key: cord-0788982-mqq6fmsp authors: Waumans, Yannick; Baerts, Lesley; Kehoe, Kaat; Lambeir, Anne-Marie; De Meester, Ingrid title: The Dipeptidyl Peptidase Family, Prolyl Oligopeptidase, and Prolyl Carboxypeptidase in the Immune System and Inflammatory Disease, Including Atherosclerosis date: 2015-08-07 journal: Front Immunol DOI: 10.3389/fimmu.2015.00387 sha: 474dc8b54f9110f60b129220549c532355fe10b2 doc_id: 788982 cord_uid: mqq6fmsp Research from over the past 20 years has implicated dipeptidyl peptidase (DPP) IV and its family members in many processes and different pathologies of the immune system. Most research has been focused on either DPPIV or just a few of its family members. It is, however, essential to consider the entire DPP family when discussing any one of its members. There is a substantial overlap between family members in their substrate specificity, inhibitors, and functions. In this review, we provide a comprehensive discussion on the role of prolyl-specific peptidases DPPIV, FAP, DPP8, DPP9, dipeptidyl peptidase II, prolyl carboxypeptidase, and prolyl oligopeptidase in the immune system and its diseases. We highlight possible therapeutic targets for the prevention and treatment of atherosclerosis, a condition that lies at the frontier between inflammation and cardiovascular disease. Research from over the past 20 years has implicated the dipeptidyl peptidase (DPP) family in various physiological processes and pathologies of the immune system. Usually only four prolyl-specific peptidases are considered: DPPIV (EC 3.4.14.5), fibroblast activation protein α (FAP; EC 3.4.21.B28), and the more recently discovered DPP8 and DPP9 (EC 3.4.14). However, due to similarities in substrate specificity and structural homology, it is more relevant to consider a broader family that also includes prolyl oligopeptidase (PREP; EC 3.4.21.26), dipeptidyl peptidase II (DPPII) (EC 3.4.14.2), and prolyl carboxypeptidase (PRCP; EC 3.4.16.2). First, DPPII and PRCP share the α/β hydrolase fold with the other DPPs and the catalytic triad is completely conserved in both enzymes (2) . Moreover, DPPII can cleave several DPPIV substrates in vitro (3) . Conversely, due to its substrate preference for tripeptides (4) , DPPII could actually be considered as a prolyl carboxytripeptidase, emphasizing its similarities to PRCP. Another argument for considering a broader family stems from the fact that functional studies on the role of peptidases rely heavily on the use of enzyme inhibitors and many of the inhibitors used in earlier studies are now known to inhibit more than one family member. For example, early studies on DPPIV used inhibitors which we now know also inhibit DPPII, DPP8, DPP9, FAP, and/or PREP due to their sequential and/or structural similarity [e.g., Ref. (5) (6) (7) (8) (9) ]. PRCP is known to be inhibited by KYP-2047 and Z-Pro-Prolinal at higher concentrations, which have often been used for the functional study of PREP [e.g., Ref. (10) (11) (12) ]. Table 1 summarizes the most commonly used DPP inhibitors and their selectivity compared to DPPIV. In view of the aforementioned reasons and for the sake of simplicity, we will use "DPP family" as a blanket term, which includes DPPII, PRCP, and PREP even though strictly speaking they are not DPPs. Figure 1 provides a general overview of this broadly defined DPP family. The roles of various family members in certain aspects of the immune system or immune dysfunction have been reviewed in the past [e.g., Ref. (13) (14) (15) ]. In this review, we provide a comprehensive discussion and update on the roles of DPPIV, DPPII, DPP8, DPP9, FAP, PREP, and PRCP in the immune system and inflammatory disease. We highlight the role of these enzymes in atherosclerosis, a condition that lies at the frontier between inflammation and cardiovascular disease, as the DPP family encompasses possible therapeutic targets for the prevention and treatment of this disease. The prototypical DPP, DPPIV (often DPP4 in medical jargon) cleaves off an N-terminal dipeptide from peptides with Pro or Ala on the penultimate position. Its localization as a soluble enzyme in body fluids, or anchored in the plasma membrane of cells provides it with the necessary access to cleave a wide range of bioactive peptides. As such, it can modify their biological activity. Glucagon-like peptide (GLP)-1 and -2, and glucosedependent insulinotropic peptide (GIP) (29, 30), substance P (31), neuropeptide Y (NPY) (32), stromal cell-derived factor-1α/β (SDF-1α/β or CXCL12) (33), granulocyte macrophage colony-stimulating factor (GM-CSF) (1), CXCL10 (34-36), and high-mobility group box 1 (HMGB1) (37) have been identified as physiological substrates, while others, such as RANTES, have been proposed based on in vitro experiments [e.g., Ref. (38)]. DPPIV also performs many of its physiological functions through interactions with other proteins, such as collagen, fibronectin, adenosine deaminase (ADA), caveolin-1, and the mannose-6-phosphate/insulin-like growth factor II receptor (M6P/IGFIIR) (39-41). Some of those will be discussed in more detail below. Dipeptidyl peptidase IV is well known for its role in glucose homeostasis. It has become a validated therapeutic target for the treatment of type 2 diabetes (T2D) (46). DPPIV inhibitors reduce the rate of GLP-1 inactivation (Boxes 1 and 2). It has also been shown to be involved in cancer biology. The role of the DPP family in cancer has been addressed in several other reviews (39, 47-51). Finally, DPPIV has recently come back into the center of attention as the receptor for the MERS coronavirus (52). The incretins are a group of glucose-lowering molecules produced by the intestines. The best known incretin is glucagon-like peptide-1 (GLP-1). This incretin is derived from proglucagon and secreted after a meal from L-cells in the distal ileum and colon. In the pancreas, it induces insulin secretion and biosynthesis while lowering glucagon secretion. In addition, GLP-1 increases the β-cell mass, thereby restoring insulin production. It is clear that GLP-1 also has functions outside glucose metabolism. Its receptor, GLP-1-R, is not only found in the pancreas but also expressed in brain, lung, kidney, stomach, and heart (42, 43). Recently, it was shown that stimulation after myocardial infarction reduces the infarct size (44, 45). Currently, GLP-1 agonists are approved for the treatment of type 2 diabetes. These incretin mimetics seem to have a slightly better efficacy as DPPIV inhibitors and lead more frequently to weight loss. Unfortunately, an important drawback for their therapeutic use is that they can only be administered by subcutaneous injection (46). Fibroblast activation protein α, also known as seprase can present itself as a type II transmembrane protein or as a shedded plasma protease (57). In the latter case, it is also known as antiplasmincleaving enzyme, which converts α2-antiplasmin into a more active form, suppressing fibrinolysis (58). Some of the known DPPIV substrates were later found to be cleaved in vitro by FAP as well (59), though any physiological relevance remains unclear. Unlike DPPIV, FAP also possesses a gelatinase activity. This enables FAP to degrade proteins of the extracellular matrix (60). This is of particular interest with regard to its involvement in a number of pathological processes (47). FAP is highly induced during inflammation, activation of hepatic stellate cells in liver cirrhosis and strongly expressed by mesenchymal cells of remodeling tissue (47, 61). FAP is also a key regulator during tumor growth and metastasis (47). As all these processes require degradation of the extracellular matrix, FAP's involvement in these pathologies is most likely associated with its gelatinase activity (51). Its role in cancer biology has been reviewed before (47, 62). It is interesting to note that, so far, in clinical trials Talabostat has shown minimal or no clinical benefit for the treatment of metastatic colorectal cancer, advanced non-small cell lung cancer, or stage IV melanoma (63-65). It should be mentioned, however, that Talabostat is a broad-range inhibitor also targeting DPPIV, DPP8, and DPP9. Dipeptidyl peptidases 8 and DPP9 show DPPIV-like activity and share a very high-sequence similarity to each other (77% aa similarity, 57% aa identity) (24). These cytoplasmic enzymes have several isoforms. It has been a matter of debate whether all are expressed as protein in cells and, if so, whether they are active (66-69). Interestingly, the N-terminal extension of the longer DPP9 variant contains a nuclear localization signal and, indeed, this form localizes to the nucleus (69). DPP8 has been shown to cleave a number of DPPIV chemokine substrates in vitro (70). Another DPPIV substrate, NPY, has indirectly been shown to be BOX 2 DPPIV inhibitors. Dipeptidyl peptidase IV inhibitors prolong the biological half-life of the incretins and are therefore used for the treatment of type 2 diabetes. Sitagliptin, vildagliptin, saxagliptin, linagliptin, and alogliptin are DPPIV inhibitors currently available on the market for treatment of type 2 diabetes. Sitagliptin and alogliptin are highly selective toward DPPIV in vitro, whereas vildagliptin and saxagliptin are less selective with regard to DPP8 and 9, and linagliptin with regard to FAP (28). Their clinical efficacy and safety in the use of type 2 diabetes seem comparable as far as can be judged from the data available. There is a growing interest toward a use outside type 2 diabetes as it has become clear that DPPIV inhibitors have pleiotropic effects. While negative effects have been found in heart failure (53), some studies suggest them as a possible therapeutic strategy in cardiovascular pathologies (28, 54). The SITAGRAMI trial and follow-up studies revealed that the combination of a DPPIV inhibitor with granulocyte-colony-stimulating factor or in monotherapy presents a therapeutic option after myocardial infarction (55, 56). As stated above, the mechanism is not yet clear but may be explained by a longer biological half-life of DPPIV substrates, glucagon-like peptide-1, B-type natriuretic peptide, and stromal cell-derived factor-1 α/β. All three peptides have a cardioprotective effect that is abolished by DPPIV-mediated cleavage. a DPP8 and DPP9 substrate as well (71). Efforts have been made to find intracellular DPP8 and 9 substrates using a peptidomic approach (72), but so far it has been hard to attribute physiological relevance to the possible substrates beyond the role of DPP8 and 9 in intracellular peptide turnover (73). The physiological functions of DPP8 and DPP9 are still not properly understood. Mainly, a lack of available knockout animals, specific inhibitors, and substrates has hampered progress (24). A mouse model has been established with a targeted inactivation of DPP9 enzymatic activity (74), but homozygous DPP9inactive neonates die within 8-24 h after birth. Despite these limitations, some indications toward their role are surfacing. Using immunohistochemistry, DPP8 and 9 were found associated with spermatozoids and spermatids and the short mRNA of DPP8 is predominantly expressed in testes (75, 76), suggesting a role in spermatogenesis and male fertility. Recent work has found SUMO1 to be an allosteric activator of DPP9 (77), whereas a small peptide corresponding to the interaction surface of SUMO1 is a non-competitive inhibitor of DPP8 and DPP9 (78). A genomewide association study has linked DPP9 to idiopathic pulmonary fibrosis (79). Finally, a number of studies have shown a role for DPP8 and DPP9 in apoptosis (71, 80-83). Two studies showed that overexpression enhanced induced apoptosis and impaired cell adhesion and migration (80, 81). Conversely, DPP8/9 inhibition in tumor cells decreased the number of viable cells because of a decreased cleavage of pro-apoptotic NPY (71). In macrophages, inhibition caused a marginal, yet significant increase in apoptosis, independent of NPY cleavage (82). Interestingly, vildagliptin, a DPPIV inhibitor already on the market to treat type 2 diabetes, but with poorer selectivity toward DPP8 and 9, was shown to enhance parthenolide's anti-leukemic activity through its inhibition of DPP8 and 9, and not DPPIV (83). Prolyl carboxypeptidase, also called angiotensinase C or lysosomal Pro-X carboxypeptidase, is a lysosomal carboxypeptidase sharing strong sequence homology with the likewise lysosomal DPPII (4, 84) . PRCP preferentially cleaves off the C-terminal amino acid when Ala or Pro is in the penultimate position, while DPPII targets N-terminal X-Pro or X-Ala dipeptides (85, 86). In addition to a structural similarity, PRCP and DPPII have partially overlapping substrate specificities due to DPPII's preference for tripeptide substrates (4) . Perhaps surprisingly, Gly-Pro-pNA and Ala-Pro-pNA, two typical synthetic DPP substrates, have actually been used to perform PRCP activity measurements (87). Prolyl carboxypeptidase is particularly known as one of the key enzymes of the renin-angiotensin system (RAS). It inactivates the vasoactive peptides angiotensin II and angiotensin III by cleaving off the C-terminal Phe (88). α-Melanocyt-stimulating hormone 1-13, an anorexigenic neuromodulator, is inactivated by PRCP, implying a role in body weight control (89). Based on the involvement of PRCP in the conversion of these peptide hormones, the enzyme has also been associated with diseases, such as hypertension, diabetes mellitus, obesity, inflammation, and cardiovascular dysfunction (90, 91). Dipeptidyl peptidase II has no known natural substrates. The DPPIV substrate substance P has been shown to be cleaved by DPPII in vitro (3), but much less efficiently, casting doubt over any physiological relevance. It has been shown that inhibition or silencing of DPPII causes apoptosis of quiescent G 0 lymphocytes (92-94). On the other hand, a highly specific DPPII inhibitor, UAMC00039, did not induce apoptosis, autophagy, or necrosis in human leukocytes (25, 95), but this study did not specifically look at quiescent cells or lymphocytes. Finally, changes in DPPII activity levels have been observed in a number of pathologies, such as neurodegenerative disorders, myopathies, cancer, and gastro-intestinal disorders (4). Prolyl oligopeptidase is an oligopeptidase with endopeptidase activity. It has been shown to be localized in the cytoplasm (96) (97) (98) (99) , but given its ability to inactivate several neuropeptides in vitro by limited proteolysis (100) (101) (102) (103) (104) (105) (106) (107) (108) (109) (110) (111) (112) (113) (114) (115) , its involvement in the in vivo generation of immunoactive peptides N-acetyl-prolyl-glycylproline and N-acetyl-seryl-aspartyl-lysyl-proline (116, 117) , and its presence in plasma (118, 119) , it most likely also has an extracellular role. Initial interest for PREP derived from the positive effects of PREP inhibitors on scopolamine-induced amnesia in rats (120) (121) (122) (123) . PREP inhibition was also found to promote neuronal survival and neurite outgrowth of cerebellar granule cells (124) . However, a recent study in mice shows that the lack of PREP in vivo causes a reduction of synaptic spine density in the hippocampal region along with reduced long-term potentiation and memory functions (125) . Many of PREP's functions are mediated through its interactions with other proteins. PREP is known to interact with GAP-43 (126, 127) , α-tubulin (96) , and GADPH (128) . Its most studied interaction is with α-synuclein (126), reviewed in Ref. (129) . PREP and α-synuclein have been shown to co-localize in cell models of stress and in the substantia nigra of post-mortem Parkinson's disease brain (11, 130) . In vitro, the aggregation rate of α-synuclein increases in the presence of high concentrations of PREP, which is abolished through active site inhibitors of PREP and absent with a catalytically impaired PREP mutant (131) . In vivo, PREP inhibition reduces α-synuclein aggregates in a cellular and animal model for Parkinson's disease (11) . The role of DPPIV in monocytes and macrophages has been somewhat contested. Whereas DPPIV's presence on monocytes and macrophages has been shown repeatedly in mice and rats (132) (133) (134) , its expression in human monocytes and macrophages is less obvious. Figure 2 shows an overview of the expression of DPPIV throughout the immune system. In visceral obesity, DPPIV expression is low on peripheral blood monocytes, macrophages, and dendritic cells, but it is upregulated in vitro Frontiers in Immunology | www.frontiersin.org August 2015 | Volume 6 | Article 387 after differentiation and activation of isolated monocytes into macrophages or dendritic cells, and in vivo locally in adipose tissue (135) . Interestingly, the authors showed that macrophage-or dendritic cell-associated DPPIV most likely binds ADA, promoting local degradation of adenosine, a T-cell proliferation suppressor, thereby inducing T-cell proliferation (135) . Three other studies also found no to low DPPIV expression or activity associated with human monocytes and/or macrophages (82, [136] [137] [138] . Others have investigated DPPIV in monocyte-or macrophage-like cell lines (136, 137, (139) (140) (141) (142) (143) (144) . In HL-60 cells, its expression has been found to be regulated by differentiation into macrophage-like cells (139) . DPPIV inhibitor alogliptin can affect ERK activation, MMP1 and IL-6 secretion in U937 cells (140, 141) . However, these studies employed alogliptin at concentrations lower than its IC 50 for DPPIV. It is therefore questionable whether the observed effects were mediated by DPPIV at all. On the other hand, proliferation is reduced in the presence of a DPP inhibitor in U937 cells expressing high levels of DPPIV, but not in the same cell type expressing low levels of DPPIV (144) . Moreover, the same inhibitor causes the former cells to secrete lower amounts of IL-1β, but higher amounts of TNFα (144) . It could be that inhibition merely increases TNFα's half-life, as DPPIV has been implicated in its degradation in U937 cells (137) . In THP-1 cells, DPPIV inhibitors alogliptin and sitagliptin both reduced these cells' chemotactic potential (142) . DPPIV inhibitors sitagliptin and NVPDPP728 also reduced NLRP3, TLR4, and IL-1β expression and increased GLP-1R expression in THP-1 cells and this effect was blocked through PMA differentiation (143) . Importantly, such cell lines have been derived from different types of myeloid leukemia, and as it is known that DPPIV expression is often dysregulated in cancer (47-51), the physiological relevance of these findings remains uncertain. FAP has been shown on tumor-associated macrophages in human breast cancer (145) . Dipeptidyl peptidase 8/9 activity has been found in human monocytes and U937 cells (136) . DPP8 was found associated with activated microglia/macrophages in a rat model of cerebral ischemia (146) . DPP8 and 9 are abundantly present in macrophage-rich regions of atherosclerotic plaques (82). Interestingly, DPP9 is upregulated after in vitro monocyte-to-macrophage differentiation. Moreover, inhibition or RNA silencing of DPP9 attenuates pro-inflammatory M1, but not M2, macrophage activation (82). In rats, DPPII is expressed in tissue-resident macrophages (147, 148) . Humans show DPPII activity in monocytes as well as U937 cells (25, 136) . Human blood derived alveolar macrophages show high-PRCP activity (138, 149) . Interestingly, in a mouse in vivo angiogenesis assay, macrophage infiltration into the wound was increased in mice with a PRCP deletion (150) . Prolyl oligopeptidase activity has been shown in mouse and rat peritoneal macrophages and in rat pulmonary macrophages (134, 151, 152) . Its activity in mouse peritoneal macrophages is increased after thioglycollate ellicitation (134) . In addition, PREP has been identified as a neurotoxic component in the supernatant of activated THP-1 cells, which are monocyte-like cells (153) . Apparently, these cells secrete PREP upon activation with IFNγ and LPS and partly because of this, their supernatant is toxic to neuroblastoma SH-SY5Y cells, as shown through the use of PREPspecific inhibitors (153) . PREP's mode of action in this remains unclear. Recently, a study showed that DPPIV acts as a chemorepellent for human and murine neutrophils (154) . Adding recombinant DPPIV to purified human neutrophils in an Insall chamber causes the neutrophils to migrate away from the higher concentration of DPPIV. This effect is blocked by DPPIV inhibitors, meaning that the effect is mediated through DPPIV's enzymatic activity, although a candidate substrate is not obvious. Moreover, in a mouse model of acute respiratory distress syndrome, oropharyngeal aspiration of DPPIV prevented accumulation of neutrophils in the lung (154) . By contrast, PREP is involved in the generation of prolyl-glycyl-proline, a collagen fragment that is an efficient neutrophil chemoattractant (155) . Human peripheral blood neutrophils contain PREP activity and are themselves capable of generating prolyl-glycyl-proline after LPS-activation, alluding to a self-sustaining pathway of neutrophil inflammation (116) . PRCP is also abundantly expressed in human neutrophils (90). The recruitment of eosinophils is affected by DPPIV activity. CCL11, also known as eotaxin, is a DPPIV substrate and cleavage by DPPIV prevents the activation of its receptor CCR3 (156) . In rats, it was shown that administration of CCL11 results in eosinophil recruitment and this recruitment is significantly more effective in DPPIV-deficient F344 mutants (156) . Finally, DPPII activity has been reported in the granules of mast cells in several publications (147, 148, 157) . It is released from peritoneal mast cells upon degranulation and is apparently inhibited by histamine and Zn 2+ at concentrations present in the granules of mast cells (157) . Dipeptidyl peptidase 4 is present in low amounts on freshly isolated human NK cells and its expression is only upregulated in a small subpopulation after IL-2 stimulation (158) . In that study, it was also shown that DPPIV inhibition suppresses DNA synthesis and cell cycle progression of NK cells, but these effects may be DPP8/9 mediated as the inhibitors used in that study are now known to also inhibit DPP8/9 activity (159) . Another study shows that DPPIV is actually only expressed by a small subpopulation of peripheral NK cells (160) . The natural cytotoxicity of NK cells is not influenced by the presence or absence of DPPIV on their cell surface (158, 160) . However, DPPIV-negative NK cells show significantly less CD16-dependent lysis than DPPIV-positive NK cells (160) . Interestingly, NK cytolytic function against tumor cells was diminished in DPPIV-deficient rats in a model for lung metastasis (161) . Figure 3 shows an overview of published data on the DPP family in the innate immune system. Only about 5% of freshly isolated CD20-positive B cells express DPPIV, but this fraction grows significantly upon pokeweed mitogen (PWM) or S. aureus protein stimulation (162) . Similar to NK cells, DPPIV inhibitors significantly suppress DNA synthesis in B-lymphocytes (162) , but again these inhibitors are now known to also inhibit DPP8 and 9 (159) . Mouse spleen-derived B-lymphocytes only express low amounts of DPPIV mRNA (163) . DPP8 and 9 mRNA, on the other hand, are expressed at much greater levels in these cells, and they are upregulated in Raji cells, a B-lymphocyte-like cell line, after PWM, LPS stimulation or mitomycin c treatment, and downregulated after DTT treatment (163) . DPP8 and 9 have also been shown immunohistochemically in human lymph follicular lymphocytes (164) . DPPII activity has also been shown in human B-lymphocytes (25). Dipeptidyl peptidase IV was originally described as a surface marker for T-lymphocytes, in which case it is better known as CD26, and later more specifically for a subset of CD4-positive memory cells, CD4 + CD45RO + CD29 + cells, which respond maximally to recall antigen tetanus toxoid and induce B-cell IgG synthesis (165, 166) . Indeed, CD26 surface expression is augmented along with the antigen sensitivity of a particular CD4 + T-cell clone (167) . CD26 high CD8 + T-cells belong to the early effector memory T-cell subset (168) . CD26 is also a marker for T-cell activation (165, (169) (170) (171) . CD26 expression on CD4 + Tcells correlates with T H 1 responses. Stimuli that typically induce a T H 1 phenotype tend to induce CD26 expression (172) . Additionally, the CD4 + T cells capable of transendothelial migration in vitro are characterized by a bright expression of CD26 (173, 174) , but CD26 does not seem to be actually involved in T-cell adhesion to endothelial cells or fibroblasts (175) . Recently, it was shown that up to 98% of all T H 17 cells show very high CD26 expression, with mean fluorescent intensity on these cells almost twice as high as on T H 1 or T H 2 cells. Therefore, the authors of this study suggest CD26 as a marker for T H 17 cells (176) . Conversely, CD26 has been proposed as a negative marker for the selection Treg cells due to its very low-surface expression on these cells (176) (177) (178) . CD26 is also a costimulatory molecule for T-cell activation. Crosslinking of CD26, along with CD3, stimulates T-cell activation and proliferation (168, 179, 180) . CD26 can also directly activate T-cells in an alternative activation pathway, but this requires the presence of the TCR/CD3 complex (181) (182) (183) . During costimulation, CD26 is mannose-6 phosphorylated and internalized, the latter of which is mediated in part by its interaction with M6P/IGFIIR (184) . It then localizes to lipid rafts where it might interact with CD45, required for TCR signaling, facilitating colocalization of this molecule with TCR signaling molecules (185, 186) . A number of candidate binding partners for costimulation have been proposed. ADA and CD26 are known binding partners (187) . Even though ADA binding to CD26 does not seem to be essential for immune functions in humans (188) , the nanomolar affinity of this interaction probably reflects its importance (189) . Indeed, association with free ADA or ADA presented by ADA-anchoring proteins on dendritic cells seems to costimulate T-cells through CD26 binding (190, 191) . On the other hand, it has been shown that soluble DPPIV enhances T-cell proliferation independent of its enzyme activity or ADA-binding capability (192) . Interestingly, the ADA-CD26 interaction can be inhibited by HIV-1 external envelope protein gp120 and this requires interaction of gp120 with CXCR4 (189) . In fact, evidence suggests a physical association between CXCR4 and CD26 on peripheral blood lymphocytes (193) . Fibronectin is another known binding partner of CD26 involved in T-cell costimulation (194) (195) (196) . Finally, CD26 interacts with caveolin-1 on monocytes. This interaction causes an upregulation of CD86 on these cells, which potentiates antigen-specific T-cell activation (197) . Most studies seem to find no need for DPPIV's enzymatic activity for succesful costimulation, as evidenced through the use of inhibitors and catalytically impaired DPPIV mutants (198) (199) (200) (201) . Dipeptidyl peptidase 8 and 9 are present in baboon spleen interfollicular T-lymphocytes and Jurkat T cells (164) . They are upregulated in the latter after PWM and LPS, but not PHA, stimulation (163, 202, 203) . Activation of PWM-stimulated T-cells is suppressed after DPPIV/8/9 inhibition. Moreover, DNA synthesis and T-cell proliferation are reduced, as well as production of IL-2, -10, -12, and IFN-γ. This is due to an induction of TGF-β secretion (159, (204) (205) (206) (207) . Inhibition also upregulates CTLA-4 and downregulates DPPIV expression (206, 208) . These observations might be physiologically relevant as endogenous inhibitors of DPPs are known which have similar effects in cell-based experiments as the synthetic inhibitors (209, 210) . Dipeptidyl peptidase II activity is higher in T-lymphocytes than in B-lymphocytes (25) and absence of DPPII steers T-lymphocytes toward a T H 17 phenotype. T-lymphocytes of DPPII KO mice hyperproliferate and secrete IL-17 after CD3 crosslinking or after in vivo priming and in vitro antigen-specific restimulation (211) . PREP activity has also been shown in mouse T-lymphocytes (212) . Its activity is significantly higher in immature, double-positive thymocytes compared to mature, single-positive thymocytes, or peripheral T-cells. T-cells stimulated with Con A followed by IL-2 show a time-dependent increase in PREP activity and pretreatment of cells with a PREP inhibitor renders them resistant to activation-induced cell death (212) . Figure 4 shows an overview of in vitro data on DPP involvement in primary human T cell activation. The DPP family has been reported to be dysregulated or even involved in a number of inflammatory disorders. Expression levels of a number of family members are modulated in rheumatoid arthritis. Whereas the density of CD26 on peripheral T cells is increased in patients, it is low on synovial fluid T cells (213) (214) (215) . DPPIV activity in plasma, serum, or synovial fluid of patients has also been found to be decreased, similar to results in several rat models of arthritis (216) (217) (218) (219) (220) (221) (222) . Interestingly, rats resistant to induction of arthritis show higher plasma DPPIV levels (222) . By contrast, DPPII and PREP activity are increased in serum or synovial fluid of arthritis patients (219) (220) (221) . Likewise, FAP immunoreactivity is much higher in fibroblast-like synoviocytes of rheumatoid arthritis patients compared to osteoarthritis controls (223) . DPPIV's involvement in rheumatoid arthritis has been studied, but remains unclear. On the one hand, inhibition can suppress development of arthritis in rats (224) . Note, however, that effects mediated through other DPPs are hard to exclude as these inhibitors were developed before DPP8 and DPP9 were discovered. On the other hand, induced arthritis is more severe in DPPIV-deficient mice (216) . This may be due to increased levels of circulating CXCL12 (216), a DPPIV substrate shown to be involved in rheumatoid arthritis. Several case reports in patients seem to suggest a link between the development of rheumatoid arthritis and the use of DPPIV inhibitors (225) (226) (227) . PRCP has also been associated with rheumatoid arthritis as its activity was shown in synovial fluid isolated from arthritic joints (149) . Inflammatory bowel disease shows a distinct expression pattern of the DPP family. DPPIV serum or plasma activity seems to be lower in patients, whereas there is an increase of circulating CD26 + CD25 + cells with a higher CD26 surface expression (228, 229) . FAP is heavily expressed by myofibroblasts in the submucosa strictures in Crohn's disease, and is upregulated after stimulation with TNFα or TGFβ (230) . In a mouse model, colonic DPPII and DPP8 mRNA and DPPII activity are increased, while colonic DPP8/9 activity only increases significantly in mice that are also DPPIV knockouts (231) . In mouse models, inhibition or abrogation of DPPIV seems to at least partially ameliorate symptoms, possibly by increasing circulating GLP-2, impairing neutrophil recruitment, and maintaining Treg populations (231) (232) (233) (234) (235) (236) . Some of those beneficial effects may be mediated in part by the other DPPs, as additive effects were found for DPPIV KO and the DPP inhibitors (231, 234, 237) . A recent study suggests that the ameliorative effects of DPP inhibitors are most likely not mediated through GLP-2 protection (238) . The DPP family has also been studied in neuroinflammation. Ischemia-induced neuroinflammation in rats prompts a distinct expression and activity pattern of the DPPs. In the days following ischemia, the brain of these rats undergoes a complex reorganization of DPP expression with changes in mRNA, protein, and activity levels of DPPII, 4, 8, and 9 in cortical neurons, microglia, and macrophages (146) . Similarly, PREP seems to be associated with astrocytes and microglia in lesioned inflamed brains of rats (239) . DPPIV and PREP also may be involved in multiple sclerosis. CD26 + T cells were found to correlate with disease scores (240) . Soluble DPPIV levels are elevated in cerebrospinal fluid of patients (241) . Plasma PREP activity, on the other hand, is lower in patients with relapsing-remitting or primary progressive multiple sclerosis and in clinically isolated syndrome (118, 119) . Interestingly, PREP inhibition seems to aggravate symptoms in a mouse model of multiple sclerosis (118) . In systemic lupus erythematosus, DPPs also seem to be dysregulated. In mouse models, DPPII and PREP activities are increased in plasma, spleen, kidney, and liver, whereas DPPIV activity is decreased (221, 242) . Human patients also show elevated DPPII and reduced DPPIV activities in serum, along with reduced numbers of CD26 + T cells (221, 243) . Interestingly, serum DPPIV levels are inversely correlated with disease score (243) . FAP immunoreactivity is decreased in the synovium of lupus patients (244) . Finally, DPPIV has been studied in psoriasis, an immunemediated chronic inflammatory disorder with primary involvement of skin and joints. Its mRNA, protein levels and activity are higher in psoriatic skin samples (245, 246) . By contrast, serum DPPIV levels and activity seem to be lower in patients (247, 248) , accompanied by a reduction of peripheral CD8 + CD26 + T cells (249, 250) . Two case reports suggest a link between the use of DPPIV inhibitor sitagliptin and psoriasis. While one woman developed a psoriaform eruption 6 days after starting sitagliptin treatment (251), another patient's psoriatic lesions gradually diminished and were effectively gone 3 months after the start of sitagliptin treatment (252) . Dipeptidyl peptidase IV has recently received much attention for its potential as a therapeutic target for the treatment of atherosclerosis (Box 3) (253). This is not surprising considering the current use of DPPIV inhibitors in the treatment of T2D and the fact that T2D is associated with a higher risk for atherosclerosis (28, 254). In the ApoE −/− mouse model of atherosclerosis, DPPIV inhibition generally reduces plaque area and monocyte and macrophage plaque infiltration (255) (256) (257) . A reduction in the number of plaque lesions or in smooth muscle cell content have also been observed (255, 256) , as well as lower plaque MMP9 and higher plaque collagen levels, suggesting increased plaque stability (258) . One study reported effects of DPPIV inhibition on atherosclerotic plaques of only diabetic ApoeE −/− mice (141), but more recently, Terasaki et al. found similar effects in non-diabetic and diabetic ApoE −/− mice (259) . Likely, such differences can be explained by the fact that different DPPIV inhibitors were employed. Effects of DPPIV on atherogenesis similar to those observed in ApoE −/− mice have been reproduced in LDLR −/− mice (142, 260) . In human atherosclerotic plaques, Atherosclerosis is the most common underlying cause of cardiovascular diseases and should be regarded as an inflammatory disease. It starts with dysfunction of the endothelium leading to the expression of leukocyte adhesion molecules, such as selectins and integrins. Locally produced proinflammatory cytokines attract the immune cells into the inner layer of the endothelium. However, not only leukocytes are found in the plaque but also low-density lipoprotein particles (LDL) and their oxidized counterparts (oxLDL). In the plaque, monocytes differentiate into macrophages, phagocytose the oxLDL and turn into so-called pro-atherogenic foam cells. This process leads to a self-sustaining, local inflammation leading to plaque growth, and migration of smooth muscle cells into the core. A plaque is defined as stable as long as it is contained by a thick fibrous cap. However, the latter is slowly degraded by the proteolytic enzymes from the leukocytes. This eventually leads to rupture and the formation of arterial thrombi (264, 265 DPPIV immunoreactivity could only be found on endothelium of neovessels (82). It was recently found that DPPIV activity may be a predictor for the onset of atherosclerosis in otherwise healthy Chinese individuals (261) . Another prospective study investigated the influence of vildagliptin or sitagliptin treatment on intima-media thickness, a surrogate marker for atherosclerosis. This study found that treatment with vildagliptin or sitagliptin reduced intima-media thickness, suggesting that DPPIV inhibition might be beneficial in atherosclerosis in humans as well (262) . Moreover, treatment naïve T2D patients treated with alogliptin for 3 months saw a significant decrease in their circulating atherogenic lipids (263) . It has been suggested that DPPIV inhibitors' anti-atherogenic effects are mainly mediated through decreased monocyte infiltration, as DPPIV inhibitors suppress monocyte activation and chemotaxis in vitro (142, 258) . DPPIV inhibition also reduces in vitro foam cell formation in exudate peritoneal macrophages from ApoE −/− mice (255) . Moreover, soluble DPPIV stimulates in vitro proliferation of smooth muscle cells and this can be reduced through the addition of a DPPIV inhibitor (256, 260) . Finally, active circulating GLP-1 levels are augmented and this improves endothelial dysfunction (259, 266) . Probably, DPPIV inhibition improves atherosclerosis through a combination of all these mechanisms. Indeed, incretin antagonists only partially attenuate the anti-atherogenic effects of DPPIV inhibition, suggesting that other mechanisms beyond incretin preservation are in play (259) . Interestingly, monocyte-endothelial cell adhesion is abrogated by an anti-SDF-1α antibody in vitro (267) . LDL seems to induce SDF-1α expression and leads to smooth muscle cell proliferation and inhibition of cell apoptosis (267, 268) . SDF-1α is a DPPIV substrate, which loses its biological activity after cleavage (216) . As DPPIV inhibition seems to improve atherosclerosis, whereas intact SDF-1α appears to be deletorious, it could be argued that SDF-1α cleavage by DPPIV does not play a major role in atherosclerosis. Dipeptidyl peptidase 8 and 9 have been found to be abundantly present in the macrophage-rich regions of human atherosclerotic plaques and considering DPP9's role in macrophage activation, it might potentially be involved in atherogenesis (82). FAP expression is enhanced in some, but not all types of human atheromata. It is found on smooth muscle cells, and its expression correlates with macrophage burden, probably due to the fact that TNFα upregulates FAP in smooth muscle cells in vitro. As it is mainly associated with collagen-poor regions and can digest type I collagen and gelatin in vitro, FAP probably contributes to plaque instability (269) . Interestingly, many of the studies reviewed above show the potential of targeting DPP family members for the treatment of atherosclerosis (see Figure 5 ). FAP inhibition might reduce plaque instability by decreasing collagen breakdown; DPP9 inhibition is likely to attenuate M1 macrophage activation, reducing the local inflammatory cascade; DPPIV inhibition may decrease monocyte infiltration, foam cell formation, improve endothelial dysfunction, and reduce smooth muscle cell proliferation; and finally, PREP inhibition might reduce neutrophil infiltration, preventing endothelial dysfunction, and monocyte infiltration. All of this shows the possibilities of repositioning DPPIV inhibitors, currently being used to treat type 2 diabetes, as well as the potential of targeting other members of the DPP family. Caution should be taken when interpreting results from literature data based on DPP inhibitors, especially from older studies. It is now known that, under the experimental conditions used, many of these inhibitors are not specific for one particular family member. The reported findings, however, remain interesting. This review has shown extensive involvement of members of the DPP family in the immune system. It is clear that these enzymes hold great potential as targets for the treatment of certain inflammatory disorders. Particularly, the possibility of targeting DPP family members for the prevention and treatment of atherosclerosis warrants further investigation. In vivo effects of a potent, selective DPPII inhibitor: UAMC00039 is a possible tool for the elucidation of the physiological function of DPPII. Adv Exp Med Dipeptidylpeptidase 4 negatively regulates colony-stimulating factor activity and stress hematopoiesis Structures of human DPP7 reveal the molecular basis of specific inhibition and the architectural diversity of proline-specific peptidases Purification of two dipeptidyl aminopeptidases II from rat brain and their action on proline-containing neuropeptides Dipeptidyl peptidase II (DPPII), a review Thioxo amino acid pyrrolidides and thiazolidides: new inhibitors of proline specific peptidases Structure-activity relationships of boronic acid inhibitors of dipeptidyl peptidase IV. 1. Variation of the P2 position of Xaa-boroPro dipeptides Structure-activity relationship of diaryl phosphonate esters as potent irreversible dipeptidyl peptidase IV inhibitors Rapid parallel synthesis of dipeptide diphenyl phosphonate esters as inhibitors of dipeptidyl peptidases Fluoro-olefins as peptidomimetic inhibitors of dipeptidyl peptidases Prolyl oligopeptidase induces angiogenesis both in vitro and in vivo in a novel regulatory manner A prolyl oligopeptidase inhibitor, KYP-2047, reduces α-synuclein protein levels and aggregates in cellular and animal models of Parkinson's disease Subcellular localization suggests novel functions for prolyl endopeptidase in protein secretion Subcellular distribution of prolyl endopeptidase and cation-sensitive neutral endopeptidase in rabbit brain Expression and traffic of cellular prolyl oligopeptidase are regulated during cerebellar granule cell differentiation, maturation, and aging Distribution of prolyl oligopeptidase in the mouse whole-body sections and peripheral tissues Partial purification and characterization of post-proline cleaving enzyme: enzymatic inactivation of neurohypophyseal hormones by kidney preparations of various species Changes in prolyl endopeptidase during maturation of rat brain and hydrolysis of substance P by the purified enzyme Porcine muscle prolyl endopeptidase and its endogenous substrates Purification and characterization of prolyl endopeptidase from pig brain Purification and characterization of prolyl endopeptidase from rat skin Effect of S 17092, a novel prolyl endopeptidase inhibitor, on substance P and alpha-melanocyte-stimulating hormone breakdown in the rat brain An evaluation of the role of a pyroglutamyl peptidase, a post-proline cleaving enzyme and a post-proline dipeptidyl amino peptidase, each purified from the soluble fraction of guinea-pig brain, in the degradation of thyroliberin in vitro Prolyl oligopeptidase: a potential target for the treatment of cognitive disorders On the role of prolyl oligopeptidase in health and disease Degradation of neurotensin by rabbit brain endo-oligopeptidase A and endo-oligopeptidase B (prolineendopeptidase) Proline specific peptidases Brain endo-oligopeptidase B: a post-proline cleaving enzyme that inactivates angiotensin I and II Inactivation of thyrotropin-releasing hormone (TRH) and (3Me-His) TRH by brain peptidases studied by highperformance liquid chromatography Evaluation of the role of prolyl endopeptidase and pyroglutamyl peptidase I in the metabolism of LHRH and TRH in brain Proline-specific proteases in cultivated neuronal and glial cells Effect of a novel prolyl endopeptidase inhibitor, JTP-4819, on prolyl endopeptidase activity and substance Pand arginine-vasopressin-like immunoreactivity in the brains of aged rats Neutrophils contain prolyl endopeptidase and generate the chemotactic peptide, PGP, from collagen Prolyl oligopeptidase is involved in release of the antifibrotic peptide Ac-SDKP Alteration of prolyl oligopeptidase and activated α-2-macroglobulin in multiple sclerosis subtypes and in the clinically isolated syndrome Prolyl oligopeptidase is inhibited in relapsing-remitting multiple sclerosis Specific inhibitors for prolyl endopeptidase and their anti-amnesic effect JTP-4819: a novel prolyl endopeptidase inhibitor with potential as a cognitive enhancer Pharmacological studies of a novel prolyl endopeptidase inhibitor, JTP-4819, in rats with middle cerebral artery occlusion Effects of prolyl endopeptidase inhibitors and neuropeptides on delayed neuronal death in rats ONO-1603, a potential antidementia drug, delays age-induced apoptosis and suppresses overexpression of glyceraldehyde-3-phosphate dehydrogenase in cultured central nervous system neurons Prolyl endopeptidase-deficient mice have reduced synaptic spine density in the CA1 region of the hippocampus, impaired LTP, and spatial learning and memory Prolyl oligopeptidase binds to GAP-43 and functions without its peptidase activity GAP43 shows partial co-localisation but no strong physical interaction with prolyl oligopeptidase Prolyl oligopeptidase is a glyceraldehyde-3-phosphate dehydrogenase-binding protein that regulates genotoxic stress-induced cell death Interaction of prolyl oligopeptidase with α-synuclein Prolyl oligopeptidase colocalizes with α-synuclein, β-amyloid, tau protein and astroglia in the post-mortem brain samples with Parkinson's and Alzheimer's diseases Prolyl oligopeptidase stimulates the aggregation of alpha-synuclein The anti-inflammatory effect of neuropeptide Y (NPY) in rats is dependent on dipeptidyl peptidase 4 (DP4) activity and age Soluble DPP4 originates in part from bone marrow cells and not from the kidney Representative aminopeptidases and prolyl endopeptidase from murine macrophages: comparative activity levels in resident and elicited cells A potential role for dendritic cell/macrophage-expressing DPP4 in obesity-induced visceral inflammation Dipeptidyl peptidase 8/9-like activity in human leukocytes Human U937 cell surface peptidase activities: characterization and degradative effect on tumor necrosis factor-alpha Plasma membrane-bound and lysosomal peptidases in human alveolar macrophages Divergent regulation of cell surface protease expression in HL-60 cells differentiated into macrophages with granulocyte macrophage colony stimulating factor or neutrophils with retinoic acid DPP-4 (CD26) inhibitor alogliptin inhibits TLR4-mediated ERK activation and ERKdependent MMP-1 expression by U937 histiocytes DPP-4 (CD26) inhibitor alogliptin inhibits atherosclerosis in diabetic apolipoprotein Edeficient mice Longterm dipeptidyl-peptidase 4 inhibition reduces atherosclerosis and inflammation via effects on monocyte recruitment and chemotaxis DPP-4 inhibitors repress NLRP3 inflammasome and interleukin-1beta via GLP-1 receptor in macrophages through protein kinase C pathway Inhibitors of dipeptidyl peptidase IV (DP IV, CD26) specifically suppress proliferation and modulate cytokine production of strongly CD26 expressing U937 cells Fibroblast activation protein expression by stromal cells and tumor-associated macrophages in human breast cancer Dipeptidyl peptidase IV, aminopeptidase N and DPIV/APN-like proteases in cerebral ischemia Cytochemical localization and biochemical characterization of dipeptidyl aminopeptidase II in macrophages and mast cells Cytochemical localization and biochemical evaluation of a lysosomal serine protease in lung: dipeptidyl peptidase II in the normal rat Prolylcarboxypeptidase (angiotensinase C) in human lung and cultured cells Prolylcarboxypeptidase promotes angiogenesis and vascular repair A prolyl endopeptidase from murine macrophages, its assay and specific inactivation Cathepsin B and prolyl endopeptidase activity in rat peritoneal and alveolar macrophages. Stimulation of peritoneal macrophages by saline lavage Prolyl endopeptidase is revealed following SILAC analysis to be a novel mediator of human microglial and THP-1 cell neurotoxicity Dipeptidyl peptidase IV is a human and murine neutrophil chemorepellent A novel proteolytic cascade generates an extracellular matrix-derived chemoattractant in chronic neutrophilic inflammation Inhibition of CD26/dipeptidyl peptidase IV enhances CCL11/eotaxinmediated recruitment of eosinophils in vivo Rat peritoneal mast cells release dipeptidyl peptidase II Expression and functional role of dipeptidyl peptidase IV (CD26) on human natural killer cells Dipeptidyl peptidase IV inhibition for the treatment of type 2 diabetes, potential importance of selectivity over dipeptidyl peptidases 8 and 9 The CD26 antigen is coupled to protein tyrosine phosphorylation and implicated in CD16-mediated lysis in natural killer cells CD26 expression determines lung metastasis in mutant F344 rats: involvement of NK cell function and soluble CD26 Functional role of CD26 on human B lymphocytes Regulation of dipeptidyl peptidase 8 and 9 expression in activated lymphocytes and injured liver The in vivo expression of dipeptidyl peptidases 8 and 9 1F7, a novel cell surface molecule, involved in helper function of CD4 cells Dipeptidyl peptidase IV of human lymphocytes -evidence for specific hydrolysis of glycylproline p-nitroanilide in T-lymphocytes Influence of CD26 and integrins on the antigen sensitivity of human memory T cells CD26-mediated co-stimulation in human CD8(+) T cells provokes effector function via proinflammatory cytokine production Dipeptidyl peptidase IV in human T lymphocytes. An approach to the role of a membrane peptidase in the immune system Dipeptidyl peptidase IV in the immune system A novel pathway of human T cell activation via a 103 kD T cell activation antigen Cell surface characterization of T lymphocytes and allergen-specific T cell clones: correlation of CD26 expression with T(H1) subsets Phenotypic characterization of CD4+ T cells that exhibit a transendothelial migratory capacity Characterization of the 4C8 antigen involved in transendothelial migration of CD26hi T cells after tight adhesion to human umbilical vein endothelial cell monolayers CD26 (dipeptidyl peptidase IV) on human T lymphocytes does not mediate adhesion of these cells to endothelial cells or fibroblasts Human Th17 cells express high levels of enzymatically active dipeptidylpeptidase IV (CD26) CD26: a negative selection marker for human Treg cells Human treg cells are characterized by low/negative CD6 expression Cloning and functional expression of the T cell activation antigen CD26 Costimulation of CD4+ and CD8+ T cells through CD26: the ADA-binding epitope is not essential for complete signaling Triggering of cytotoxic T lymphocytes and NK cells via the Tp103 pathway is dependent on the expression of the T cell receptor/CD3 complex Function of dipeptidyl peptidase IV (CD26, Tp103) in transfected human T cells FcR-mediated crosslinking of Ta1 (CDw26) induces human T lymphocyte activation Internalization of CD26 by mannose 6-phosphate/insulin-like growth factor II receptor contributes to T cell activation CD26-mediated signaling for T cell activation occurs in lipid rafts through its association with CD45RO Coassociation of CD26 (dipeptidyl peptidase IV) with CD45 on the surface of human T lymphocytes Direct association of adenosine deaminase with a T cell activation antigen The binding site of human adenosine deaminase for CD26/dipeptidyl peptidase IV: the Arg142Gln mutation impairs binding to cd26 but does not cause immune deficiency The HIV-1 gp120 inhibits the binding of adenosine deaminase to CD26 by a mechanism modulated by CD4 and CXCR4 expression CD26, adenosine deaminase, and adenosine receptors mediate costimulatory signals in the immunological synapse Expression of ecto-adenosine deaminase and CD26 in human T cells triggered by the TCR-CD3 complex. Possible role of adenosine deaminase as costimulatory molecule Soluble CD26/dipeptidyl peptidase IV enhances human lymphocyte proliferation in vitro independent of dipeptidyl peptidase enzyme activity and adenosine deaminase binding Comodulation of CXCR4 and CD26 in human lymphocytes A novel consensus motif in fibronectin mediates dipeptidyl peptidase IV adhesion and metastasis Fibronectin promotes proliferation of naive and memory T cells by signaling through both the VLA-4 and VLA-5 integrin molecules VLA-4 mediates CD3-dependent CD4+ T cell activation via the CS1 alternatively spliced domain of fibronectin CD26 up-regulates expression of CD86 on antigen-presenting cells by means of caveolin-1 The costimulatory activity of the CD26 antigen requires dipeptidyl peptidase IV enzymatic activity Enzymatic activity of CD26 (dipeptidylpeptidase IV) is not required for its signalling function in T cells Molecular analysis of CD26-mediated signal transduction in T cells Unchanged signaling capacity of mutant CD26/dipeptidylpeptidase IV molecules devoid of enzymatic activity Biochemical properties and expression profile of human prolyl dipeptidase DPP9 Cloning, expression and chromosomal localization of a novel human dipeptidyl peptidase (DPP) IV homolog, DPP8 Role of dipeptidyl peptidase IV (DP IV)-like enzymes in T lymphocyte activation: investigations in DP IV/CD26-knockout mice Inhibitors of dipeptidyl peptidase IV induce secretion of transforming growth factor-ß1 in PWM-stimulated PBMC and T cells Dipeptidyl peptidase IV (DP IV/CD26) mRNA expression in PWM-stimulated Tcells is suppressed by specific DP IV inhibition, an effect mediated by TGFbeta(1) Dipeptidyl peptidase IV on activated T cells as a target molecule for therapy of rheumatoid arthritis The expression of T-cell surface antigens CTLA-4, CD26, and CD28 is modulated by inhibition of dipeptidylpeptidase IV (DPP IV, CD26) activity in murine stress-induced abortions Downregulation of T cell activation following inhibition of dipeptidyl peptidase IV/CD26 by the N-terminal part of the thromboxane A2 receptor Non-substrate peptides influencing dipeptidyl peptidase IV/CD26 activity and immune cell function Th17 differentiation is the default program for DPP2-deficient T cell differentiation Murine T cells expressing high activity of prolyl endopeptidase are susceptible to activationinduced cell death Expression and functional role of 1F7 (CD26) antigen on peripheral blood and synovial fluid T cells in rheumatoid arthritis patients CD26 surface molecule involvement in T cell activation and lymphokine synthesis in rheumatoid and other inflammatory synovitis In active chronic rheumatoid arthritis, dipeptidyl peptidase IV density is increased on monocytes and CD4(+) T lymphocytes Circulating CD26 is negatively associated with inflammation in human and experimental arthritis Serum levels of soluble CD26 and CD30 and their clinical significance in patients with rheumatoid arthritis Levels of dipeptidyl peptidase IV/CD26 substrates neuropeptide Y and vasoactive intestinal peptide in rheumatoid arthritis patients Activities of dipeptidyl peptidase II and dipeptidyl peptidase IV in synovial fluid from patients with rheumatoid arthritis and osteoarthritis Activities of dipeptidyl peptidase II, dipeptidyl peptidase IV, prolyl endopeptidase, and collagenaselike peptidase in synovial membrane from patients with rheumatoid arthritis and osteoarthritis Activities of dipeptidyl peptidase II and dipeptidyl peptidase IV in mice with lupus erythematosus-like syndrome and in patients with lupus erythematosus and rheumatoid arthritis Neutral aminopeptidase and dipeptidyl peptidase IV in the development of collagen II-induced arthritis Fibroblast activation protein is expressed by rheumatoid myofibroblast-like synoviocytes Antiarthritic effects of the novel dipeptidyl peptidase IV inhibitors TMC-2A and TSL-225 Polyarthropathy in type 2 diabetes patients treated with DPP4 inhibitors Sitagliptin (DPP-4 inhibitor)-induced rheumatoid arthritis in type 2 diabetes mellitus: a case report Acute onset of rheumatoid arthritis associated with administration of a dipeptidyl peptidase-4 (DPP-4) inhibitor to patients with diabetes mellitus Dipeptidyl peptidase IV (DP IV, CD26) in patients with inflammatory bowel disease Dipeptidyl peptidase-4 expression is reduced in Crohn's disease Fibroblast activation protein expression in Crohn's disease strictures Dipeptidyl peptidase expression during experimental colitis in mice The DPP-IV inhibitor ER-319711 has a proliferative effect on the colonic epithelium and a minimal effect in the amelioration of colitis Dipeptidyl peptidase IV (DP IV, CD26) and aminopeptidase N (APN, CD13) as regulators of T cell function and targets of immunotherapy in CNS inflammation Inhibiting dipeptidyl peptidase activity partially ameliorates colitis in mice Contribution of dipeptidyl peptidase IV to the severity of dextran sulfate sodium-induced colitis in the early phase Dipeptidyl peptidase-4 inhibitor anagliptin facilitates restoration of dextran sulfate sodium-induced colitis Biochemical and histological changes in the small intestine of mice with dextran sulfate sodium colitis The effects of a TGR5 agonist and a dipeptidyl peptidase IV inhibitor on dextran sulfate sodium-induced colitis in mice Prolyl oligopeptidase: a rising star on the stage of neuroinflammation research A longitudinal study of the T cell activation marker CD26 in chronic progressive multiple sclerosis Soluble CD26 and CD30 levels in CSF and sera of patients with relapsing neuromyelitis optica Activities of dipeptidyl peptidases in BXSB mice and MRL/lpr mice with lupus erythematosus-like syndrome Reduction of serum soluble CD26/dipeptidyl peptidase IV enzyme activity and its correlation with disease activity in systemic lupus erythematosus Identification of distinct gene expression profiles in the synovium of patients with systemic lupus erythematosus CD26/dipeptidyl-peptidase IV in psoriatic skin: upregulation and topographical changes Distribution of dipeptidyl-peptidase IV on keratinocytes in the margin zone of a psoriatic lesion: a comparison with hyperproliferation and aberrant differentiation markers CD26/dipeptidyl-peptidase IV and adenosine deaminase serum levels in psoriatic patients treated with cyclosporine, etanercept, and psoralen plus ultraviolet A phototherapy Serum soluble CD26 levels: diagnostic efficiency for atopic dermatitis, cutaneous T-cell lymphoma and psoriasis in combination with serum thymus and activation-regulated chemokine levels Expression of dipeptidyl-peptidase IV (CD26) on CD8+ T cells is significantly decreased in patients with psoriasis vulgaris and atopic dermatitis Reduced CD26bright expression of peripheral blood CD8+ Tcell subsets in psoriatic patients Psoriasiform eruption triggered by a dipeptidyl peptidase IV inhibitor Sitagliptin, a dipeptidyl peptidase-IV inhibitor, improves psoriasis DPP-4 inhibitors and atherosclerosis: the promise Primary prevention of cardiovascular diseases in people with diabetes mellitus: a scientific statement from the American Heart Association and the American Diabetes Association Effects of PKF275-055, a dipeptidyl peptidase-4 inhibitor, on the development of atherosclerotic lesions in apolipoprotein E-null mice DPP-4 inhibitor, suppresses proliferation of vascular smooth muscles and monocyte inflammatory reaction and attenuates atherosclerosis in male apo E-deficient mice Dipeptidyl peptidase-4 inhibitor, sitagliptin, improves endothelial dysfunction in association with its anti-inflammatory effects in patients with coronary artery disease and uncontrolled diabetes Sitagliptin reduces plaque macrophage content and stabilises arteriosclerotic lesions in Apoe (-/-) mice Preventive effect of dipeptidyl peptidase-4 inhibitor on atherosclerosis is mainly attributable to incretin's actions in nondiabetic and diabetic apolipoprotein E-null mice Dipeptidyl-peptidase-4 inhibitor, alogliptin, attenuates arterial inflammation and neointimal formation after injury in low-density lipoprotein (LDL) receptor-deficient mice Increased plasma DPP4 activities predict new-onset atherosclerosis in association with its proinflammatory effects in Chinese over a four year period: a prospective study Decreased carotid atherosclerotic process by control of daily acute glucose fluctuations in diabetic patients treated by DPP-IV inhibitors Alogliptin: a new dipeptidyl peptidase-4 inhibitor with potential anti-atherogenic properties Inflammation and atherosclerosis The immune response in atherosclerosis: a doubleedged sword A dipeptidyl peptidase-4 inhibitor, des-fluoro-sitagliptin, improves endothelial function and reduces atherosclerotic lesion formation in apolipoprotein E-deficient mice Upregulation of SDF-1 is associated with atherosclerosis lesions induced by LDL concentration polarization SDF-1 promotes ox-LDL induced vascular smooth muscle cell proliferation Fibroblast activation protein is induced by inflammation and degrades type I collagen in thin-cap fibroatheromata