Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jan 1.
Published in final edited form as: J Immunol. 2020 Jan 1;204(1):3–11. doi: 10.4049/jimmunol.1900868

CD13/aminopeptidase N is a potential therapeutic target for inflammatory disorders

Chenyang Lu *,, Mohammad A Amin *, David A Fox *
PMCID: PMC6997018  NIHMSID: NIHMS1549646  PMID: 31848300

Abstract

CD13/aminopeptidase N is a widely expressed ectoenzyme with multiple functions. As an enzyme, CD13 regulates activities of numerous cytokines by cleaving their N-terminals and is involved in antigen processing by trimming the peptides bound to MHC II. Independent of its enzymatic activity, cell membrane CD13 functions by crosslinking-induced signal transduction, regulation of receptor recycling, enhancement of FcγR-mediated phagocytosis, and acting as a receptor for cytokines. Moreover, soluble CD13 has multiple pro-inflammatory roles mediated by binding to G protein-coupled receptors. CD13 not only modulates development and activities of immune-related cells, but also regulates functions of inflammatory mediators. Therefore, CD13 is important in the pathogenesis of various inflammatory disorders. Inhibitors of CD13 have shown impressive anti-inflammatory effects, but none of them has yet been used for clinical therapy of human inflammatory diseases. We reevaluate CD13’s regulatory role in inflammation and suggest that CD13 could be a potential therapeutic target for inflammatory disorders.


CD13, also known as aminopeptidase N or membrane alanyl aminopeptidase, is a type II membrane 150kDa metalloprotease with an extracellular oriented catalytic domain. It is a seahorse-shaped molecule and usually forms a head-to-head homodimer by means of hydrophobic interactions(1, 2). Each monomeric molecule of CD13 possesses a 7-domain organization, which is characteristic of M1 metallopeptidases(1, 3). CD13 has been termed a “moonlighting enzyme” because of its multiple functions(4). Increasing evidence points to crucial regulatory functions for CD13 during normal and pathologic immune responses. CD13 has been suggested to play a role in antigen processing(5, 6), cell trafficking(710), and processing of inflammatory mediators, which are important features of immune responses. Here we review evidence concerning the involvement of CD13, including a soluble form of the molecule, in inflammation, and its potential as a therapeutic target in inflammatory disorders.

Mechanisms of CD13 functions

CD13 was named aminopeptidase N because of its preference for binding neutral amino acids, and because it removes N-terminal amino acids from unsubstituted oligopeptides, with the exception of peptides with proline in the penultimate position(11). By cleaving N-termini, CD13 regulates activity of numerous hormones, cytokines and chemokines that participate in inflammation. Moreover, CD13 is co-expressed by major histocompatibility complex class (MHC) II-bearing antigen-presenting cells(12), and is involved in the trimming of MHC II-associated peptides on the surface of antigen-presenting cells(5, 13, 14) (Fig. 1).

Figure 1. Mechanisms of CD13 functions.

Figure 1.

CD13 acts by enzyme-dependent mechanisms and enzyme-independent mechanisms. CD13 can cleave the N-terminus of numerous cytokines, hormones, and chemokines, and trim peptides that bind to MHC II. Antibody crosslinking of CD13 induces clustering of CD13, tyrosine phosphorylation of CD13, activation of Src kinase, FAK and ERK kinases, a calcium flux and potentially other components of the Ras/MAPK and PI-3K pathways, resulting in increased adhesion of monocytes to endothelial or monolayer CD13. CD13 also tethers the IQGAP1-ARF6-EFA6 complex on the plasma membrane to promote ARF6 activation, β1 integrin recycling and cell migration. CD13 functionally interacts with FcγRs and enhances phagocytosis by increasing the level and duration of Syk phosphorylation. CD13 is a cell surface receptor for some cytokines such as 14–3-3 proteins, signaling via p38 MAPK and JNK. CD13 can be shed from the cell membrane and soluble CD13 functions through engagement of GPCRs. MAPK: mitogen-activated protein kinases; IQGAP1: IQ motif containing GTPase activating protein 1; ARF6: adenosine diphosphate ribosylation factor; EFA6: exchange factor for ARF6; GPCRs: G protein coupled receptors; ECM: extracellular matrix; ER: endoplasmic reticulum.

Some CD13 functions in the immune system are independent of its enzymatic activity. These mechanisms include crosslinking-induced signal transduction(15); enhancement of FcγR-mediated phagocytosis(16); acting as a receptor(17, 18); and binding of soluble CD13 (sCD13) to G protein-coupled receptors (GPCRs)(19) (Fig. 1).

Various cellular responses, including homotypic aggregation, cell-cell adhesion, and migration, have been observed after crosslinking CD13 with monoclonal antibodies (mAbs). Interestingly, CD13, which includes a 7-aa cytoplasmic tail that was previously assumed to be inert, is itself phosphorylated in a Src-dependent manner(20). MAb crosslinking of CD13 induces clustering and phosphorylation of tyrosine 6 in the cytoplasmic domain of CD13, activation of Src, FAK, ERK, and potentially other components of the Ras/MAPK (JNK and p38) and PI-3K pathways, and initiation of a Ca2+ flux, resulting in increased adhesion of monocytes (MNs) to endothelial cells (ECs) or to a monolayer of CD13(20), and upregulation of cytokines including IL-8(15). Anti-CD13 mAbs also induce integrin-independent homotypic aggregation of monocytic U937 cells independent of their effects on CD13’s enzymatic activity(7). Such CD13-induced cell-cell adhesion is also dependent on the carbohydrate-binding protein galectin-3. A possible mechanism could be that antibody crosslinking of CD13 triggers a signaling cascade leading to release of galectin-3, which then binds to another receptor and triggers adhesion, or in oligomeric form binds to glycoconjugates on neighboring cells(21). CD13 can constitutively complex with(20) or be phosphorylated at Tyr6(10) (depending on cell types) to interact with IQ motif containing GTPase activating protein 1 (IQGAP1), a scaffold protein that interacts with over 90 other proteins to assemble complexes with defined biologic functions and transmits receptor-mediated signals to trigger functional outputs(22). CD13 serves to properly position IQGAP1 to perform its functions, thus, it was expected that CD13 participates in numerous functions mediated by IQGAP1, including cell motility, endocytosis, filopodia formation, angiogenesis, and phagocytosis(10). A recent study demonstrated that CD13 can tether IQGAP1, active adenosine diphosphate ribosylation factor (ARF) 6, exchange factor for ARF6 (EFA6), and β1 integrin at the plasma membrane, to allow β1 integrin recycling and cell migration(10). However, mAbs binding to distinct epitopes of CD13 mediate different consequences for cell adhesion. For example, cross-linking CD13 by some mAbs inhibits cell adhesion while mAb 452 induces it(23).

CD13 co-localizes with FcγRs including FcγR II/CD32 and FcγR I/CD64 after Fc receptor ligation with specific antibodies(24). During FcγR-mediated phagocytosis, CD13 redistributes to the phagocytic cup and is simultaneously internalized into phagosomes with FcγRs, rendering ingestion more efficient by increasing the level and duration of Syk phosphorylation induced by FcγRs themselves(16). There is a possibility that CD13 positively modulates FcγR-mediated signals and functions when CD13 and FcγR bind to their ligands, as CD13 is a well-known receptor for viral proteins and other pathogen-derived molecules. Moreover, CD13 was reported to be a cell surface receptor for some cytokines, including 14–3-3ε and 14–3-3σ (a.k.a stratifin)(17, 18). By binding to CD13 on cells such as fibroblasts and chondrocytes, 14–3-3ε regulates production of matrix-degrading enzymes such as matrix metalloproteinases (MMP)-3 and 13 through p38 MAPKs and JNK signaling cascades, while 14–3-3σ induces expression of MMP-1 through p38 MAPK activation(17, 18). Phosphorylation of the ectodomains of CD13 augments the binding affinity of 14–3-3 proteins to CD13, regulating the downstream signaling cascade. It has been proposed that enzymatic activity and binding to extracellular matrix proteins through Asn-Gly-Arg (NGR) motifs is mediated through the catalytic site involving domains IV to VII; cell-cell adhesion is mediated by the C terminal part, involving half of domain VI and VII; while signal transduction depends on the transmembrane and cytoplasmic domains (I and II)(8, 23).

CD13 can also be shed from the cell membrane by MMP-14(25), and the resultant sCD13 functions by engaging GPCRs on cytokine-activated lymphocytes(19), MNs, ECs, and fibroblasts(26). However, the identity of such GPCRs has not yet been reported. With binding to GPCRs, sCD13 induces phosphorylation of Src, ERK1/2, and to some extent, NF-κB, activating cell migration, angiogenesis, and other responses(26). Pertussis toxin, a GPCR inhibitor, significantly reduced chemotactic responses of cytokine-activated T cells(19) or monocytic cells as well as endothelial capillary morphogenesis in response to sCD13(26).

CD13 modulates development and function of immune-related cells

CD13 is widely expressed. Within the hematopoietic system, CD13 is predominantly expressed on myelomonocytic lineage cells, and it has long been used as a lineage marker in the characterization and typing of leukemia or lymphoma cells(27). However, CD13 is not just a cellular marker, but also a molecule modulating development and function of immune-related cells (Table 1).

Table 1.

Regulation of immune-related cells and inflammatory mediators by CD13

Type Subjects Major functions of CD13
Immune cells Monocyte/macrophage Differentiation, adhesion, migration, and activation
Dendritic cell Regulates differentiation and antigen presentation, and restricts TLR4 signal transduction
Lymphocyte T cell proliferation, migration, and secretion of cytokines including IL-2, IL-17, IFN-γ, TNF-α, TGF-β1 and IL-1RA
Neutrophil TNF-α induced apoptosis; aggregation
Mast cell Negative regulator of production of IL-6 and TNF-α
Inflammation related non-immune cells Endothelial cell Induces tube formation; angiogenesis; invasion
Fibroblast Augments migration; proliferation; cytokine secretion
Inflammatory mediators Kinins Both kallidin and its derivative Lys-des-Arg9-BK are substrates of CD13. CD13 also facilitates BK’s biologic functions by participating in membrane protein organization.
Tuftsin CD13 can cleave tuftsin, generating an antagonist for receptor binding
Cytokines Truncates CXCL11 leading to a reduction of lymphocyte infiltration and a reduced inhibition of endothelial cell migration; cleaves synthetic oligopeptides corresponding to the N-terminal of human IL-1β, IL-2, TNF-β and IL-6. Contradictory results were found for IL-8.

IL: interleukin; TNF-α: tumor necrosis factor alpha; TGF-β1: transforming growth factor beta-1; IL-1RA: IL-1 receptor antagonist; BK: bradykinin; CXCL11: C-X-C motif chemokine 11; CXCR: C-X-C motif chemokine receptor.

1). Monocytes/macrophages

CD13 has been implicated in MN/macrophage activation and differentiation(28). It is expressed on the surface of blood MNs and is upregulated by interferon-γ (IFN-γ), lipopolysaccharides (LPS), C5a, IL-4 and transforming growth factor (TGF)-β but downregulated by IL-10(2831). There is also an increase of CD13 expression during the differentiation from MNs to macrophages(28). Meanwhile, blocking CD13 by mAb or by binding of human cytomegalovirus (HCMV) inhibits macrophage differentiation(32).

CD13 mediates monocyte activation either by crosslinking of cell membrane CD13 or by binding of sCD13 to a GPCR. Crosslinking membrane CD13 by mAbs activates signaling pathways involving MAPK and PI-3K, and a Ca2+ flux, resulting in homotypic aggregation, cytokine secretion and increased adhesive capacity(7, 15, 20, 21). SCD13, binding to a GPCR, increases phosphorylation of multiple downstream signaling molecules and activates MNs(26). Moreover, CD13 interacts with FcγRs on the monocytic cell membrane and regulates FcγR-mediated phagocytosis(16, 33). CD13 also regulates monocyte migration and contributes to inflammation. The expression of CD13 is enriched specifically on the pro-inflammatory subset of MNs, and blocking CD13 by antibody or knockdown of CD13 significantly decreases monocyte infiltration, suggesting that CD13 may regulate trafficking and function of these cells(8). In addition, CD13 expression by ECs is also essential for MN infiltration(8, 34). CD13 is up-regulated on activated ECs (35), and mediates MN/EC adhesion by homotypic interactions, that involve the C-terminal domain(8). In CD13−/− mice, lack of monocytic CD13 ablates anti-CD13-dependent MN adhesion to ECs, yet CD13 appears to be dispensable for phagocytosis, proliferation and antigen presentation of macrophages(36). Meanwhile, assessment of four inflammatory disease models (thioglycollate-induced peritonitis, collagen antibody-induced arthritis, DSS-induced colitis, and croton oil-induced contact hypersensitivity) showed that lack of CD13 had little effect on disease onset or progression(36). These models are, however, relatively independent of autoimmune T cell responses.

2). Dendritic cells

CD13 is involved in the development of dendritic cells from CD34+ hematopoietic progenitor cells(37). CD13 is highly expressed on dendritic cells and participates in cell-surface antigen processing through trimming of MHC II or MHC I-bound peptides on antigen-presenting cells(12, 13, 38). Furthermore, CD13 is highly and specifically expressed on the CD8+ splenic dendritic cells, which are responsible for cross-presentation(39). It negatively regulates receptor-mediated, dynamin-dependent endocytosis of antigens to control T cell activation in adaptive immunity(39). CD13 restricts toll-like receptor 4 endocytic signal transduction, balancing the innate response by maintaining the inflammatory equilibrium critical to innate immune regulation(40). However, development, maturation, antigen processing and presentation of dendritic cells are normal in CD13−/− mice(39).

3). Lymphocytes

Lymphocytes including T cells and B cells are important in inflammation and autoimmunity. However, the expression and function of CD13 on lymphocytes remain controversial. Generally, it is accepted that CD13 is absent on resting lymphocytes but may be inducible by stimulation or contact with some CD13 positive cells. CD13 expression is observed on hematopoietic stem cells and at the earliest stages of B or T cell differentiation but is then lost upon maturation(41, 42). Consistent with this, some malignant B cells were CD13 positive(43, 44). Human T cell lines HuT78 and H9 contained both Ala-pNA hydrolyzing activity and CD13 mRNA; however, CD13 protein was undetectable(45). Mature B and resting T cells lack CD13 expression detectable by standard flow cytometry(46). In contrast, other studies suggested that CD13 mRNA is consistently detected in resting T cells and its surface expression is markedly up-regulated in response to T cell activation(4751). For instance, CD13 expression on T cells can be upregulated by concanavalin A(50) or other mitogens such as PHA and PMA(49). However, induction of CD13 in lymphocytes was not dominantly dependent on de novo protein biosynthesis or glycosylation, as inhibitors of protein biosynthesis and glycosylation only partly prevented mitogen-induced CD13 expression(49). CD13+ T cells have also been found outside the peripheral blood, for instance, in the synovial fluid of patients with arthritis(52), in the pericardial fluid of patients undergoing cardiac surgery(53), or in tumor microenvironment(54). The upregulation of CD13 in lymphocytes was mediated by adhesion to CD13+ cells including fibroblast-like synoviocytes (FLS), MNs/macrophages, and ECs(55), which was attributed to increased CD13 promoter activity in lymphocytes through cell-cell contact(56). However, the possibility that lymphocytes increase CD13 on the cell surface by trogocytosis from non-lymphoid cells has not been excluded.

The CD13 inhibitors, actinonin and probestin, suppress inflammation through anti-proliferation and reduced production of proinflammatory cytokines such as IL-1β and IL-2, accompanied by augmented release of anti-inflammatory cytokines including TGF-β1 and IL-1 receptor antagonist (IL-1RA)(46, 57). Although actinonin has no effect on chemotaxis or unstimulated cell migration, it selectively suppresses lymphocyte functions including proliferation and production of IFN-γ, IL-17, as well as TNF-α, and ameliorates autoimmunity in vivo(58). Additionally, CD13 inhibitors could preserve and promote immunosuppressive functions of CD4+CD25+ Treg cells. Inhibition of CD13 on CD4+CD25+ Treg cells by phebestin enhanced expression of FoxP3 and TGF-β1, and ameliorated acute colitis in mice(59, 60). Simultaneous application of dipeptidyl peptidase IV (DP IV, a.k.a DPP-4 or CD26) and CD13 inhibitors significantly suppressed DNA synthesis and increased TGF-β1 production in mitogen- or anti-CD3-stimulated human T cells in vitro(61, 62). In response to CD13 inhibitors, there is a marked increase of expression and activity of p42/ERK2(63); meanwhile, a reduction of expression and phosphorylation of glycogen synthase kinase-3β (GSK-3β) occurs(64). However, another study showed that the CD13+CD4+CD25hi Treg subpopulation exhibits stronger suppressive function among CD4+CD25hi Treg cells by expressing a higher level of Foxp3, CTLA-4, membrane-bound TGF-β1,and B7H1(65). A CD13 crosslinking antibody, WM15, suppresses expression of Foxp3, CTLA-4, B7H1, the secretion of TGF-β1 and IL-10 by CD13+CD4+CD25hi Treg cells, and their ability to suppress CD25 expression and proliferation of CD4+CD25 T cells(65).

4). Neutrophils

CD13 is present on neutrophils and can be upregulated by proinflammatory agonists including fMLF, IL-8 and TNF-α(66). Anti-CD13 mAbs, WM15 and MY7, impair IL-8 mediated neutrophil migration in type I collagen gels, and induce significant homotypic aggregation of neutrophils, which is dependent on CD13 cross-linking(66). Neutrophil apoptosis is critical to resolution of acute inflammation and prevention of granulocyte-mediated tissue injury(67). TNF-α was found as a priming agonist to enhance the rate of neutrophil apoptosis, while CD13 could regulate TNFα-induced apoptosis in human neutrophils(68). Inhibition of cell surface CD13 using CD13 inhibitors such as actinonin, or bestatin significantly enhanced TNF-α-induced apoptosis of neutrophils(68). A possible mechanism was that CD13 inhibition interfered with shedding of TNF receptor 1, resulting in augmented TNFα-induced apoptosis, cell polarization, and respiratory burst(68).

5). Mast cells

Evidence of CD13’s role in mast cells is limited. One study claimed that CD13 is a negative regulator of mast cell activation, because antigen stimulation of CD13-deficient bone marrow-derived mast cells increased degranulation and proinflammatory cytokine production(69). This study also identified a functional interaction between FcεRI and CD13 on mast cells as antibody-mediated cross-linking of CD13 caused IL-6 production in an FcεRI-dependent manner(69). However, the pathophysiological or physical connection between CD13 cross-linking and FcεRI activation remains uncertain.

6). Endothelial cells

Angiogenesis is a critical step for tumor growth as well as metastasis and an integral component of the pathologic inflammatory response in arthritis. CD13 was exclusively expressed by angiogenic or activated ECs but not resting ECs(35, 70). Hypoxia, angiogenic growth factors, and signals mediating angiogenesis potently induce CD13 transcription in primary ECs, while CD13 inhibitors interfered with tube formation but not EC proliferation, suggesting that CD13 is a crucial regulator of endothelial morphogenesis during angiogenesis(35, 71). Otherwise, addition of exogenous CD13 is sufficient to restore arrested endothelial migration and morphogenesis resulting from inhibition of Ras/MAPK or PI-3K signaling(72). In vivo, CD13−/− mice showed a severely impaired angiogenic response under pathological conditions(73). One possible mechanism is that CD13 mediates galectin-3-induced angiogenesis through carbohydrate-recognition interactions(74). Alternatively, CD13, through its peptidase activity, can regulate endothelial invasion and filopodia formation by facilitating bradykinin (BK)-bradykinin receptor B2 (B2R) internalization and plasma membrane protein organization(75). Pharmacological inhibitors of CD13 and anti-CD13 mAbs blocked angiogenesis and endothelial invasion(75). Moreover, recent work demonstrated that sCD13 can induce angiogenesis through GPCRs(26).

7). Fibroblasts

Synovial fibroblasts (a.k.a FLS) reside in the synovial lining of joints and act as key contributors to arthritis(76). In RA, activated FLS are a major source of inflammatory cytokines and catabolic enzymes that promote joint damage(76). CD13 was found to be highly expressed on RA FLS(19, 77), and upregulated by pro-inflammatory cytokines including IL-17, IFN-γ, and TNF-α(25). CD13 is more abundant in synovial fluids of RA compared to osteoarthritis (OA)(19). Moreover, CD13 is present as both a soluble molecule and on extracellular vesicles in biological fluids including plasma, synovial fluid, and FLS culture supernatant(25). SCD13 arises from the shedding of membrane CD13 on the FLS surface by MMPs such as MMP14(25). CD13 is localized predominantly in caveolae or lipid rafts on RA FLS, which are hotspot regions for functional cell-cell interactions(78). Inhibition of CD13 by either inhibitors of enzymatic activity or mAb resulted in decreased proliferation and diminished migration of RA FLS(25). Similarly, there was elevated expression of CD13 in dermal fibroblasts at wound sites(79). Blocking CD13 with mAbs (WM15, 3D8, and H300) reduced the migration activity of dermal fibroblast in a dose-dependent manner without any antiproliferative or cytotoxic effect(79). Recent work has found that soluble CD13 increases expression of some proinflammatory cytokines including IL-6 and monocyte chemoattractant protein-1 (MCP-1) in RA FLS(26). Moreover, soluble CD13 activated signaling pathways in RA FLS through GPCRs, and pertussis toxin blocked this effect(26).

CD13 regulates functions of inflammatory mediators

Biological activities of inflammatory mediators can change robustly following short deletions or mutations at the N-terminus. By cleaving N-terminal amino acids, CD13 can regulate the activity of numerous hormones, cytokines and chemokines that participate in inflammation (Table 1). Kinins are naturally occurring vasoactive peptide hormones, influencing inflammation, nociception, and cardiovascular homeostasis. The kinin family contains BK, kallidin (Lys-BK), and their des-Arg derivatives des-Arg9-BK and Lys-des-Arg9-BK, respectively)(80), and both kallidin and its derivative Lys-des-Arg9-BK are CD13 substrates. N-terminal Lys residue of Lys-des-Arg9-BK can be cleaved by CD13, yielding the much less potent des-Arg9-BK(81). Des-Arg9-BK or BK is not as susceptible to CD13 degradation because the peptide bond preceding a proline is resistant to this enzyme(27, 82). Nevertheless, CD13 controls EC invasion in response to BK by facilitating signal transduction at plasma membrane, after BK binding but prior to ligand-receptor internalization; inhibition of CD13 enzymatic activity abrogates B2R internalization, leading to the attenuation of downstream events such as BK-induced activation of Cdc42 and filopodia formation, thus affecting endothelial cell motility(75). Interestingly, inhibitors of two BK receptors directly inhibited CD13 as well(83, 84).

Tuftsin, a natural tetrapeptide (TKPA) located in the Fc-domain of the IgG heavy chain, can enhance phagocytosis, immune responses, bactericidal, and anti-tumor activities. It is hydrolyzed by CD13, generating an antagonist that competes for receptor binding and regulates tuftsin functions(82).

CXCL11 selectively recruits immune cells at sites of inflammation and inhibits angiogenesis, upon binding to its receptors CXCR3 and CXCR7(85). In combination with DP IV, CD13 rapidly processed CXCL11 to generate truncated forms, which may bind and desensitize CXCR3 as well as CXCR7, leading to a reduction of lymphocyte infiltration and an increase of EC migration(86). In vitro, CD13 cleaves synthetic oligopeptides corresponding to the N-terminus of human IL-1β, IL-2, TNF-β (lymphotoxin) and murine IL-6, although natural IL-1α, IL-1β, IL-2 and G-CSF are resistant to CD13(87). This suggests that proteolysis of cytokines by cellular exopeptidases may require one or more preceding steps. CD13 degraded IL-8 and inactivated its chemotactic activity(88), while other groups found that IL-8 was resistant to degradation by CD13(86, 89). More studies are needed to clarify knowledge about inflammatory mediators processed by CD13.

The role of CD13 in the pathogenesis of inflammatory disorders

1). Rheumatoid arthritis

Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by persistent synovitis, systemic inflammation, and autoantibodies, causing cartilage and bone damage as well as disability(90). CD13 is strongly expressed in RA synovial tissue(28, 91), specifically, it is highly expressed by RA FLS and synovial MNs, T cells and is also present in synovial fluid(19, 52, 77). In addition, CD13 is also found in serum and FLS culture supernatant, which contains sCD13 cleaved from FLS by MMP-14 or secreted in extracellular vesicles(25). However, measurement of sCD13 in RA sera did not show higher levels than in normal sera(19). Complementary DNA microarray analysis showed that the gene encoding laeverin (an enzyme with sequence homology to CD13) was found to be the most significantly overexpressed gene in RA twins’ lymphoblastoid B cell lines compared to those from healthy twins(92).

CD13 facilitates cell trafficking and cytokine secretion of immune-related cells that play an important role in the pathogenesis of RA. SCD13 induced chemotaxis of lymphocytes, especially cytokine activated T cells, a T cell population similar to that found in RA synovium(19, 77). This chemotactic activity was mediated through GPCRs are inhibited by pertussis toxin(19). Immunodepleting CD13 from RA synovial fluid removed a substantial fraction of the total cytokine-activated T cell(19) and MN(26) chemotactic activity from that fluid. Inhibitors of CD13 enzymatic activity and anti-CD13 antibodies also decreased growth and migration of FLS(25). Neutralizing antibodies against CD13 inhibited secretion of proinflammatory cytokines such as IL-6, IL-8, and MCP-1 from FLS(93) and RA synovial tissue ex vivo(26). Remarkably, sCD13 also increased MN migration, angiogenesis, and cytokine expression by RA FLS, which are key processes for joint inflammation(26). Injection of sCD13 into mouse knees caused severe joint inflammation in 24 hours while upregulating cytokines such as IL-1β, IL-6, and MCP-1, and augmenting MN/macrophage infiltration(26).

2). Psoriasis

Psoriasis is an immune-mediated disease manifesting in skin or joints or both(94). The skin plaques are characterized by epidermal hyperplasia with hyperproliferation, epidermal differentiation impairment, angiogenesis and a brisk infiltration of MNs/macrophages, dendritic cells, and activated T cells in the dermis and the epidermis(95). CD13 has been identified as an activation marker in immune cells(27, 57) and keratinocytes(96, 97). Moreover, it is over-expressed in psoriatic skin and skin fibroblasts compared to skin of healthy control(98). Calcitonin gene related peptide and IL-4 were implicated in the regulation of CD13 expression and activity(98). Inhibiting CD13 with actinonin, bestatin, or PAC-22 (a cytosol alanyl aminopeptidase-specific inhibitor) led to dose-dependent suppression of DNA synthesis in keratinocytes, correlating well with the simultaneous decrease in enzyme activity(97). In a mouse tail model of psoriasis, actinonin dose-dependently restored the stratum granulosum and ameliorated the impaired keratinocyte differentiation(97). In clinical situations, epidermal CD13 was especially up-regulated in psoriatic plaques, and successful treatments were associated with a reduction of CD13 expression(99).

3). Multiple sclerosis

Multiple sclerosis (MS) is the most prevalent chronic inflammatory disease of the central nervous system, and it is currently incurable(100). A complex immune process, involving T cells, B cells, antibodies, and cells of the innate immune system such as bloodborne macrophages and microglia, is found in lesions of MS(100). Increased expression of CD13 on lymphocytes and PBMC of MS patients was found during acute exacerbation and chronic progression, compared to MS remission or other neurological diseases, suggesting that CD13 can be a cell activation and migration marker in MS(101103). Compared to use of a single ectopeptidase inhibitor, combined application of DP IV and CD13 inhibitors increased suppression of DNA synthesis as well as increased TGF-β1 production in human PBMC and isolated T cells(62). In experimental autoimmune encephalomyelitis (EAE), an animal model of MS, combined DP IV and CD13 inhibition dramatically ameliorated clinical severity of EAE compared to the response to each of the inhibitors alone(62). Therefore, a dual inhibitor of DP IV and CD13, PETIR-001, exhibits a therapeutic effect in EAE(104). Although inhibitors of DP IV and CD13 have no effect on migration of pathogenic effector T cells into target tissues, they induce release of TGF-β by T cells at the site of inflammation, and suppress lymphocyte proliferation and production of IFN-γ, IL-17 and TNF-α(58). The amount of IL-17 and the number of CD13+IL-17R+ myeloid cells in peripheral blood were steadily decreased in MS patients with effective treatment(105).

4). Inflammatory bowel diseases

Inflammatory bowel diseases (IBD), comprising Crohn’s disease and ulcerative colitis, result from an inappropriate inflammatory response to intestinal microbes(106). In a mouse model of colitis, simultaneously inhibiting DP IV and CD13 reduced colitis activity significantly(107). Endogenous opioid peptides such as Met- and Leu-enkephalin have beneficial impact on the function of the digestive system and exert anti-inflammatory effects through direct influence on immune cells. However, those peptides are rapidly degraded by endogenous enkephalinases including neutral endopeptidase (NEP) and CD13. Blocking NEP and CD13 with Pal-KKQHNPR (an analog of natural enkephalin) or sialorphin (a natural blocker) attenuated colitis in mice(108, 109).

It has been noticed that HCMV infection induces production of CD13-specific autoantibodies, which may promote inflammation and tissue damage(110112). Evidence suggests an association between HCMV infection and IBD, but the mechanism is unclear. However, cytotoxic CD13-specific autoantibodies were identified in 66% of the sera obtained from HCMV-IgG positive patients with ulcerative colitis and in 58% of the sera obtained from HCMV-IgG positive patients with Crohn’s disease, but not in control individuals(113). These cytotoxic autoantibodies may interfere with cell functions and could thereby contribute to chronic inflammation in patients with IBD.

5). Osteoarthritis

Osteoarthritis (OA), once viewed as mechanical cartilage degradation, is now known to be a complex condition affecting the entire joint, in which matrix proteases, synovium, and systemic inflammation have key roles(114). OA shares some features with RA, and CD13 has also been detected in OA synovial tissue(19, 91). CD13 is expressed on articular chondrocytes, and a proinflammatory cytokine 14–3-3ε might directly bind to CD13, which transmits a signal in chondrocytes to induce a catabolic phenotype similar to that observed in OA(17). Therefore, 14–3-3ε-CD13 interaction could be a new therapeutic target in OA.

7). Others

Significantly higher aminopeptidase activity was detected in bronchoalveolar lavage fluid from patients with interstitial lung diseases (ILD) due to RA, polymyositis/dermomyositis, systemic sclerosis, and Sjogren’s syndrome than from normal volunteers or control patients who were free of ILD, and increased aminopeptidase activity and increased expression of CD13 were found in alveolar macrophages from patients with ILD(115). CD13 may also be a target for acne. DP IV and CD13 inhibitors upregulate IL-1RA in sebocytes and keratinocyte cell line, suppress propionibacterium acnes-stimulated T cell proliferation and IL-2 production, and enhance TGF-β1 expression(116). CD13+CD33+ B cells were more numerous in Behcet’s disease and sepsis compared to healthy controls and RA or systemic lupus erythematosus (SLE) patients(117), but the authors failed to further explore functions of this subset of B cells. Recently, another research group identified a distinct CD13+CD33+ population of leukemic cells contributing to a proinflammatory microenvironment that may be detrimental to long-term normal hematopoiesis within acute lymphoblastic leukemia bone marrow(118). With the capacity to secrete many proinflammatory cytokines, such as IL-1α, IL-1β, TNF-α, and GM-CSF, the CD13+ B cells may contribute to autoimmune diseases such as Behcet’s disease. Activated immune cells such as MNs are important factors in the systemic inflammatory response syndrome (SIRS) following trauma or sepsis. Increased number of tissue factor (TF)+CD13+ microparticles were detected in peripheral blood from patients in the acute phase of trauma and severe sepsis, which correlated significantly with severity, suggesting that TF+CD13+ microparticles are important in the pathogenesis of early SIRS following trauma or sepsis(119). A role for CD13 has been suggested in SLE, but the data are still preliminary(115, 120122). CD13 is also a receptor for a strain of human coronavirus. A broad spectrum of variations in the CD13 domain were found critical for coronavirus binding(123). Some studies pointed out that single site polymorphisms and alternative splicing of human CD13 are characteristic for acute myeloid leukemia, implying that CD13 could be a disease marker and target for therapy(124126). However, to our knowledge, there is no data concerning CD13 polymorphisms in autoimmune diseases/inflammatory diseases.

CD13 inhibitors

Almost 100 CD13 inhibitors have been reported and tested(127, 128). Mina-Osorio divided them into 7 categories: natural inhibitors (betatin, amastatin), synthetic peptidomimetic inhibitors, synthetic non-peptide inhibitors, tumor-homing peptides (NGR), cholesterol-lowering drugs (ezetimibe), recombinant antibodies (single-chain Fv antibody fragments), and monoclonal antibodies (WM15, mAb 452)(4). Most of the clinical studies with CD13 enzymatic inhibitors, such as bestatin and tosedostat, were performed in cancers including leukemia, lymphomas, and carcinomas of lung, bladder, esophagus and stomach. Inhibition of aminopeptidase yielded a higher efficacy in some cancer types(129). Several other compounds inhibiting CD13 enzymatic activity are under preclinical development, for instance, LYP, a bestatin dimethylaminoethyl ester(130). Curcumin, a phenolic natural product, has been described as an irreversible inhibitor of CD13 and is currently being investigated for its effects on patients with cancer(131) as well as patients with inflammation (available from ClinicalTrials.gov URL: https://clinicaltrials.gov/ct2/show/NCT04032132?term=Curcumin&draw=2&rank=14; https://clinicaltrials.gov/ct2/show/NCT03122613?term=Curcumin&draw=2&rank=12). However, there is no available data concerning the clinical use of CD13 mAbs. To our knowledge, no clinical trial has been completed to test CD13 inhibitors in inflammatory diseases, except one ongoing phase II trial on psoriasis studying IP10.C8, a combined DP IV and CD13 inhibitor. The possible reason for this could be the complexity and extensiveness of CD13 functions. CD13 is essential for proper trafficking of inflammatory cells which is necessary to prime and sustain the reparative responses(9). Therefore, the role of CD13 as well as the potential use of CD13 inhibitors in inflammatory diseases requires further investigation.

Conclusions

In summary, CD13 functions though enzyme-dependent mechanisms and enzyme-independent mechanisms. It can not only modulate development and activities of immune-related cells, but also regulate functions of inflammatory mediators. Furthermore, CD13 plays an important role in various inflammatory disorders, including rheumatoid arthritis, psoriasis, multiple sclerosis, and inflammatory bowel diseases. Many inhibitors against CD13 have been found and tested, yet none of them has been used for the clinical treatment of human inflammatory disorders. The contribution of CD13 to various diseases and its significance as a therapeutic target need further study.

Acknowledgements

We thank Jonatan Hervoso and Mikel Gurrea from the University of Michigan for their generous help in reviewing the manuscript. Special thanks are due to the University of Michigan librarians for their assistance with access to literature.

Abbreviations used in this article

sCD13

soluble CD13

MHC

major histocompatibility complex class

GPCR

G-protein coupled receptor

MN

monocyte

MAPK

mitogen-activated protein kinases

PI-3K

phosphatidylinositol 3-kinase

IQGAP1

IQ motif containing GTPase activating protein 1

ARF

adenosine diphosphate ribosylation factor

EFA6

exchange factor for ARF6

NGR

Asn-Gly-Arg

PBMC

peripheral blood mononuclear cells

FLS

fibroblast-like synoviocytes

EC

endothelial cell

RA

rheumatoid arthritis

OA

osteoarthritis

SLE

systemic lupus erythematosus

ILD

interstitial lung diseases

MMP

matrix metalloproteinase

DP IV

dipeptidyl peptidase IV

EAE

experimental autoimmune encephalomyelitis

LPS

lipopolysaccharides

fMLF

formyl-methionyl-leucyl-phenylalanine

PHA

phytohemagglutinin

PMA

phorbol myristate acetate

TNF

tumor necrosis factor

IFN

interferon

IL

interleukin

IL-1RA

interleukin 1 receptor antagonist

SIRS

systemic inflammatory response syndrome

TF

tissue factor

MCP-1

monocyte chemoattractant protein-1

TGF

transforming growth factor

mAb

monoclonal antibody

BK

bradykinin

B2R

bradykinin B2 receptor

Footnotes

Disclosure: The authors have no financial conflicts of interest.

References

  • 1.Wong AH, Zhou D, and Rini JM. 2012. The X-ray crystal structure of human aminopeptidase N reveals a novel dimer and the basis for peptide processing. J. Biol. Chem 287: 36804–36813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Chen L, Lin Y, Peng G, and Li F. 2012. Structural basis for multifunctional roles of mammalian aminopeptidase N. Proc. Natl. Acad. Sci. U. S. A 109: 17966–17971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Sjöström H, Norén O, and Olsen J. 2000. Structure and function of aminopeptidase N. Adv. Exp. Med. Biol 477: 25–34. [DOI] [PubMed] [Google Scholar]
  • 4.Mina-Osorio P 2008. The moonlighting enzyme CD13: old and new functions to target. Trends Mol. Med 14: 361–371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mouritsen S, Meldal M, Werdelin O, Hansen A, and Buus S. 1992. MHC molecules protect T cell epitopes against proteolytic destruction. J. Immunol 149: 1987–1993. [PubMed] [Google Scholar]
  • 6.Falk K, Rötzschke O, Stevanović S, Jung G, and Rammensee H. 1994. Pool sequencing of natural HLA-DR, DQ, and DP ligands reveals detailed peptide motifs, constraints of processing, and general rules. Immunogenetics. 39: 230–242. [DOI] [PubMed] [Google Scholar]
  • 7.Mina-Osorio P, Shapiro L, and Ortega E. 2006. CD13 in cell adhesion: aminopeptidase N (CD13) mediates homotypic aggregation of monocytic cells. J. Leukoc. Biol 79: 719–730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ghosh M, Gerber C, Rahman MM, Vernier KM, Pereira FE, Subramani J, Caromile LA, and Shapiro LH. 2014. Molecular mechanisms regulating CD13-mediated adhesion. Immunology. 142: 636–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pereira FE, Cronin C, Ghosh M, Zhou SY, Agosto M, Subramani J, Wang R, Shen JB, Schacke W, Liang B, Yang TH, McAulliffe B, Liang BT, and Shapiro LH. 2013. CD13 is essential for inflammatory trafficking and infarct healing following permanent coronary artery occlusion in mice. Cardiovasc. Res. 100: 74–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ghosh M, Lo R, Ivic I, Aguilera B, Qendro V, Devarakonda C, and Shapiro L. 2019. CD13 tethers the IQGAP1-ARF6-EFA6 complex to the plasma membrane to promote ARF6 activation, β1 integrin recycling, and cell migration. Sci Signal. 12: pii: eaav5938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hooper N 1994. Families of zinc metalloproteases. FEBS Lett. 354: 1–6. [DOI] [PubMed] [Google Scholar]
  • 12.Hansen A, Norén O, Sjöström H, and Werdelin O. 1993. A mouse aminopeptidase N is a marker for antigen-presenting cells and appears to be co-expressed with major histocompatibility complex class II molecules. Eur. J. Immunol 23: 2358–2364. [DOI] [PubMed] [Google Scholar]
  • 13.Larsen SL, Pedersen LO, Buus S, and Stryhn A. 1996. T cell responses affected by aminopeptidase N (CD13)-mediated trimming of major histocompatibility complex class II-bound peptides. J. Exp. Med 184: 183–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Dong X, An B, Salvucci Kierstead L, Storkus WJ, Amoscato AA, and Salter RD. 2000. Modification of the Amino Terminus of a Class II Epitope Confers Resistance to Degradation by CD13 on Dendritic Cells and Enhances Presentation to T Cells. The Journal of Immunology. 164: 129–135. [DOI] [PubMed] [Google Scholar]
  • 15.Santos AN, Langner J, Herrmann M, and Riemann D. 2000. Aminopeptidase N/CD13 is directly linked to signal transduction pathways in monocytes. Cell. Immunol 201: 22–32. [DOI] [PubMed] [Google Scholar]
  • 16.Mina-Osorio P, and Ortega E. 2005. Aminopeptidase N (CD13) functionally interacts with FcgammaRs in human monocytes. J. Leukoc. Biol 77: 1008–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nefla M, Sudre L, Denat G, Priam S, Andre-Leroux G, Berenbaum F, and Jacques C. 2015. The pro-inflammatory cytokine 14–3-3ε is a ligand of CD13 in cartilage. J. Cell Sci 128: 3250–3262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ghaffari A, Li Y, Kilani R, and Ghahary A. 2010. 14–3-3 sigma associates with cell surface aminopeptidase N in the regulation of matrix metalloproteinase-1. J. Cell Sci: 2996–3005. [DOI] [PubMed] [Google Scholar]
  • 19.Morgan R, Endres J, Behbahani-Nejad N, Phillips K, Ruth JH, Friday SC, Edhayan G, Lanigan T, Urquhart A, Chung KC, and Fox DA. 2015. Expression and function of aminopeptidase N/CD13 produced by fibroblast-like synoviocytes in rheumatoid arthritis: role of CD13 in chemotaxis of cytokine-activated T cells independent of enzymatic activity. Arthritis Rheumatol. 67: 74–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Subramani J, Ghosh M, Rahman MM, Caromile LA, Gerber C, Rezaul K, Han DK, and Shapiro LH. 2013. Tyrosine phosphorylation of CD13 regulates inflammatory cell-cell adhesion and monocyte trafficking. J. Immunol 191: 3905–3912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mina-Osorio P, Soto-Cruz I, and Ortega E. 2007. A role for galectin-3 in CD13-mediated homotypic aggregation of monocytes. Biochem. Biophys. Res. Commun 353: 605–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.White CD, Erdemir HH, and Sacks DB. 2012. IQGAP1 and its binding proteins control diverse biological functions. Cell. Signal 24: 826–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Garay-Canales CA, Licona-Limon I, and Ortega E. 2018. Distinct Epitopes on CD13 Mediate Opposite Consequences for Cell Adhesion. Biomed Res Int. 2018: 4093435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Riemann D, Tcherkes A, Hansen GH, Wulfaenger J, Blosz T, and Danielsen EM. 2005. Functional co-localization of monocytic aminopeptidase N/CD13 with the Fc gamma receptors CD32 and CD64. Biochem. Biophys. Res. Commun 331: 1408–1412. [DOI] [PubMed] [Google Scholar]
  • 25.Morgan RL, Behbahani-Nejad N, Endres J, Amin MA, Lepore NJ, Du Y, Urquhart A, Chung KC, and Fox DA. 2016. Localization, Shedding, Regulation and Function of Aminopeptidase N/CD13 on Fibroblast like Synoviocytes. PLoS One. 11: e0162008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Du Y, Lu C, Morgan R, Stinson W, Campbell P, Cealey E, and Fu W. 2019. Angiogenic and Arthritogenic Properties of the Soluble Form of CD13. J. Immunol 203: 360–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Riemann D, Kehlen A, and Langner J. 1999. CD13--not just a marker in leukemia typing. Immunol. Today 20: 83–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Koch AE, Burrows JC, Skoutelis A, Marder R, Domer PH, Anderson B, and Leibovich SJ. 1991. Monoclonal antibodies detect monocyte/macrophage activation and differentiation antigens and identify functionally distinct subpopulations of human rheumatoid synovial tissue macrophages. Am. J. Pathol 138: 165–173. [PMC free article] [PubMed] [Google Scholar]
  • 29.Kehlen A, Geisler M, Olsen J, Sablotzki A, Langner J, and Riemann D. 2004. IL-10 and TGF-beta differ in their regulation of aminopeptidase N/CD13 expression in monocytes. Int. J. Mol. Med 13: 877–882. [DOI] [PubMed] [Google Scholar]
  • 30.van Hal P, Hopstaken-Broos J, Prins A, Favaloro E, Huijbens R, Hilvering C, Figdor C, and Hoogsteden H. 1994. Potential indirect anti-inflammatory effects of IL-4. Stimulation of human monocytes, macrophages, and endothelial cells by IL-4 increases aminopeptidase-N activity (CD13; EC 3.4.11.2). J. Immunol 153: 2718–2728. [PubMed] [Google Scholar]
  • 31.Werfel T, Sonntag G, Weber M, and Götze O. 1991. Rapid increases in the membrane expression of neutral endopeptidase (CD10), aminopeptidase N (CD13), tyrosine phosphatase (CD45), and Fc gamma-RIII (CD16) upon stimulation of human peripheral leukocytes with human C5a. J. Immunol 147: 3909–3914. [PubMed] [Google Scholar]
  • 32.Gredmark S, Britt WB, Xie X, Lindbom L, and Soderberg-Naucler C. 2004. Human cytomegalovirus induces inhibition of macrophage differentiation by binding to human aminopeptidase N/CD13. J. Immunol 173: 4897–4907. [DOI] [PubMed] [Google Scholar]
  • 33.MacIntyre E, Roberts P, Jones M, Van der Schoot C, Favalaro E, Tidman N, and Linch D. 1989. Activation of human monocytes occurs on cross-linking monocytic antigens to an Fc receptor. J. Immunol 142: 2377–2383. [PubMed] [Google Scholar]
  • 34.Mina-Osorio P, Winnicka B, O’Conor C, Grant C, Vogel L, Rodriguez-Pinto D, Holmes K, Ortega E, and Shapiro L. 2008. CD13 is a novel mediator of monocytic/endothelial cell adhesion. J. Leukoc. Biol 84: 448–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bhagwat S, Lahdenranta J, Giordano R, Arap W, Pasqualini R, and Shapiro L. 2001. CD13/APN is activated by angiogenic signals and is essential for capillary tube formation. Blood. 97: 652–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Winnicka B, O’Conor C, Schacke W, Vernier K, Grant C, Fenteany F, Pereira F, and Liang B. 2010. CD13 is dispensable for normal hematopoiesis and myeloid cell functions in the mouse. J. Leukoc. Biol 88: 347–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rosenzwajg M, Tailleux L, and Gluckman J. 2000. CD13/N-aminopeptidase is involved in the development of dendritic cells and macrophages from cord blood CD34(+) cells. Blood. 95: 453–460. [PubMed] [Google Scholar]
  • 38.Amoscato A, Prenovitz D, and Lotze M. 1998. Rapid extracellular degradation of synthetic class I peptides by human dendritic cells. J. Immunol 161: 4023–4032. [PubMed] [Google Scholar]
  • 39.Ghosh M, McAuliffe B, Subramani J, Basu S, and Shapiro LH. 2012. CD13 regulates dendritic cell cross-presentation and T cell responses by inhibiting receptor-mediated antigen uptake. J. Immunol 188: 5489–5499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ghosh M, Subramani J, Rahman MM, and Shapiro LH. 2015. CD13 restricts TLR4 endocytic signal transduction in inflammation. J. Immunol 194: 4466–4476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Spits H, Lanier L, and Phillips J. 1995. Development of human T and natural killer cells. Blood. 85: 2654–2670. [PubMed] [Google Scholar]
  • 42.Syrjälä M, Ruutu T, and Jansson S. 1994. A flow cytometric assay of CD34-positive cell populations in the bone marrow. Br. J. Haematol 88: 679–684. [DOI] [PubMed] [Google Scholar]
  • 43.Pinto A, Del Vecchio L, Carbone A, Roncadin M, Volpe R, Serraino D, Monfardini S, Colombatti A, and Zagonel V. 1991. Expression of myelomonocytic antigens is associated with unfavourable clinicoprognostic factors in B-cell chronic lymphocytic leukaemia. Ann. Oncol. 2: 107–113. [DOI] [PubMed] [Google Scholar]
  • 44.Drexler H, Thiel E, and Ludwig W. 1991. Review of the incidence and clinical relevance of myeloid antigen-positive acute lymphoblastic leukemia. Leukemia. 5: 637–645. [PubMed] [Google Scholar]
  • 45.Lendeckel U, Wex T, Kähne T, Frank K, Reinhold D, and Ansorge S. 1994. Expression of the aminopeptidase N (CD13) gene in the human T cell lines HuT78 and H9. Cell. Immunol. 153: 214–226. [DOI] [PubMed] [Google Scholar]
  • 46.Lendeckel U, Bukowska A, Lättig J, and Brandt W. 2004. Alanylaminopeptidases in human T cells. Plenum Publishers, New York: Kluwer Academic. [Google Scholar]
  • 47.Lendeckel U, Wex T, Reinhold D, Kähne T, Frank K, Faust J, Neubert K, and Ansorge S. 1996. Induction of the membrane alanyl aminopeptidase gene and surface expression in human T-cells by mitogenic activation. Biochem. J. 319: 817–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lendeckel U, Wex T, Reinhold D, Arndt M, Ittenson A, Frank K, and Ansorge S. 1997. Activation-dependent induction of T cell alanyl aminopeptidase and its possible involvement in T cell growth. Adv. Exp. Med. Biol 421: 59–66. [DOI] [PubMed] [Google Scholar]
  • 49.Lendeckel U, Wex T, Ittenson A, Arndt M, Frank K, Mayboroda O, Schubert W, and Ansorge S. 1997. Rapid mitogen-induced aminopeptidase N surface expression in human T cells is dominated by mechanisms independent of de novo protein biosynthesis. Immunobiology. 197: 55–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kunz D, Bühling F, Hütter H, Aoyagi T, and Ansorge S. 1993. Aminopeptidase N (CD13, EC 3.3.4.11.2) occurs on the surface of resting and concanavalin A-stimulated lymphocytes. Biol. Chem. Hoppe Seyler 374: 291–296. [DOI] [PubMed] [Google Scholar]
  • 51.Bukowska A, Tadje J, Arndt M, Wolke C, Kähne T, Bartsch J, Faust J, Neubert K, Hashimoto Y, and Lendeckel U. 2003. Transcriptional regulation of cytosol and membrane alanyl-aminopeptidase in human T cell subsets. Biol. Chem 384: 657–665. [DOI] [PubMed] [Google Scholar]
  • 52.Riemann D, Schwachula A, Hentschel M, and Langner J. 1993. Demonstration of CD13/aminopeptidase N on synovial fluid T cells from patients with different forms of joint effusions. Immunobiology. 187: 24–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Riemann D, Wollert H, Menschikowski J, Mittenzwei S, and Langner J. 1994. Immunophenotype of lymphocytes in pericardial fluid from patients with different forms of heart disease. Int. Arch. Allergy Immunol 104: 48–56. [DOI] [PubMed] [Google Scholar]
  • 54.Riemann D, Göhring B, and Langner J. 1994. Expression of aminopeptidase N/CD13 in tumour-infiltrating lymphocytes from human renal cell carcinoma. Immunol. Lett 42: 19–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Riemann D, Kehlen T A K, Löhn M, and Langner J. 1997. Induction of aminopeptidase N/CD13 on human lymphocytes after adhesion to fibroblast-like synoviocytes, endothelial cells, epithelial cells, and monocytes/macrophages. J. Immunol 158: 3425–3432. [PubMed] [Google Scholar]
  • 56.Kehlen A, Olsen J, Langner J, and Riemann D. 2000. Increased lymphocytic aminopeptidase N/CD13 promoter activity after cell-cell contact. J. Cell. Biochem 80: 115–123. [DOI] [PubMed] [Google Scholar]
  • 57.Lendeckel U, Arndt M, Frank K, Wex T, and Ansorge S. 1999. Role of alanyl aminopeptidase in growth and function of human T cells (review). Int. J. Mol. Med 4: 17–27. [PubMed] [Google Scholar]
  • 58.Biton A, Ansorge S, Bank U, Tager M, Reinhold D, and Brocke S. 2011. Divergent actions by inhibitors of DP IV and APN family enzymes on CD4+ Teff cell motility and functions. Immunobiology. 216: 1295–1301. [DOI] [PubMed] [Google Scholar]
  • 59.Bank U, Tadje J, Täger M, Wolke C, Bukowska A, Ittenson A, and Reinhold D. 2007. Inhibition of alanyl-aminopeptidase on CD4+CD25+ regulatory T-cells enhances expression of FoxP3 and TGF-beta1 and ameliorates acute colitis in mice. Int. J. Mol. Med. 20: 483–492. [PubMed] [Google Scholar]
  • 60.Lendeckel U, Bukowska A, Lättig J, and Brandt W. 2004. Alanyl-aminopeptidases in human T cells. Kluwer Academic/Plenum Publishers, New York. [Google Scholar]
  • 61.Reinhold D, Biton A, Goihl A, Pieper S, Lendeckel U, Faust J, Neubert K, Bank U, Tager M, Ansorge S, and Brocke S. 2007. Dual inhibition of dipeptidyl peptidase IV and aminopeptidase N suppresses inflammatory immune responses. Ann. N. Y. Acad. Sci 1110: 402–409. [DOI] [PubMed] [Google Scholar]
  • 62.Reinhold D, Biton A, Pieper S, Lendeckel U, Faust J, Neubert K, Bank U, Tager M, Ansorge S, and Brocke S. 2006. Dipeptidyl peptidase IV (DP IV, CD26) and aminopeptidase N (APN, CD13) as regulators of T cell function and targets of immunotherapy in CNS inflammation. Int. Immunopharmacol 6: 1935–1942. [DOI] [PubMed] [Google Scholar]
  • 63.Lendeckel U, Kähne T, Arndt M, Frank K, and Ansorge S. 1998. Inhibition of alanyl aminopeptidase induces MAP-kinase p42/ERK2 in the human T cell line KARPAS-299. Biochem. Biophys. Res. Commun. 252: 5–9. [DOI] [PubMed] [Google Scholar]
  • 64.Lendeckel U, Scholz B, Arndt M, Frank K, Spiess A, Chen H, Roques BP, and Ansorge S. 2000. Inhibition of alanyl-aminopeptidase suppresses the activation-dependent induction of glycogen synthase kinase-3beta (GSK-3beta) in human T cells. Biochem. Biophys. Res. Commun 273: 62–65. [DOI] [PubMed] [Google Scholar]
  • 65.Ju S, Qiu H, Zhou X, Zhu B, Lv X, Huang X, Li J, and Zhang Y. 2009. CD13+CD4+CD25hi regulatory T cells exhibit higher suppressive function and increase with tumor stage in non-small cell lung cancer patients. Cell Cycle. 8: 2578–2585. [DOI] [PubMed] [Google Scholar]
  • 66.Fiddler C, Parfrey H, Cowburn A, Luo D, Nash G, Murphy G, and Chilvers E. 2016. The Aminopeptidase CD13 Induces Homotypic Aggregation in Neutrophils and Impairs Collagen Invasion. PLoS One. 11: e0160108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Fox S, Leitch AE, Duffin R, Haslett C, and Rossi AG. 2010. Neutrophil apoptosis: relevance to the innate immune response and inflammatory disease. J. Innate Immun. 2: 216–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cowburn AS, Sobolewski A, Reed BJ, Deighton J, Murray J, Cadwallader KA, Bradley JR, and Chilvers ER. 2006. Aminopeptidase N (CD13) regulates tumor necrosis factor-alpha-induced apoptosis in human neutrophils. J. Biol. Chem 281: 12458–12467. [DOI] [PubMed] [Google Scholar]
  • 69.Zotz JS, Wolbing F, Lassnig C, Kauffmann M, Schulte U, Kolb A, Whitelaw B, Muller M, Biedermann T, and Huber M. 2016. CD13/aminopeptidase N is a negative regulator of mast cell activation. FASEB J. 30: 2225–2235. [DOI] [PubMed] [Google Scholar]
  • 70.Fukasawa K, Fujii H, Saitoh Y, Koizumi K, Aozuka Y, Sekine K, Yamada M, Saiki I, and Nishikawa K. 2006. Aminopeptidase N (APN/CD13) is selectively expressed in vascular endothelial cells and plays multiple roles in angiogenesis. Cancer Lett. 243: 135–143. [DOI] [PubMed] [Google Scholar]
  • 71.Shim JS, Kim JH, Cho HY, Yum YN, Kim SH, Park H-J, Shim BS, Choi SH, and Kwon HJ. 2003. Irreversible Inhibition of CD13/Aminopeptidase N by the Antiangiogenic Agent Curcumin. Chem. Biol 10: 695–704. [DOI] [PubMed] [Google Scholar]
  • 72.Bhagwat S, Petrovic N, Okamoto Y, and Shapiro L. 2003. The angiogenic regulator CD13/APN is a transcriptional target of Ras signaling pathways in endothelial morphogenesis. Blood. 101: 1818–1826. [DOI] [PubMed] [Google Scholar]
  • 73.Rangel R, Sun Y, Guzman-Rojas L, Ozawa M, Sun J, Giordano R, Van Pelt C, Tinkey P, Behringer R, Sidman R, Arap W, and Pasqualini R. 2007. Impaired angiogenesis in aminopeptidase N-null mice. Proc. Natl. Acad. Sci. U. S. A 104: 4588–4593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yang E, Shim JS, Woo HJ, Kim KW, and Kwon HJ. 2007. Aminopeptidase N/CD13 induces angiogenesis through interaction with a pro-angiogenic protein, galectin-3. Biochem. Biophys. Res. Commun 363: 336–341. [DOI] [PubMed] [Google Scholar]
  • 75.Petrovic N, Schacke W, Gahagan J, O’Conor C, Winnicka B, Conway R, Mina-Osorio P, and Shapiro L. 2007. CD13/APN regulates endothelial invasion and filopodia formation. Blood. 110: 142–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Neumann E, Lefevre S, Zimmermann B, Gay S, and Muller-Ladner U. 2010. Rheumatoid arthritis progression mediated by activated synovial fibroblasts. Trends Mol. Med 16: 458–468. [DOI] [PubMed] [Google Scholar]
  • 77.Shimizu T, Tani K, Hase K, Ogawa H, Huang L, Shinomiya F, and Sone S. 2002. CD13/aminopeptidase N-induced lymphocyte involvement in inflamed joints of patients with rheumatoid arthritis. Arthritis Rheum. 46: 2330–2338. [DOI] [PubMed] [Google Scholar]
  • 78.Riemann D, Hansen G, Niels-Christiansen L, Thorsen E, Immerdal L, Santos A, Kehlen A, Langner J, and Danielsen E. 2001. Caveolae/lipid rafts in fibroblast-like synoviocytes: ectopeptidase-rich membrane microdomains. Biochem. J. 354: 47–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lai A, Ghaffari A, and Ghahary A. 2010. Inhibitory effect of anti-aminopeptidase N/CD13 antibodies on fibroblast migration. Mol. Cell. Biochem. 343: 191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Leeb-Lundberg L, Marceau F, Müller-Esterl W, Pettibone D, and Zuraw B. 2005. International union of pharmacology. XLV. Classification of the kinin receptor family: from molecular mechanisms to pathophysiological consequences. Pharmacol. Rev 57: 27–77. [DOI] [PubMed] [Google Scholar]
  • 81.Fortin JP, Gera L, Bouthillier J, Stewart JM, Adam A, and Marceau F. 2005. Endogenous aminopeptidase N decreases the potency of peptide agonists and antagonists of the kinin B1 receptors in the rabbit aorta. J. Pharmacol. Exp. Ther. 314: 1169–1176. [DOI] [PubMed] [Google Scholar]
  • 82.Xu Y, Wellner D, and Scheinberg D. 1995. Substance P and bradykinin are natural inhibitors of CD13/aminopeptidase N. Biochem. Biophys. Res. Commun 208: 664–674. [DOI] [PubMed] [Google Scholar]
  • 83.Bawolak MT, Fortin JP, Vogel LK, Adam A, and Marceau F. 2006. The bradykinin B2 receptor antagonist icatibant (Hoe 140) blocks aminopeptidase N at micromolar concentrations: off-target alterations of signaling mediated by the bradykinin B1 and angiotensin receptors. Eur. J. Pharmacol. 551: 108–111. [DOI] [PubMed] [Google Scholar]
  • 84.Gera L, Fortin JP, Adam A, Stewart JM, and Marceau F. 2006. Discovery of a dual-function peptide that combines aminopeptidase N inhibition and kinin B1 receptor antagonism. J. Pharmacol. Exp. Ther 317: 300–308. [DOI] [PubMed] [Google Scholar]
  • 85.Lacotte S, Brun S, Muller S, and Dumortier H. 2009. CXCR3, inflammation, and autoimmune diseases. Ann. N. Y. Acad. Sci 1173: 310–317. [DOI] [PubMed] [Google Scholar]
  • 86.Proost P, Mortier A, Loos T, Vandercappellen J, Gouwy M, Ronsse I, and Schutyser E. 2007. Proteolytic processing of CXCL11 by CD13/aminopeptidase N impairs CXCR3 and CXCR7 binding and signaling and reduces lymphocyte and endothelial cell migration. Blood. 110: 37–44. [DOI] [PubMed] [Google Scholar]
  • 87.Hoffmann T, Faust J, Neubert K, and Ansorge S. 1993. Dipeptidyl peptidase IV (CD 26) and aminopeptidase N (CD 13) catalyzed hydrolysis of cytokines and peptides with N-terminal cytokine sequences. FEBS Lett. 336: 61–64. [DOI] [PubMed] [Google Scholar]
  • 88.Kanayama N, Kajiwara Y, Goto J, el Maradny E, Maehara K, Andou K, and Terao T. 1995. Inactivation of interleukin-8 by aminopeptidase N (CD13). J. Leukoc. Biol. 57: 129–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Kehlen A, Egbert I, Thiele K, Fischer K, Riemann D, and Langner J. 2001. Increased expression of interleukin-8 and aminopeptidase N by cell-cell contact: interleukin-8 is resistant to degradation by aminopeptidase N/CD13. Eur. Cytokine Netw. 12: 316–324. [PubMed] [Google Scholar]
  • 90.Smolen J, Aletaha D, and McInnes I. 2016. Rheumatoid arthritis. Lancet. 388: 2023–2038. [DOI] [PubMed] [Google Scholar]
  • 91.Haringman JJ, Smeets TJ, Reinders-Blankert P, and Tak PP. 2006. Chemokine and chemokine receptor expression in paired peripheral blood mononuclear cells and synovial tissue of patients with rheumatoid arthritis, osteoarthritis, and reactive arthritis. Ann. Rheum. Dis. 65: 294–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Haas CS, Creighton CJ, Pi X, Maine I, Koch AE, Haines GK, Ling S, Chinnaiyan AM, and Holoshitz J. 2006. Identification of genes modulated in rheumatoid arthritis using complementary DNA microarray analysis of lymphoblastoid B cell lines from disease-discordant monozygotic twins. Arthritis Rheum. 54: 2047–2060. [DOI] [PubMed] [Google Scholar]
  • 93.Chomarat P, Rissoan MC, Pin JJ, Banchereau J, and Miossec P. 1995. Contribution of IL-1, CD14, and CD13 in the increased IL-6 production induced by in vitro monocyte-synoviocyte interactions. J. Immunol 155: 3645–3652. [PubMed] [Google Scholar]
  • 94.Boehncke W, and Schön M. 2015. Psoriasis. Lancet. 386: 983–994. [DOI] [PubMed] [Google Scholar]
  • 95.Perera GK, Di Meglio P, and Nestle FO. 2012. Psoriasis. Annu. Rev. Pathol 7: 385–422. [DOI] [PubMed] [Google Scholar]
  • 96.Hunyadi J, Simon M J, Kenderessy A, and Dobozy A. 1993. Expression of monocyte/macrophage markers (CD13, CD14, CD68) on human keratinocytes in healthy and diseased skin. J. Dermatol 20: 341–345. [DOI] [PubMed] [Google Scholar]
  • 97.Thielitz A, Bukowska A, Wolke C, Vetter R, Lendeckel U, Wrenger S, Hashimoto Y, Ansorge S, Gollnick H, and Reinhold D. 2004. Identification of extra- and intracellular alanyl aminopeptidases as new targets to modulate keratinocyte growth and differentiation. Biochem. Biophys. Res. Commun 321: 795–801. [DOI] [PubMed] [Google Scholar]
  • 98.Gerbaud P, Guibourdenche J, Jarray R, Conti M, Palmic P, Leclerc-Mercier S, Bruneau J, Hermine O, Lepelletier Y, and Raynaud F. 2018. APN/CD13 is over-expressed by Psoriatic fibroblasts and is modulated by CGRP and IL-4 but not by retinoic acid treatment. J. Cell. Physiol 233: 958–967. [DOI] [PubMed] [Google Scholar]
  • 99.Bonnekoh B, Pommer AJ, Bockelmann R, Hofmeister H, Philipsen L, and Gollnick H. 2007. Topo-proteomic in situ analysis of psoriatic plaque under efalizumab treatment. Skin Pharmacol. Physiol 20: 237–252. [DOI] [PubMed] [Google Scholar]
  • 100.Reich DS, Lucchinetti CF, and Calabresi PA. 2018. Multiple Sclerosis. N. Engl. J. Med. 378: 169–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ziaber J, Tchórzewski H, Chmielewski H, Baj Z, and Paśnik J. 1999. Increased expression of aminopeptidase N on lymphocytes in multiple sclerosis. Neurol. Neurochir. Pol. 33: 1025–1032. [PubMed] [Google Scholar]
  • 102.Ziaber J, Baj Z, Paśnik J, Chmielewski H, and Tchórzewski H. 2000. Increased expression of neutral endopeptidase (NEP) and aminopeptidase N (APN) on peripheral blood mononuclear cells in patients with multiple sclerosis. Immunol. Lett. 71: 127–129. [DOI] [PubMed] [Google Scholar]
  • 103.Ziaber J, Baj Z, Paśnik J, Chmielewski H, and Tchórzewski H. 2000. Expression of aminopeptidase N (APN) on peripheral blood mononuclear cells’ surface as a marker of these cells’ transendothelial migration properties in the course of multiple sclerosis. Mediators Inflamm. 9: 45–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Reinhold D, Bank U, Entz D, Goihl A, Stoye D, Wrenger S, Brocke S, Thielitz A, Stefin S, Nordhoff K, Heimburg A, Tager M, and Ansorge S. 2011. PETIR-001, a dual inhibitor of dipeptidyl peptidase IV (DP IV) and aminopeptidase N (APN), ameliorates experimental autoimmune encephalomyelitis in SJL/J mice. Biol. Chem 392: 233–237. [DOI] [PubMed] [Google Scholar]
  • 105.Esendagli G, Kurne AT, Sayat G, Kilic AK, Guc D, and Karabudak R. 2013. Evaluation of Th17-related cytokines and receptors in multiple sclerosis patients under interferon beta-1 therapy. J. Neuroimmunol. 255: 81–84. [DOI] [PubMed] [Google Scholar]
  • 106.Abraham C, and Cho J. 2009. Inflammatory bowel disease. N. Engl. J. Med. 361: 2066–2078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Bank U, Heimburg A, Helmuth M, Stefin S, Lendeckel U, Reinhold D, Faust J, Fuchs P, Sens B, Neubert K, Tager M, and Ansorge S. 2006. Triggering endogenous immunosuppressive mechanisms by combined targeting of Dipeptidyl peptidase IV (DPIV/CD26) and Aminopeptidase N (APN/ CD13)--a novel approach for the treatment of inflammatory bowel disease. Int. Immunopharmacol 6: 1925–1934. [DOI] [PubMed] [Google Scholar]
  • 108.Salaga M, Mokrowiecka A, Jacenik D, Cygankiewicz AI, Malecka-Panas E, Kordek R, Krajewska WM, Sobocinska MK, Kamysz E, and Fichna J. 2017. Systemic Administration of Sialorphin Attenuates Experimental Colitis in Mice via Interaction With Mu and Kappa Opioid Receptors. J Crohns Colitis. 11: 988–998. [DOI] [PubMed] [Google Scholar]
  • 109.Kamysz E, Sałaga M, Sobocińska M, Giełdoń A, and Fichna J. 2016. Anti-inflammatory effect of novel analogs of natural enkephalinase inhibitors in a mouse model of experimental colitis. Future Med. Chem 8: 2231–2243. [DOI] [PubMed] [Google Scholar]
  • 110.Soderberg C, Sumitran-Karuppan S, Ljungman P, and Moller E. 1996. CD13-specific autoimmunity in cytomegalovirus-infected immunocompromised patients. Transplantation. 61: 594–600. [DOI] [PubMed] [Google Scholar]
  • 111.Soderberg C, Larsson S, Rozell B, Sumitran-Karuppan S, Ljungman P, and Moller E. 1996. Cytomegalovirus-induced CD13-specific autoimmunity--a possible cause of chronic graft-vs-host disease. Transplantation. 61: 600–609. [DOI] [PubMed] [Google Scholar]
  • 112.Kasman L 2005. CD13/aminopeptidase N and murine cytomegalovirus infection. Virology. 334: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Rahbar A, Bostrom L, and Soderberg-Naucler C. 2006. Detection of cytotoxic CD13-specific autoantibodies in sera from patients with ulcerative colitis and Crohn’s disease. J. Autoimmun. 26: 155–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Glyn-Jones S, Palmer A, Agricola R, Price A, Vincent T, Weinans H, and Carr A. 2015. Osteoarthritis. Lancet. 386: 376–387. [DOI] [PubMed] [Google Scholar]
  • 115.Dan H, Tani K, Hase K, Shimizu T, Tamiya H, Biraa Y, Huang L, Yanagawa H, and Sone S. 2003. CD13/aminopeptidase N in collagen vascular diseases. Rheumatol. Int 23: 271–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Thielitz A, Reinhold D, Vetter R, Bank U, Helmuth M, Hartig R, Wrenger S, and Wiswedel I. 2007. Inhibitors of dipeptidyl peptidase IV and aminopeptidase N target major pathogenetic steps in acne initiation. J. Invest. Dermatol 127: 1042–1051. [DOI] [PubMed] [Google Scholar]
  • 117.Ekşioglu-Demiralp E, Kibaroglu A, Direskeneli H, Yavuz S, Karsli F, Yurdakul S, Yazici H, and Akoglu T. 1999. Phenotypic characteristics of B cells in Behçet’s disease: increased activity in B cell subsets. J. Rheumatol 26: 826–832. [PubMed] [Google Scholar]
  • 118.Vilchis-Ordoñez A, Contreras-Quiroz A, Vadillo E, Dorantes-Acosta E, Reyes-López A, Quintela-Nuñez del Prado H, and Venegas-Vázquez J. 2015. Bone Marrow Cells in Acute Lymphoblastic Leukemia Create a Proinflammatory Microenvironment Influencing Normal Hematopoietic Differentiation Fates. Biomed Res Int. 2015: 386165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Matsumoto H, Yamakawa K, Ogura H, Koh T, Matsumoto N, and Shimazu T. 2017. Clinical Significance of Tissue Factor and CD13 Double-Positive Microparticles in Sirs Patients with Trauma and Severe Sepsis. Shock. 47: 409–415. [DOI] [PubMed] [Google Scholar]
  • 120.Tsokos G 2011. Systemic lupus erythematosus. N. Engl. J. Med 365: 2110–2121. [DOI] [PubMed] [Google Scholar]
  • 121.Mitic B, Lazarevic G, Vlahovic P, Rajic M, and Stefanovic V. 2008. Diagnostic value of the aminopeptidase N, N-acetyl-beta-D-glucosaminidase and dipeptidylpeptidase IV in evaluating tubular dysfunction in patients with glomerulopathies. Ren. Fail 30: 896–903. [DOI] [PubMed] [Google Scholar]
  • 122.Behzadi M, Ahmadzadeh A, Valizadeh M, Haji Molla Hoseini M, and Yeganeh F. 2017. CD13/aminopeptidase N mRNA expression and enzyme activity in Systemic Lupus Erythematosus. Acta Reumatol Port. 42: 162–167. [PubMed] [Google Scholar]
  • 123.Vijgen L, Keyaerts E, Zlateva K, and Van Ranst M. 2004. Identification of six new polymorphisms in the human coronavirus 229E receptor gene (aminopeptidase N/CD13). Int. J. Infect. Dis 8: 217–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Alfalah M, Krahn MP, Wetzel G, von Hörsten S, Wolke C, Hooper N, Kalinski T, Krueger S, Naim HY, and Lendeckel U. 2006. A mutation in aminopeptidase N (CD13) isolated from a patient suffering from leukemia leads to an arrest in the endoplasmic reticulum. J. Biol. Chem 281: 11894–11900. [DOI] [PubMed] [Google Scholar]
  • 125.Dybkaer K, Kristensen JS, and Pedersen FS. 2001. Single site polymorphisms and alternative splicing of the human CD13 gene--different splicing frequencies among patients with acute myeloid leukaemia and healthy individuals. Br. J. Haematol 112: 691–696. [DOI] [PubMed] [Google Scholar]
  • 126.Adamia S, Haibe-Kains B, Pilarski PM, Bar-Natan M, Pevzner S, Avet-Loiseau H, Lode L, Verselis S, and Fox EA. 2014. A genome-wide aberrant RNA splicing in patients with acute myeloid leukemia identifies novel potential disease markers and therapeutic targets. Clin. Cancer Res 20: 1135–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Bauvois B, and Dauzonne D. 2006. Aminopeptidase-N/CD13 (EC 3.4.11.2) inhibitors: chemistry, biological evaluations, and therapeutic prospects. Med. Res. Rev 26: 88–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Su L, Fang H, and Xu W. 2011. Aminopeptidase N (EC 3.4.11.2) inhibitors (2006 – 2010): a patent review. Expert Opin. Ther. Pat 21: 1241–1265. [DOI] [PubMed] [Google Scholar]
  • 129.Wickström M, Larsson R, Nygren P, and Gullbo J. 2011. Aminopeptidase N (CD13) as a target for cancer chemotherapy. Cancer Sci. 102: 501–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Gao JJ, Gao ZH, Zhao CR, Yuan Y, Cui SX, Zhang XF, Cheng YN, Xu WF, Tang W, and Qu XJ. 2011. LYP, a novel bestatin derivative, inhibits cell growth and suppresses APN/CD13 activity in human ovarian carcinoma cells more potently than bestatin. Invest. New Drugs 29: 574–582. [DOI] [PubMed] [Google Scholar]
  • 131.Allegra A, Innao V, Russo S, Gerace D, Alonci A, and Musolino C. 2017. Anticancer Activity of Curcumin and Its Analogues: Preclinical and Clinical Studies. Cancer Invest. 35: 1–22. [DOI] [PubMed] [Google Scholar]

RESOURCES