In animals Notum acts to modify heparan sulfate proteoglycans and thereby alters the extracellular gradient of Wg/Wnt protein. Kinetic and mass spectrometric analyses of human proteins show that Notum is a carboxylesterase that removes an essential palmitoleate moiety from Wnt proteins and thus constitutes the first known extracellular protein deacylase (Kakugawa et al.). Notum also deacylates octanoylated Ghrelin
(Below N is a link to NCBI taxonomic web page and E link to ESTHER at designed phylum.) > cellular organisms: NE > Eukaryota: NE > Opisthokonta: NE > Metazoa: NE > Eumetazoa: NE > Bilateria: NE > Deuterostomia: NE > Chordata: NE > Craniata: NE > Vertebrata: NE > Gnathostomata: NE > Teleostomi: NE > Euteleostomi: NE > Sarcopterygii: NE > Dipnotetrapodomorpha: NE > Tetrapoda: NE > Amniota: NE > Mammalia: NE > Theria: NE > Eutheria: NE > Boreoeutheria: NE > Euarchontoglires: NE > Primates: NE > Haplorrhini: NE > Simiiformes: NE > Catarrhini: NE > Hominoidea: NE > Hominidae: NE > Homininae: NE > Homo: NE > Homo sapiens: NE
A85-EsteraseD-FGH : human-ESD Homo sapiens (Human) esterase D (EC 3.1.1.1) formylglutathione hydrolase. ABHD6-Lip : human-ABHD6 Homo sapiens (Human) ABHD6 Monoacylglycerol lipase EC: 3.1.1.23. ABHD8 : human-ABHD8Homo sapiens (Human) Abhydrolase domain containing 8 (ABHD8) cDNA FLJ11743 fis, clone HEMBA1005517. ABHD10 : human-ABHD10Homo sapiens (Human) ABHDA ABHD10 Abhydrolase domain-containing protein 10, Mycophenolic acid acyl-glucuronide esterase, mitochondrial. ABHD11-Acetyl_transferase : human-ABHD11Homo sapiens (Human) (EC 3.3.2.3) Abhydrolase domain-containing protein 11 williams-beuren syndrome critical region protein 21. ABHD12-PHARC : human-ABHD12Homo sapiens (Human) abhydrolase domain-containing protein 12. Protein C20orf22, flj90542, CT022, 2-arachidonoylglycerol hydrolase, Monoacylglycerol lipase, human-ABHD12BHomo sapiens (Human) Abhydrolase domain-containing protein 12B ABHD12B protein c14orf29. ABHD13-BEM46 : human-ABHD13Homo sapiens (Human) C13orf6 Q7L211 ABHDD_HUMAN ABHD13 Abhydrolase domain-containing protein 13. ABHD16 : human-ABHD16AHomo sapiens (Human) Abhydrolase domain-containing protein 16A BAT5 (HLA-B-associated transcript 5) (NG26 protein) (G5) (PP199), human-ABHD16BHomo sapiens (Human) ABHD16B PS-PLA1 lipase activity. ABHD17-depalmitoylase : human-ABHD17AHomo sapiens (Human) Abhydrolase domain-containing protein FAM108A1, C19orf27 ABHD17A, human-ABHD17BHomo sapiens (Human) CGI-67 C9orf77 FAM108B1 protein Abhydrolase domain-containing protein FAM108B1, human-ABHD17CHomo sapiens (Human) Abhydrolase domain-containing protein FAM108C1 Q6PCB6 F108C_HUMAN. ABHD18 : human-ABHD18Homo sapiens (Human) ABHD18 C4orf29 CD029 hypothetical protein. abh_upf0017 : human-ABHD1Homo sapiens (Human) lung alpha/beta hydrolase 1, human-ABHD2Homo sapiens (Human) Monoacylglycerol lipase ABHD2 LABH2 LBH2 protein phps1-2, human-ABHD3Homo sapiens (Human) hypothetical 49.3 kda protein, human-ABHD15Homo sapiens (Human) ABH15 Abhydrolase domain-containing protein 15. ACHE : human-ACHE Homo sapiens (Human) acetylcholinesterase. Acidic_Lipase : human-LIPA Homo sapiens (Human) lysosomal acid lipase LICH_HUMAN gene LIPA, Lysosomal acid lipase/cholesteryl ester hydrolase (EC:3.1.1.13) LAL cholesterol esterase (wolman disease) Sebelipase, human-LIPF Homo sapiens (Human) human gastric lipase, human-LIPJHomo sapiens (Human) Lipase member J lipase-like, ab-hydrolase domain containing 1, human-LIPKHomo sapiens (Human) Lipase member K lipase-like, ab-hydrolase domain containing 2 LIPL2, human-LIPMHomo sapiens (Human) LIPM LIPL3 ba304i5.1, human-LIPNHomo sapiens (Human) lipase-like, Lipase-like abhydrolase domain-containing protein 4. ACPH_Peptidase_S9 : human-APEHHomo sapiens (Human) acylamino acid-releasing enzyme APH APEH. Acyl-CoA_Thioesterase : human-ACOT1Homo sapiens (Human) Inducible cytosolic acyl-coenzyme A thioester hydrolase Long chain Acyl-CoA hydrolase) (cte-i) (cte-ib), human-ACOT2 Homo sapiens (Human) peroxisomal long-chain Acyl-CoA thioesterase 2 (zap128) (protein for mgc:3983) mitochondrial (EC 3.1.2.2) CTE-1a, human-ACOT4 Homo sapiens (Human) Q8N9L9 Acyl-coenzyme A thioesterase 4, inducible (EC 3.1.2.2), human-ACOT6Homo sapiens (Human) Acyl-CoA thioesterase 6 (EC 3.1.2.2), human-BAATHomo sapiens (Human) bile acid CoA: amino acid n-acyltransferase (EC 3.1.2.2). Arb2_FAM172A : human-f172aHomo sapiens (Human).Cotranscriptional regulator Protein FAM172A. Arylacetamide_deacetylase : human-AADACHomo sapiens (Human) arylacetamide deacetylase, human-AADACL2Homo sapiens (Human) similar to arylacetamide deacetylase (aadac), human-AADACL3Homo sapiens (Human) AADACL3 arylacetamide deacetylase-like 3 ADCL3, human-AADACL4Homo sapiens (Human) Arylacetamide deacetylase-like 4, human-NCEH1Homo sapiens (Human) NCEH1 KIAA1363 AADACL1 neutral cholesterol ester hydrolase 1. BCHE : human-BCHE Homo sapiens (Human) butyrylcholinesterase. Carboxypeptidase_S10 : human-CPVLHomo sapiens (Human) carboxypeptidase, vitellogenic-like CP-Mac ou CPVL carboxypeptidase WUG, human-CTSA Homo sapiens (Human) protective protein associated with lysosomal beta-galactosidase ppt2 protein CTSA Cathepsin A, PPGB, human-SCPEP1Homo sapiens (Human) serine Retinoid-inducible serine carboxypeptidase RISC SCP1 (EC 3.4.16.-). Carb_B_Chordata : human-CES1 Homo sapiens (Human) carboxylesterase CES1 hCE1 & for monocyte/macrophage serine-esterase 1 egasyn, human-CES2Homo sapiens (Human) carboxylesterase hCE-2,iCE, hiCE, CES2 gene cDNA FLJ76104 Cocaine esterase, human-CES3Homo sapiens (Human) Carboxylesterase 3 (Brain) Liver carboxylesterase 31 homolog, human-CES4AHomo sapiens (Human) Carboxylesterase 4A Carboxylesterase 8, human-CES5AHomo sapiens (Human) est5a CES7 Cauxin Carboxylesterase-like urinary excreted protein homolog. CGI-58_ABHD5_ABHD4 : human-ABHD4Homo sapiens (Human) abhydrolase domain-containing protein 4 FLJ12816 similar to 2-hydroxymuconic semialdehyde hydrolase (EC 3.1.1.-), human-ABHD5 Homo sapiens (Human) 39.1 kDa Comparative gene identification 58 (CGI-58)/Alpha Beta Hydrolase Domain 5 (ABHD5). Cholesterol_esterase : human-CEL Homo sapiens (Human) bile-salt-activated lipase, BSSL BAL CEL CEH carboxyl ester lipase chr 9. CIB-CCG1-interacting-factor-B : human-ABHD14AHomo sapiens (Human) Abhydrolase domain-containing protein 14A srsq1913, human-CIB Homo sapiens (Human) Ccg1/TafII250-Interacting Factor B CIB MGC15429 Abhydrolase domain-containing protein 14B ABHD14B. lysine deacetylase. CMBL : human-CMBLHomo sapiens (Human) Carboxymethylenebutenolidase homolog. DPP4N_Peptidase_S9 : human-DPP4 Homo sapiens (Human) dipeptidyl peptidase IV (DPP4), T-cell activation antigen CD26, human-DPP6 Homo sapiens (Human) (dipeptidylpeptidase VI) (dppx), human-DPP8 Homo sapiens (Human) dipeptidyl peptidase 8 (DPP8), human-DPP9 Homo sapiens (Human) dipeptidyl peptidase 9 DPP9 DPRP2, human-DPP10 Homo sapiens (Human) DPP-10 Dipeptidyl peptidase IV-related protein-3 KIAA1492 protein (fragment), human-FAP Homo sapiens (Human) fibroblast activation protein alpha FAPalpha, integral membrane serine protease seprase FAPA, FAP, SEPR. Duf_676 : human-FAM135AHomo sapiens (Human) F135A DKFZp781H2319 FLJ20176 fis KIAA1411 previously human-F135A, human-FAM135BHomo sapiens (Human) F135B loc51059 c8orfk32 protein. Duf_726 : human-TMCO4Homo sapiens (Human) Transmembrane and coiled-coil domain-containing protein 4. Duf_829 : human-TMEM53Homo sapiens (Human) Transmembrane protein 53, FLJ22353, NET4. Epoxide_hydrolase : human-EPHX1Homo sapiens (Human) microsomal epoxide hydrolase HYEP mEH, epoxide hydratase EPHX1, human-EPHX2 Homo sapiens (Human) epoxide hydrolase 2, Bifunctional epoxide hydrolase 2 cytosolic (EPHX2) (EC 3.3.2.3) Lipid-phosphate phosphatase (EC 3.1.3.76), human-EPHX3Homo sapiens (Human) Epoxide hydrolase 3 (EPHX3) Abhydrolase domain-containing protein 9 (ABHD9) FLJ22408, human-EPHX4Homo sapiens (Human) Epoxide hydrolase 4 EPHX4 ABHD7 EPHXRP Abhydrolase domain-containing protein 7. FSH1 : human-OVCA2Homo sapiens (Human) Candidate tumor suppressor in ovarian cancer. Hepatic_Lipase : human-LIPCHomo sapiens (Human) LIPC hepatic triacylglycerol lipase HTGL. Hormone-sensitive_lipase_like : human-LIPEHuman mRNA (Human) hormone sensitive lipase HSL. Hydrolase_RBBP9_YdeN : human-RBBP9 Homo sapiens (Human) Retinoblastoma-binding protein 9 and 10 (rbbp-10) (b5t overexpressed gene protein) (bog protein). Kynurenine-formamidase : human-AFMIDHomo sapiens (Human) Kynurenine formamidase. LIDHydrolase : human-LDAHHomo sapiens (Human) lipid droplet-associated hydrolase (LDAH) C2orf43. Lipase_3 : human-DAGLAHomo sapiens (Human) DAGLA Sn1-specific diacylglycerol lipase alpha DGL-alpha, neural stem cell-derived dendrite regulator KIAA0659, human-DAGLBHomo sapiens (Human) DAGLB Sn1-specific diacylglycerol lipase beta kccr13l FLJ36639. Lipoprotein_Lipase : human-LIPGHomo sapiens (Human) endothelial lipase LIPE_HUMAN flj43354, human-LPL Homo sapiens (Human) Lipoprotein lipase LPL, LIPD. LYsophospholipase_carboxylesterase : human-LYPLA1 Homo sapiens (Human) lysophospholipase I (LYPLA1) APT1, acyl-protein thioesterase 1 S-depalmitoylase, human-LYPLA2 Homo sapiens (Human) acyl-protein thioesterase dJ886K2.4 lysophospholipase II APT2, human-LYPLAL1 Homo sapiens (Human) LYPLAL1 26.3 kda protein lysophospholipase-like 1. Maspardin-ACP33-SPG21_like : human-SPG21Homo sapiens (Human) Maspardin spg21 acid cluster protein 33 ACP33 sbm-019 (gl010)flj24010 Maspardin. MEST-like : human-MESTHomo sapiens (Human) MEST mesoderm-specific transcript. Monoglyceridelipase_lysophospholip : human-MGLL Homo sapiens (Human) Monoglyceride lipase (MAGL) lysophospholipase homolog. Ndr_family : human-NDRG1 Homo sapiens (Human) N-myc downstream-regulated gene 1 protein (cap43,rit42, ndr1 DRG1, PROXY1, RTP, TDD5), human-NDRG2 Homo sapiens (Human) ndrg2 protein N-myc downstream-regulated gene 2 protein (syld709613 protein) ndr1-related protein 2, human-NDRG3 Homo sapiens (Human) ndrg3 protein ndr1-related development protein ndr3 otthump00000030883 otthump00000030882, human-NDRG4Homo sapiens (Human) NDRG4, N-myc downstream-regulated gene 4 protein (smap-8) flj42011 flj16174 flj44611. Neuroligin : human-NLGN1 Homo sapiens (Human) Neuroligin 1 KIAA1070 protein, human-NLGN2 Homo sapiens (Human) neuroligin 2 (KIAA1366), human-NLGN3Homo sapiens (Human) Neuroligin 3 KIAA1480, human-NLGN4X Homo sapiens (Human) Neuroligin-4, X-linked (HNLX) Neuroligin4 KIAA0951, human-NLGN4YHomo sapiens (Human) Neuroligin-4, Y-linked precursor (Neuroligin Y) KIAA0951. NLS3-Tex30 : human-KANSL3Homo sapiens (Human) KAT8 regulatory NSL complex subunit 3, Testis development protein PRTD, KIAA1310, PRTD, SI1, FLJ10081, NSL3, Rcd1, human-TEX30Homo sapiens (Human) testis expressed 30 C13orf27 chromosome 13 open reading frame 27. PAF-Acetylhydrolase : human-PAFAH2Homo sapiens (Human) (EC 3.1.1.47) platelet-activating factor acetylhydrolase 2, cytoplasmic (serine dependent phospholipase a2) (hsd-pla2), PAFAH2, PAFA2 PAF-AH, human-PLA2G7 Homo sapiens (Human) plasma PAF acetylhydrolase Phospholipase A2 groupe 7 PLA2G7 PAFAH PAF-AH Lp-PLA(2). Palmitoyl-protein_thioesterase : human-PPT1 Homo sapiens (Human) palmitoyl-protein thioesterase (PPT), human-PPT2 Homo sapiens (Human) 34.9 kda protein (palmitoyl-protein thioesterase-2). Pancreatic_lipase : human-PNLIP Homo sapiens (Human) triacylglycerol lipase (pancreatic lipase), human-PNLIPRP1 Homo sapiens (Human) pancreatic lipase related protein 1, human-PNLIPRP2 Homo sapiens (Human) pancreatic lipase related protein 2 PLRP2, human-PNLIPRP3Homo sapiens (Human) Pancreatic lipase-related protein 3. PC-sterol_acyltransferase : human-LCAT Homo sapiens (Human) phosphatidylcholine-sterol acyltransferase. Lecithin-cholesterol acyltransferase, human-PLA2G15 Homo sapiens (Human) Group XV phospholipase A2 lcat-like lysophospholipase (llpl) (unq341/pro540). PGAP1 : human-PGAP1Homo sapiens (Human)GPI inositol-deacylase PGAP1 117.8 kd protein in ste2-frs2 intergenic region, human-SERAC1Homo sapiens (Human) Protein SERAC1. Phospholipase : human-LIPHHomo sapiens (Human) membrane-bound phosphatidic acid-selective phospholipase a1-alpha, LPD lipase-related protein mPA-PLA1 alpha, human-LIPIHomo sapiens (Human) membrane-associated phosphatidic acid-selective phospholipase a1 beta mPA-PLA1 beta (LPD lipase) Cancer/testis antigen 17 CT17, human-PLA1AHomo sapiens (Human) Phospholipase A1 member A, phosphatidylserine-specific phospholipase A1 deltaC. PPase_methylesterase_euk : human-PPME1 Homo sapiens (Human) protein phosphatase PP2A methylesterase-1 (EC 3.1.1.-) (pme-1). Prolylcarboxypeptidase : human-DPP7 Homo sapiens (Human), Dipeptidyl peptidase 2, quiescent cell proline dipeptidase precursor, DPP7, DPP2, QPP, human-PRCP Homo sapiens (Human) Lysosomal Pro-X carboxypeptidase C prolylcarboxypeptidase , Angiotensinase C, Proline carboxypeptidase (EC3.4.16.2), human-PRSS16Homo sapiens (Human) PRSS16 protease, serine, 16 (thymus) TSSP thymus-specific serine protease precursor (EC 3.4.-.-). S9N_PPCE_Peptidase_S9 : human-PREP Homo sapiens (Human) Prolyl endopeptidase PE, Post-proline cleaving enzyme PPCE, prolyl oligopeptidase POP. S9N_PREPL_Peptidase_S9 : human-PREPL Homo sapiens (Human) PREPL Prolylendopeptidase-like KIAA0436. SERHL : human-SERHL2Homo sapiens (Human) serine hydrolase-like protein 2 SERHL2 chomosome 22. Thioesterase : human-FASN Homo sapiens (Human) FAS FASN Fatty acid synthase Thioesterase domain (EC 2.3.1.85), human-OLAH Homo sapiens (Human) s-acyl fatty acid synthase thioesterase, medium chain OLAH THEDC1 SAST (EC 3.1.2.14). Thyroglobulin : human-TG Homo sapiens (Human) Thyroglobulin TG Tg. Valacyclovir-hydrolase : human-BPHL Homo sapiens (Human) biphenyl hydrolase-like DJ40E16.6.3, breast epithelial mucin-associated antigen AG BPHL (mcnaa), Valacyclovir hydrolase VACVase
Molecular evidence
Database
No mutation 141 structures(e.g. : 4UYU, 4UYW, 4UYZ... more)(less) 4UYU: Structure of the wnt deacetylase notum - Crystal form I iodide complex - 2.3A, 4UYW: Structure of the wnt deacetylase notum - Crystal form I heparin complex - 1.7A, 4UYZ: Structure of the wnt deacetylase notum - Crystal form II - 2.8A, 4UZ1: Structure of the wnt deacetylase notum - Crystal form III - 1.4A, 4UZ5: Structure of the wnt deacetylase notum - Crystal form IV - 2.1A, 4UZ6: Structure of the wnt deacetylase notum - Crystal form V SOS complex - 1.9A, 4UZ7: Structure of the wnt deacetylase notum - Crystal form VI - 2.2A, 4UZ9: Structure of the wnt deacetylase notum - Crystal form VII SOS complex - 2.2A, 4UZA: Structure of the wnt deacetylase notum - Crystal form VIII - Phosphate complex 2.4A, 4UZL: Structure of the wnt deacetylase notum - Crystal form I myristoleate complex 2.1A, 4UZQ: Structure of the wnt deacetylase notum in complex with O-Palmitoleoyl serine- Crystal form IX - 1.5A, 4WBH: Structure of the wnt deacetylase notum - Crystal form I Apo - 2.2A, 6R8P: Structure of the Wnt deacetylase notum with inhibitor fragment 723 JVB Pyridine, 6R8Q: Structure of the Wnt deacylase Notum in complex with a benzotriazole fragment JV5, 6R8R: Structure of the Wnt deacylase Notum in complex with isoquinoline 45 JV8, 6T2H: Furano[2,3-d]prymidine amides as Notum inhibitors 1 (M9N), 6T2K: Furano[2,3-d]prymidine amides as Notum inhibitors 2 (LP-922056), 6TR5: Melatonin-Notum complex, 6TR6: N-acetylserotonin-Notum complex, 6TR7: Structure of the Wnt deacylase Notum in complex with N-[2-(5-fluoro-1H-indol-3-yl)ethyl]acetamide (6QCQ withdrawn), 6TUZ: Theophylline-Notum complex, 6TV4: Caffeine-Notum complex, 6YSK: 1-phenylpyrroles and 1-enylpyrrolidines as inhibitors of Notum, 6YUW: Structure of the Wnt deacylase Notum in complex with a pyrrole-3-carboxylic acid fragment 454, 6YUY: Structure of the Wnt deacylase Notum in complex with a pyrrole-3-carboxylic acid fragment 471, 6YV0: Structure of the Wnt deacylase Notum in complex with a pyrrolidine-3-carboxylic acid fragment 587, 6YV2: Structure of the Wnt deacylase Notum in complex with a pyrrolidine-3-carboxylic acid fragment 598, 6YV4: Structure of the Wnt deacylase Notum in complex with a pyrrole-3-carboxylic acid fragment 454, 6YXI: Structure of Notum in complex with a 1-(3-Chlorophenyl)-2,5-dimethyl-1H-pyrrole-3-carboxylic acid inhibitor, 6ZUV: Structure of the Wnt deacylase Notum in complex with fragment 286 (Compound 7, 4F-329S), 6ZVL: Structure of the Wnt deacylase Notum in complex with SCHEMBL21776992 (ARUK3000263 Compound23dd 7a), 6ZYF: Notum (S232A) inactive mutant Ghrelin complex, 7ARG: Notum in complex with ARUK3002704 compound 1 Methyl-, 7B2V: Notum in complex with ARUK3003906 compound 3; ethyl-, 7B2Y: Notum in complex with ARUK3003910 compound 4; 2,2 difluoroethyl-, 7B2Z: Notum in complex with ARUK3003907 compound 6; iso-propanol, 7B37: Notum in complex with ARUK3003718 compound 2 acid, 7B3F: Notum S232A in complex with ARUK3003718 compound 2 acid, 7B3G: Notum in complex with ARUK3003902, 7B3H: Notum in complex with ARUK3003909, 7B3I: Notum in complex with ARUK3003776, 7B3P: Notum in complex with ARUK3003775, 7B3X: Notum in complex with ARUK3003748, 7B45: Notum in complex with ARUK3003934, 7B4X: Notum in complex with ARUK3002697, 7B50: Notum in complex with ARUK3003778, 7B7W: Structure of the Wnt deacylase Notum in complex with fragment 049, 7B7X: Structure of the Wnt deacylase Notum in complex with fragment 063, 7B7Y: Structure of the Wnt deacylase Notum in complex with fragment 064, 7B84: Structure of the Wnt deacylase Notum in complex with fragment 065, 7B86: Structure of the Wnt deacylase Notum in complex with fragment 067, 7B87: Structure of the Wnt deacylase Notum in complex with fragment 074, 7B89: Structure of the Wnt deacylase Notum in complex with fragment 077, 7B8A: Structure of the Wnt deacylase Notum in complex with fragment 110, 7B8C: Structure of the Wnt deacylase Notum in complex with fragment 147, 7B8D: Structure of the Wnt deacylase Notum in complex with (fragment 151) Sulfapyridine, 7B8F: Structure of the Wnt deacylase Notum in complex with fragment 154, 7B8G: Structure of the Wnt deacylase Notum in complex with fragment 159, 7B8J: Structure of the Wnt deacylase Notum in complex with fragment 163 Felbinac, 7B8K: Structure of the Wnt deacylase Notum in complex with fragment 173, 7B8L: Structure of the Wnt deacylase Notum in complex with fragment 174, 7B8M: Structure of the Wnt deacylase Notum in complex with fragment 193, 7B8N: Structure of the Wnt deacylase Notum in complex with fragment 197, 7B8O: Structure of the Wnt deacylase Notum in complex with fragment 199, 7B8U: Structure of the Wnt deacylase Notum in complex with fragment 201, 7B8X: Structure of the Wnt deacylase Notum in complex with fragment 210, 7B8Y: Structure of the Wnt deacylase Notum in complex with fragment 276, 7B8Z: Structure of the Wnt deacylase Notum in complex with fragment 277, 7B98: Structure of the Wnt deacylase Notum in complex with fragment 282, 7B99: Structure of the Wnt deacylase Notum in complex with fragment 283, 7B9D: Structure of the Wnt deacylase Notum in complex with fragment 290, 7B9I: Structure of the Wnt deacylase Notum in complex with fragment 297, 7B9N: Structure of the Wnt deacylase Notum in complex with fragment 588, 7B9U: Structure of the Wnt deacylase Notum in complex with fragment 609, 7BA1: Structure of the Wnt deacylase Notum in complex with fragment 634, 7BAC: Structure of the Wnt deacylase Notum in complex with fragment 646, 7BAP: Structure of the Wnt deacylase Notum in complex with fragment 648, 7BC8: Structure of the Wnt deacylase Notum in complex with fragment 658, 7BC9: Structure of the Wnt deacylase Notum in complex with fragment 690, 7BCC: Structure of the Wnt deacylase Notum in complex with fragment 705, 7BCD: Structure of the Wnt deacylase Notum in complex with fragment 714, 7BCE: Structure of the Wnt deacylase Notum in complex with fragment 718, 7BCF: Structure of the Wnt deacylase Notum in complex with fragment 722, 7BCH: Structure of the Wnt deacylase Notum in complex with fragment 772, 7BCI: Structure of the Wnt deacylase Notum in complex with fragment 784, 7BCK: Structure of the Wnt deacylase Notum in complex with fragment 791, 7BCL: Structure of the Wnt deacylase Notum in complex with fragment 792, 7BD2: Structure of the Wnt deacylase Notum in complex with fragment 810, 7BD3: Structure of the Wnt deacylase Notum in complex with fragment 823, 7BD4: Structure of the Wnt deacylase Notum in complex with fragment 828, 7BD5: Structure of the Wnt deacylase Notum in complex with fragment 830, 7BD6: Structure of the Wnt deacylase Notum in complex with fragment 863, 7BD8: Structure of the Wnt deacylase Notum in complex with fragment 872, 7BD9: Structure of the Wnt deacylase Notum in complex with fragment 886, 7BDA: Structure of the Wnt deacylase Notum in complex with fragment 900, 7BDB: Structure of the Wnt deacylase Notum in complex with fragment 916, 7BDC: Structure of the Wnt deacylase Notum in complex with fragment 923, 7BDD: Structure of the Wnt deacylase Notum in complex with fragment 924, 7BDF: Structure of the Wnt deacylase Notum in complex with fragment 927, 7BDG: Structure of the Wnt deacylase Notum in complex with fragment 934, 7BDH: Structure of the Wnt deacylase Notum in complex with fragment 955, 7BLI: Notum-Bepridil complex, 7BLS: Notum-maybridge_18, 7BLT: Notum-maybridge_4, 7BLU: Notum-maybridge_56, 7BLW: Notum_Piperine complex, 7BM1: Notum_Valsartan complex, 7BM3: Notum Rosmarinic acid complex, 7BM7: Notum complexed with fragment 5e, 1-Naphthalenepentanoic acid, 7BMB: Notum PPOH complex, 7BMD: Notum TDZD8 complex, 7BN5: Notum fragment_1 (2-(isoquinolin-1-ylsulfanyl)acetic acid), 7BN8: Notum fragment_3 (4H,5H-naphtho[1,2-b]thiophene-2-carboxylic acid), 7BNB: Notum fragment_50 (3-(quinazolin-4-ylsulfanyl)propanoic acid), 7BNC: Notum fragment_126 (2-(1,2-dihydroacenaphthylen-5-ylsulfanyl)acetic acid), 7BND: Notum_Fragment41 (N-methyl-4,5-dihydronaphtho,2-bthiophene-2-carboxamide), 7BNE: Notum Nicotine, 7BNF: Notum Cotinine, 7BNJ: Notum Riluzole, 7BNL: Notum ARUK3003710, 7BO1: Notum Fragment_274 [(4-fluorophenyl)amino]thiourea ((4-fluoroanilino)thiourea), 7BO2: Notum Fragment_130 (4H-thieno[3,2-c]chromene-2-carboxylic acid), 7BO5: Notum Fragment_130_methyEster (methyl 4H-thieno[3,2-c]chromene-2-carboxylate), 7PJR: Structure of the Wnt deacylase Notum in complex with ARUK3000438, 7PK3: Structure of the Wnt deacylase Notum in complex with ARUK3001185, 7PKV: Structure of the Wnt deacylase Notum in complex with ARUK3000223 Compound 7b, 7QVZ: Structure of the Wnt deacylase Notum in complex with ARUK3001043, 8BSP: Structure of the Wnt deacylase Notum in complex with ARUK3006560, 8BSQ: Structure of the Wnt deacylase Notum in complex with ARUK3006561, 8BSR: Structure of the Wnt deacylase Notum in complex with ARUK3006562, 8BSZ: Structure of the Wnt deacylase Notum in complex with ARUK3005522, 8BT0: Structure of the Wnt deacylase Notum in complex with ARUK3005518, 8BT2: Structure of the Wnt deacylase Notum in complex with ARUK3004876, 8BT5: Structure of the Wnt deacylase Notum in complex with ARUK3004877, 8BT7: Structure of the Wnt deacylase Notum in complex with ARUK3004903, 8BT8: Structure of the Wnt deacylase Notum in complex with ARUK3004048, 8BTA: Structure of the Wnt deacylase Notum in complex with ARUK3004308, 8BTC: Structure of the Wnt deacylase Notum in complex with ARUK3004558, 8BTE: Structure of the Wnt deacylase Notum in complex with ARUK3004470, 8BTH: Structure of the Wnt deacylase Notum in complex with ARUK3004552, 8BTI: Structure of the Wnt deacylase Notum in complex with ARUK3004556 No kinetic
LegendThis sequence has been compared to family alignement (MSA) red => minority aminoacid blue => majority aminoacid color intensity => conservation rate title => sequence position(MSA position)aminoacid rate Catalytic site Catalytic site in the MSA MGRGVRVLLLLSLLHCAGGSEGRKTWRRRGQQPPPPPRTEAAPAAGQPVE SFPLDFTAVEGNMDSFMAQVKSLAQSLYPCSAQQLNEDLRLHLLLNTSVT CNDGSPAGYYLKESRGSRRWLLFLEGGWYCFNRENCDSRYDTMRRLMSSR DWPRTRTGTGILSSQPEENPYWWNANMVFIPYCSSDVWSGASSKSEKNEY AFMGALIIQEVVRELLGRGLSGAKVLLLAGSSAGGTGVLLNVDRVAEQLE KLGYPAIQVRGLADSGWFLDNKQYRHTDCVDTITCAPTEAIRRGIRYWNG VVPERCRRQFQEGEEWNCFFGYKVYPTLRCPVFVVQWLFDEAQLTVDNVH LTGQPVQEGLRLYIQNLGRELRHTLKDVPASFAPACLSHEIIIRSHWTDV QVKGTSLPRALHCWDRSLHDSHKASKTPLKGCPVHLVDSCPWPHCNPSCP TVRDQFTGQEMNVAQFLMHMGFDMQTVAQPQGLEPSELLGMLSNGS
Notum is a negative regulator of Wnt signaling acting through the hydrolysis of a palmitoleoylate ester, which is required for Wnt activity. Inhibitors of Notum could be of use in diseases where dysfunctional Notum activity is an underlying cause. A docking-based virtual screen (VS) of a large commercial library was used to shortlist 952 compounds for experimental validation as inhibitors of Notum. The VS was successful with 31 compounds having an IC(50) < 500 nM. A critical selection process was then applied with two clusters and two singletons (1-4d) selected for hit validation. Optimization of 4d guided by structural biology identified potent inhibitors of Notum activity that restored Wnt/beta-catenin signaling in cell-based models. The [1,2,4]triazolo[4,3-b]pyradizin-3(2H)-one series 4 represent a new chemical class of Notum inhibitors and the first to be discovered by a VS campaign. These results demonstrate the value of VS with well-designed docking models based on X-ray structures.
Notum is a carboxylesterase that suppresses Wnt signaling through deacylation of an essential palmitoleate group on Wnt proteins. There is a growing understanding of the role Notum plays in human disease such as colorectal cancer and Alzheimer'ss disease supporting the need to discover improved inhibitors, especially for use in models of neurodegeneration. Here, we describe the discovery and profile of 8l (ARUK3001185) as a potent, selective and brain pentrant inhibitor of Notum activity suitable for oral dosing in rodent models of disease. Crystallographic fragment screening of the Diamond-SGC Poised Library for binding to Notum, supported by a biochemical enzyme assay to rank inhibition activity, identifed 6a and 6b as a pair of outstanding hits. Fragment development of 6 delivered 8l that restored Wnt signaling in the presence of Notum in a cell-based reporter assay. Assessment in pharmacology screens showed 8l to be selective against serine hydrolases, kinases and drug targets.
The Wnt signaling suppressor Notum is a promising target for osteoporosis, Alzheimer's disease, and colorectal cancers. To develop novel Notum inhibitors, we used an X-ray crystallographic fragment screen with the Diamond-SGC Poised Library (DSPL) and identified 59 fragment hits from the analysis of 768 data sets. Fifty-eight of the hits were found bound at the enzyme catalytic pocket with potencies ranging from 0.5 to >1000 microM. Analysis of the fragments' diverse binding modes, enzymatic inhibitory activities, and chemical properties led to the selection of six hits for optimization, and five of these resulted in improved Notum inhibitory potencies. One hit, 1-phenyl-1,2,3-triazole 7, and its related cluster members, have shown promising lead-like properties. These became the focus of our fragment development activities, resulting in compound 7d with IC(50) 0.0067 microM. The large number of Notum fragment structures and their initial optimization provided an important basis for further Notum inhibitor development.
Notum is a negative regulator of Wnt signaling acting through the hydrolysis of a palmitoleoylate ester, which is required for Wnt activity. Inhibitors of Notum could be of use in diseases where dysfunctional Notum activity is an underlying cause. A docking-based virtual screen (VS) of a large commercial library was used to shortlist 952 compounds for experimental validation as inhibitors of Notum. The VS was successful with 31 compounds having an IC(50) < 500 nM. A critical selection process was then applied with two clusters and two singletons (1-4d) selected for hit validation. Optimization of 4d guided by structural biology identified potent inhibitors of Notum activity that restored Wnt/beta-catenin signaling in cell-based models. The [1,2,4]triazolo[4,3-b]pyradizin-3(2H)-one series 4 represent a new chemical class of Notum inhibitors and the first to be discovered by a VS campaign. These results demonstrate the value of VS with well-designed docking models based on X-ray structures.
Notum is a carboxylesterase that suppresses Wnt signaling through deacylation of an essential palmitoleate group on Wnt proteins. There is a growing understanding of the role Notum plays in human disease such as colorectal cancer and Alzheimer'ss disease supporting the need to discover improved inhibitors, especially for use in models of neurodegeneration. Here, we describe the discovery and profile of 8l (ARUK3001185) as a potent, selective and brain pentrant inhibitor of Notum activity suitable for oral dosing in rodent models of disease. Crystallographic fragment screening of the Diamond-SGC Poised Library for binding to Notum, supported by a biochemical enzyme assay to rank inhibition activity, identifed 6a and 6b as a pair of outstanding hits. Fragment development of 6 delivered 8l that restored Wnt signaling in the presence of Notum in a cell-based reporter assay. Assessment in pharmacology screens showed 8l to be selective against serine hydrolases, kinases and drug targets.
The Wnt signaling suppressor Notum is a promising target for osteoporosis, Alzheimer's disease, and colorectal cancers. To develop novel Notum inhibitors, we used an X-ray crystallographic fragment screen with the Diamond-SGC Poised Library (DSPL) and identified 59 fragment hits from the analysis of 768 data sets. Fifty-eight of the hits were found bound at the enzyme catalytic pocket with potencies ranging from 0.5 to >1000 microM. Analysis of the fragments' diverse binding modes, enzymatic inhibitory activities, and chemical properties led to the selection of six hits for optimization, and five of these resulted in improved Notum inhibitory potencies. One hit, 1-phenyl-1,2,3-triazole 7, and its related cluster members, have shown promising lead-like properties. These became the focus of our fragment development activities, resulting in compound 7d with IC(50) 0.0067 microM. The large number of Notum fragment structures and their initial optimization provided an important basis for further Notum inhibitor development.
Regulation of the Wnt signaling pathway is critically important for a number of cellular processes in both development and adult mammalian biology. This Perspective will provide a summary of current and emerging therapeutic opportunities in modulating Wnt signaling, especially through inhibition of Notum carboxylesterase activity. Notum was recently shown to act as a negative regulator of Wnt signaling through the removal of an essential palmitoleate group. Inhibition of Notum activity may represent a new approach to treat disease where aberrant Notum activity has been identified as the underlying cause. Reliable screening technologies are available to identify inhibitors of Notum, and structural studies are accelerating the discovery of new inhibitors. A selection of these hits have been optimized to give fit-for-purpose small molecule inhibitors of Notum. Three noteworthy examples are LP-922056 (26), ABC99 (27), and ARUK3001185 (28), which are complementary chemical tools for exploring the role of Notum in Wnt signaling.
Wnt signaling plays a vital role in the cell biology of skeletal patterning, differentiation, and maintenance. Notum is a secreted member of the alpha/beta-hydrolase superfamily that hydrolyzes the palmitoleoylate modification on Wnt proteins, thereby disrupting Wnt signaling. As a secreted inhibitor of Wnt, Notum presents an attractive molecular target for improving skeletal health. To determine the cell type of action for Notum's effect on the skeleton, we generated mice with Notum deficiency globally (Notum(-/-) ) and selectively (Notum(f/f) ) in limb bud mesenchyme (Prx1-Cre) and late osteoblasts/osteocytes (Dmp1-Cre). Late-stage deletion induced increased cortical bone properties, similar to global mutants. Notum expression was enhanced in response to sclerostin inhibition, so dual inhibition (Notum/sclerostin) was also investigated using a combined genetic and pharmacologic approach. Co-suppression increased cortical properties beyond either factor alone. Notum suppressed Wnt signaling in cell reporter assays, but surprisingly, also enhanced Shh signaling independent of effects on Wnt. Notum is an osteocyte-active suppressor of cortical bone formation that is likely involved in multiple signaling pathways important for bone homeostasis.
The tumour suppressor APC is the most commonly mutated gene in colorectal cancer. Loss of Apc in intestinal stem cells drives the formation of adenomas in mice via increased WNT signalling(1), but reduced secretion of WNT ligands increases the ability of Apc-mutant intestinal stem cells to colonize a crypt (known as fixation)(2). Here we investigated how Apc-mutant cells gain a clonal advantage over wild-type counterparts to achieve fixation. We found that Apc-mutant cells are enriched for transcripts that encode several secreted WNT antagonists, with Notum being the most highly expressed. Conditioned medium from Apc-mutant cells suppressed the growth of wild-type organoids in a NOTUM-dependent manner. Furthermore, NOTUM-secreting Apc-mutant clones actively inhibited the proliferation of surrounding wild-type crypt cells and drove their differentiation, thereby outcompeting crypt cells from the niche. Genetic or pharmacological inhibition of NOTUM abrogated the ability of Apc-mutant cells to expand and form intestinal adenomas. We identify NOTUM as a key mediator during the early stages of mutation fixation that can be targeted to restore wild-type cell competitiveness and provide preventative strategies for people at a high risk of developing colorectal cancer.
Osteoporosis is a common skeletal disease, with increased risk of fractures. Currently available osteoporosis treatments reduce the risk of vertebral fractures, mainly dependent on trabecular bone, whereas the effect on non-vertebral fractures, mainly dependent on cortical bone, is less pronounced. WNT signaling is a crucial regulator of bone homeostasis, and the activity of WNTs is inhibited by NOTUM, a secreted WNT lipase. We previously demonstrated that conditional inactivation of NOTUM in all osteoblast lineage cells increases the cortical but not the trabecular bone mass. The aim of the present study was to determine if NOTUM increasing cortical bone is derived from osteoblast precursors/early osteoblasts or from osteocytes/late osteoblasts. First, we demonstrated Notum mRNA expression in Dmp1-expressing osteocytes and late osteoblasts in cortical bone using in situ hybridization. We then developed a mouse model with inactivation of NOTUM in Dmp1 expressing osteocytes and late osteoblasts (Dmp1-creNotum(flox/flox) mice). We observed that the Dmp1-creNotum(flox/flox) mice displayed a substantial reduction of Notum mRNA in cortical bone, resulting in increased cortical bone mass and decreased cortical porosity in femur, but no change in trabecular bone volume fraction (BV/TV) in femur or in the lumbar vertebrae L5 in Dmp1-creNotum(flox/flox) mice as compared to control mice. In conclusion, osteocytes and late osteoblasts are the principal source of NOTUM in cortical bone, and NOTUM derived from osteocytes/late osteoblasts reduces cortical bone mass. These findings demonstrate that inhibition of osteocyte/late osteoblast-derived NOTUM might be an interesting pharmacological target to increase cortical bone mass and reduce non-vertebral fracture risk.
Notum has recently been identified as a negative regulator of Wnt signaling through the removal of an essential palmitoleate group from Wnt proteins. There are emerging reports that Notum plays a role in human disease, with published data suggesting that targeting Notum could represent a new therapeutic approach for treating cancer, osteoporosis and neurodegenerative disorders. Complementary hit-finding strategies have been applied with successful approaches that include high-throughput screening, activity-based protein profiling, screening of fragment libraries and virtual screening campaigns. Structural studies are accelerating the discovery of new inhibitors of Notum. Three fit-for-purpose examples are LP-922056, ABC99 and ARUK3001185. The application of these small-molecule inhibitors is helping to further advance an understanding of the role Notum plays in human disease.
OBJECTIVES: The only proteins known to be modified by O-linked lipidation are Wnts and ghrelin, and enzymatic removal of this post-translational modification inhibits ligand activity. Indeed, the Wnt-deacylase activity of Notum is the basis of its ability to act as a feedback inhibitor of Wnt signalling. Whether Notum also deacylates ghrelin has not been determined. METHODS: We used mass-spectrometry to assay ghrelin deacylation by Notum and co-crystallisation to reveal enzyme-substrate interactions at the atomic level. CRISPR/Cas technology was used to tag endogenous Notum and assess its localisation in mice while liver-specific Notum knock-out mice allowed us to investigate the physiological role of Notum in modulating the level of ghrelin deacylation. RESULTS: Mass-spectrometry detected the removal of octanoyl from ghrelin by purified active Notum, but not by an inactive mutant. The 2.2 A resolution crystal structure of the Notum-ghrelin complex shows the octanoyl lipid is accommodated in the hydrophobic pocket of Notum. The knock-in allele expressing HA-tagged Notum reveals that Notum is produced in the liver and present in the bloodstream, albeit at a low level. Liver-specific inactivation of Notum in animals fed with a high fat diet leads to a small but significant increase in acylated ghrelin in the circulation, while no such increase is seen in wildtype animals on the same diet. CONCLUSIONS: Overall our data demonstrate Notum can act as a ghrelin deacylase, and that this may be physiologically relevant under high fat diet conditions. Our work therefore adds Notum to the list of enzymes, including butylcholineasterase and other carboxylesterases, that modulate the acylation state of ghrelin. The contribution of multiple enzymes could help tune the activity of this important hormone to a wide range of physiological conditions.
The Wnt family of proteins are secreted signaling proteins that play key roles in regulating cellular functions. Recently, carboxylesterase Notum was shown to act as a negative regulator of Wnt signaling by mediating the removal of an essential palmitoleate. Here we disclose two new chemical scaffolds that inhibit Notum enzymatic activity. Our approach was to create a fragment library of 250 acids for screening against Notum in a biochemical assay followed by structure determination by X-ray crystallography. Twenty fragments were identified as hits for Notum inhibition, and 14 of these fragments were shown to bind in the palmitoleate pocket of Notum. Optimization of 1-phenylpyrrole 20, guided by structure-based drug design, identified 20z as the most potent compound from this series. Similarly, the optimization of 1-phenylpyrrolidine 8 gave acid 26. This work demonstrates that inhibition of Notum activity can be achieved by small, drug-like molecules possessing favorable in vitro ADME profiles.
In the adult ventricular-subventricular zone (V-SVZ), neural stem cells (NSCs) generate new olfactory bulb (OB) neurons and glia throughout life. To map adult neuronal lineage progression, we profiled >56,000 V-SVZ and OB cells by single-cell RNA sequencing (scRNA-seq). Our analyses reveal the molecular diversity of OB neurons, including fate-mapped neurons, lineage progression dynamics, and an NSC intermediate enriched for Notum, which encodes a secreted WNT antagonist. SCOPE-seq technology, which links live-cell imaging with scRNA-seq, uncovers cell-size transitions during NSC differentiation and preferential NOTUM binding to proliferating neuronal precursors. Consistently, application of NOTUM protein in slice cultures and pharmacological inhibition of NOTUM in slice cultures and in vivo demonstrated that NOTUM negatively regulates V-SVZ proliferation. Timely, context-dependent neurogenesis demands adaptive signaling among neighboring progenitors. Our findings highlight a critical regulatory state during NSC activation marked by NOTUM, which attenuates WNT-stimulated proliferation in NSC progeny.
        
Title: Caffeine inhibits Notum activity by binding at the catalytic pocket Zhao Y, Ren J, Hillier J, Lu W, Jones EY Ref: Commun Biol, 3:555, 2020 : PubMed
Notum inhibits Wnt signalling via enzymatic delipidation of Wnt ligands. Restoration of Wnt signalling by small molecule inhibition of Notum may be of therapeutic benefit in a number of pathologies including Alzheimer's disease. Here we report Notum activity can be inhibited by caffeine (IC(50) 19 microM), but not by demethylated caffeine metabolites: paraxanthine, theobromine and theophylline. Cellular luciferase assays show Notum-suppressed Wnt3a function can be restored by caffeine with an EC(50) of 46 microM. The dissociation constant (K(d)) between Notum and caffeine is 85 microM as measured by surface plasmon resonance. High-resolution crystal structures of Notum complexes with caffeine and its minor metabolite theophylline show both compounds bind at the centre of the enzymatic pocket, overlapping the position of the natural substrate palmitoleic lipid, but using different binding modes. The structural information reported here may be of relevance for the design of more potent brain-accessible Notum inhibitors.
The carboxylesterase Notum is a key negative regulator of the Wnt signaling pathway by mediating the depalmitoleoylation of Wnt proteins. Our objective was to discover potent small molecule inhibitors of Notum suitable for exploring the regulation of Wnt signaling in the central nervous system. Scaffold-hopping from thienopyrimidine acids 1 and 2, supported by X-ray structure determination, identified 3-methylimidazolin-4-one amides 20-24 as potent inhibitors of Notum with activity across three orthogonal assay formats (biochemical, extra-cellular, occupancy). A preferred example 24 demonstrated good stability in mouse microsomes and plasma, and cell permeability in the MDCK-MDR1 assay albeit with modest P-gp mediated efflux. Pharmacokinetic studies with 24 were performed in vivo in mouse with single oral administration of 24 showing good plasma exposure and reasonable CNS penetration. We propose that 24 is a new chemical tool suitable for cellular studies to explore the fundamental biology of Notum.
NOTUM is a carboxylesterase that has been shown to act by mediating the O-depalmitoleoylation of Wnt proteins resulting in suppression of Wnt signaling. Here, we describe the development of NOTUM inhibitors that restore Wnt signaling for use in in vitro disease models where NOTUM over activity is an underlying cause. A crystallographic fragment screen with NOTUM identified 2-phenoxyacetamide 3 as binding in the palmitoleate pocket with modest inhibition activity (IC50 33 muM). Optimization of hit 3 by SAR studies guided by SBDD identified indazole 38 (IC50 0.032 muM) and isoquinoline 45 (IC50 0.085 muM) as potent inhibitors of NOTUM. The binding of 45 to NOTUM was rationalized through an X-ray co-crystal structure determination which showed a flipped binding orientation compared to 3. However, it was not possible to combine NOTUM inhibition activity with metabolic stability as the majority of the compounds tested were rapidly metabolized in an NADPH-independent manner.
The disability, mortality and costs caused by non-vertebral osteoporotic fractures are enormous. Existing osteoporosis therapies are highly effective at reducing vertebral but not non-vertebral fractures. Cortical bone is a major determinant of non-vertebral bone strength. To identify novel osteoporosis drug targets, we phenotyped cortical bone of 3 366 viable mouse strains with global knockouts of druggable genes. Cortical bone thickness was substantially elevated in Notum (-/-) mice. NOTUM is a secreted WNT lipase and we observed high NOTUM expression in cortical bone and osteoblasts but not osteoclasts. Three orally active small molecules and a neutralizing antibody inhibiting NOTUM lipase activity were developed. They increased cortical bone thickness and strength at multiple skeletal sites in both gonadal intact and ovariectomized rodents by stimulating endocortical bone formation. Thus, inhibition of NOTUM activity is a potential novel anabolic therapy for strengthening cortical bone and preventing non-vertebral fractures.
        
Title: Notum attenuates HBV-related liver fibrosis through inhibiting Wnt 5a mediated non-canonical pathways Li W, Yu X, Zhu C, Wang Z, Zhao Z, Li Y, Zhang Y Ref: Biol Res, 52:10, 2019 : PubMed
BACKGROUND: Non-canonical Wnt pathways play important roles in liver fibrosis. Notum is a newly discovered inhibitor to Wnt proteins. This study was to investigate anti-fibrotic effects of Notum. METHODS: 53 patients with hepatitis B virus (HBV) infection as well as a cell co-culture system of LX-2 and Hep AD38 cells were engaged in this study. Clinical, biological and virological data of each patient were analyzed. Cell viability was detected at different time points. mRNA and protein levels of NFATc1 (Nuclear factor of activated T-cells), Jnk, alpha-SMA, Col1A1 and TIMP-1 were detected both in LX-2 and liver tissue. Protein levels of NFATc1 and Jnk in liver tissue and their correlations with fibrosis score were analyzed. RESULTS: Hepatitis B virus replication up-regulated Wnt5a induced NFATc1 and Jnk activity in Hep AD38. Notum suppressed NFATc1, Jnk and fibrosis genes expression, reduced cell viability in co-cultured LX-2 cells induced by HBV. Interestingly, Patients with HBV DNA > 5log copies/ml had higher mRNA levels of NFATc1 and fibrosis genes than patients with HBV DNA < 5log copies/ml. Most importantly, protein expressions of NFATc1 and pJnk have positive correlations with liver fibrosis scores in HBV-infected patients. CONCLUSIONS: Our data showed that Notum inhibited HBV-induced liver fibrosis through down-regulating Wnt 5a mediated non-canonical pathways. This study shed light on anti-fibrotic treatment.
Osteoporosis is a common skeletal disease, affecting millions of individuals worldwide. Currently used osteoporosis treatments substantially reduce vertebral fracture risk, whereas nonvertebral fracture risk, mainly caused by reduced cortical bone mass, has only moderately been improved by the osteoporosis drugs used, defining an unmet medical need. Because several wingless-type MMTV integration site family members (WNTs) and modulators of WNT activity are major regulators of bone mass, we hypothesized that NOTUM, a secreted WNT lipase, might modulate bone mass via an inhibition of WNT activity. To characterize the possible role of endogenous NOTUM as a physiologic modulator of bone mass, we developed global, cell-specific, and inducible Notum-inactivated mouse models. Notum expression was high in the cortical bone in mice, and conditional Notum inactivation revealed that osteoblast lineage cells are the principal source of NOTUM in the cortical bone. Osteoblast lineage-specific Notum inactivation increased cortical bone thickness via an increased periosteal circumference. Inducible Notum inactivation in adult mice increased cortical bone thickness as a result of increased periosteal bone formation, and silencing of Notum expression in cultured osteoblasts enhanced osteoblast differentiation. Large-scale human genetic analyses identified genetic variants mapping to the NOTUM locus that are strongly associated with bone mineral density (BMD) as estimated with quantitative ultrasound in the heel. Thus, osteoblast-derived NOTUM is an essential local physiologic regulator of cortical bone mass via effects on periosteal bone formation in adult mice, and genetic variants in the NOTUM locus are associated with BMD variation in adult humans. Therapies targeting osteoblast-derived NOTUM may prevent nonvertebral fractures.-Moverare-Skrtic, S., Nilsson, K. H., Henning, P., Funck-Brentano, T., Nethander, M., Rivadeneira, F., Coletto Nunes, G., Koskela, A., Tuukkanen, J., Tuckermann, J., Perret, C., Souza, P. P. C., Lerner, U. H., Ohlsson, C. Osteoblast-derived NOTUM reduces cortical bone mass in mice and the NOTUM locus is associated with bone mineral density in humans.
A decline in stem cell function impairs tissue regeneration during ageing, but the role of the stem-cell-supporting niche in ageing is not well understood. The small intestine is maintained by actively cycling intestinal stem cells that are regulated by the Paneth cell niche(1,2). Here we show that the regenerative potential of human and mouse intestinal epithelium diminishes with age owing to defects in both stem cells and their niche. The functional decline was caused by a decrease in stemness-maintaining Wnt signalling due to production of Notum, an extracellular Wnt inhibitor, in aged Paneth cells. Mechanistically, high activity of mammalian target of rapamycin complex 1 (mTORC1) in aged Paneth cells inhibits activity of peroxisome proliferator activated receptor alpha (PPAR-alpha)(3), and lowered PPAR-alpha activity increased Notum expression. Genetic targeting of Notum or Wnt supplementation restored function of aged intestinal organoids. Moreover, pharmacological inhibition of Notum in mice enhanced the regenerative capacity of aged stem cells and promoted recovery from chemotherapy-induced damage. Our results reveal a role of the stem cell niche in ageing and demonstrate that targeting of Notum can promote regeneration of aged tissues.
Background: The carboxylesterase Notum has been shown to act as a key negative regulator of the Wnt signalling pathway by mediating the depalmitoleoylation of Wnt proteins. LP-922056 (1) is an orally active inhibitor of Notum. We are investigating the role of Notum in modulating Wnt signalling in the central nervous system and wished to establish if 1 would serve as a peripherally restricted control. An accessible and improved synthetic route would allow 1 to become more readily available as a chemical tool to explore the fundamental biology of Notum and build target validation to underpin new drug discovery programs. Results: An improved, scalable synthesis of 1 is reported. Key modifications include: (1) the introduction of the C7-cyclopropyl group was most effectively achieved with a Suzuki-Miyaura cross-coupling reaction with MIDA-boronate 11 (5 --> 6), and (2) C6 chlorination was performed with 1-chloro-1,2-benziodoxol-3-one (12) (6 --> 7) as a mild and selective electrophilic chlorination agent. This 7-step route from 16 has been reliably performed on large scale to produce multigram quantities of 1 in good efficiency and high purity. Pharmacokinetic studies in mouse showed CNS penetration of 1 is very low with a brain/plasma concentration ratio of just 0.01. A small library of amides 17 were prepared from acid 1 to explore if 1 could be modified to deliver a CNS penetrant tool by capping off the acid as an amide. Although significant Notum inhibition activity could be achieved, none of these amides demonstrated the required combination of metabolic stability along with cell permeability without evidence of P-gp mediated efflux. Conclusion: Mouse pharmacokinetic studies demonstrate that 1 is unsuitable for use in models of disease where brain penetration is an essential requirement of the compound but would be an ideal peripherally restricted control. These data will contribute to the understanding of drug levels of 1 to overlay with appropriate in vivo efficacy endpoints, i.e., the PK-PD relationship. The identification of a suitable analogue of 1 (or 17) which combines Notum inhibition with CNS penetration would be a valuable chemical probe for investigating the role of Notum in disease models.
        
Title: Structural characterisation of melatonin as an inhibitor of the Wnt deacylase Notum Zhao Y, Ren J, Hillier J, Jones M, Lu W, Jones EY Ref: J Pineal Res, :e12630, 2019 : PubMed
The hormone melatonin, secreted from the pineal gland, mediates multiple physiological effects including modulation of Wnt/beta-catenin signalling. The Wnt palmitoleate lipid modification is essential for its signalling activity, while the carboxylesterase Notum can remove the lipid from Wnt and inactivate it. Notum enzyme inhibition can therefore upregulate Wnt signalling. While searching for Notum inhibitors by crystallographic fragment screening, a hit compound N-[2-(5-fluoro-1H-indol-3-yl)ethyl]acetamide that is structurally similar to melatonin, came to our attention. We then soaked melatonin and its precursor N-acetylserotonin into Notum crystals and obtained high resolution structures (<= 1.5 A) of their complexes. In each of the structures, two compound molecules bind with Notum: one at the enzyme's catalytic pocket, overlapping the space occupied by the acyl tail of the Wnt palmitoleate lipid; and the other at the edge of the pocket opposite the substrate entrance. Although the inhibitory activity of melatonin shown by in vitro enzyme assays is low (IC50 75 microM), the structural information reported here provides a basis for the design of potent and brain accessible drugs for neurodegenerative diseases such as Alzheimer's disease, in which up-regulation of Wnt signalling may be beneficial.
        
Title: Selective Irreversible Inhibitors of the Wnt-Deacylating Enzyme NOTUM Developed by Activity-Based Protein Profiling Suciu RM, Cognetta AB, 3rd, Potter ZE, Cravatt BF Ref: ACS Med Chem Lett, 9:563, 2018 : PubMed
Wnt proteins are secreted morphogens that play critical roles in embryonic development and tissue remodeling in adult organisms. Aberrant Wnt signaling contributes to diseases such as cancer. Wnts are modified by an unusual O-fatty acylation event (O-linked palmitoleoylation of a conserved serine) that is required for binding to Frizzled receptors. O-Palmitoleoylation of Wnts is introduced by the porcupine (PORCN) acyltransferase and removed by the serine hydrolase NOTUM. PORCN inhibitors are under development for oncology, while NOTUM inhibitors have potential for treating degenerative diseases. Here, we describe the use of activity-based protein profiling (ABPP) to discover and advance a class of N-hydroxyhydantoin (NHH) carbamates that potently and selectively inhibit NOTUM. An optimized NHH carbamate inhibitor, ABC99, preserves Wnt-mediated cell signaling in the presence of NOTUM and was also converted into an ABPP probe for visualizing NOTUM in native biological systems.
Activation of Wnt signaling due to Wnt overexpression or mutations of Wnt pathway components is associated with various cancers. Blocking Wnt secretion by inhibiting PORCN enzymatic activity has shown efficacy in a subset of cancers with elevated Wnt signaling. Predicting response to upstream Wnt inhibitors and monitoring response to therapeutics is challenging due to the paucity of well-defined biomarkers. In this study we identify Notum as a potential biomarker for Wnt driven cancers and show that coordinate regulation of NOTUM and AXIN2 expression may be a useful predictor of response to PORCN inhibitors. Most importantly, as NOTUM is a secreted protein and its levels in blood correlate with tumor growth, it has potential as a pharmacodynamic biomarker for PORCN and other Wnt pathway inhibitors.
A group of small molecule thienopyrimidine inhibitors of Notum Pectinacetylesterase are described. We explored both 2-((5,6-thieno[2,3-d]pyrimidin-4-yl)thio)acetic acids and 2-((6,7-thieno[3,2-d]pyrimidin-4-yl)thio)acetic acids. In both series, highly potent, orally active Notum Pectinacetylesterase inhibitors were identified.
        
Title: Genetic Approaches To Identifying Novel Osteoporosis Drug Targets Brommage R Ref: Journal of Cellular Biochemistry, 116:2139, 2015 : PubMed
During the past two decades effective drugs for treating osteoporosis have been developed, including anti-resorptives inhibiting bone resorption (estrogens, the SERM raloxifene, four bisphosphonates, RANKL inhibitor denosumab) and the anabolic bone forming daily injectable peptide teriparatide. Two potential drugs (odanacatib and romosozumab) are in late stage clinical development. The most pressing unmet need is for orally active anabolic drugs. This review describes the basic biological studies involved in developing these drugs, including the animal models employed for osteoporosis drug development. The genomics revolution continues to identify potential novel osteoporosis drug targets. Studies include human GWAS studies and identification of mutant genes in subjects having abnormal bone mass, mouse QTL and gene knockouts, and gene expression studies. Multiple lines of evidence indicate that Wnt signaling plays a major role in regulating bone formation and continued study of this complex pathway is likely to lead to key discoveries. In addition to the classic Wnt signaling targets DKK1 and sclerostin, LRP4, LRP5/LRP6, SFRP4, WNT16, and NOTUM can potentially be targeted to modulate Wnt signaling. Next-generation whole genome and exome sequencing, RNA-sequencing and CRISPR/CAS9 gene editing are new experimental techniques contributing to understanding the genome. The International Knockout Mouse Consortium efforts to knockout and phenotype all mouse genes are poised to accelerate. Accumulating knowledge will focus attention on readily accessible databases (Big Data). Efforts are underway by the International Bone and Mineral Society to develop an annotated Skeletome database providing information on all genes directly influencing bone mass, architecture, mineralization or strength.
The connection between colorectal cancer (CRC) and Wnt signaling pathway activation is well known, but full elucidation of the underlying regulation of the Wnt/beta-catenin pathway and its biological functions in CRC pathogenesis is still needed. Here, the azoxymethane/dextran sulfate sodium salt (AOM/DSS) murine model has been used as an experimental platform able to mimic human sporadic CRC development with predictable timing. We performed genome-wide expression profiling of AOM/DSS-induced tumors and normal colon mucosa to identify potential novel CRC biomarkers. Remarkably, the enhanced expression of Notum, a conserved feedback antagonist of Wnt, was observed in tumors along with alterations in Glypican-1 and Glypican-3 levels. These findings were confirmed in a set of human CRC samples. Here, we provide the first demonstration of significant changes in Notum and glypicans gene expression during CRC development and present evidence to suggest them as potential new biomarkers of CRC pathogenesis.
Signalling by Wnt proteins is finely balanced to ensure normal development and tissue homeostasis while avoiding diseases such as cancer. This is achieved in part by Notum, a highly conserved secreted feedback antagonist. Notum has been thought to act as a phospholipase, shedding glypicans and associated Wnt proteins from the cell surface. However, this view fails to explain specificity, as glypicans bind many extracellular ligands. Here we provide genetic evidence in Drosophila that Notum requires glypicans to suppress Wnt signalling, but does not cleave their glycophosphatidylinositol anchor. Structural analyses reveal glycosaminoglycan binding sites on Notum, which probably help Notum to co-localize with Wnt proteins. They also identify, at the active site of human and Drosophila Notum, a large hydrophobic pocket that accommodates palmitoleate. Kinetic and mass spectrometric analyses of human proteins show that Notum is a carboxylesterase that removes an essential palmitoleate moiety from Wnt proteins and thus constitutes the first known extracellular protein deacylase.
Chromosome 17 is unusual among the human chromosomes in many respects. It is the largest human autosome with orthology to only a single mouse chromosome, mapping entirely to the distal half of mouse chromosome 11. Chromosome 17 is rich in protein-coding genes, having the second highest gene density in the genome. It is also enriched in segmental duplications, ranking third in density among the autosomes. Here we report a finished sequence for human chromosome 17, as well as a structural comparison with the finished sequence for mouse chromosome 11, the first finished mouse chromosome. Comparison of the orthologous regions reveals striking differences. In contrast to the typical pattern seen in mammalian evolution, the human sequence has undergone extensive intrachromosomal rearrangement, whereas the mouse sequence has been remarkably stable. Moreover, although the human sequence has a high density of segmental duplication, the mouse sequence has a very low density. Notably, these segmental duplications correspond closely to the sites of structural rearrangement, demonstrating a link between duplication and rearrangement. Examination of the main classes of duplicated segments provides insight into the dynamics underlying expansion of chromosome-specific, low-copy repeats in the human genome.
The National Institutes of Health's Mammalian Gene Collection (MGC) project was designed to generate and sequence a publicly accessible cDNA resource containing a complete open reading frame (ORF) for every human and mouse gene. The project initially used a random strategy to select clones from a large number of cDNA libraries from diverse tissues. Candidate clones were chosen based on 5'-EST sequences, and then fully sequenced to high accuracy and analyzed by algorithms developed for this project. Currently, more than 11,000 human and 10,000 mouse genes are represented in MGC by at least one clone with a full ORF. The random selection approach is now reaching a saturation point, and a transition to protocols targeted at the missing transcripts is now required to complete the mouse and human collections. Comparison of the sequence of the MGC clones to reference genome sequences reveals that most cDNA clones are of very high sequence quality, although it is likely that some cDNAs may carry missense variants as a consequence of experimental artifact, such as PCR, cloning, or reverse transcriptase errors. Recently, a rat cDNA component was added to the project, and ongoing frog (Xenopus) and zebrafish (Danio) cDNA projects were expanded to take advantage of the high-throughput MGC pipeline.
        
Title: HSPG modification by the secreted enzyme Notum shapes the Wingless morphogen gradient Giraldez AJ, Copley RR, Cohen SM Ref: Dev Cell, 2:667, 2002 : PubMed
The secreted signaling protein Wingless acts as a morphogen to pattern the imaginal discs of Drosophila. Here we report identification of a secreted repressor of Wingless activity, which we call Notum. Loss of Notum function leads to increased Wingless activity by altering the shape of the Wingless protein gradient. When overexpressed, Notum blocks Wingless activity. Notum encodes a member of the alpha/beta-hydrolase superfamily, with similarity to pectin acetylesterases. We present evidence that Notum influences Wingless protein distribution by modifying the heparan sulfate proteoglycans Dally-like and Dally. High levels of Wingless signaling induce Notum expression. Thus, Wingless contributes to shaping its own gradient by regulating expression of a protein that modifies its interaction with cell surface proteoglycans.