drug Drugs 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 238000013518 transcription Methods 0.000 description 1
- 230000035897 transcription Effects 0.000 description 1
- 230000002103 transcriptional effect Effects 0.000 description 1
- 230000002463 transducing effect Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- QORWJWZARLRLPR-UHFFFAOYSA-H tricalcium bis(phosphate) Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 210000003934 vacuole Anatomy 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 230000029663 wound healing Effects 0.000 description 1
Landscapes
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Description
WO 2006/015426 PCT/AU2005/001188 COMPOSITIONS AND METHODS FOR ANGIOGENESIS-RELATED MOLECULES AND TREATMENTS Technical Field 5 The present invention relates to isolated nucleic acid molecules and their encoded polypeptides that are involved in the process of angiogenesis. In view of their involvement in angiogenesis, the invention is also concerned with the therapy of angiogenesis-related 10 disorders, the screening of compounds for pro- and anti-angiogenic activity, and the diagnosis and prognosis of angiogenesis-related disorders. The invention is also concerned with siRNA molecules targeted to the nucleic acid molecules of the invention and their use for 15 therapeutic application in the treatment of angiogenesis related disorders. Background Art It will be clearly understood that, although a number 20 of prior art publications are referred to herein to describe background information, this reference does not constitute an admission that any of these documents forms part of the common general knowledge in the art, in Australia or in any other country. The discussion of the 25 references states what their authors assert, and the applicants reserve the right to challenge the accuracy and pertinency of the cited documents. Angiogenesis, the formation of new blood vessels from pre-existing vessels, plays a critical role in many 30 physiological and pathologic processes including embryogenesis, wound healing, tumour growth and metastasis (Augustin, 1998; Hanahan, 1997). Thus, the angiogenic process is considered an excellent target for therapeutic intervention. Identification of key regulatory molecules 35 has principally used in vitro models in which endothelial cells (EC) are cultured on extracellular matrix (ECM) components such.as collagen, fibrinogen or fibronectin, WO 2006/015426 PCT/AU2005/001188 -2 with identification of targets that are involved in events such as migration or proliferation which are elements of angiogenesis but are not specific for it. However, one problem in these investigations is the fact that the 5 assays are generally performed on a flat or two dimensional (2D) environment, whereas EC morphogenesis to form capillary tubes requires a 3D matrix, allowing the establishment of important polarity cues. The use of such 3D assays, which include matrices of collagen type 1, 10 fibrin, or Matrigel, recapitulates many of the events in angiogenesis, and has allowed dissection of the cellular and molecular events in angiogenesis (Gamble et al., 1993; 1999; Bayless and Davis, 2002; Meyer et al., 1997). Data using these assays to define genes altered during 15 angiogenesis has supported the ideas firstly that there are fundamental differences between cells responding on 3D versus 2D matrices, and secondly that genes specific for angiogenesis might exist. The mammalian Rho family of small GTPases has been 20 implicated in diverse cellular functions, including reorganisation of the actin cytoskeleton, cell growth control, transcription regulation and membrane trafficking (Van Aelst and D'Souza-Schorey, 1997). The Rho family of small GTPases consists of at least 20 members: Rho 25 (A,B,C), Rac (1,2,3), Cdc42, TC10, TCL, Chp (1,2), RhoG, Rnd (1,2,3), RhoBTB (1,2), RhoD, Rif and TTF (Etienne Manneville and Hall, 2002). Like other members of the Ras superfamily, Rho proteins act as molecular switches to control cellular processes by cycling between active GTP 30 bound and inactive GDP-bound states. Regulation of these GTPases occurs via three major classes of regulatory proteins. The guanine nucleotide exchange factors (GEF) regulate activation through GDP-GTP exchange, GTPase activating proteins (GAPs), which promote hydrolysis of 35 the GTP to GDP-bound form, since the Rho proteins themselves display little if any basal GTPase activity and guanine nucleotide dissociation inhibitors (GDIs) which WO 2006/015426 PCT/AU2005/001188 -3 stabilise the inactive GDP-bound form of the protein (Mackay and Hall, 1998). At least 134 of these regulatory proteins have now been defined (Etienne-Manneville and Hall, 2002). 5 The function of the Rho family in endothelial morphogenesis is only now being elucidated, and it appears that different Rho family members play specific roles. Rho and Rac are important for regulation of permeability and cell migration (Wojciak-Stothard et al., 2001; Nobes and 10 Hall, 1999). Rho is also important for EC attachment and apoptosis (Chrzanowska-Wodnicka and Burridge, 1996; Hippenstiel et al., 2002), while Cdc42 and Racl are implicated in vacuole and subsequent lumen formation (Bayless and Davis, 2002). Given the limited number of 15 RhoGTPases and the seeming over-abundance of RhoGAPs, it is likely that the RhoGAPs may partly provide the specificity in control of function of the RhoGTPases. Summary of the Invention 20 A novel RhoGAP called BNO69 which is essential for angiogenesis has now been cloned and characterised as described in our co-pending International Application No. PCT/AU02/01282 the contents of which are incorporated herein by reference, and novel therapies for inhibition of 25 BN069 have been devised. Therapies which inhibit the expanding vasculature are desirable for the treatment of angiogenesis-related disorders which result in uncontrolled or enhanced angiogenesis, or a disorder in which a decreased 30 vasculature is of benefit. These include, but are not limited to, cancer; inflammatory disorders including arthritis; corneal, retinal or choroidal neovascularization including macular degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases. 35 In the present instance this involves reduction of the expression and activity of the BN069 gene product. Inhibiting the function of a gene or protein can be - 4 achieved in a variety of ways. Antisense nucleic acid methodologies generally represent one approach to inactivation of genes whose altered expression is causative of a disorder. In particular, RNA interference 5 using short interfering RNA (siRNA) molecules. As will be understood by the person skilled in the art, an siRNA is a short sequence of RNA which is the complement of a segment of a transcribed RNA and consequently binds thereto, and, in so doing, modulates or silences expression of the gene 10 in question. Accordingly, in a first aspect of the invention there is provided an isolated or synthetic short interfering RNA (siRNA) molecule comprising a complement of a segment of the mRNA transcribed from the BNO69 gene, wherein said 15 siRNA molecule modulates the expression of BNO69 by RNA interference and said siRNA molecule comprises SEQ ID NO:3. As will be appreciated by the person skilled in the art, a siRNA molecule used to modulate or silence the 20 expression of BNO69 may be in the form of single-stranded antisense, double-stranded antisense or double-stranded antisense with chemical modifications. The siRNA molecule comprises the following sequence: 5'-GUAGUCGUGCCACCAGUAG-3' (SEQ ID NO: 3) 25 In a further embodiment, a siRNA molecule of the 2579748_1 (GMafters) - 5 invention is incorporated into a short hairpin RNA (shRNA) molecule. In a preferred embodiment, the shRNA molecule comprises a nucleotide sequence corresponding to the siRNA molecule, followed by a generic nucleotide linker 5 sequence, typically a 9 nucleotide sequence (advantageously with the sequence TTCAAGAGA as represented by SEQ ID NO: 5), followed by the reverse complement of the nucleotide sequence corresponding to the siRNA molecule. Upon integration in the host cell genome the 10 nucleotide sequence corresponding to the siRNA molecule forms a double stranded structure by annealing to its reverse complement. The generic sequence forms a loop at one end of the double stranded molecule. In an embodiment the shRNA has the following 15 sequence: 5'- GATCCCCGTAGTCGTGCCACCAGTAGTTCAAGAGACTACTGGTGGCACGACTA CTTTTTGGAAA-3' (SEQ ID NO: 6) The siRNA, shRNA molecules or nucleic acid molecules of the present invention may be used to modulate the 20 expression of BNO69, or may be administered to a subject to treat or prevent an angiogenesis-related disorder, or a disorder in which a decreased vasculature is of benefit. In a further aspect there is provided a method of treating an angiogenesis-related disorder, comprising 25 administering one or more of a siRNA molecule comprising a complement of a segment of the mRNA transcribed from BN069 or a shRNA molecule targeted to BN069 2548012_1 (GHMatters) - 6 as described above, to a subject. In a further aspect of the invention there is provided the use of one or more of a siRNA molecule comprising a complement of a segment of the mRNA 5 transcribed from BNO69 or a shRNA molecule targeted to BNO69, as described above, in the manufacture of a medicament for the treatment of an angiogenesis-related disorder, or a disorder in which a decreased vasculature is of benefit. 10 The siRNA molecules, shRNA molecules, or nucleic acid molecules of the invention may be cloned into a vector. Vector systems may be plasmid, cosmid or viral in origin, as would be appreciated by the person skilled in the art. Many methods for introducing vectors into cells 15 or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for autologous transplant back into that same patient. Delivery by transfection, by 20 liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (See for example Goldman et al., 1997). In a further aspect there is provided a method of treating an angiogenesis-related disorder, or a disorder 25 in which a decreased vasculature is of benefit, comprising administering a vector comprising either a siRNA molecule, 2548012_1 (GHMatters) WO 2006/015426 PCT/AU2005/001188 -7 shRNA molecule, or nucleic acid molecule of the invention, to a subject. In a further aspect of the invention there is provided the use of a vector comprising a siRNA molecule, 5 a shRNA, or a nucleic acid molecule of the invention in the manufacture of a medicament for the treatment of an angiogenesis-related disorder, or a disorder in which a decreased vasculature is of benefit. Any of the siRNA molecules, shRNA molecules, nucleic 10 acid molecules, or vectors comprising such molecules form a part of the present invention, as do pharmaceutical compositions containing these and a pharmaceutically acceptable carrier. Any of the therapeutic methods described above may be 15 applied to any subject, including, for example, a mammal. The mammal may be a human, or may be a domestic, companion or zoo animal. While it is particularly contemplated that the pharmaceutical compositions of the invention are suitable for use in medical treatment of humans, they are 20 also applicable to veterinary treatment, including treatment of companion animals such as dogs and cats, and domestic animals such as horses, cattle and sheep, or zoo animals such as non-human primates, felids, canids, bovids, and ungulates. 25 Isoforms of the BN069 gene have been identified'. The siRNA or shRNA molecules of the invention may be targeted to any one or more of these isoforms. Specifically, any one or more of isoform I, II or III may be targeted. The nucleotide and amino acid sequence of isoform I is 30 represented by SEQ ID Numbers: 8 and 9 respectively. Isoform II is the molecule identified as BNO69 in PCT/AUO2/01282 and its nucleotide and amino acid sequences are represented by SEQ ID Numbers: 10 and 11 respectively. The nucleotide and amino acid sequence of isoform III is 35 represented by SEQ ID Numbers: 12 and 13 respectively. The BNO69 isoforms share a common region of sequence identity, the nucleotide and amino acid sequence of which is - 8 represented by SEQ ID Numbers: 14 and 15 respectively. This region of identity includes a GAP domain, the nucleotide and amino acid sequence of which is represented by SEQ ID NO: 16 and 17 respectively. The siRNA or shRNA 5 molecules of the invention may target the common region shared by all isoforms of BN069, including the GAP domain, or may bind specifically to one isoform alone. Isolated nucleic acid molecules comprising the sequence set forth in one of SEQ ID Numbers: 8, 12, 14 or 10 16, or fragments thereof, that play a role in diseases associated with the angiogenic process are disclosed. Such diseases include, but are not restricted to, cancer; inflammatory disorders including arthritis; corneal, retinal or choroidal neovascularization including macular 15 degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases. Useful fragments may include those which are unique and which do not overlap any previously identified genes, unique fragments which do overlap with a known sequence, and fragments which span 20 alternative splice junctions etc. Isolated nucleic acid molecules may be at least 70% identical to any one of SEQ ID Numbers: 8, 12, 14 or 16 and which encodes a polypeptide that plays a role in an angiogenic process. Such variants will have preferably at 25 least about 85%, and 2548012_1 (GHMatters) - 9 most preferably at least about 95% sequence identity to these sequences. Sequence identity is typically calculated using the BLAST algorithm, described in Altschul et al (1997) with 5 the BLOSUM62 default matrix. Also encompassed are isolated nucleic acid molecules which hybridize under stringent conditions with any one of SEQ ID Numbers: 8, 12, 14 or 16, and which plays a role in an angiogenic process. 10 Hybridization with PCR probes is contemplated. The specificity of the probe, whether it is made from a highly specific region, e.g., the 5' regulatory region, or from a less specific region, e.g., a conserved motif such as the GAP domain, and the stringency of the hybridization or 15 amplification will determine whether the probe identifies only naturally occurring sequences, allelic variants, or related sequences. Probes used for the detection of related sequences, should preferably have at least 50% sequence identity to 20 any of SEQ ID Numbers: 8, 12, 14 or 16. The hybridization probes of the present invention may be DNA or RNA. Means for producing specific hybridization probes for any of SEQ ID Numbers: 8, 12, 14 or 16 include the cloning of these sequences into vectors for the production of mRNA 25 probes. Such vectors are known in the art, and are commercially available. Hybridization probes may be labelled by radionuclides such as 32 P or 3S, or by enzymatic labels, such as alkaline phosphatase coupled to the probe via avidin/biotin coupling systems, or other 30 methods known in the art. Under stringent conditions, hybridization with 32 P labelled probes will most preferably occur at 42 0 C in 750 mM NaCl, 75 mM trisodium citrate, 2% SDS, 50% formamide, 1X Denhart's, 10% (w/v) dextran sulphate and 100 pg/ml 35 denatured salmon sperm DNA. Useful variations on these conditions will be readily apparent to those skilled in the art. The washing steps which follow hybridization most 2548012_1 (GHMatters) WO 2006/015426 PCT/AU2005/001188 - 10 preferably occur at 65'C in 15 mM NaCl, 1.5 mM trisodium citrate, and 1% SDS. Additional variations on these conditions will be readily apparent to those skilled in the art. 5 The nucleic acid molecules, or fragments thereof, of the present invention have a nucleotide sequence obtainable from a natural source. They therefore include naturally occurring normal, naturally occurring mutant, naturally occurring polymorphic alleles, differentially 10 spliced transcripts, splice variants etc. Natural sources include animal cells and tissues, body fluids, tissue culture cells etc. The nucleic acid molecules of the present invention can also be engineered using methods accepted in the art 15 so as to alter the gene-encoding sequences for a variety of purposes. These include, but are not limited to, modification of the cloning, processing, and/or expression of the gene product. PCR reassembly of gene fragments and the use of synthetic oligonucleotides allow the 20 engineering of the nucleic acid molecules of the present invention. For example, oligonucleotide-mediated site-directed mutagenesis can introduce mutations that create new restriction sites, alter glycosylation patterns and produce splice variants etc. 25 As a result of the degeneracy of the genetic code, a number of nucleic acid sequences representing the nucleic acid molecules of the present invention, some that may have minimal similarity to the naturally occurring sequence, may be produced. Thus, the invention includes 30 each and every possible variation of polynucleotide sequence that could be made by selecting combinations based on possible codon choices. These combinations are made in accordance with the standard triplet genetic code as applied to the polynucleotide sequence of the naturally 35 nucleic acid molecule, and all such variations are to be considered as being specifically disclosed. The nucleic acid molecules of this invention are WO 2006/015426 PCT/AU2005/001188 - 11 typically DNA molecules, and include cDNA, genomic DNA, synthetic forms, and mixed polymers, both sense and antisense strands, and may be chemically or biochemically modified, or may contain non-natural or derivatised 5 nucleotide bases as will be appreciated by those skilled in the art. Such modifications include labels, methylation, intercalators, alkylators and modified linkages. In some instances it may be advantageous to produce nucleotide sequences that represent the nucleic 10 acid molecules of the present invention, which possess a substantially different codon usage than that of the naturally occurring molecule. For example, codons may be selected to increase the rate of expression of the encoded peptide in a particular prokaryotic or eukaryotic host 15 corresponding with the frequency that the host utilizes particular codons. Other reasons to alter the nucleotide sequence without altering the encoded amino acid sequence include the production of RNA transcripts having more desirable properties, such as a greater half-life, than 20 transcripts produced from the naturally occurring molecule. The invention also encompasses production of the nucleic acid molecules of the invention, entirely by synthetic chemistry. Synthetic sequences may be inserted 25 into expression vectors and cell systems that contain the necessary elements for transcriptional and translational control of the inserted coding sequence in a suitable host. These elements may include regulatory sequences, promoters, 5' and 3' untranslated regions and specific 30 initiation signals (such as an ATG initiation codon and Kozak consensus sequence) which allow more efficient translation of the nucleotide sequence. In cases where the complete coding sequence including its initiation codon and upstream regulatory sequences are inserted into the 35 appropriate expression vector, additional control signals may not be needed. However, in cases where only coding sequence, or a fragment thereof, is inserted, exogenous - 12 translational control signals as described above should be provided by the vector. Such signals may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers 5 appropriate for the particular host cell system used (Scharf et al., 1994). The present invention allows for the preparation of purified polypeptides or proteins. In order to do this, host cells may be transfected with a nucleic acid molecule 10 as described above. Typically, said host cells are transfected with an expression vector comprising a nucleic acid molecule according to the invention. A variety of expression vector/host systems may be utilized to contain and express the sequences. These include, but are not 15 limited to, microorganisms such as bacteria transformed with plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with viral expression vectors (e.g., baculovirus); or mouse or other animal or human tissue 20 cell systems. Mammalian cells can also be used to express a polypeptide that is encoded by a nucleic acid molecule of the invention using various expression vectors including plasmid, cosmid and viral systems such as a vaccinia virus expression system. The invention is not 25 limited by the host cell or vector employed. The nucleic acid molecules, or variants thereof, of the present invention can be stably expressed in cell lines to allow long term production of recombinant proteins in mammalian systems. The nucleic acid sequences 30 of the invention can be transformed into cell lines using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. The selectable marker confers resistance to a 2548012_1 (GHMatters) - 13 selective agent, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be propagated using tissue culture 5 techniques appropriate to the cell type. The protein produced by a transformed cell may be secreted or retained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing 10 polynucleotides which encode a protein may be designed to contain signal sequences which direct secretion of the protein through a prokaryotic or eukaryotic cell membrane. In addition, a host cell strain may be chosen for its ability to modulate expression of the inserted sequences 15 or to process the expressed protein in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, glycosylation, phosphorylation, and acylation. Post-translational cleavage of a "prepro" form of the protein may also be 20 used to specify protein targeting, folding, and/or activity. Different host cells having specific cellular machinery and characteristic mechanisms for post translational activities (e.g., CHO or HeLa cells), are available from the American Type Culture Collection (ATCC) 25 and may be chosen to ensure the correct modification and processing of the foreign protein. When large quantities of protein are needed such as for antibody production, vectors which direct high levels of expression may be used such as those containing the T5 2548012.1 (GHMatters) - 14 or T7 inducible bacteriophage promoter. The present invention also includes the use of the expression systems described above in generating and isolating fusion proteins which contain important functional domains of the 5 protein. These fusion proteins are used for binding, structural and functional studies as well as for the generation of appropriate antibodies. In order to express and purify the protein as a fusion protein, the appropriate polynucleotide sequences 10 of the present invention are inserted into a vector which contains a nucleotide sequence encoding another peptide (for example, glutathionine succinyl transferase). The fusion protein is expressed and recovered from prokaryotic or eukaryotic cells. The fusion protein can then be 15 purified by affinity chromatography based upon the fusion vector sequence and the relevant protein can subsequently be obtained by enzymatic cleavage of the fusion protein. Fragments of polypeptides of the present invention may also be produced by direct peptide synthesis using 20 solid-phase techniques. Automated synthesis may be achieved by using the ABI 431A Peptide Synthesizer (Perkin-Elmer). Various fragments of polypeptide may be synthesized separately and then combined to produce the full length molecule. 25 Isolated polypeptide set forth in one of SEQ ID Numbers: 9, 13, 15 or 17, or fragments thereof are provided, that play a role 2548012_1 (GHMatter) - 15 in diseases associated with the angiogenic process. Diseases may include, but are not restricted to, cancer; inflammatory disorders including arthritis; corneal, retinal or choroidal neovascularization including macular 5 degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases. Also encompassed are isolated polypeptides having at least 70%, preferably 85%, and more preferably 95%, identity to any one of SEQ ID Numbers: 9, 13, 15 or 17, 10 and which play a role in an angiogenic process. Sequence identity is typically calculated using the BLAST algorithm, described in Altschul et al (1997) with the BLOSUM62 default matrix. In a further aspect there is provided a method of 15 preparing a polypeptide as described above, comprising the steps of: (a) culturing cells as described above under conditions effective for production of the polypeptide; and 20 (b) harvesting the polypeptide. According to still another aspect there is provided a polypeptide which is the product of the process described above. Substantially purified protein or fragments thereof 25 can be used in further biochemical analyses to establish secondary and tertiary structure. Such methodology is known in the art and includes, but is not restricted to, X-ray crystallography of crystals of the proteins or by nuclear magnetic resonance (NMR). Determination of 30 structure allows for the rational design of pharmaceuticals to interact with the protein, alter protein charge configuration or charge interaction with other proteins, or to alter its function in the cell. 2548012_1 (GHMatters) - 16 As angiogenesis is critical in a number of pathological processes, the invention therefore also provides therapeutic methods for the treatment of angiogenesis-related disorders, and provides the diagnosis 5 or prognosis of angiogenesis-related disorders associated with abnormalities in expression and/or function of BN069. Examples of such disorders include, but are not limited to, cancer; inflammatory disorders including arthritis; corneal, retinal or choroidal 10 neovascularization including macular degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases. Therapeutic Applications In a further aspect, the invention provides a method 15 of treating an angiogenesis-related disorder as described above, comprising administering a siRNA, shRNA or a vector of the invention to a subject. For the treatment of angiogenesis-related disorders which result in uncontrolled or enhanced angiogenesis, 20 including but not limited to, cancer; inflammatory disorders including arthritis; corneal, retinal or choroidal neovascularization including macular degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases, therapies which inhibit the 25 expanding vasculature are desirable. This would involve inhibition of the nucleic acid molecules or polypeptides 2548012_1 (GHMattes) - 17 of the invention. For the treatment of angiogenesis-related disorders which are characterised by inhibited or decreased angiogenesis, including but not limited to, ischaemic limb 5 disease and coronary artery disease, therapies which enhance or promote vascular expansion are desirable. This would involve enhancement, stimulation or re-activation of the nucleic acid molecules or polypeptides of the invention. 10 Inhibiting gene or protein function Inhibiting the function of a gene or protein can be achieved in a variety of ways. As mentioned above, antisense nucleic acid methodologies represent one 15 approach to inactivate genes that are causative of a disorder. Antisense or gene-targeted silencing strategies may include, but are not limited to, the use of antisense oligonucleotides, injection of antisense RNA, transfection of antisense RNA expression vectors, and the use of RNA 20 interference (RNAi) or short interfering RNA (siRNA) as described above. Still further, catalytic nucleic acid molecules such as DNAzymes and ribozymes may be used for gene silencing (Breaker and Joyce, 1994; Haseloff and Gerlach, 1988). These molecules function by cleaving their 25 target mRNA molecule rather than merely binding to it as in traditional antisense approaches. An isolated nucleic acid molecule, which is the complement of any one of the nucleic acid molecules described above may be administered to a subject. 30 Typically, a complement is administered to a subject to treat or prevent an angiogenesis-related disorder. In a further aspect the complement is an RNA molecule that hybridizes with the mRNA encoded by a nucleic acid molecule of the invention, a short interfering RNA (siRNA) 35 that hybridizes with the mRNA encoded by a nucleic acid molecule of the invention, or a catalytic nucleic acid molecule that is targeted to a 2548012_1 (GHMafters) - 18 nucleic acid molecule of the invention. Typically, a vector expressing the complement may be administered to a subject to treat or prevent an angiogenesis-related disorder including, but not limited 5 to, those described above. Many methods for introducing vectors into cells or tissues are available and equally suitable for use in vivo, in vitro, and ex vivo. For ex vivo therapy, vectors may be introduced into stem cells taken from the patient and clonally propagated for 10 autologous transplant back into that same patient. Delivery by transfection, by liposome injections, or by polycationic amino polymers may be achieved using methods which are well known in the art. (For example, see Goldman et al., 1997). 15 In a further aspect purified protein according to the invention may be used to produce antibodies which specifically bind to a polypeptide of the invention. These antibodies may be used directly as an antagonist or indirectly as a targeting or delivery mechanism for 20 bringing a pharmaceutical agent (such as a cytotoxic agent) to cells or tissues that express the polypeptide. Such antibodies may include, but are not limited to, polyclonal, monoclonal, chimeric and single chain antibodies as would be understood by the person skilled in 25 the art. For the production of antibodies, various hosts including rabbits, rats, goats, mice, humans, and others may be immunized by injection with a polypeptide of the invention or with any fragment or oligopeptide thereof, 2548012_i (GHMatters) WO 2006/015426 PCT/AU2005/001188 - 19 which has immunogenic properties. Various adjuvants may be used to increase immunological response, and include, but are not limited to, Freund's adjuvant, mineral gels such as aluminium hydroxide, and surface-active substances such 5 as lysolecithin. Adjuvants used in humans include BCG (bacillus Calmette-Guerin) and Corynebacterium parvum. It is preferred that the oligopeptides, peptides, or fragments used to induce antibodies to a.polypeptide have an amino acid sequence consisting of at least about 5 10 amino acids, and, more preferably, of at least about 10 amino acids. It is also preferable that these oligopeptides, peptides, or fragments are identical to a portion of the amino acid sequence of the polypeptide and contain the entire amino acid sequence of a small, 15 naturally occurring molecule. Short stretches of amino acids from these proteins may be fused with those of another protein, such as KLH, and antibodies to the chimeric molecule may be produced. Monoclonal antibodies to a polypeptide of the 20 invention may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique. (For 25 example, see Kohler and Milstein, 1975; Kozbor et al., 1985; Cote et al., 1983; Cole et al., 1984). Monoclonal antibodies produced may include, but are not limited to, mouse-derived antibodies, humanised antibodies and fully-human antibodies. For example, 30 antibodies are obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge. In one example of this technique, elements of the human heavy and light chain loci are introduced into strains of mice derived from 35 embryonic stem cell lines that contain targeted disruptions of the endogenous heavy and light chain loci. These transgenic mice can synthesise human antibodies WO 2006/015426 PCT/AU2005/001188 - 20 specific for human antigens and can be used to produce human antibody-secreting hybridomas. Methods for obtaining human antibodies from transgenic mice are described for example in Lonberg et al., 1994; Green et al., 1994; 5 Taylor et al., 1994. Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed in the literature. (For 10 example, see Orlandi et al., 1989; Winter et al., 1991). Antibody fragments which contain specific binding sites for a polypeptide of the invention may also be generated. For example, such fragments include, F(ab')2 fragments produced by pepsin digestion of the antibody 15 molecule and Fab fragments generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity. (For example, see Huse et al., 20 1989). Various immunoassays may be used for screening to identify antibodies having the desired specificity. Numerous protocols fo.r competitive binding or immunoradiometric assays using either polyclonal or 25 monoclonal antibodies with established specificities are well known in the art. Such immunoassays typically involve the measurement of complex formation between a protein and its specific antibody. A two-site, monoclonal-based immunoassay utilizing monoclonal antibodies reactive to 30 two non-interfering epitopes is preferred, but a competitive binding assay may also be employed. In a further aspect, antagonists may include peptides, phosphopeptides or small organic or inorganic compounds. These antagonists should disrupt the function 35 of a nucleic acid molecule or polypeptide of the invention so as to provide the necessary therapeutic effect. Peptides, phosphopeptides or small organic or - 21 inorganic compounds suitable for therapeutic applications may be identified using nucleic acid molecules and polypeptides of the invention in drug screening applications as described below. 5 Enhancing gene or protein function Enhancing, stimulating or re-activating a gene's or protein's function can be achieved in a variety of ways. Typically, a nucleic acid molecule as described above can 10 be administered to a subject to treat or prevent an angiogenesis-related disorder. Typically, a vector capable of expressing a polypeptide of the invention, or a fragment or derivative thereof, may be administered to a subject to treat or 15 prevent a disorder including, but not limited to, those described above. Transducing retroviral vectors are often used for somatic cell gene therapy because of their high efficiency of infection and stable integration and expression. A nucleic acid molecule of the invention, or 20 portions thereof, can be cloned into a retroviral vector and expression may be driven from its endogenous promoter or from the retroviral long terminal repeat or from a promoter specific for the target cell type of interest. Other viral vectors can be used and include, as is known 25 in the art, adenoviruses, adeno-associated viruses, vaccinia viruses, papovaviruses, lentiviruses and retroviruses of avian, murine and human origin. Gene therapy may be carried out according to established methods (See for example Friedman, 1991; 30 Culver, 1996). A vector containing a nucleic acid molecule of the invention linked to expression control elements and 2548012_1 (GHMatters) - 22 capable of replicating inside the cells is prepared. Alternatively the vector may be replication deficient and may require helper cells for replication and use in gene therapy. 5 Gene transfer using non-viral methods of infection in vitro can also be used. These methods include direct injection of DNA, uptake of naked DNA in the presence of calcium phosphate, electroporation, protoplast fusion or liposome delivery. Gene transfer can also be achieved by 10 delivery as a part of a human artificial chromosome or receptor-mediated gene transfer. This involves linking the DNA to a targeting molecule that will bind to specific cell-surface receptors to induce endocytosis and transfer of the DNA into mammalian cells. One such technique uses 15 poly-L-lysine to link asialoglycoprotein to DNA. An adenovirus is also added to the complex to disrupt the lysosomes and thus allow the DNA to avoid degradation and move to the nucleus. Infusion of these particles intravenously has resulted in gene transfer into 20 hepatocytes. In a further aspect, a suitable agonist may also include peptides, phosphopeptides or small organic or inorganic compounds that can mimic the function of a polypeptide of the invention, or may include an antibody 25 specific for a polypeptide of the invention that is able to restore function to a normal level. Peptides, phosphopeptides or small organic or inorganic compounds suitable for therapeutic applications 2548012_1 (GHMatters) - 23 may be identified using nucleic acids and polypeptides of the invention in drug screening applications as described below. In further embodiments, any of the agonists, 5 antagonists, complementary sequences, siRNA molecules, shRNA molecules, nucleic acid molecules, polypeptides, antibodies, or vectors of the invention may be administered in combination with other appropriate pharmaceutical or therapeutic agents, or treatment 10 methods. Selection of the appropriate agents and treatment methods may be made by those skilled in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents and treatment methods may act synergistically to effect the treatment or prevention of 15 the various disorders described above. Using this approach, therapeutic efficacy with lower dosages of each agent may be possible, thus reducing the potential for adverse side effects. Any of the therapeutic methods described above may be 20 applied to any subject, including, for example, mammals such as dogs, cats, cows, horses, rabbits, monkeys, and most preferably, humans. Modulation of angiogenesis 25 Any of the methods described above used for the treatment of an angiogenesis-related disorder may be used for the modulation of angiogenesis in any system comprising cells. These systems may include but are not limited to, in vitro assay systems (e.g. Matrigel assays, 30 proliferation assays, migration assays, collagen assays, bovine capillary endothelial cell assay etc), in vivo assay systems (e.g. in vivo Matrigel-type assays, chicken chorioallantoic membrane assay, isolated organs, tissues or cells etc), animal models (e.g. in vivo 35 neovascularisation assays, tumour angiogenesis models etc) or hosts in need of treatment (e.g. hosts suffering from angiogenesis-related 25480121 (GHMatters) - 24 disorders as previously described). Drug screening Nucleic acid molecules as well as polypeptides, or 5 fragments thereof described herein, and cells expressing these, are useful for the screening of candidate pharmaceutical compounds in a variety of techniques for the treatment of angiogenesis-related disorders. Still further, it provides the use wherein high 10 throughput screening techniques are employed. Molecules that interact with the nucleic acid molecules and polypeptides of the invention have been identified. Still further, a mutant of BN069 in which the putative GAP activity of BN069 is eliminated has been 15 identified, specifically a mutant in which Arg82 of SEQ ID NO: 11 is replaced, more specifically in which Arg82 is replaced by Ala (hence an R82A mutation). Accordingly, in still another aspect of the invention, nucleic acid molecules encoding these as well as the polypeptides and 20 cells and animals expressing these are useful for the screening of candidate pharmaceutical compounds in a variety of techniques for the treatment of angiogenesis related disorders. Compounds that can be screened in accordance with the 25 invention include, but are not limited, to peptides (such as soluble peptides), phosphopeptides and small organic or inorganic molecules (such as natural product or synthetic chemical libraries and peptidomimetics). In one embodiment, a screening assay may include a 30 cell-based assay utilising eukaryotic or prokaryotic host cells that are stably transformed with recombinant nucleic acid molecules expressing the polypeptides, or fragments thereof, of the invention, in competitive binding assays. Binding assays will measure for the formation of complexes 35 between the polypeptide, or fragments thereof, and the 2548012_1 (GHMatters) WO 2006/015426 PCT/AU2005/001188 - 25 compound being tested, or will measure the degree to which a compound being tested will interfere with the formation of a complex between the polypeptide, or fragments thereof, and its interactor or ligand. 5 For example BNO69 is able to interact with multiple members of the Rho family in a cell-specific manner given that the effects of BN069 knockdown on the activity of Rho, Rac and Cdc42 differs dependent upon the cell type. BN069 knockdown increases Rho activity in endothelial 10 cells but leads to no change in active Rac or Cdc42. This is'in contrast to results in NIH3T3 cells where inhibition of BN069 expression leads to an increase in Rac activity. This suggests that the GAP domain of BNO69 is able to bind to multiple members of the Rho family in a cell-specific 15 manner. Accordingly the interactors of BN069 may be a RhoGTPase as in endothelial cells or could be a RacGTPase as in NIH3T3 cells. Furthermore, disruption of the pathway that BNO69 is a part of (through introduction of an siRNA according to the invention or manipulation to introduce a 20 mutant BN069 in which GAP activity is reduced or eliminated in accordance with the invention) will alter the expression of other proteins in the pathway which allows identification of further drug targets in the pathway. Thus the interactor for polypeptides of the 25 invention may be Rho, Rac, Cdc42 or other members of the Rho family, or other proteins in the pathway that BN069 is a part of. Non cell-based assays may also be used for identifying compounds that interrupt binding between the 30 polypeptides of the invention and their interactors. Such assays are known in the art and include for example AlphaScreen technology (PerkinElmer Life Sciences, MA, USA). This application relies on the use of beads such that each interaction partner is bound to a separate bead 35 via an antibody. Interaction of each partner will bring the beads into proximity, such that laser excitation initiates a number of chemical reactions ultimately - 26 leading to fluorophores emitting a light signal. Candidate compounds that disrupt the binding of the polypeptide with its interactor will result in loss of light emission enabling identification and isolation of the responsible 5 compound. High-throughput drug screening techniques may also employ methods as described in W084/03564. Small peptide test compounds synthesised on a solid substrate can be assayed through polypeptide binding and washing. The bound 10 polypeptide is then detected by methods well known in the art. In a variation of this technique, purified polypeptides can be coated directly onto plates to identify interacting test compounds. An additional method for drug screening involves the 15 use of host eukaryotic cell lines that carry mutations in a nucleic acid molecule of the invention. The host cell lines are also defective at the polypeptide level. Other cell lines may be used where the expression of the nucleic acid molecule of the invention can be regulated (i.e. 20 over-expressed, under-expressed, or switched off). The host cell lines or cells are grown in the presence of various drug compounds and the rate of growth of the host cells is measured to determine if the compound is capable of regulating the growth of defective cells. 25 The polypeptides described herein may also be used for screening compounds developed as a result of combinatorial library technology. This provides a way to test a large number of different substances for their ability to modulate activity of a polypeptide. A substance 30 identified as a modulator of polypeptide function may be peptide or non-peptide in nature. Non-peptide "small molecules" are often preferred for many in vivo pharmaceutical applications. In addition, a mimic or mimetic of the substance may be designed for 35 pharmaceutical use. The design of mimetics based on a known pharmaceutically active compound ("lead" compound) is a common approach to the development of novel 2548012_1 (GHMatters) WO 2006/015426 PCT/AU2005/001188 - 27 pharmaceuticals. This is often desirable where the original active compound is difficult or expensive to synthesise or where it provides an unsuitable method of administration. In the design of a mimetic, particular 5 parts of the original active compound that are important in determining the target property are identified. These parts or residues constituting the active region of the compound are known as its pharmacophore. Once found, the pharmacophore structure is modelled according to its 10 physical properties using data from a range of sources including x-ray diffraction data and NMR. A template molecule is then selected onto which chemical groups that mimic the pharmacophore can be added. The selection can be made such that the mimetic is easy to synthesise, is 15 likely to be pharmacologically acceptable, does not degrade in vivo and retains the biological activity of the lead compound. Further optimisation or modification can be carried out to select one or more final mimetics useful for in vivo or clinical testing. 20 It is also possible to isolate a target-specific antibody and then solve its crystal structure. In principle, this approach yields a pharmacophore upon which subsequent drug design can be based as described above. It may be possible to avoid protein crystallography 25 altogether by generating anti-idiotypic antibodies (anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analogue of the original binding site. The anti-id could then be 30 used to isolate peptides from chemically or biologically produced peptide banks. Another alternative method for drug screening relies on structure-based rational drug design. Determination of the three dimensional structure of the polypeptides of the 35 invention, or the three dimensional structure of the protein complexes which may incorporate these polypeptides - 28 allows for structure-based drug design to identify biologically active lead compounds. Three-dimensional structural models can be generated by a number of applications, some of which include 5 experimental models such as x-ray crystallography and NMR and/or from in silico studies using information from structural databases such as the Protein Databank (PDB). In addition, three dimensional structural models can be determined using a number of known protein structure 10 prediction techniques based on the primary sequences of the polypeptides (e.g. SYBYL - Tripos Associated, St. Louis, MO), de novo protein structure design programs (e.g. MODELER - MSI Inc., San Diego, CA, or MOE - Chemical Computing Group, Montreal, Canada) or ab initio methods 15 (e.g. see US Patent Numbers 5331573 and 5579250). Once the three dimensional structure of a polypeptide or polypeptide complex has been determined, structure based drug discovery techniques can be employed to design biologically active compounds based on these three 20 dimensional structures. Such techniques are known in the art and include examples such as DOCK (University of California, San Francisco) or AUTODOCK (Scripps Research Institute, La Jolla, California). A computational docking protocol will identify the active site or sites that are 25 deemed important for protein activity based on a predicted protein model. Molecular databases, such as the Available Chemicals Directory (ACD) are then screened for molecules that complement the protein model. Using methods such as these, potential clinical drug 30 candidates can be identified and computationally ranked in order to reduce the time and expense associated with typical 'wet lab' drug screening methodologies. 2548012_1 (GHMattler) WO 2006/015426 PCT/AU2005/001188 - 29 Pharmaceutical Preparations Compounds identified from screening assays as indicated above, as well as siRNA and shRNA molecules of the invention can be administered to a patient at a 5 therapeutically effective dose to treat or ameliorate a disorder associated with angiogenesis. A therapeutically effective dose refers to that amount of the compound, siRNA, or shRNA molecules sufficient to result in amelioration of symptoms of the disorder. 10 Toxicity and therapeutic efficacy of such compounds or molecules can be determined by standard pharmaceutical procedures in cell cultures or experimental animals. The data obtained from these studies can then be used in the formulation of a range of dosages for use in humans. 15 Pharmaceutical compositions for use in accordance with the present invention can be formulated in a conventional manner using one or more physiological acceptable carriers, excipients or stabilisers which are well known. Acceptable carriers, excipients or stabilizers 20 are non-toxic at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including absorbic acid; low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or 25 immunoglobulins; binding agents including hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents 30 such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forming counterions such as sodium; and/or non-ionic surfactants such as Tween, Pluronics or polyethylene glycol (PEG). The formulation of pharmaceutical compositions for 35 use in accordance with the present invention will be based on the proposed route of administration. Routes of administration may include, but are not limited to, WO 2006/015426 PCT/AU2005/001188 - 30 inhalation, insufflation (either through the mouth or nose), oral, buccal, rectal or parental administration. Diagnostic and prognostic applications 5 The nucleic acid molecules and polypeptides of the invention enable the diagnosis or prognosis of angiogenesis-related disorders, or a predisposition to such disorders. Examples of such disorders include, but are not limited to, cancer; inflammatory disorders 10 including arthritis; corneal, retinal or choroidal neovascularization including macular degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases. Diagnosis or prognosis may be used to determine the severity, type or stage of the disease state in order to 15 initiate an appropriate therapeutic intervention. In another embodiment of the invention, the nucleic acid molecules that may enable diagnosis or prognosis include polynucleotides such as oligonucleotides, genomic DNA and complementary RNA and DNA molecules corresponding 20 to the nucleic acid molecules of the invention. The polynucleotides may be used to detect and quantitate gene expression in biopsied tissues in which abnormal expression of, or mutations in, a nucleic acid molecule of the invention may be correlated with disease. Genomic DNA 25 used for the diagnosis or prognosis may be obtained from body cells, such as those present in the blood, tissue biopsy, surgical specimen, or autopsy material. The DNA may be isolated and used directly for detection or may be amplified by the polymerase chain reaction (PCR) prior to 30 analysis. Similarly, RNA or cDNA may also be used, with or without PCR amplification. To detect a specific nucleic acid molecule, direct nucleotide sequencing, reverse transcriptase PCR (RT-PCR), hybridization using specific oligonucleotides, restriction enzyme digest and mapping, 35 PCR mapping, RNAse protection, and various other methods may be employed. Oligonucleotides specific to a specific nucleic acid molecule can be chemically synthesized and WO 2006/015426 PCT/AU2005/001188 - 31 labelled radioactively or nonradioactively and hybridized to individual samples immobilized on membranes or other solid-supports or in solution. The presence, absence or excess expression of a specific nucleic acid molecule may 5 then be visualized using methods such as autoradiography, fluorometry, or colorimetry. In a particular aspect, a polynucleotide corresponding to a nucleic acid molecule of the invention, as described above, may be useful in hybridisation assays 10 that detect the presence of associated disorders, particularly those mentioned previously. The polynucleotide may be labelled by standard methods and added to a fluid or tissue sample from a patient under conditions suitable for the formation of hybridisation 15 complexes. After a suitable incubation period, the sample is washed and the signal is quantitated and compared with a standard value. If the amount of signal in the patient sample is significantly altered in comparison to a control sample then the presence of altered levels of the nucleic 20 acid molecule in the sample indicates the presence of the associated disorder. Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies, in clinical trials, or to monitor the treatment of an individual patient. 25 In order to provide a basis for the diagnosis or prognosis of an angiogenesis-related disorder associated with a mutation in a nucleic acid molecule of the invention, the nucleotide sequence of the nucleic acid molecule can be compared between normal tissue and 30 diseased tissue in order to establish whether the patient expresses a mutant gene. In order to provide a basis for the diagnosis or prognosis of a disorder associated with abnormal expression of a nucleic acid molecule of the invention, a 35 normal or standard profile for expression is established. This may be accomplished by combining body fluids or cell extracts taken from normal subjects, either animal or - 32 human, with a nucleic acid molecule of the invention, under conditions suitable for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained from normal subjects with 5 values from an experiment in which a known amount of a substantially purified polynucleotide is used. Another method to identify a normal or standard profile for expression of a nucleic acid molecule of the invention is through quantitative RT-PCR studies. RNA isolated from 10 body cells of a normal individual, particularly RNA isolated from endothelial cells, is reverse transcribed and real-time PCR using oligonucleotides specific for the nucleic acid molecule is conducted to establish a normal level of expression of the gene. Standard values obtained 15 in both these examples may be compared with values obtained from samples from patients who are symptomatic for a disorder. Deviation from standard values is used to establish the presence of a disorder. Once the presence of a disorder is established and a 20 treatment protocol is initiated, hybridization assays or quantitative RT-PCR studies may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in the normal subject. The results obtained from successive 25 assays may be used to show the efficacy of treatment over a period ranging from several days to months. When a diagnostic or prognostic assay is to be based upon a polypeptide of the invention, a variety of approaches are possible. For example, diagnosis or 30 prognosis can be achieved by monitoring differences in the electrophoretic mobility of normal and mutant polypeptides. Such an approach will be particularly useful 2548012_1 (GHMatiers) - 33 in identifying mutants in which charge substitutions are present, or in which insertions, deletions or substitutions have resulted in a significant change in the electrophoretic migration of the resultant polypeptide. 5 Alternatively, diagnosis or prognosis may be based upon differences in the proteolytic cleavage patterns of normal and mutant polypeptides, differences in molar ratios of the various amino acid residues, or by functional assays demonstrating altered function of the polypeptides. 10 Antibodies that specifically bind the polypeptides of the invention may be used for the diagnosis or prognosis of angiogenesis-related disorders, or in assays to monitor patients being treated with a polypeptide of the invention or agonists, antagonists, or inhibitors thereof. 15 Antibodies useful for diagnostic or prognostic purposes may be prepared in the same manner as described above for therapeutics. Diagnostic or prognostic assays may include methods that utilize the antibody and a label to detect the relevant polypeptide in human body fluids or in 20 extracts of cells or tissues. The antibodies may be used with or without modification, and may be labelled by covalent or non-covalent attachment of a reporter molecule. A variety of assays for measuring the polypeptide 25 based on the use of antibodies specific for the polypeptide are known in the art and provide a basis for diagnosing altered or abnormal levels of expression. Normal or standard values for expression are established by combining body fluids or cell extracts taken from 30 normal mammalian subjects, preferably human, with antibody to the polypeptide under conditions suitable for complex formation. The amount of standard complex formation may be quantitated by various methods which are known in the art. Examples include, but are not limited to, enzyme-linked 35 immunosorbent assays (ELISAs), radioimmunoassays (RIAs), immunofluorescence, flow cytometry, histology, electron microscopy, in situ assays, immunoprecipitation, Western 2548012_1 (GHMatters) - 34 blot etc. For example, using the ELISA technique an enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in 'such a manner as to produce a chemical moiety that can 5 be detected for example by spectrophotomeric, fluorimetric or by visual means. Detection may also be accomplished by using other assays such as RIAs where the antibodies or antibody fragments are radioactively labelled. It is also possible to label the antibody with a fluorescent 10 compound. When the fluorescently labelled antibody is exposed to light of a certain wavelength, its presence can then be detected due to fluorescence. The antibody can also be detectably labelled by coupling it to a chemiluminescent compound. The presence of the 15 chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Quantities of polypeptide expressed in subject, control, and disease samples from biopsied tissues are 20 compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing or prognosing disease. Once an individual has been diagnosed or prognosed with a disorder, effective treatments can be initiated, as 25 described above. In the treatment of angiogenesis-related diseases which are characterised by uncontrolled or enhanced angiogenesis, the expanding vasculature needs to be inhibited. This would involve inhibiting the nucleic acid molecules or polypeptides of the invention. 30 In the treatment of angiogenesis-related diseases which are characterised by inhibited or decreased angiogenesis, approaches which enhance or promote vascular expansion are desirable. This may be achieved by enhancing, stimulating or re-activating the expression of 35 the nucleic acid molecules or polypeptides described herein. 2548033_1 (GHMatters) - 35 Microarray In further embodiments, complete cDNAs, oligonucleotides or longer fragments derived from any of the nucleic acid molecules described herein may be used as 5 probes in a microarray. The microarray can be used to monitor the expression level of large numbers of genes simultaneously and to identify genetic variants, mutations, and polymorphisms. This information may be used to determine gene function, to understand the genetic 10 basis of angiogenesis-related disorders, to diagnose or prognose angiogenesis-related disorders, and to develop and monitor the activities of therapeutic agents. Microarrays may be prepared, used, and analysed using methods known in the art. (For example, see Schena et al., 15 1996; Heller et al., 1997). Transformed hosts The production of genetically modified (knock-out, knock-in and transgenic), non-human animal models 20 comprising the nucleic acid molecules described herein is possible. These animals are useful for the study of the function of the nucleic acid molecule, to study the process of angiogenesis, to study the mechanisms of angiogenic disease as related to these molecules, for the 25 screening of candidate pharmaceutical compounds for the treatment of angiogenesis-related disorders, for the creation of explanted mammalian cell cultures which express the encoded polypeptide or mutant polypeptide, and for the evaluation of potential therapeutic interventions. 30 Animal species which are suitable for use in the animal models of the present invention include, but are not limited to, rats, mice, hamsters, guinea pigs, rabbits, dogs, cats, goats, sheep, pigs, and non-human primates such as monkeys and chimpanzees. For initial 35 studies, genetically modified mice and rats are highly desirable due to the relative ease in generating knock-in, 2548012_1 (GHMatters) WO 2006/015426 PCT/AU2005/001188 - 36 knock-out or transgenics of these animals, their ease of maintenance and their shorter life spans. For certain studies, transgenic yeast or invertebrates may be suitable and preferred because they allow for rapid screening and 5 provide for much easier handling. For longer term studies, non-human primates may be desired due to their similarity with humans. To create an animal model based on the nucleic acid molecules of the invention, several methods can be 10 employed. These include, but are not limited to, generation of a specific mutation in a homologous animal gene, insertion of a wild type human gene and/or a humanized animal gene by homologous recombination, insertion of a mutant (single or multiple) human gene as 15 genomic or minigene cDNA constructs using wild type, mutant or artificial promoter elements, or insertion of artificially modified fragments of the endogenous gene by homologous recombination. The modifications include insertion of mutant stop codons, the deletion of DNA 20 sequences, or the inclusion of recombination elements (lox p sites) recognized by enzymes such as Cre recombinase. To create transgenic mice in order to study gain of gene function in vivo, a nucleic acid molecule of the invention can be inserted into a mouse germ line using 25 standard techniques such as oocyte microinjection. Gain of gene function can mean the overexpression of a nucleic acid molecule and its encoded polypeptide product, or the genetic complementation of a mutation of the nucleic acid molecule under investigation. For oocyte injection, one or 30 more copies of the wild type or mutant nucleic acid molecule can be inserted into the pronucleus of a just fertilized mouse oocyte. This oocyte is then reimplanted into a pseudo-pregnant foster mother. The liveborn mice can then be screened for integrants using analysis of tail 35 DNA for the presence of the nucleic acid molecule. The transgene can be either a complete genomic sequence injected as a YAC, BAC, PAC or other chromosome DNA WO 2006/015426 PCT/AU2005/001188 - 37 fragment, a cDNA with either the natural promoter or a heterologous promoter, or a minigene containing all of the coding region and other elements found to be necessary for optimum expression. 5 To generate knock-out mice or knock-in mice, gene targeting through homologous recombination in mouse embryonic stem (ES) cells may be applied. Knock-out mice are generated to study loss of gene function in vivo while knock-in mice allow the study of gain of function or to 10 study the effect of specific mutations. Knock-in mice are similar to transgenic mice however the integration site and copy number are defined in the former. For knock-out mouse generation, gene targeting vectors can be designed such that they disrupt (knock-out) 15 the, protein coding sequence of the relevant nucleic acid molecule in the mouse genome. Knock-out animals of the invention will comprise a functional disruption of a relevant nucleic acid molecule of the invention such that the gene does not express a biologically active product. 20 It can be substantially deficient in at least one functional activity coded for by the nucleic acid molecule. Expression of the polypeptide encoded by the nucleic acid molecule can be substantially absent (i.e. essentially undetectable amounts are made) or may be 25 deficient in activity such as where only a portion of the polypeptide product is produced. In contrast, knock-in mice can be produced whereby a gene targeting vector containing the relevant nucleic acid molecule can integrate into a defined genetic locus in the mouse 30 genome. For both applications, homologous recombination is catalysed by specific DNA repair enzymes that recognise homologous DNA sequences and exchange them via double crossover. Gene targeting vectors are usually introduced into ES 35 cells using electroporation. ES cell integrants are then isolated via an antibiotic resistance gene present on the targeting vector and are subsequently genotyped to - 38 identify those ES cell clones in which the nucleic acid molecule under investigation has integrated into the locus of interest. The appropriate ES cells are then transmitted through the germline to produce a novel mouse strain. 5 In instances where gene ablation results in early embryonic lethality, conditional gene targeting may be employed. This allows genes to be deleted in a temporally and spatially controlled fashion. As above, appropriate ES cells are transmitted through the germline to produce a 10 novel mouse strain, however the actual deletion of the relevant nucleic acid molecule is performed in the adult mouse in a tissue specific or time controlled manner. Conditional gene targeting is most commonly achieved by use of the cre/lox system. The enzyme cre is able to 15 recognise the 34 base pair loxP sequence such that loxP flanked (or floxed) DNA is recognised and excised by cre. Tissue specific cre expression in transgenic mice enables the generation of tissue specific knock-out mice by mating gene targeted floxed mice with cre transgenic mice. Knock 20 out can be conducted in every tissue (Schwenk et al., 1995) using the 'deleter' mouse or using transgenic mice with an inducible cre gene (such as those with tetracycline inducible cre genes), or knock-out can be tissue specific for example through the use of the 25 CD19-cre mouse (Rickert et al., 1997). In the claims of this application and in the description of the invention, except where the context requires otherwise due to express language or necessary implication, the words "comprise" or variations such as 30 "comprises" or "comprising" are used in an inclusive sense, i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention. 2548012.1 (GHMatters) WO 2006/015426 PCT/AU2005/001188 - 39 Brief Description of the Drawings Figure 1: BNO69 splicing isoforms identified by in silico analysis of representative EST sequences in dbest. Figure 2: Real-time RT-PCR expression analysis of 5 BNO69 isoforms I and III in a range of human normal and tumour tissue samples. BN069 isoform III exhibits preferential expression in HUVECs. Figure 3: Effect of BNO69 knock-down in HUVECs on the activity of Rho, Rac and Cdc42. BN069 has activity for Rho 10 but not Rac and Cdc42 in HUVECs. HUVECs were adenoviral infected with empty vector (EV), BNO69R and BNO69 mutant (R82A) and incubated without serum for 20h. The cells were lysed and assayed for detection of active Rho-GTP, Rac-GTP and Cdc42-GTP, and the average fold increase compared to 15 empty vector (EV) is shown. Each result represents at least 4 independent experiments. Figure 4: Effect of BNO69 knock-down in NIH3T3 cells on the activity Rac. BNO69 has activity for Rac in NIH3T3 cells. 20 Figure 5: BNO69 regulates stress fibre formation. HUVECs were adenoviral infected with empty vector (EV) (A) or BNO69R (B & C) or were retrovirally infected with empty vector (D) or BN069.3 siRNA (SEQ ID NO: 3) (E), 24h prior to plating on fibronectin. BNO69R infected HUVECs were 25 treated without (B) or with (C) C3 exoenzyme (30pg/ml) for ,32h and stained with rhodamine-phalloidin. Retrovirally infected HUVECs (empty vector and BNO69.3 siRNA) were stained with FITC-phalloidin and the nuclei were stained with DAPI. 30 Figure 6: Real-Time RT-PCR analysis of BNO69 expression knock-down in HUVECs mediated by BNO69.3 (SEQ ID NO: 3) siRNA. BN069 mRNA expression in shRNA infected cells is expressed as a percentage of this gene's expression in HUVEC infected with the vector control. 35 BNO69.3 siRNA silenced total BNO69 mRNA expression by approximately 50%.
WO 2006/015426 PCT/AU2005/001188 - 40 Figure 7: Silencing BNO69 mRNA expression inhibits HUVEC tube formation. HUVECs infected with BNO69.3 (SEQ ID NO: 3) siRNA or a vector control were plated on Matrigel for 24hrs. Vector control infected cells formed 5 tube structures whereas cells infected with BNO69.3 siRNA failed to form tubes. Figure 8: BNO69 expression silencing results in HUVEC enlargement in culture. Images of cells infected with BNO69.3 (SEQ ID NO: 3) siRNA and a vector control are 10 shown. Figure 9: BNO69 expression silencing inhibits HUVEC proliferation. HUVECs infected with BN069.3 (SEQ ID NO: 3) siRNA or a vector control, were cultured for 72hrs in complete medium and cell numbers were measured using an 15 MTT assay. Cells infected with BN069.3 siRNA exhibited substantially reduced growth compared to cells infected with the vector control. Figure 10: BNO69 expression silencing inhibits HUVEC migration. HUVECs infected with BN069.3 (SEQ ID NO: 3) 20 siRNA or a vector control, were allowed to migrate towards fibronectin for 24hrs. Cells infected with BNO69.3 siRNA exhibited substantially reduced migration ability as compared to cells infected with the vector control. Figure 11: Proliferation induction signalling. HUVECs 25 infected with BNO69.3 (SEQ ID NO: 3) siRNA or a vector control were cultured in basic medium in the presence or absence of the pro-angiogenic growth factors VEGF and bFGF. These growth factors induced proliferation increase in vector-infected cells but failed to activate 30 proliferation in cells infected with a BNO69.3 siRNA. Figure 12: Real-time RT-PCR expression analysis of BNO69 isoforms I and III in a range of tumour cell line samples. Figure 13: Analysis of the effect of BNO69.3 35 (SEQ ID NO: 3) siRNA on proliferation in a range of tumour cell lines.
WO 2006/015426 PCT/AU2005/001188 - 41 Figure 14: Analysis of the effect of BN069.4 (SEQ ID NO: 4) siRNA on proliferation in a range of tumour cell lines. Figure 15: Analysis of the effect of BN069.3 5 (SEQ ID NO: 3) siRNA and BNO69.4 (SEQ ID NO: 4) siRNA on migration in a range of tumour cell lines. Figure 16: Analysis of the effect of BN069.3 (SEQ ID NO: 3) siRNA on MDA-MB-231 orthotopic xenografts in nude mice. 10 Figure 17: Analysis of the effect of BNO69.4 (SEQ ID NO: 4) siRNA on MDA-MB-231 orthotopic xenografts in nude mice. The invention will now be described in detail by way of reference to the following non-limiting example. 15 Example: Identification and functional analysis of BN069 isoforms Materials and Methods 20 Endothelial cell culture and infection Human umbilical vein endothelial cells (HUVECs) were purchased from Clonetics (cc-2519) and cultured in EGM-2 Bullet kit (Clonetics cc-3162). Cells were subcultured weekly up to a maximum of 6 passages. 25 Tumour cells and cell culture The MDA-MB-231 breast adenocarcinoma cell line was obtained from ATCC (Cat. HBT-26), and cultured in RPMI medium (Gibco, Cat. 21870-076) supplemented with 10% 30 foetal bovine serum (FBS; JRH Biosciences), 10mM HEPES (Gibco, Cat. 15630-080), 1mM sodium pyruvate (Gibco, Cat. 11360-070), and 1% penicillin/streptomycin/glutamine (PSG; Gibco, Cat. 10378-016). Media was replaced with fresh media every 2-3 days and cells were subcultured once a 35 week at a subcultivation ratio of 1:4 for up to three months. The U-87MG brain glioblastoma cell line was obtained from ATCC (Cat. HTB-14), and cultured in EMEM WO 2006/015426 PCT/AU2005/001188 - 42 medium (Eagles minimum essential medium, MultiCel, Cat. 11-050-0500V) supplemented with 10% foetal bovine serum (FBS; JRH Biosciences), 10mM HEPES (Gibco, Cat. 15630 080), 1mM sodium pyruvate (Gibco, Cat. 11360-070), 0.1mM 5 non-essential amino acids (NEAA; Gibco, Cat. 11140-05) and 1% penicillin/streptomycin/glutamine (PSG; Gibco, Cat. 10378-016). Media was replaced with fresh media every 2-3 days and cells were subcultured once a week at a subcultivation ratio of 1:5 for up to three months. All 10 tumour cells used (see Figure 12 for complete cell line details) were maintained in a humidified incubator at 37 0 C in 5% CO 2 in air. -Generation of recombinant BN069 clones 15 HUVEC RNA was isolated and cDNA generated by reverse transcription. Primers were designed to amplify BN069 5' and 3' cDNA fragments. Both PCR products were cloned into the pGEM-Teasy vector (Promega), confirmed by sequencing and ligated together via a common internal BamHI site. To 20 generate the BN069 mutant (R82A), the arginine codon (CGA) at position 82 of BN069 isoform II was changed to alanine (GCA) using a PCR mutagenesis approach. The BN069 or R82A mutant cDNA were excised from pGEM-Teasy with NotI and subcloned (in both orientations for BN069) into the 25 shuttle vector pAdTrack-CMV (Qbiogene). 5'-FLAG-tagged BN069 was generated by PCR and cloned into the EcoRV site of pAdTrack-CMV. Recombinant adenovirus was made using the pAdEasy system (Qbiogene). Viral titres were determined using the TCID 50 method and viral particle number 30 quantified at OD 600 nm. For gene transfer experiments, cells were grown to 80% confluence and infected with an amount of pAdEasy empty vector (EV), pAdEasy-BNO69 antisense (BNO69R), pAdEasy-BN069 sense (BNO69F) or pAdEasy-BN069R82A (R82A) 35 mutant virus particles which yielded a similar level of GFP expression.
WO 2006/015426 PCT/AU2005/001188 - 43 Generation of a retroviral vector expressing the BN069.3 siRNA and BN069.4 siRNA sequences The pMSCVpuro (BD Biosciences) was modified to create a short hairpin RNA (shRNA)-generating retroviral vector. 5 To do this, the 3'LTR of pMSCVpuro was inactivated by removal of a XbaI/NheI fragment. A H1-RNA Polymerase III promoter cassette was then inserted into the multiple cloning site (MCS) of the vector. The BN069.3 siRNA sequence (SEQ ID NO: 3) or the BN069.4 siRNA sequence 10 (SEQ ID NO: 4) was cloned into the modified pMSCVpuro vector in a short hairpin sequence format. The shRNA format comprises the siRNA sequence followed by a generic 9 nucleotide sequence, followed by the reverse complement of the siRNA. The shRNA sequences corresponding to BNO69.3 15 and BNO69.4 are represented by SEQ ID Numbers: 6 and 7 respectively. Upon integration in the host cell genome the siRNA forms a double stranded structure by annealing to its reverse complement. The 9 nucleotide generic sequence forms a loop at one end of the double stranded molecule. 20 Negative controls included the sequence 5' AGGCATCAGCGGACCTCAT-3' (SEQ ID NO: 18) as this contained similar GC content as to BN069.3 siRNA and BNO69.4 siRNA,. or a vector-only construct. 25 Retroviral particle production 293T cells (ATCC Cat. CRL-11268) were plated at a density of 1.7 x 107 cells per T175 flask 18-24 hours pre-transfection in RPMI media supplemented with 10% FCS without antibiotics. Cells were co-transfected with 28pg 30 retroviral vector (DNA), 23pg pVPack VSV-G, 23pg pVPack GP using LF2000 (Invitrogen) reagent. Transfected cells were incubated overnight at 37 0 C in 5% CO 2 . The following day media containing the DNA/LF2000 complexes was removed and replaced with RPMI supplemented with 10% FCS, 1M HEPES and 35 1% PSG. Virus containing supernatants were collected 48-72 hours post-transfection, centrifuged to pellet cell debris and then filtered using a sterile 0.45pm filter. Virus was WO 2006/015426 PCT/AU2005/001188 - 44 aliquoted and stored at -800C. Viral titre was determined utilizing viral supernatants in serial dilution to infect NIH-3T3 cells, which were then cultured under puromycin selection for 7 days. 5 Retroviral Infection of HUVE cells HUVE cells were plated in EGM-2 complete media at a density of 1.3 x 103 cells per well of a 6 well plate. 500pl of virus supernatant was combined with 500pl of EGM-2 10 complete media. Polybrene was added to a final concentration of 8ptg/ml. Cells were incubated with the viral mix at 37 0 C 5% Co 2 . Following 3 hours incubation an additional 1.0ml of EGM-2 media was added and cells were incubated for a further 24 hours. Subsequently, cells 15 were incubated in EGM-2 complete medium containing puromycin (0.35tg/ml final concentration). Cells were incubated until uninfected cells treated with puromycin had died and infected resistant cells had grown to confluence. Media containing puromycin was replaced every 20 48 hours to replenish puromycin and remove cell debris. Once resistant cells were grown to confluence (approximately 4-5 days after starting selection), cells were washed in PBS and trypsinised prior to their use. 25 Retroviral infection of tumour cells Tumour cells (for each cell line) were plated out at a density of 2 x 106 cells per T175 flask 18-24 hours pre infection in supplemented RPMI media as described earlier. At infection, media was aspirated from the flasks and 30 viral supernatant was added to an m.o.i. of 1:10 in a total of 15mls supplemented RPMI media containing polybrene at a final concentration of 8pg/ml. Cells were incubated with the viral mix at 37 0 C for 4 hours in humidified conditions in 5% CO 2 in air. Following this 35 incubation an additional 25mls of media was added to each flask and the cells were incubated for a further 24 hours. Fresh media was subsequently added to the cells for the WO 2006/015426 PCT/AU2005/001188 - 45 next 24 hours and then cells were grown under puromycin selection (0.6pg/ml) until uninfected cells treated with puromycin had died and infected resistant cells had grown to confluence. Media containing puromycin was replaced 5 every 48 hours in order to replenish puromycin and also remove cell debris. Once resistant cells were selected (4-5 days after starting selection), cells were washed in PBS, trypsinized and either resuspended in Dulbecco's PBS with Calcium and Magnesium (SIGMA, Cat. D8662) for 10 injection into animals or plated out for the proliferation assay or the migration assay as outlined below. Cells were infected with viral particles depending on the experiment conducted: pMSCVpuro-vector alone (no DNA insert), pMSCVpuro-BNO69.3 siRNA or pMSCVpuro-BNO69.4 15 siRNA. Additionally untreated cells were cultured and harvested on the same day as the infected cells contributing as another control group to each experiment. Expression analysis in HUVE cells 20 Real-time RT-PCR was utilized to perform expression analysis. Total RNA was isolated from HUVE cells using the RNeasy Mini kit (Qiagen) as per manufacturer's instructions including the on-column DNase treatment. Total RNA was visualised on a 1.2% TBE agarose gel 25 containing ethidium bromide to check for quality and purity. Total RNA concentration was determined by A260 on a spectrophotometer. Total RNA (lug/ul) was reverse transcribed using M-MLV (Promega) as per manufacturer's directions. Real-Time PCR was run on the RotorGeneTM 2000 30 (Corbett Research). Reactions used AmpliTaq Gold enzyme (Applied Biosystems) and followed manufacturer's instructions. Cycling conditions were typically 94*C for 12 minutes, 35 cycles of 94'C for 15 s, 57*C for 15 s, 72*C for 20 s. All data were normalised to the expression of 35 the housekeeping gene POLR2K (RNA polymerase II).
WO 2006/015426 PCT/AU2005/001188 - 46 Expression analysis in tumour cells Real-time RT-PCR was utilized to perform expression analysis. Total RNA was isolated from all cell lines using the RNeasy Mini Kit (Qiagen Cat. 74103) as per 5 manufacturer's instructions including the on-column DNase treatment. Total RNA was visualised on a 1.2% TBE agarose gel containing ethidium bromide to check for quality and purity of the RNA. Total RNA concentration was determined by A260 on a spectrophotometer. Total RNA (1pg/ll) was 10 reverse transcribed using M-MLV(Promega) as per manufacturer's instructions. Real-time PCR was run on the RotorGene M 2000 (Corbett research). Reactions used AmpliTaq Gold enzyme (Applied Biosystems) and followed the manufacturer's directions. Cycling conditions were 15 typically 94*C for 12 minutes, 35 cycles of 94'C for 15secs, 57 0 C for 15 secs, 72'C for 20 secs with some optimization required for selected primer pairs. All data were normalised to the expression of the housekeeping gene POLR2K (RNA polymerase II). 20 Proliferation assay - HUVE cells Infected cells were plated at 1000 cells/well in EBM + 0.5%FBS in a 96 well plate in triplicate. Cells were cultured overnight at 37*C 5% C0 2 . Proliferation was 25 induced by addition of the angiogenic growth factors VEGF (10ng/ml) and bFGF (10ng/ml) or EBM + 0.5% FBS alone as a negative control. Medium was replaced every 48 hours and MTT assays were performed at time points. Briefly, 2 0pl of MTT reagent was added to cells containing 100ptl of EBM + 30 0.5% FBS and incubated at 37 0 C for 2 hours. Absorbance was measured at 492nm. Proliferation assay - tumour cells For short term assays (time points up-to 96 hours 35 post seeding) infected cells were plated at 1000 cells/well in 200gl supplemented RPMI media (as described above), in a 96 well plate in triplicate. MTT assays were WO 2006/015426 PCT/AU2005/001188 - 47 performed at 24 hour time points up-to 96 hours post seeding of cells. For long term assays (associated with xenograft experiments) infected cells were plated at 200 cells/well in 200pl supplemented RPMI media (as described 5 above), in a 96 well plate in triplicate. Medium was replaced every 7 days and MTT assays were performed at each measurement time point as specified below for each xenograft experiment. To conduct the proliferation assay the following steps were performed: initially media was 10 aspirated from the wells and 100pl of fresh media was added followed by 20Al of MTT reagent (Promega Cat. G3581). Cells were incubated for 2 hours at 37 0 C. Absorbance was measured at 492nm. 15 Matrigel assay - HUVE cells Infected HUVECs were plated in 96 well plates at 2.5 x 104 cells/well. Wells were pre-coated with 50ul Matrigel (Becton Dickinson) in EGM-2 media. HUVECs were allowed to form tubes by incubation at 37 0 C at 5% CO 2 for 22hrs. 20 Migration assay - HUVE cells The migration assays were carried out in Neuroprobe ChemoTX 96 well plates with Bum pore size filter. The underside of the filter was coated with 40ul of Sug/ml 25 fibronectin (or NIH3T3 conditioned medium for the MDA MB231 assay) and incubated at RT for 1 hour. HUVECs were harvested and washed in MCDB 131 media + 0.1% BSA. 5x10 4 cells were seeded per well in 40ul media and incubated at 37 0 C for 24 hours. Cells on the underside of the filter 30 were fix/stained with 1% Crystal Violet/20% methanol. Stain was quantified by addition of 100ul 33% acetic acid and absorbance read at 540nm. Migration assay - tumour cells The migration assay was carried out in Neuroprobe 35 ChemoTX 96 well plates with Oum pore size filter. Cells were harvested and washed in media + 0.1% BSA. 5x10 4 cells WO 2006/015426 PCT/AU2005/001188 - 48 were seeded per well in 40ul media and incubated at 37 0 C for 12 hours. Lower chambers were filled with NIH 3T3 conditioned media. Following the 12hr incubation period cells on the underside of the chemotactic filter were 5 fix/stained with 1% Crystal Violet/20% methanol. Stain was quantified by addition of 100ul 33% acetic acid and absorbance read at 540nm. Picture files of the underside of each chemotactic membrane were captured using a 4x objective on an Olympus BX51 microscope with a CCD 10 Optronics high resolution camera. All samples were tested in triplicate. F-actin staining Endothelial cells were plated on fibronectin coated LAB-TEK* Chamber Slides (Nalge, Nunc Int.). The cells were 15 grown with or without C3 exoenzyme (30pLg/ml) for 32h and then serum starved overnight also in the presence and absence of C3 exoenzyme. Cells infected with adenovirus containing antisense BNO69 were stained with rhodamine phalloidin (Molecular Probes Inc.) while cells infected 20 with retrovirus containing BNO69.3 siRNA were stained with FITC-phalloidin (Sigma) and nuclei were stained with DAPI (Vector Laboratories Inc.) The actin filaments were observed using an epi-fluorescence microscope. Rho-, Rac- and Cdc42-GTP activity assays 25 Cells were serum depleted (EBM, Clonetics) overnight. Rho, Rac and Cdc42 activity was measured using the EZ Detect" Rho, Rac or Cdc42 Activation Kit (Pierce Biotechnology). Active protein was detected by western blotting using the monoclonal antibodies against Rho, Rac 30 or Cdc42. Rac-GTP activity assay NIH3T3 cell lines transduced with retrovirus containing antisense BN069 (BNO69R) or empty vector (EV) 35 were serum starved overnight and lysed. Active RacGTP was WO 2006/015426 PCT/AU2005/001188 - 49 pulled down by binding to the p21 binding domain (from p21 activated kinase) GST fusion protein and glutathione sepharose. The sepharose-bound fraction and total cell lysate were Western blotted and probed with anti-Rac 5 antibody. Soft agar assay Cells (5x10 5 ) were placed in a DMEM medium/0.3% agarose suspension and cultured over 2 weeks in the 10 presence of either BNO69.3 siRNA or vector only control on plates pre-coated with DMEM/0.7% agarose at 37 0 C/5%CO2 Athymic nude mice Female athymic BALB/c-nu/nu mice were used for this 15 study. Mice were between 6-8 weeks old and were purchased from ARC Western Australia and allowed to acclimatize for a couple of days. All the animals were housed under pathogen-free conditions and cared for in accordance with Flinders University of SA and NH&MRC guidelines and the 20 Australian Code of Practise for the care and use of animals for scientific purposes. Orthotopic tumour model MDA-MB-231 cells untreated or infected and puromycin 25 selected as described earlier were injected into each female athymic mouse. Each mouse was injected with 2 x 106 cells in 50p1 Dulbecco's PBS subcutaneously just above the mammary fat pad, below the right forward limb. Each group of mice were housed in a separate cage and tumour growth 30 was measured using digital calipers and animals checked for health, three times a week for approximately 5 weeks. Tumour volume were calculated as a product of length x width x height. At the end of the experiment, the mice were sacrificed, the tumours resected, photographed and 35 then frozen in OCT Compound (Tissue-Tek; Sakura) in liquid nitrogen and stored at -80 0 C until sectioned.
WO 2006/015426 PCT/AU2005/001188 - 50 Histology Frozen tumours from all mice were cut utilizing a cryostat; sections of 10pm thick were cut. Sections were 5 subsequently fixed, stained with biotin conjugated rat a mouse CD31 primary antibody (1:200 dilution, Pharmingen) for 4 hours followed by a 20 minute incubation with Extra Avidin (1:80 dilution, SIGMA Cat. E-4889). Further staining was performed with a 10 minute incubation with 10 Fast Red (SIGMA FAST m Fast Red TR/Napthol AS-MX, Cat. F 4648), followed by a final step of a 5 minute incubation with Mayer's Haematoxylin (SIGMA Cat. MHS-1). Slides were mounted and sections viewed under an Olympus BX51 microscope. Sections were photographed (5 fields per 15 section) and area representing capillaries captured and analysed by image analysis software (ImageJ). Statistical Analysis - tumour cells and xenograft models Proliferation and migration data are presented as 20 mean + SD, and analysis done utilising unpaired two-tailed t-tests to determine differences between each treatment group (pMSCV-BNO69.3 siRNA or pMSCV-BN069.4 siRNA) and the control group (pMSCVpuro-vector alone). Xenograft data are presented as mean + SEM. Analysis of variance (ANOVA). 25 was performed to determine differences between groups at each time point with Tukey's as the post-hoc test. All statistical analyses utilized GraphPad PRISM software (Version 4; GraphPad Software Inc.) A value of p<0.05 was considered statistically significant. 30 Results/Discussion Identification of BN069 isoforms and analysis of their GAP activity Identification of regulated genes during angiogenesis 35 may result in characterisation of novel targets for therapeutic drug development. To this end, we have utilised a model of in vitro angiogenesis, where the WO 2006/015426 PCT/AU2005/001188 - 51 morphological events and the time course of changes have been well characterised (Gamble et al., 1999; Meyer et al., 1997). Human umbilical vein endothelial cells, plated onto a 3-D collagen gel in the presence of growth factors, 5 PMA and an antibody to the integrin, a2pl, are induced to make capillary tubes over a 24 hour period. Isolation of these cells at critical time points, namely 0, 0.5, 3, 6 and 24 hours, and utilising a PCR based suppression subtractive hybridisation approach allowed the isolation 10 of regulated genes. One of these genes was BNO69 which encodes a novel protein that contains a GTPase Activating Protein (GAP) domain suggesting that the gene may have GAP activity. In silico analysis of EST sequences corresponding to BNO69 15 retrieved from the dbest database using BLAST (National Centre for Biotechnology Information http://www.ncbi.nlm.nih.gov/) enabled the assembly of three major BNO69 mRNA transcripts that appear to constitute the products of alternative exon splicing 20 events (Figure 1). We performed real time RT-PCR analyses to confirm the existence of these isoforms. Our data indicate that the predominantly expressed isoforms are I and III. Isoform I was found to be expressed at low levels in HUVECs, however in contrast, isoform III exhibited 25 preferential expression in HUVECs (Figure 2). All three isoforms contained the GAP domain. A comparison of the GAP domain of BN069 with those of other GAP proteins revealed considerable homology to other known RhoGAP containing proteins such as Bcr, N-chimerin, 30 p5ORhoGAP and p190RhoGAP. This region of homology comprises -160 amino acid residues with 10 residues critical to the structural integrity of the GAP domain, 3 residues (Arg85, Asn188 and Lys172 with respect to the p50RhoGAP domain) catalytically crucial to GAP activity, 35 and 5 residues (Gly82, Leu132, Leul78, Met190 and Asn194) that promote GTP hydrolysis (Rittinger et al., 1997; Barrett et al., 1997; Musacchio et al., 1996). The BN069 WO 2006/015426 PCT/AU2005/001188 - 52 protein has 9 of the 10 residues involved in the structural integrity of the GAP domain, with one conservative change from Leu to Ile. All three catalytic amino acids and four of the five GTP hydrolysis-promoting 5 residues are identical with one conservative change of Leu to Val. The highly conserved identity suggests that BNO69 may have GAP activity. In order to test this hypothesis, adenovirus expressing antisense constructs of BN069 (BNO69R) were 10 delivered to endothelial cells in order to examine the effects of BNO69 knockdown on the activity of Rho, Rac and Cdc42. The results in Figure 3 show that BNO69R increased Rho activity in endothelial cells compared to empty vector (EV) infected control cells. In 5 experiments performed, 15 there was a 3.1 ± 0.5 fold increase in Rho activity with BNO69R. No change in active Rac or Cdc42 (fold increase of 1.1 ± 0.3 in both cases) with BNO69R was seen confirming that the BN069 protein contains an active RhoGAP domain that has specificity in endothelialcells for Rho and not 20 Rac or Cdc42. This is in contrast to results observed in NIH3T3 cells (Figure 4) where inhibition of BNO69 expression using retroviral vectors expressing antisense BNO69 (BNO69R) lead to an approximately 3-fold increase in Rac activity. This suggests that the GAP domain of BNO69 25 is able to bind to multiple members of the Rho family in a cell-specific manner. From protein structural analysis, the highly conserved Arg residue corresponding to Arg85 in p50RhoGAP is critical for the binding to Rho target proteins and 30 increasing hydrolysis of Rho-bound GTP (Rittinger et al., 1997; Barrett et al., 1997; Musacchio et al., 1996). This amino acid residue corresponds to Arg82 in BN069 (based on the numbering of BNO69 isoform II) which we mutated to Ala (R82A). Expression of this mutant in endothelial cells 35 showed a 2.6 ± 0.7 fold increase (n=4 experiments) in Rho activity (Figure 3), consistent with this mutation both eliminating the GAP activity of BN069 and generating a WO 2006/015426 PCT/AU2005/001188 - 53 dominant negative form, further confirming that BNO69 indeed is a RhoGAP family member. Consistent with Rho activity in endothelial cells, BNO69R and BN069.3 siRNA infected endothelial cells showed 5 increased stress fibre formation compared to empty vector control cells which displayed the classic cortical actin type morphology with minimal stress fibres, characteristic of unstimulated endothelial cells (Figure 5A and Figure 5D). In contrast, BNO69R and BNO69.3 siRNA infected 10 endothelial cells displayed prominent, thick F-actin bundles (stress fibres), aligned in parallel arrays (Figure 5B and Figure 5E respectively). To confirm the effects of stress fibre formation were mediated by Rho, BNO69R infected HUVECs were treated with the Rho specific 15 inhibitor, C3 transferase and then stained for F-actin. In the presence of C3 transferase, stress fibre formation was abolished (Figure 5C), suggesting that the stress fibres are induced in the BNO69R infected cells, through an alteration in Rho. 20 BN069.3 siRNA inhibits HUVE cell function To determine the role of BN069 in angiogenic processes and on endothelial cell function, studies of the effect of siRNA-mediated silencing of BN069 gene 25 expression were conducted. Initially, HUVECs were infected with retroviral vectors expressing a range of short hairpin RNA (shRNA) sequences specific for BNO69. BNO69 mRNA levels were then determined by quantitative real-time RT-PCR and compared to BNO69 levels in cells infected with 30 a vector-only control. Expression of RNA polymerase was used for data normalisation. These studies assessed the efficiency of the shRNA probes in silencing expression of the BN069 mRNA. BNO69.3 siRNA silenced expression of BN069 by approximately 50% (Figure 6) and was used for 35 subsequent experiments. These studies incorporated systems for examination of changes to endothelial cell function such as determination of cell proliferation changes, cell WO 2006/015426 PCT/AU2005/001188 - 54 migration changes and effects on capillary tube formation, which are all essential features of the angiogenic process. These experiments established that HUVECs infected 5 with the BN069.3 siRNA were unable to form capillary tubes when cultured on Matrigel. As can be seen in Figure 7, vector-only infected cells formed tube structures (right panel) while cells infected with BN069.3 siRNA failed to form tubes (left panel). In addition, BN069.3 siRNA 10 infected cells became enlarged as shown in Figure 8 (right panel) as compared to vector-only infected cells (left panel). BNO69.3 siRNA infected cells also lost their ability to proliferate (Figure 9) and migrate (Figure 10). The ability of the pro-angiogenic growth factors VEGF and 15 bFGF to rescue HUVECs from the anti-proliferative effects of BN069.3 siRNA mediated silencing was also assessed. As can be seen in Figure 11, HUVECs infected with BNO69.3 remained unable to proliferate despite the presence of these growth factors in the culture medium. This 20 observation suggests that BNO69 functions at a point where the VEGF and bFGF signalling pathways converge. This is of significance given the demonstrated ability of tumours to switch between production of angiogenic stimuli like VEGF and bFGF. As a result the tumour has the ability to 25 develop resistance to any drugs targeting either of these two signalling pathways. However drugs targeting BNO69 would account for a tumour's ability to switch between these two angiogenic stimuli. 30 BN069.3 siRNA and BNO69.4 siRNA inhibit tumour cell growth and migration The expression of BN069 isoforms in tumour cell lines (Figure 12) prompted us to investigate the role of this gene in tumour cell growth. A number of tumour cell lines 35 were infected with virus producing RNAi molecules that silence both BN069 Isoforms I and III (BNO69.3 siRNA) or silence only the BNO69 Isoform I (BNO69.4 siRNA). These WO 2006/015426 PCT/AU2005/001188 - 55 cell lines are representative of different tumour types including breast and brain (Figure 12). Infected cell lines were selected for resistance to puromycin (a marker expressed by the viral vector 5 containing the sequence that codes for the shRNA molecules) in order to enrich for cells that express the shRNA molecules under consideration. Following 4 days of selection the cells were transferred to 96 well plates and cultured for 48hrs with their growth assessed following 10 this culture period. All cancer cells types infected with BNO69.3 siRNA exhibited reduced growth rates (Figure 13) whereas only a proportion of cancer types exhibited reduced growth in response to the influence of BNO69.4 siRNA (Figure 14). 15 Furthermore, we examined the possible influence of BN069.3 siRNA and BNO69.4 siRNA on the ability of tumour cell's to migrate. Migratory behaviour is a hallmark of tumour cell behaviour and underlies tumour metastasis. The potential GAP function of BN069 would implicate it in 20 signalling pathways that control cell shape and movement. Consequently, it is reasonable to hypothesise that targeting this gene may interfere with cell migration. The breast cancer cell lines (MDA-MB-231 and MDA-MB-468)' as well as the U-87 brain glioblastoma cell line infected 25 with BN069.3 siRNA exhibited significant reduced migration rates whereas only a proportion of cancer types exhibited significant reduced migration in response to the influence of BNO69.4 (Figure 15). 30 BN069.3 siRNA and BN069.4 siRNA inhibit the growth of solid breast tumours in mice The observation that BNO69.3 siRNA and BNO69.4 siRNA curtail the growth and migratory potential of tumour cells in vitro prompted us to investigate the effects of these 35 molecules in the growth of solid tumours in animals. Breast cancer cells (MDA-MB-231) were infected with the viral vectors coding for BNO69.3 siRNA and BN069.4 siRNA.
WO 2006/015426 PCT/AU2005/001188 - 56 Following a four day selection in puromycin, to enrich for cells expressing the shRNA molecules under consideration, the cells were injected in the mammary fat pad of immunocompromised mice (nu/nu mice). Cells infected with 5 the viral vector without the shRNA coding sequences were used as control. The experiment comprised 3 groups of 5 mice per group. Tumour growth was monitored over 34 days, by tumour size measurements every 2-3 days. The data obtained indicate that BN069.3 dramatically curtails the 10 ability of MDA-MB-231 cells in forming tumours in mice. Comparison with the tumours arising from cells infected with the vector control showed up to 94% reduction in tumour growth as a result of the influence of BNO69.3 siRNA (Figure 16). 15 Infection of MDA-MB231 cells with vector coding for BNO69.4 siRNA resulted in statistically significant retardation of solid tumour growth in nude mice but not to the same extent as that seen with BN069.3 siRNA (Figure 17). 20 Together, these results indicate that BN069 may play a role both in tumour cell growth and tumour angiogenesis. The use of an siRNA to BN069, and particularly BN069.3 siRNA and/or BNO69.4 siRNA, in the treatment of angiogenesis-related disorders would therefore encompass 25 the targeting of both tumour cells and endothelial cells.
WO 2006/015426 PCT/AU2005/001188 - 57 References The contents of the following documents are incorporated herein by reference: 5 Altschul, SF. et al. (1997) . Nucleic Acids Res. 25: 3389 3402. Augustin, HG. (1998) . Trends Pharmacol. Sci. 19: 216-222. Barrett, T., Xiao, B., Dodson, EJ., Dodson, G., Ludbrook, SB., Nurmahomed, K., Gamblin, SJ., Musacchio, A., 10 Smerdon, SJ., and Eccleston, JF. (1997). Nature 385: 458-461. Bayless, KJ. and Davis, GE., (2002). J. Cell Sci. 115: 1123-1136. Breaker, RR. and Joyce, GF. (1995). Chem. Biol. 15 2: 655-600. Chrzanowska-Wodnicka, M., and Burridge, K. (1996). J. Cell Biol. 133: 1403-1415. Cole, SP. et al. (1984). Mol. Cell Biol. 62: 109-120. Cote, RJ. et al. (1983). Proc. Natl. Acad. Sci. USA 80: 20 2026-2030. Culver, K. (1996). Gene Therapy : A Primer for Physicians.. Second Edition. (Mary Ann Liebert). Etienne-Manneville, S., and Hall, A. (2002). Nature 420: 629-635. 25 Friedman, T. (1991) . In Therapy for Genetic Diseases. (T Friedman (Ed) Oxford University Press. pp 105-121. Gamble, JR., Matthias, LJ., Meyer, G., Kaur, P., Russ, G., Faull, R., Berndt, MC., and Vadas, MA. (1993). J. Cell Biol. 121: 931-943. 30 Gamble, J., Meyer, G., Noack, L., Furze, J., Matthias, L., Kovach, N., Harlant, J., and Vadas, M. (1999). Endothelium 7: 23-34. Goldman, CK. et al. (1997). Nature Biotechnology 15: 462 466. 35 Green, LL. et al. (1994) . Nature Genet. 7: 13-21. Hanahan, D. (1997). Science 277: 48-50. Haseloff, J. and Gerlach, WL. (1988) . Nature 334: 585-591.
WO 2006/015426 PCT/AU2005/001188 - 58 Heller, RA. et al. (1997). Proc. Natl. Acad. Sci. USA 94: 2150-2155. Hippenstiel, S., Schmeck, B., N'Guessan, PD., Seybold, J., Krull, M., Preissner, K., Eichel-Streiber, CV., and 5 Suttorp, N. (2002). Am. J. Physiol. Lung Cell. Mol. Physiol. 283: L830-L838. Huse, WD. et al. (1989). Science 246: 1275-1281. Kohler, G. and Milstein, C. (1975). Nature 256: 495-497. Kozbor, D. et al. (1985). J. Immunol. Methods 81:31-42. 10 Lonberg, N. et al. (1994). Nature 368: 856-859. Mackay, DJ., and Hall, A. (1998). J. Biol. Chem. 273: 20685-20688. Meyer, GT., Matthias, LJ., Noack, L., Vadas, MA. and Gamble, JR., (1997). Anat. Rec. 249: 327-340. 15 Musacchio, A., Cantley, LC., and Harrison, SC. (1996). Proc. Natl. Acad. Sci. USA. 93: 14373-14378. Nobes, CD., and Hall, A. (1999). J. Cell Biol. 144: 1235 1244. Orlandi, R. et al. (1989). Proc. Natl. Acad. Sci. USA 86: 20 3833-3837. Rickert, RC. et al. (1997). Nucleic Acids Res. 25: 1317 1318. Rittinger, K., Walker, PA., Eccleston, JF., Smerdon, SJ., and Gamblin, SJ. (1997). Nature 389: 758-762. 25 Scharf, D. et al. (1994)'. Results Probl. Cell Differ. 20: 125-162. Schena, M. et al. (1996). Proc. Natl. Acad. Sci. USA 93: 10614-10619. Schwenk, F. et al. (1995). Nucleic Acids Res. 23: 5080 30 5081. Van Aelst, L., and D'Souza-Schorey, C. (1997). Genes Dev. 11: 2295-2322. Winter, G. et al. (1991). Nature 349: 293-299. Wojciak-Stothard, B., Potempa, S., Eichholtz, T., and 35 Ridley, A. J. (2001). J. Cell Sci. 114: 1343-1355.
Claims (12)
1. An isolated or synthetic short interfering RNA (siRNA) molecule comprising a complement of a segment of 5 an mRNA transcribed from the BNO69 gene, wherein said siRNA molecule modulates the expression of BNO69 by RNA interference and said siRNA molecule comprises SEQ ID NO:3. 10
2. An isolated or synthetic short hairpin RNA (shRNA) molecule which comprises the siRNA molecule according to claim 1.
3. The isolated or synthetic shRNA molecule according to 15 claim 2, further comprising a linker sequence and a sequence which is the reverse complement of the siRNA molecule.
4. The isolated or synthetic shRNA molecule according to 20 claim 3, wherein the linker sequence is a nucleotide sequence as set forth in SEQ ID NO: 5.
5. The isolated or synthetic shRNA molecule according to any one of claims 2 to 4, comprising SEQ ID NO: 6. 25
6. A vector comprising one or more of: (a) an isolated or synthetic siRNA molecule according to claim 1; or (b) an isolated or synthetic shRNA molecule 30 according to any one of claims 2 to 5.
7. A pharmaceutical composition comprising any one or more of: (a) an isolated or synthetic siRNA molecule 35 according to claim 1; (b) an isolated or synthetic shRNA molecule according to any one of claims 2 to 5; 2579740_1 (GHMatters) - 60 (c) a vector according to claim 6, and a pharmaceutically acceptable carrier. 5
8. A method of treating an angiogenesis-related disorder comprising administering to a subject an effective amount of any one or more of: (a) an isolated or synthetic siRNA molecule according to claim 1; 10 (b) an isolated or synthetic shRNA molecule according to any one of claims 2 to 5; or (c) a vector according to claim 6.
9. The method according to claim 8, wherein the siRNA 15 molecule, shRNA molecule or vector is administered to a subject in combination with at least one of: (a) a pharmaceutical agent for treating angiogenesis-related disorders; or (b) another therapeutic method to treat the 20 angiogenesis-related disorder.
10. The method according to claim 8 or claim 9, wherein the disorder is selected from the group consisting of cancer; inflammatory disorders including arthritis; 25 corneal, retinal or choroidal neovascularization including macular degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases.
11. Use of (a) an isolated or synthetic siRNA according 30 to claim 1, (b) an isolated or synthetic shRNA according to any one of claims 2 to 5, (c) a vector according to claim 6, in the manufacture of a medicament for the treatment of an angiogenesis-related disorder. 35
12. Use as claimed in claim 11, wherein the disorder is selected from the group consisting of cancer; inflammatory 257974_1 (GilMatters) - 61 disorders including arthritis; corneal, retinal or choroidal neovascularization including macular degeneration and diabetic retinopathy; psoriasis; cardiovascular diseases 5 2579748_1 (GHMulters)
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU2005270734A AU2005270734B2 (en) | 2004-08-09 | 2005-08-09 | Compositions and methods for angiogenesis-related molecules and treatments |
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| AU2004904441A AU2004904441A0 (en) | 2004-08-09 | siRNA molecules | |
| AU2004904441 | 2004-08-09 | ||
| AU2005902344A AU2005902344A0 (en) | 2005-05-10 | siRNA molecules | |
| AU2005902344 | 2005-05-10 | ||
| AU2005270734A AU2005270734B2 (en) | 2004-08-09 | 2005-08-09 | Compositions and methods for angiogenesis-related molecules and treatments |
| PCT/AU2005/001188 WO2006015426A1 (en) | 2004-08-09 | 2005-08-09 | Compositions and methods for angiogenesis-related molecules and treatments |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| AU2005270734A1 AU2005270734A1 (en) | 2006-02-16 |
| AU2005270734B2 true AU2005270734B2 (en) | 2011-03-24 |
Family
ID=37846299
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| AU2005270734A Ceased AU2005270734B2 (en) | 2004-08-09 | 2005-08-09 | Compositions and methods for angiogenesis-related molecules and treatments |
Country Status (1)
| Country | Link |
|---|---|
| AU (1) | AU2005270734B2 (en) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2003027285A1 (en) * | 2001-09-27 | 2003-04-03 | Bionomics Limited | Dna sequences for human angiogenesis genes |
-
2005
- 2005-08-09 AU AU2005270734A patent/AU2005270734B2/en not_active Ceased
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2003027285A1 (en) * | 2001-09-27 | 2003-04-03 | Bionomics Limited | Dna sequences for human angiogenesis genes |
Also Published As
| Publication number | Publication date |
|---|---|
| AU2005270734A1 (en) | 2006-02-16 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP1135485B1 (en) | Promotion or inhibition of angiogenesis and cardiovascularization | |
| JP4486815B2 (en) | DNA sequence for human angiogenic genes | |
| JP2001521382A (en) | Novel human delta-3 compositions and methods for their use in therapy and diagnosis | |
| AU3381600A (en) | Promotion or inhibition of angiogenesis and cardiovascularization | |
| JP2003531811A5 (en) | ||
| US7273927B2 (en) | Mdm2 splice variants | |
| JP2006524492A (en) | Methods for identifying nucleic acid molecules associated with angiogenesis | |
| US7667028B2 (en) | Compositions and methods for angiogenesis-related molecules and treatments | |
| CA2398775A1 (en) | Inhibitor of hepatocyte growth factor activator for use in modulation of angiogenesis and cardiovascularization | |
| EP1212417B1 (en) | Promotion or inhibition of angiogenesis and cardiovascularization | |
| US7538088B2 (en) | Method for inhibiting angiogenesis by administration of the extracellular domain of D1-1 polypeptide | |
| AU2005270734B2 (en) | Compositions and methods for angiogenesis-related molecules and treatments | |
| US20100266615A1 (en) | D1-1 nucleic acids, polypeptides and related methods | |
| AU2003259607B2 (en) | Promotion or inhibition of angiogenesis and cardiovascularization | |
| JP2005528434A (en) | Semaphorin-like proteins and methods of use | |
| WO2004028342A2 (en) | Endogenous granzyme b in human non-hematopoietic cells | |
| AU2002328200A1 (en) | DNA sequences for human angiogenesis genes | |
| ZA200103707B (en) | Promotion or inhibition of angiogenesis and cardio-vascularization. | |
| AU2008201496A1 (en) | DNA sequences for human angiogenesis genes | |
| NZ545534A (en) | Promotion or inhibition of angiogenesis and cardiovascularization | |
| NZ532803A (en) | Promotion or inhibition of angiogenesis and cardiovascularization |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| FGA | Letters patent sealed or granted (standard patent) | ||
| MK14 | Patent ceased section 143(a) (annual fees not paid) or expired |