Homo sapiens (Human) dipeptidyl peptidase 8 (DPP8)
Comment
DPP8 and DPP9 are cytosolic peptidases. Have dpp activity similar to dpp4. The first identified endogenous DPP9 substrate is the tumor epitope RU13442 (VPYGSFKHV). Inhibition or silencing of DPP9, but not DPP8, led to increased presentation of this antigen on MHC class I alleles to cytotoxic T-cells, linking DPP9 to the MHC class I antigen presentation pathway. Small ubiquitin-like protein modifier SUMO1 acts as an allosteric activator of DPP9. By binding to an armlike motif in DPP9; DPP9 functions as an endogenous inhibitor of NLRP1 inflammasome. DPP9 interacts with a unique autoproteolytic domain (Function to Find Domain (FIIND)) found in NLRP1 and CARD8. Previously called DRP1 Qi et al . Has dpp activity as dpp4 . Q7Z4D3 mstp135 very short sequence with some diff in N-term ?!! Q7Z4C8 mstp141 Q7Z4E1 mstp097
(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-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). Pectinacetylesterase-Notum : human-NOTUM Homo sapiens (Human) Protein notum homolog. 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 19 structures(e.g. : 6EOO, 6EOP, 6EOS... more)(less) 6EOO: Human dipeptidyl peptidases 8 - DPP8 - Apo, space group 20, 6EOP: Human dipeptidyl peptidases 8 - DPP8 - SLRFLYEG, space group 20, 6EOS: Human dipeptidyl peptidases 8 - DPP8 - Apo, space group 19, 6EOT: Human dipeptidyl peptidases 8 - DPP8 - SLRFLYEG, space group 19, 6HP8: Human dipeptidyl peptidases 8 - DPP8 - bound to Val-BoroPro, 6QZW: DPP8 bound to a dipeptide (MP) from the N-terminus of BRCA2, 6TRW: Crystal structure of DPP8 in complex with the EIL peptide (SLRFLFEGQRIADNH), 6TRX: Crystal structure of DPP8 in complex with 1G244, 7A3G: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor91, 7A3I: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor LMC375, 7A3J: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor A272, 7A3K: Crystal structure of DPP8 in complex with a b-lactam based inhibitor, A296.1, 7A3L: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor A241, 7AYQ: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor, B114 (replaces 6T6T withdrawn), 7AYR: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor, B115 (replaces 6T6U withdrawn), 7OR4: Crystal structure of DPP8 in complex with a b-lactam based inhibitor, B142, 7OZ7: Crystal structure of DPP8 in complex with a 4-oxo-b-lactam based inhibitor, L84, 7SVM: Human Dipeptidyl peptidase 8 (DPP8) - ICeD-2 (inducer of cell death-2) complex, 7SVO: Human Dipeptidyl peptidase 8 (DPP8) - ICeD-1 (inducer of cell death-1) complex 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 MAAAMETEQLGVEIFETADCEENIESQDRPKLEPFYVERYSWSQLKKLLA DTRKYHGYMMAKAPHDFMFVKRNDPDGPHSDRIYYLAMSGENRENTLFYS EIPKTINRAAVLMLSWKPLLDLFQATLDYGMYSREEELLRERKRIGTVGI ASYDYHQGSGTFLFQAGSGIYHVKDGGPQGFTQQPLRPNLVETSCPNIRM DPKLCPADPDWIAFIHSNDIWISNIVTREERRLTYVHNELANMEEDARSA GVATFVLQEEFDRYSGYWWCPKAETTPSGGKILRILYEENDESEVEIIHV TSPMLETRRADSFRYPKTGTANPKVTFKMSEIMIDAEGRIIDVIDKELIQ PFEILFEGVEYIARAGWTPEGKYAWSILLDRSQTRLQIVLISPELFIPVE DDVMERQRLIESVPDSVTPLIIYEETTDIWINIHDIFHVFPQSHEEEIEF IFASECKTGFRHLYKITSILKESKYKRSSGGLPAPSDFKCPIKEEIAITS GEWEVLGRHGSNIQVDEVRRLVYFEGTKDSPLEHHLYVVSYVNPGEVTRL TDRGYSHSCCISQHCDFFISKYSNQKNPHCVSLYKLSSPEDDPTCKTKEF WATILDSAGPLPDYTPPEIFSFESTTGFTLYGMLYKPHDLQPGKKYPTVL FIYGGPQVQLVNNRFKGVKYFRLNTLASLGYVVVVIDNRGSCHRGLKFEG AFKYKMGQIEIDDQVEGLQYLASRYDFIDLDRVGIHGWSYGGYLSLMALM QRSDIFRVAIAGAPVTLWIFYDTGYTERYMGHPDQNEQGYYLGSVAMQAE KFPSEPNRLLLLHGFLDENVHFAHTSILLSFLVRAGKPYDLQIYPQERHS IRVPESGEHYELHLLHYLQENLGSRIAALKVI
N-terminal sequences are important sites for post-translational modifications that alter protein localization, activity, and stability. Dipeptidyl peptidase 9 (DPP9) is a serine aminopeptidase with the rare ability to cleave off N-terminal dipeptides with imino acid proline in the second position. Here, we identify the tumor-suppressor BRCA2 as a DPP9 substrate and show this interaction to be induced by DNA damage. We present crystallographic structures documenting intracrystalline enzymatic activity of DPP9, with the N-terminal Met1-Pro2 of a BRCA21-40 peptide captured in its active site. Intriguingly, DPP9-depleted cells are hypersensitive to genotoxic agents and are impaired in the repair of DNA double-strand breaks by homologous recombination. Mechanistically, DPP9 targets BRCA2 for degradation and promotes the formation of RAD51 foci, the downstream function of BRCA2. N-terminal truncation mutants of BRCA2 that mimic a DPP9 product phenocopy reduced BRCA2 stability and rescue RAD51 foci formation in DPP9-deficient cells. Taken together, we present DPP9 as a regulator of BRCA2 stability and propose that by fine-tuning the cellular concentrations of BRCA2, DPP9 alters the BRCA2 interactome, providing a possible explanation for DPP9's role in cancer.
        
Title: New insights into the role of dipeptidyl peptidase 8 and dipeptidyl peptidase 9 and their inhibitors Cui C, Tian X, Wei L, Wang Y, Wang K, Fu R Ref: Front Pharmacol, 13:1002871, 2022 : PubMed
Dipeptidyl peptidase 8 (DPP8) and 9 (DPP9) are widely expressed in mammals including humans, mainly locate in the cytoplasm. The DPP8 and DPP9 (DPP8/9) belong to serine proteolytic enzymes, they can recognize and cleave N-terminal dipeptides of specific substrates if proline is at the penultimate position. Because the localization of DPP8/9 is different from that of DPP4 and the substrates for DPP8/9 are not yet completely clear, their physiological and pathological roles are still being further explored. In this article, we will review the recent research advances focusing on the expression, regulation, and functions of DPP8/9 in physiology and pathology status. Emerging research results have shown that DPP8/9 is involved in various biological processes such as cell behavior, energy metabolism, and immune regulation, which plays an essential role in maintaining normal development and physiological functions of the body. DPP8/9 is also involved in pathological processes such as tumorigenesis, inflammation, and organ fibrosis. In recent years, related research on immune cell pyroptosis has made DPP8/9 a new potential target for the treatment of hematological diseases. In addition, DPP8/9 inhibitors also have great potential in the treatment of tumors and chronic kidney disease.
Soaking of macromolecular crystals allows the formation of complexes via diffusion of molecules into a preformed crystal for structural analysis. Soaking offers various advantages over co-crystallization, e.g. small samples and high-throughput experimentation. However, this method has disadvantages, such as inducing mechanical stress on crystals and reduced success rate caused by low affinity/solubility of the ligand. To bypass these issues, the Picodropper was previously developed in the authors' laboratory. This technique aimed to deliver small volumes of compound solution in response to crystal dehydration supported by the Free Mounting System humidity control or by IR-laser-induced protein crystal transformation. Herein, a new related soaking development, the Aerosol-Generator, is introduced. This device delivers compounds onto the solution-free surface of protein crystals using an ultrasonic technique. The produced aerosol stream enables an easier and more accurate control of solution volumes, reduced crystal handling, and crystal-size-independent soaking. The Aerosol-Generator has been used to produce complexes of DPP8 crystals, where otherwise regular soaking did not achieve complex formation. These results demonstrate the potential of this device in challenging ligand-binding scenarios and contribute to further understanding of DPP8 inhibitor binding.
N-terminal sequences are important sites for post-translational modifications that alter protein localization, activity, and stability. Dipeptidyl peptidase 9 (DPP9) is a serine aminopeptidase with the rare ability to cleave off N-terminal dipeptides with imino acid proline in the second position. Here, we identify the tumor-suppressor BRCA2 as a DPP9 substrate and show this interaction to be induced by DNA damage. We present crystallographic structures documenting intracrystalline enzymatic activity of DPP9, with the N-terminal Met1-Pro2 of a BRCA21-40 peptide captured in its active site. Intriguingly, DPP9-depleted cells are hypersensitive to genotoxic agents and are impaired in the repair of DNA double-strand breaks by homologous recombination. Mechanistically, DPP9 targets BRCA2 for degradation and promotes the formation of RAD51 foci, the downstream function of BRCA2. N-terminal truncation mutants of BRCA2 that mimic a DPP9 product phenocopy reduced BRCA2 stability and rescue RAD51 foci formation in DPP9-deficient cells. Taken together, we present DPP9 as a regulator of BRCA2 stability and propose that by fine-tuning the cellular concentrations of BRCA2, DPP9 alters the BRCA2 interactome, providing a possible explanation for DPP9's role in cancer.
        
Title: New insights into the role of dipeptidyl peptidase 8 and dipeptidyl peptidase 9 and their inhibitors Cui C, Tian X, Wei L, Wang Y, Wang K, Fu R Ref: Front Pharmacol, 13:1002871, 2022 : PubMed
Dipeptidyl peptidase 8 (DPP8) and 9 (DPP9) are widely expressed in mammals including humans, mainly locate in the cytoplasm. The DPP8 and DPP9 (DPP8/9) belong to serine proteolytic enzymes, they can recognize and cleave N-terminal dipeptides of specific substrates if proline is at the penultimate position. Because the localization of DPP8/9 is different from that of DPP4 and the substrates for DPP8/9 are not yet completely clear, their physiological and pathological roles are still being further explored. In this article, we will review the recent research advances focusing on the expression, regulation, and functions of DPP8/9 in physiology and pathology status. Emerging research results have shown that DPP8/9 is involved in various biological processes such as cell behavior, energy metabolism, and immune regulation, which plays an essential role in maintaining normal development and physiological functions of the body. DPP8/9 is also involved in pathological processes such as tumorigenesis, inflammation, and organ fibrosis. In recent years, related research on immune cell pyroptosis has made DPP8/9 a new potential target for the treatment of hematological diseases. In addition, DPP8/9 inhibitors also have great potential in the treatment of tumors and chronic kidney disease.
Soaking of macromolecular crystals allows the formation of complexes via diffusion of molecules into a preformed crystal for structural analysis. Soaking offers various advantages over co-crystallization, e.g. small samples and high-throughput experimentation. However, this method has disadvantages, such as inducing mechanical stress on crystals and reduced success rate caused by low affinity/solubility of the ligand. To bypass these issues, the Picodropper was previously developed in the authors' laboratory. This technique aimed to deliver small volumes of compound solution in response to crystal dehydration supported by the Free Mounting System humidity control or by IR-laser-induced protein crystal transformation. Herein, a new related soaking development, the Aerosol-Generator, is introduced. This device delivers compounds onto the solution-free surface of protein crystals using an ultrasonic technique. The produced aerosol stream enables an easier and more accurate control of solution volumes, reduced crystal handling, and crystal-size-independent soaking. The Aerosol-Generator has been used to produce complexes of DPP8 crystals, where otherwise regular soaking did not achieve complex formation. These results demonstrate the potential of this device in challenging ligand-binding scenarios and contribute to further understanding of DPP8 inhibitor binding.
BACKGROUND: DPP8 and DPP9 have been demonstrated to play important roles in multiple diseases. Evidence for increased gene expression of DPP8 and DPP9 in tubulointerstitium was found to be associated with the decline of kidney function in chronic kidney disease (CKD) patients, which was observed in the Nephroseq human database. To examine the role of DPP8 and DPP9 in the tubulointerstitial injury, we determined the efficacy of DPP8 and DPP9 on epithelial-to-mesenchymal transition (EMT) and tubulointerstitial fibrosis (TIF) as well as the underlying mechanisms. METHODS: We conducted the immunofluorescence of DPP8 and DPP9 in kidney biopsy specimens of CKD patients, established unilateral ureteral obstruction (UUO) animal model, treated with TC-E5007 (a specific inhibitor of both DPP8 and DPP9) or Saxagliptin (positive control) or saline, and HK-2 cells model. RESULTS: We observed the significantly increased expression of DPP8 and DPP9 in the renal proximal tubule epithelial cells of CKD patients compared to the healthy control subjects. DPP8/DPP9 inhibitor TC-E5007 could significantly attenuate the EMT and extracellular matrix (ECM) synthesis in UUO mice, all these effects were mediated via interfering with the TGF-beta1/Smad signaling. TC-E5007 treatment also presented reduced renal inflammation and improved renal function in the UUO mice compared to the placebo-treated UUO group. Furthermore, the siRNA for DPP8 and DPP9, and TC-E5007 treatment decreased EMT- and ECM-related proteins in TGF-beta1-treated HK-2 cells respectively, which could be reversed significantly by transduction with lentivirus-DPP8 and lentivirus-DPP9. CONCLUSION: These data obtained provide evidence that the DPP8 and DPP9 could be potential therapeutic targets against TIF.
Several cytosolic pattern-recognition receptors (PRRs) form multiprotein complexes called canonical inflammasomes in response to intracellular danger signals. Canonical inflammasomes recruit and activate caspase-1 (CASP1), which in turn cleaves and activates inflammatory cytokines and gasdermin D (GSDMD), inducing pyroptotic cell death. Inhibitors of the dipeptidyl peptidases DPP8 and DPP9 (DPP8/9) activate both the human NLRP1 and CARD8 inflammasomes. NLRP1 and CARD8 have different N-terminal regions but have similar C-terminal regions that undergo autoproteolysis to generate two non-covalently associated fragments. Here, we show that DPP8/9 inhibition activates a proteasomal degradation pathway that targets disordered and misfolded proteins for destruction. CARD8's N terminus contains a disordered region of -160 amino acids that is recognized and destroyed by this degradation pathway, thereby freeing its C-terminal fragment to activate CASP1 and induce pyroptosis. Thus, CARD8 serves as an alarm to signal the activation of a degradation pathway for disordered and misfolded proteins.
Dipeptidyl peptidase 9 (DPP9) is a serine protease cleaving N-terminal dipeptides preferentially post-proline with (patho)physiological roles in the immune system and cancer. Only few DPP9 substrates are known. Here we identify an association of human DPP9 with the tumour suppressor BRCA2, a key player in repair of DNA double-strand breaks that promotes the formation of RAD51 filaments. This interaction is triggered by DNA-damage and requires access to the DPP9 active-site. We present crystallographic structures documenting the N-terminal Met1-Pro2 of a BRCA21-40 peptide captured in the DPP9 active-site. Mechanistically, DPP9 targets BRCA2 for degradation by the N-degron pathway, and promotes RAD51 foci formation. Both processes are phenocopied by BRCA2 N-terminal truncation mutants, indicating that DPP9 regulates both stability and the cellular stoichiometric interactome of BRCA2. Consistently, DPP9-deprived cells are hypersensitive to DNA-damage. Together, we identify DPP9 as a regulator of BRCA2, providing a possible explanation for DPP9 involvement in cancer development.
Dipeptidyl peptidase 9 (DPP9) was recently identified as fusion gene in ovarian high-grade serous carcinoma (HGSC). The aim of this study was to analyze the expression and clinical relevance of DPP8 and DPP9 in ovarian carcinoma, with focus on HGSC. mRNA expression by qRT-PCR of DPP8 and DPP9 was analyzed in 232 carcinomas, including 114 effusions and 118 surgical specimens (89 ovarian, 29 solid metastases). DPP8 and DPP9 protein expression was analyzed in 92 effusions. DPP8 and DPP9 mRNA was overexpressed in effusions compared to solid lesions in analysis of all histotypes (p < 0.001 both), as well as in analysis limited to HGSC (p < 0.001 for DPP9, p = 0.002 for DPP8). DPP9 mRNA was additionally overexpressed in HGSC compared to other histotypes (p = 0.021). DPP8 and DPP9 protein was expressed in carcinoma cells in 31/92 (37%) and 81/92 (88%) effusions, respectively. DPP8 protein expression in HGSC effusions was significantly related to better (complete) chemoresponse at diagnosis (p = 0.005). DPP8 and DPP9 mRNA and protein expression was unrelated to survival in analysis of the entire effusion cohort. However, higher DPP9 mRNA levels were significantly related to longer overall survival in pre-chemotherapy effusions (p = 0.049). In conclusion, DPP8 and DPP9 mRNA is frequently expressed in ovarian carcinoma, whereas DPP9 is more frequently expressed at the protein level. DPP8 and DPP9 may be related to less aggressive disease in advanced-stage HGSC.
Intracellular pathogenic structures or activities stimulate the formation of inflammasomes, which recruit and activate caspase-1 and trigger an inflammatory form of cell death called pyroptosis. The well-characterized mammalian inflammasome sensor proteins all detect one specific type of signal, for example double-stranded DNA or bacterial flagellin. Remarkably, NLRP1 was the first protein discovered to form an inflammasome, but the pathogenic signal that NLRP1 detects has not yet been identified. NLRP1 is highly polymorphic, even among inbred rodent strains, and it has been suggested that these diverse NLRP1 alleles may have evolved to detect entirely different stimuli. Intriguingly, inhibitors of the serine proteases DPP8 and DPP9 (DPP8/9) were recently shown to activate human NLRP1, its homolog CARD8, and several mouse NLRP1 alleles. Here, we show now that DPP8/9 inhibitors activate all functional rodent NLRP1 alleles, indicating that DPP8/9 inhibition induces a signal detected by all NLRP1 proteins. Moreover, we discovered that the NLRP1 allele sensitivities to DPP8/9 inhibitor-induced and Toxoplasma gondii-induced pyroptosis are strikingly similar, suggesting that DPP8/9 inhibition phenocopies a key activity of T. gondii. Overall, this work indicates that the highly polymorphic NLRP1 inflammasome indeed senses a specific signal like the other mammalian inflammasomes.
Dipeptidyl peptidases (DPPs) are proteolytic enzymes that are ideal therapeutic targets in human diseases. Indeed, DPP4 inhibitors are widely used in clinical practice as anti-diabetic agents. In this paper, we show that DPP4 inhibitors also induced cell death in multiple human myeloma cells. Among five DPP4 inhibitors, only two of them, vildagliptin and saxagliptin, exhibited apparent cytotoxic effects on myeloma cell lines, without any difference in suppression of DPP4 activity. As these two DPP4 inhibitors are known to have off-target effects against DPP8/9, we employed the specific DPP8/9 inhibitor 1G244. 1G244 demonstrated anti-myeloma effects on several cell lines and CD138+ cells from patients as well as in murine xenograft model. Through siRNA silencing approach, we further confirmed that DPP8 but not DPP9 is a key molecule in inducing cell death induced by DPP8/9 inhibition. In fact, the expression of DPP8 in CD38+ cells from myeloma patients was higher than that of healthy volunteers. DPP8/9 inhibition induced apoptosis, as evidenced by activated form of PARP, caspases-3 and was suppressed by the pan-caspase inhibitor Z-VAD-FMK. Taken together, these results indicate that DPP8 is a novel therapeutic target for myeloma treatment.
Activating germline mutations in the human inflammasome sensor NLRP1 causes palmoplantar dyskeratosis and susceptibility to Mendelian autoinflammatory diseases. Recent studies have shown that the cytosolic serine dipeptidyl peptidases DPP8 and DPP9 suppress inflammasome activation upstream of NLRP1 and CARD8 in human keratinocytes and peripheral blood mononuclear cells. Moreover, pharmacological inhibition of DPP8/DPP9 protease activity was shown to induce pyroptosis in murine C57BL/6 macrophages without eliciting other inflammasome hallmark responses. Here, we show that DPP8/DPP9 inhibition in macrophages that express a Bacillus anthracis lethal toxin (LeTx)-sensitive Nlrp1b allele triggered significantly accelerated pyroptosis concomitant with caspase-1 maturation, ASC speck assembly, and secretion of mature IL-1beta and IL-18. Genetic ablation of ASC prevented DPP8/DPP9 inhibition-induced caspase-1 maturation and partially hampered pyroptosis and inflammasome-dependent cytokine release, whereas deletion of caspase-1 or gasdermin D triggered apoptosis in the absence of IL-1beta and IL-18 secretion. In conclusion, blockade of DPP8/DPP9 protease activity triggers rapid pyroptosis and canonical inflammasome hallmarks in primary macrophages that express a LeTx-responsive Nlrp1b allele.
Small-molecule inhibitors of the serine dipeptidases DPP8 and DPP9 (DPP8/9) induce a lytic form of cell death called pyroptosis in mouse and human monocytes and macrophages(1,2). In mouse myeloid cells, Dpp8/9 inhibition activates the inflammasome sensor Nlrp1b, which in turn activates pro-caspase-1 to mediate cell death(3), but the mechanism of DPP8/9 inhibitor-induced pyroptosis in human myeloid cells is not yet known. Here we show that the CARD-containing protein CARD8 mediates DPP8/9 inhibitor-induced pro-caspase-1-dependent pyroptosis in human myeloid cells. We further show that DPP8/9 inhibitors induce pyroptosis in the majority of human acute myeloid leukemia (AML) cell lines and primary AML samples, but not in cells from many other lineages, and that these inhibitors inhibit human AML progression in mouse models. Overall, this work identifies an activator of CARD8 in human cells and indicates that its activation by small-molecule DPP8/9 inhibitors represents a new potential therapeutic strategy for AML.
Val-boroPro (PT-100, Talabostat) induces powerful anti-tumor immune responses in syngeneic cancer models, but its mechanism of action has not yet been established. Val-boroPro is a non-selective inhibitor of post-proline-cleaving serine proteases, and the inhibition of the highly related cytosolic serine proteases Dpp8 and Dpp9 (Dpp8/9) by Val-boroPro was recently demonstrated to trigger an immunostimulatory form of programmed cell death known as pyroptosis selectively in monocytes and macrophages. Here we show that Dpp8/9 inhibition activates the inflammasome sensor protein Nlrp1b, which in turn activates pro-caspase-1 to mediate pyroptosis. This work reveals a previously unrecognized mechanism for activating an innate immune pattern recognition receptor and suggests that Dpp8/9 serve as an intracellular checkpoint to restrain Nlrp1b and the innate immune system.
Dipeptidyl peptidases 8 and 9 are intracellular N-terminal dipeptidyl peptidases (preferentially postproline) associated with pathophysiological roles in immune response and cancer biology. While the DPP family member DPP4 is extensively characterized in molecular terms as a validated therapeutic target of type II diabetes, experimental 3D structures and ligand-/substrate-binding modes of DPP8 and DPP9 have not been reported. In this study we describe crystal and molecular structures of human DPP8 (2.5 A) and DPP9 (3.0 A) unliganded and complexed with a noncanonical substrate and a small molecule inhibitor, respectively. Similar to DPP4, DPP8 and DPP9 molecules consist of one beta-propeller and alpha/beta hydrolase domain, forming a functional homodimer. However, they differ extensively in the ligand binding site structure. In intriguing contrast to DPP4, where liganded and unliganded forms are closely similar, ligand binding to DPP8/9 induces an extensive rearrangement at the active site through a disorder-order transition of a 26-residue loop segment, which partially folds into an alpha-helix (R-helix), including R160/133, a key residue for substrate binding. As vestiges of this helix are also seen in one of the copies of the unliganded form, conformational selection may contributes to ligand binding. Molecular dynamics simulations support increased flexibility of the R-helix in the unliganded state. Consistently, enzyme kinetics assays reveal a cooperative allosteric mechanism. DPP8 and DPP9 are closely similar and display few opportunities for targeted ligand design. However, extensive differences from DPP4 provide multiple cues for specific inhibitor design and development of the DPP family members as therapeutic targets or antitargets.
Val-boroPro (Talabostat, PT-100), a nonselective inhibitor of post-proline cleaving serine proteases, stimulates mammalian immune systems through an unknown mechanism of action. Despite this lack of mechanistic understanding, Val-boroPro has attracted substantial interest as a potential anticancer agent, reaching phase 3 trials in humans. Here we show that Val-boroPro stimulates the immune system by triggering a proinflammatory form of cell death in monocytes and macrophages known as pyroptosis. We demonstrate that the inhibition of two serine proteases, DPP8 and DPP9, activates the pro-protein form of caspase-1 independent of the inflammasome adaptor ASC. Activated pro-caspase-1 does not efficiently process itself or IL-1beta but does cleave and activate gasdermin D to induce pyroptosis. Mice lacking caspase-1 do not show immune stimulation after treatment with Val-boroPro. Our data identify what is to our knowledge the first small molecule that induces pyroptosis and reveals a new checkpoint that controls the activation of the innate immune system.
        
Title: Pyroptosis and Apoptosis Pathways Engage in Bidirectional Crosstalk in Monocytes and Macrophages Taabazuing CY, Okondo MC, Bachovchin DA Ref: Cell Chemical Biology, 24:507, 2017 : PubMed
Pyroptosis is a lytic form of programmed cell death mediated by the inflammatory caspase-1, -4, and -5. We recently discovered that small-molecule inhibitors of the serine peptidases DPP8 and DPP9 (DPP8/9) induce pro-caspase-1-dependent pyroptosis in monocytes and macrophages. Notably, DPP8/9 inhibitors, unlike microbial agents, absolutely require caspase-1 to induce cell death. Therefore, DPP8/9 inhibitors are useful probes to study caspase-1 in cells. Here, we show that, in the absence of the pyroptosis-mediating substrate gasdermin D (GSDMD), caspase-1 activates caspase-3 and -7 and induces apoptosis, demonstrating that GSDMD is the only caspase-1 substrate that induces pyroptosis. Conversely, we found that, during apoptosis, caspase-3/-7 specifically block pyroptosis by cleaving GSDMD at a distinct site from the inflammatory caspases that inactivates the protein. Overall, this work reveals bidirectional crosstalk between apoptosis and pyroptosis in monocytes and macrophages, further illuminating the complex interplay between cell death pathways in the innate immune system.
        
Title: The SUMO1-E67 Interacting Loop Peptide Is an Allosteric Inhibitor of the Dipeptidyl Peptidases 8 and 9 Pilla E, Kilisch M, Lenz C, Urlaub H, Geiss-Friedlander R Ref: Journal of Biological Chemistry, 288:32787, 2013 : PubMed
The intracellular peptidases dipeptidyl peptidase (DPP) 8 and DPP9 are involved in multiple cellular pathways including antigen maturation, cellular homeostasis, energy metabolism, and cell viability. Previously we showed that the small ubiquitin-like protein modifier SUMO1 interacts with an armlike structure in DPP9, leading to allosteric activation of the peptidase. Here we demonstrate that the E67-interacting loop (EIL) peptide, which corresponds to the interaction surface of SUMO1 with DPP9, acts as a noncompetitive inhibitor of DPP9. Moreover, by analyzing the sensitivity of DPP9 arm mutants to the EIL peptide, we mapped specific residues in the arm that are important for inhibition by the EIL, suggesting that the peptide acts as an allosteric inhibitor of DPP9. By modifying the EIL peptide, we constructed peptide variants with more than a 1,000-fold selectivity toward DPP8 (147 nm) and DPP9 (170 nm) over DPPIV (200 mum). Furthermore, application of these peptides to cells leads to a clear inhibition of cellular prolyl peptidase activity. Importantly, in line with previous publications, inhibition of DPP9 with these novel allosteric peptide inhibitors leads to an increase in EGF-mediated phosphorylation of Akt. This work highlights the potential use of peptides that mimic interaction surfaces for modulating enzyme activity.
This work represents the first directed study to identify modification points in the topology of a representative DPP8/9-inhibitor, capable of rendering selectivity for DPP8 over DPP9. The availability of a DPP8-selective compound would be highly instrumental for studying and untwining the biological roles of DPP8 and DPP9 and for the disambiguation of biological effects of nonselective DPP-inhibitors that have mainly been ascribed to blocking of DPPIV's action. The cell-permeable DPP8/9-inhibitor 7 was selected as a lead and dissected into several substructures that were modified separately for evaluating their potential to contribute to selectivity. The obtained results, together with earlier work from our group, clearly narrow down the most probable DPP8-selectivity imparting modification points in DPP8/9 inhibitors to parts of space that are topologically equivalent to the piperazine ring system in 7. This information can be considered of high value for future design of compounds with maximal DPP8 selectivity.
        
Title: Hydrophilic residues surrounding the S1 and S2 pockets contribute to dimerisation and catalysis in human dipeptidyl peptidase 8 (DP8) Pitman MR, Menz RI, Abbott CA Ref: Biol Chem, 391:959, 2010 : PubMed
Dipeptidyl peptidase (DP) 8 belongs to the dipeptidyl peptidase IV gene family. DP8 has been implicated in immune function and asthma, although its biological function is yet unknown. Structures of the homologs, fibroblast activation protein (FAP) and DPIV, are known but the DP8 structure is yet to be resolved. To help characterise the DP8 substrate pocket, mutants of residues lining the pocket were produced at DP8(D772), DP8(Y315), DP8(H434) and DP8(D435) and assessed by substrate kinetics and size-exclusion chromatography. Mutations of DP8(D772A/E/S/V) affected catalysis but did not confer endopeptidase activity. Mutations of DP8(H434F), DP8(D435F) and DP8(Y315F) reduced catalytic activity. Furthermore, mutations to DP8(D772A/E/S/V), DP8(H434F), DP8(D435F) and DP8(Y315F) affected dimer stabilisation. Homology modelling of DP8 using DPIV and FAP crystal structures suggested that DP8(D772), DP8(H434) and DP8(D435) were located at the edge of the S2 catalytic pocket, contributing to the junction between the alpha-beta hydrolase and beta-propeller domains. This study provides insights into how the DP8 substrate pocket and dimer interface differ from DPIV and FAP which could be utilised for designing more selective DP8 inhibitors.
The dipeptidyl peptidase IV (DPIV) enzyme family contains both potential and proven therapeutic targets. Recent reports indicate the presence of DP8 and DP9 in peripheral blood lymphocytes, testis, lung, and brain. For a more comprehensive understanding of DP8 and DP9 tissue and cellular expression, mRNA and enzyme activity were examined. Many organs from C57BL/6 wild-type and DPIV gene-knockout mice were examined; DP8/9 enzyme activity was detected in the immune system, brain, testis, muscle, and epithelia. In situ hybridization localized DP8 and DP9 mRNA to lymphocytes and epithelial cells in liver, gastrointestinal tract, lymph node, spleen, and lung. DP8 and DP9 mRNA was detected in baboon and mouse testis, and DP9 expression was elevated in human testicular cancers. DP8 and DP9 mRNA were ubiquitous in day 17 mouse embryo, with greatest expression in epithelium (skin and gastrointestinal tract) and brain. Thus, DP8 and DP9 are widely expressed enzymes. Their expression in lymphocytes and epithelia indicates potential for roles in the digestive and immune systems. This manuscript contains online supplemental material at http://www.jhc.org. Please visit this article online to view these materials.
        
Title: Investigation of the dimer interface and substrate specificity of prolyl dipeptidase DPP8 Lee HJ, Chen YS, Chou CY, Chien CH, Lin CH, Chang GG, Chen X Ref: Journal of Biological Chemistry, 281:38653, 2006 : PubMed
DPP8 belongs to the family of prolyl dipeptidases, which are capable of cleaving the peptide bond after a penultimate proline residue. Unlike DPP-IV, a drug target for type II diabetes, no information is available on the crystal structure of DPP8, the regulation of its enzymatic activity, or its substrate specificity. In this study, using analytical ultracentrifugation and native gel electrophoresis, we show that the DPP8 protein is predominantly dimeric when purified or in the cell extracts. Four conserved residues in the C-terminal loop of DPP8 (Phe(822), Val(833), Tyr(844), and His(859)), corresponding to those located at the dimer interface of DPP-IV, were individually mutated to Ala. Surprisingly, unlike DPP-IV, these single-site mutations abolished the enzymatic activity of DPP8 without disrupting its quaternary structure, indicating that dimerization itself is not sufficient for the optimal enzymatic activity of DPP8. Moreover, these mutations not only decreased k(cat), as did the corresponding DPP-IV mutations, but also dramatically increased K(m). We further show that the K(m) effect is independent of the substrate assayed. Finally, we identified the distinctive and strict substrate selectivity of DPP8 for hydrophobic or basic residues at the P2 site, which is in sharp contrast to the much less discriminative substrate specificity of DPP-IV. Our study has identified the residues absolutely required for the optimal activity of DPP8 and its unique substrate specificity. This study extends the functional importance of the C-terminal loop to the whole family of prolyl dipeptidases.
        
Title: DP8 and DP9 have extra-enzymatic roles in cell adhesion, migration and apoptosis Yu DM, Wang XM, Ajami K, McCaughan GW, Gorrell MD Ref: Advances in Experimental Medicine & Biology, 575:63, 2006 : PubMed
The dipeptidyl peptidase IV gene family contains the four peptidases dipeptidyl peptidase IV, fibroblast activation protein, dipeptidyl peptidase 8 and dipeptidyl peptidase 9. Dipeptidyl peptidase IV and fibroblast activation protein are involved in cell-extracellular matrix interactions and tissue remodeling. Fibroblast activation protein is upregulated and dipeptidyl peptidase IV is dysregulated in chronic liver disease. The effects of dipeptidyl peptidase 8 and dipeptidyl peptidase 9 on cell adhesion, cell migration, wound healing and apoptosis were measured by using green fluorescent protein fusion proteins to identify transfected cells. Dipeptidyl peptidase 9-overexpressing cells exhibited impaired cell adhesion, migration in transwells and monolayer wound healing on collagen I, fibronectin and Matrigel. Dipeptidyl peptidase 8-overexpressing cells exhibited impaired cell migration on collagen I and impaired wound healing on collagen I and fibronectin in comparison to the green fluorescent protein-transfected controls. Dipeptidyl peptidase 8 and dipeptidyl peptidase 9 enhanced induced apoptosis, and dipeptidyl peptidase 9 overexpression increased spontaneous apoptosis. Mechanistic investigations showed that neither the catalytic serine of dipeptidyl peptidase 8 or dipeptidyl peptidase 9 nor the Arg-Gly-Asp integrin-binding motif in dipeptidyl peptidase 9 were required for the impairment of cell survival, cell adhesion or wound healing. We have previously shown that the in vitro roles of dipeptidyl peptidase IV and fibroblast activation protein in cell-extracellular matrix interactions and apoptosis are similarly independent of catalytic activity. Dipeptidyl peptidase 9 overexpression reduced beta-catenin, tissue inhibitor of matrix metalloproteinases 2 and discoidin domain receptor 1 expression. This is the first demonstration that dipeptidyl peptidase 8 and dipeptidyl peptidase 9 influence cell-extracellular matrix interactions, and thus may regulate tissue remodeling.
Here we present a finished sequence of human chromosome 15, together with a high-quality gene catalogue. As chromosome 15 is one of seven human chromosomes with a high rate of segmental duplication, we have carried out a detailed analysis of the duplication structure of the chromosome. Segmental duplications in chromosome 15 are largely clustered in two regions, on proximal and distal 15q; the proximal region is notable because recombination among the segmental duplications can result in deletions causing Prader-Willi and Angelman syndromes. Sequence analysis shows that the proximal and distal regions of 15q share extensive ancient similarity. Using a simple approach, we have been able to reconstruct many of the events by which the current duplication structure arose. We find that most of the intrachromosomal duplications seem to share a common ancestry. Finally, we demonstrate that some remaining gaps in the genome sequence are probably due to structural polymorphisms between haplotypes; this may explain a significant fraction of the gaps remaining in the human genome.
DPP8 is a prolyl dipeptidase homologous to DPP-IV, which is a drug target for Type II diabetes. The biological function of DPP8 is not known. To identify potent and selective chemical compounds against DPP8, we have synthesized a series of isoquinoline and isoindoline derivatives and have tested their inhibitory activity against DPP8, DPP-IV and DPP-II. Isoindoline derivatives were found to be more potent DPP8 inhibitors than isoquinoline derivatives. Isoindoline with a 1-(4,4'-difluor-benzhydryl)-piperazine group at the P2 site was observed to be a very potent DPP8 inhibitor, having an IC(50) value of 14nM with at least a 2500-fold selectivity over either DPP-IV or DPP-II. From SAR results, we speculate that the S1 site of DPP8 may be larger than that of DPP-IV, which would allow the accommodation of larger C-terminal residues, such as isoquinoline or isoindoline.
DPP8 is a new member of the prolyl dipeptidases, many of which have important biological functions in vivo. DPP8 catalyzes the cleavage at the carboxyl side of the proline residue at the penultimate position. To study its structure and biochemical properties, we have overexpressed the human DPP8 protein in baculovirus infected Sf9 cells. The protein is soluble and can be purified to homogeneity. Using the chromogenic H-Gly-Pro-pNA as the substrate, a kinetic study shows that purified DPP8 is active and has a similar kcat value as that of DPP-IV, a prolyl dipeptidase that is a drug target for type II diabetes. The kinetic constants of DPP8 are also determined for other chromogenic substrates, and the results indicate that DPP8 has substrate preference at both the P1 and P2 sites. The expression system provides means of better understanding the structure, catalytic mechanism, and biological function of DPP8 protein.
Dipeptidyl peptidase IV (DP-IV/CD26), fibroblast activation protein (FAP), DP-like 1 (DPL1), DP8, DP9, and DPL2 comprise the CD26 gene family. CD26/DP-IV has roles in liver disease, T cell costimulation, chemokine biology, type II diabetes, and tumor biology. DPIV substrates include the glucagonlike peptides, neuropeptide Y, and the chemokines CCL3, CCL5, CCL11, CCL22, and CXCL12. We have proposed that the extracellular region of CD26 is analogous to prolyl oligopeptidase in consisting of an alpha/beta hydrolase domain contributed by both N- and C-terminal portions of the polypeptide and a seven-blade beta-propeller domain. Replacing the C-terminal portion of the predicted alpha/beta hydrolase domain of CD26 (residues 501-766) with the homologous portion of DP8 or DP9 produced intact proteins. However, these chimeric proteins lacked dimerization and peptidase activity, suggesting that CD26 dimerization requires the C-terminal portion of the alpha/beta hydrolase domain. Deleting some N-terminal residues of the alpha/beta hydrolase domain of CD26 ablated peptidase activity and greatly diminished cell surface expression. Together with previous data that CD26 peptidase activity requires the C-terminal 20 residues, this suggests that peptidase activity requires the entire alpha/beta hydrolase domain. The catalytic triad of DP8 was shown to be Ser(739)-Asp (817)-His(849). Glu(259) of DP8, a residue distant from the catalytic triad yet greatly conserved in the CD26 gene family, was shown to be required for peptidase activity. These data concord with our predicted CD26 structure, indicate that biosynthesis of a functional fragment of CD26 is difficult, and confirm the functional homology of DP8 with CD26.
        
Title: Cloning and characterization of dipeptidyl peptidase 10, a new member of an emerging subgroup of serine proteases Qi SY, Riviere PJ, Trojnar J, Junien JL, Akinsanya KO Ref: Biochemical Journal, 373:179, 2003 : PubMed
Two dipeptidyl peptidase IV (DPPIV, DPP4)-related proteins, DPP8 and DPP9, have been identified recently [Abbott, Yu, Woollatt, Sutherland, McCaughan, and Gorrell (2000) Eur. J. Biochem. 267, 6140-6150; Olsen and Wagtmann (2002) Gene 299, 185-193; Qi, Akinsanya, Riviere, and Junien (2002) Patent application WO0231134]. In the present study, we describe the cloning of DPP10, a novel 796-amino-acid protein, with significant sequence identity to DPP4 (32%) and DPP6 (51%) respectively. We propose that DPP10 is a new member of the S9B serine proteases subfamily. The DPP10 gene is located on the long arm of chromosome 2 (2q12.3-2q14.2), close to the DPP4 (2q24.3) and FAP (2q23) genes. The active-site serine residue is replaced by a glycine residue in DPP10, resulting in the loss of DPP activity. The serine residue is also replaced in DPP6, which lacks peptidase activity. DPP8 and DPP9 share an identical active site with DPP4 (Gly-Trp-Ser-Tyr-Gly). In contrast with the previous results suggesting that DPP9 is inactive, we show that DPP9 is a DPP, hydrolysing Ala-Pro-(7-amino-4-methyl-coumarin) with similar pH-specificity and protease-inhibitor-sensitivity to those of DPP4 and DPP8. Northern-blot analysis shows that whereas DPP8 and DPP9 are widely expressed, DPP10 is expressed mainly in the brain and pancreas. DPP6, which has the highest amino acid identity with DPP10, has been shown previously [Nadal, Ozaita, Amarillo, de Miera, Ma, Mo, Goldberg, Misumi, Ikehara, Neubert et al. (2003) Neuron 37, 449-461] to associate with A-type K(+) channel subunits, modulating their transport and function in somatodendritic compartments of neurons. It is possible that DPP10 is involved in similar functions in the brain. Elucidation of the physiological or pathophysiological role of DPP8, DPP9 and DPP10 and characterization of their structure-function relationships will add impetus to the development of inhibitor molecules for pharmacological or therapeutic use.
Dipeptidyl peptidase (DPP) IV has roles in T-cell costimulation, chemokine biology, type-II diabetes and tumor biology. Fibroblast activation protein (FAP) has been implicated in tumor growth and cirrhosis. Here we describe DPP8, a novel human postproline dipeptidyl aminopeptidase that is homologous to DPPIV and FAP. Northern-blot hybridization showed that the tissue expression of DPP8 mRNA is ubiquitous, similar to that of DPPIV. The DPP8 gene was localized to chromosome 15q22, distinct from a closely related gene at 19p13.3 which we named DPP9. The full-length DPP8 cDNA codes for an 882-amino-acid protein that has about 27% identity and 51% similarity to DPPIV and FAP, but no transmembrane domain and no N-linked or O-linked glycosylation. Western blots and confocal microscopy of transfected COS-7 cells showed DPP8 to be a 100-kDa monomeric protein expressed in the cytoplasm. Purified recombinant DPP8 hydrolyzed the DPPIV substrates Ala-Pro, Arg-Pro and Gly-Pro. Thus recombinant DPP8 shares a postproline dipeptidyl aminopeptidase activity with DPPIV and FAP. DPP8 enzyme activity had a neutral pH optimum consistent with it being nonlysosomal. The similarities between DPP8 and DPPIV in tissue expression pattern and substrates suggests a potential role for DPP8 in T-cell activation and immune function.