Startseite Trefoil factor family peptides – friends or foes?
Artikel Open Access

Trefoil factor family peptides – friends or foes?

  • Maike Busch und Nicole Dünker EMAIL logo
Veröffentlicht/Copyright: 17. Oktober 2015

Abstract

Trefoil factor family (TFF) peptides are a group of molecules bearing a characteristic three-loop trefoil domain. They are mainly secreted in mucous epithelia together with mucins but are also synthesized in the nervous system. For many years, TFF peptides were only known for their wound healing and protective function, e.g. in epithelial protection and restitution. However, experimental evidence has emerged supporting a pivotal role of TFF peptides in oncogenic transformation, tumorigenesis and metastasis. Deregulated expression of TFF peptides at the gene and protein level is obviously implicated in numerous cancers, and opposing functions as oncogenes and tumor suppressors have been described. With regard to the regulation of TFF expression, epigenetic mechanisms as well as the involvement of various miRNAs are new, promising aspects in the field of cancer research. This review will summarize current knowledge about the expression and regulation of TFF peptides and the involvement of TFF peptides in tumor biology and cancerogenesis.

Introduction

Trefoil factor family (TFF) peptides consist of three members of small proteins (TFF1, TFF2 and TFF3 (1), formerly breast cancer-associated peptide 2, spasmolytic peptide and intestinal trefoil factor, respectively), first described about 35 years ago (2). All three human TFF genes are clustered on chromosome 21q22.3 (3, 4). They are characterized by a P-domain or trefoil motif, a three-looped or three-leaved structure resembling a trefoil or clover leaf, held together by disulfide bonds (2, 4–6). Each trefoil domain comprises 42 or 43 amino acids containing six cysteine residues, which form disulfide bonds resulting in the characteristic trefoil structure. One such motif is found in TFF1 and TFF3, whereas TFF2 possesses two TFF domains (7, 8). TFF1 and TFF3 contain a seventh cysteine residue that facilitates homodimerization and interaction with other proteins (7). For details and diagrams depicting TFF peptides’ gene and protein structure, the readers are referred to the following reviews (5, 7, 9, 10).

TFFs have been reported to play a key role in the maintenance and protection of epithelial surface integrity. Being secreted in response to injuries, they act as motogens to facilitate cell migration into the lesion, forming a protective barrier and thus being crucial for epithelial restitution, particularly of mucosal surfaces (11, 12). TFFs have been described as potent inhibitors of apoptosis and anoikis (cell death induced by anchorage independence) (8). The signaling pathways that mediate the effects of TFFs have not been fully elucidated yet, and no definite TFF peptide receptor has been characterized so far.

Studies of the last decades, however, indicated that TFFs seem to be involved in more processes than just epithelial restitution, e.g. the development of human cancer. Depending on the context, in the current literature TFFs are presented as oncogenes but also as tumor suppressors [for review, see (12, 13)].

The aim of the present review is to present an update of what is currently known about TFF peptide expression under pathological conditions as compared to their expression in normal, healthy tissues with a special focus on the nervous system including the eye and retina. Besides, TFF signaling and the regulation of TFF expression will be outlined, highlighting new insides in epigenetic mechanisms and effects of miRNAs. Last but not least, the implications of TFF knockdown and overexpression in vitro and in vivo will be addressed with regard to their opposing functions as oncogenes and tumor suppressors.

TFF expression in normal tissues

TFF expression in mucous epithelia

TFF peptides are co-secreted with mucins, typical constituents of mucus gels, e.g. in the gastrointestinal (GI) and respiratory tracts and the uterus [for review, see (14, 15)]. Among others, TFF expression has likewise been observed in human salivary glands (16, 17), the pancreas of rodents and man (17, 18), the prostate (17), the female reproductive organs (17), the urogenital system (17), the conjunctiva [reviewed in (15)] and the lacrimal apparatus (19). In the human respiratory tract and uterus, studies by Wiede et al. (20) revealed an accumulation of TFF3 mRNA, whereas TFF1 and TFF2 transcripts were hardly detectable.

The expression of TFF peptides in mucous epithelia has already been addressed in-depth in former reviews, and for more details on TFF expression in these tissues the reader is referred to the following review articles (9, 12, 17, 21, 22).

TFF peptides in nervous tissue

Next to their prominent expression in – mostly mucous – epithelia, TFF peptides are also synthesized in the central nervous system (CNS) (14, 15, 23–26).

In the brain of rats, Tff1 mRNA is predominantly expressed in the hippocampus, followed by cerebral cortex and cerebellum (24), and rat intestinal trefoil factor (rITF) was detected in neurons of the rat hypothalamus (25). A recent study showed the expression of Tff1 in the developing and adult rat ventral mesencephalon (27). Formerly, astrocytes but not neurons have been reported to synthesize Tff1 in mice and rats (24, 28). By contrast, a more recent study on a rat model of Parkinson disease reports on the expression of Tff1 in distinct subpopulations of dopaminergic neurons of the substantia nigra (27).

The murine pituitary is a major expression site for Tff2, with high Tff2 mRNA transcript levels in the anterior lobe (23).

Numerous studies detected TFF3 mRNA and/or protein in different areas of the developing and adult murine and human brain and spinal cord (23, 25, 29, 30). Hinz et al. (23) found murine Tff3 expression to be restricted to three brain regions: the hippocampus, the temporal cortex and the cerebellum – the latter showing the strongest signal. A study investigating the expression of TFF peptides in the nervous tissue of developing mouse embryos demonstrated Tff3 expression in ganglion cell somata and neurons of the spinal cord and Tff3 staining in neurons and nuclei of different regions of the brain and medulla oblongata (26). In humans, synthesis of TFF3 has been verified for neurons of the hypothalamic nuclei and the pituitary (25, 29, 31) as well as for TFF3 in the cerebrospinal fluid (31, 32). In a most recent study on the expression of TFF3 in the adult and developing human brain, Bernstein et al. (33) revealed that this peptide is particularly enriched in midbrain and brain stem nuclei.

Initially, TFF3 expression appeared to be restricted mainly to neurons and not glial cells, and TFF3 was described as a typical neuropeptide synthesized by magnocellular neurons of the rat and human hypothalamus (25, 29, 31). In a recent study, Tff3 transcripts have, however, been detected in activated microglia of glial cell-enriched embryonic and neonatal cultures of the rat cerebral cortex and hippocampus (34). Along this line, TFF3 expression was likewise observed in human oligodendroglia neurons, though neurons are the predominant cell type to express TFF3 (33).

A developmental regulated expression was reported for Tff1 in the rat hippocampus, where Tff1 mRNA gradually decreased in the first 3 weeks after birth (24). Tff3 expression is likewise clearly developmentally regulated with a maximum expression at postnatal day (P)15 (23). This led to the hypothesis that TFFs play an important role during brain development (24, 26). Besides, cerebral TFF3 has been reported to be involved in processes such as fear, depression, learning, object recognition and opiate addiction (35–38). Furthermore, mutations of the TFF3 gene or altered expression have been linked to neurodegenerative and neuropsychiatric disorders like Alzheimer’s disease (32), Parkinson’s disease (39) and schizophrenia (40).

TFF peptides in ocular tissues and the retina

Messenger RNA and/or protein expression of TFF1 and TFF3, but not TFF2, have been detected in human and porcine conjunctival goblet cells (41–43), human cornea (44, 45), rabbit corneal and conjunctival tissue (46) and the epithelium of the nasolacrimal ducts (47). TFF3 has not only been shown to promote corneal wound healing (48) but also to be a promising therapeutic candidate for patients with dry eye syndrome (45).

TFF expression was monitored in the CNS and ocular tissues, but our group was the first to investigate retinal expression of TFF peptides and to report on Tff expression in the murine retina (49). Interestingly, Tff2 turned out to be the only Tff peptide expressed in the murine retina as in most tissues studied TFF1 and TFF3 are the predominant peptides. Recent studies by our group revealed that only TFF3, but not TFF1 and TFF2, is expressed in the healthy human retina (50, 51).

Pathological expression of TFF peptides

Role of TFFs in epithelial restitution and ulcers

Analyzing animal models of wound healing, it has been observed that in the restitution step after damage, TFFs stimulate migration of epithelial cells surrounding the wound to re-establish the continuity of the epithelium [for review, see (9, 11, 12)]. Impaired healing and its association with chronic inflammation-related injury, e.g. inflammatory bowel disease (IBD) and ulceration, is a key event in tumorigenesis and one of the key areas of TFFs’ functions, being involved in mucosal defense and regeneration or reconstitution, respectively (21, 52–57). The disturbance of TFFs’ function in mucosal healing aggravating the risk of tumorigenesis under chronic inflammatory conditions is one model for the involvement of TFFs in cancer (21). TFF-peptide expression is upregulated in GI ulcerations and IBDs (e.g. ulcerative colitis) (58). Besides, expression and upregulation of TFF1 and TFF2 mRNA and peptides has been observed in epithelial cells adjacent to ulcerative conditions of the GI tract and epithelial cells undergoing migration across the base of the lesion, termed the ulceration-associated cell lineage (UACL), and TFFs have been shown to be expressed in small intestinal Crohn’s disease (52, 59–62). Along this line, Tff2 mRNA levels increase within minutes following gastric ulceration of rats (62), and Playford et al. (63) described TFF2 as a cytoprotective agent.

For a comprehensive overview of TFFs’ role in epithelial restitution and their effects in animal models of GI ulceration, see the following reviews (10, 11, 14).

Role of TFFs in tumor progression, suppression and prognosis

TFF peptides are overexpressed in cancer progression (64). Experimental and clinical studies indicate a role of TFFs in oncogenic transformation, tumor growth and tumor metastasis of common human solid tumors [for review, see (10, 13, 21)]. TFFs are connected with multiple oncogenic pathways (65) and, depending on the context, are considered as tumor suppressor genes or potential tumor progression factors (13).

TFF1 and TFF3 but not TFF2 mRNAs are expressed in hyperplastic and neoplastic human breast epithelium as well as in MCF-7 breast carcinoma cells (66). Breast cancer is indeed a typical example of cancers overexpressing TFF1. This TFF peptide was first controversially considered as an oncogene in this carcinoma (13). Buache and colleagues (67), however, demonstrated that TFF1 is not an oncogene in the mammary epithelium but rather reduces the development of breast tumors and has a tumor suppressor function. The beneficial function of TFF1 is in agreement with clinical studies indicating a better outcome for breast cancer patients with TFF1-positive primary tumors (9, 68, 69). Both TFF1 and TFF3 mRNAs have been identified as predictive markers for micrometastatic breast cancer (70, 71). Ectopic expression of TFF1 is observed in numerous other carcinomas and GI acute inflammatory disorders. By contrast, tumors of patients with gastric cancer usually have reduced TFF1 levels, and disruption of the TFF1 gene causes animals to develop gastric carcinomas and adenomas. TFF1 is significantly increased in gastric cancer cells with greater metastatic potential compared to their less metastatic counterparts (72). Progressive loss of TFF1 and downregulation of TFF2, together with the induction of TFF3, has been suggested to be involved in multi-step gastric cancer development (73, 74).

TFF2 is upregulated in diverse pathological conditions of the GI tract. To our knowledge, no genetic or epigenetic alterations are currently known to back up a tumor supressor role for TFF2 (73).

TFF3 is upregulated by inflammatory and ulcerative conditions. Augmented TFF3 and loss of TFF1 expression was reported to precede metaplastic differentiation of gastric epithelia (75). TFF3 is highly expressed in intestinal metaplasia and is a designated marker for poor prognosis in gastric carcinoma (76, 77). TFF3 overexpression is, however, not only frequently observed in human gastric cancers but also in colon, pancreatic, breast and hepatocellular carcinomas (66, 74, 78–82) and, thus, was thought to induce cancer growth. Most recently, Morito et al. (83) reported on the value of TFF3 expression in predicting the long-term outcome and recurrence of colorectal cancer. Along this line, TFF3 expression correlates with the tumor grade in hepatocellular carcinoma (84). Moreover, TFF3 is overexpressed in prostatic carcinoma, and increased plasma levels in patients with advanced prostate cancer have been described, suggesting a predictive importance of TFF3 also in prostate cancer (64, 85).

For a more comprehensive overview, readers are referred to review articles (10, 13, 21).

TFFs in retinal and corneal diseases

Studies from our lab show that compared to the human retina, TFF1 is upregulated in retinoblastoma (Rb) cell lines established from malignant eye tumors of children (50, 51). By contrast, no TFF2 and only trace amounts of TFF3 mRNA are detectable in Rb cell lines (50). As mentioned above, virtually no expression of TFF1 is observed in the healthy human retina, whereas in all Rb cell lines TFF1 is the only TFF peptide expressed at well detectable levels.

In line with our results, it has been reported that all three TFF peptides are absent in healthy corneas, while by yet unknown stimuli the secretion of one TFF peptide, in this case TFF3, is induced in different corneal diseases, e.g. in corneas of patients suffering from keratoconus, herpetic keratitis, Fuchs’ dystrophy and pterygium (44), probably as a protection mechanism. Further along this line, Schulze et al. (86) reported that tears of patients suffering from dry eye disease contain significantly higher TFF3 levels than those from healthy volunteers. The authors demonstrated that in human corneal epithelial cells TFF3 is upregulated under experimental conditions similar to dry eye disease.

Regulation of TFF expression and signaling mechanisms conferring TFFs’ effects

Several mechanisms seem to be involved in the regulation of the TFF expression and their downstream effects on normal epithelial and cancer tissues in vivo. The following paragraphs will summarize the current knowledge about TFF-related signaling cascades with regard to (i) the regulation of TFFs by proteins and chemicals, (ii) the regulation of TFFs by receptors and pathways, (iii) mechanisms conferring TFF’s effects on cytoprotection and motility, (iv) epigenetic regulation of TFF expression and (v) effects of miRNAs.

Regulation of TFFs by proteins and chemicals

In normal tissues, TFFs are expressed in a strictly tissue-specific manner, whereby the different tissues seem to possess individual regulation mechanisms (87, 88). For example, the TFF3 promotor contains cis-regulatory enhancer and silencer regions, and nuclear proteins binding to these regions are exclusively found in intestinal goblet cells (89).

The adjacent localization of the three TFF genes on chromosome 21q23 and shared 5′ regulatory sequences lead to the assumption that their expression is coordinated (90). This assumption was supported by the finding that TFFs are auto- and cross-inducible, e.g. TFF2 and TFF3 enhance the expression of all TFFs in intestinal and gastric cell lines by binding cis-regulatory elements of their promotors in a mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK)-dependent fashion (91). Besides, in Tff3 knockout (KO) mice a decreased expression of Tff1 and Tff2 was found (91), and in Tff1 KO mice Tff2 levels are decreased (92). The cross-induction of TFFs requires activation via phosphorylation of the epidermal growth factor receptor (EGF-R), the latter being activated by all three TFF peptides (93).

TFF1 expression was shown to be regulated by estrogen in the cancer cell line MCF-7 as well as in primary breast cancers (87, 94–96). Analyses of the TFF1 promotor revealed an estrogen-response element (ERE) as well as enhancer sites responsive for epidermal growth factor (EGF) (97). In breast cancer cells, estrogen effects are mediated mainly by the estrogen receptor (ER)α but also by ERβ, which in turn binds to the ERE in the TFF1 promotor region. The binding of ERβ is enhanced in the presence of Sp1 and Sp3 binding sites (98) as well as other transcription factors like GATA3, HNF3 and XBP1 (99). A more recent study describes new mechanisms by which ERα and the insulin-like growth factor I receptor are related in breast cancer, contributing to tumor progression and resistance to anticancer treatments (100). In ER-positive breast cancer cells, Akt2 is activated via the downstream phosphatidylinositol 3-kinase (PI3-K)/Akt pathway and modulates ER transcriptional activity in a ligand-independent manner. This activation leads to the expression of the forkhead transcription factor FoxO3a, which in turn binds to forkhead-responsive sequences in the TFF1 promotor. FoxO3a itself plays a repressive role in ER activation and TFF1 expression (100). Also, other constitutively expressed estrogen-receptor related receptors of the orphan nuclear receptor family can regulate TFF1 expression in an estrogen-independent fashion (101). The regulation of the TFF1 expression is estrogen-independent in gastric mucosa, where TFF1 is highly expressed, although ERα and ERβ are present (102, 103).

Like TFF1, TFF3 has two ERE located in the promotor region, and in breast cancer cell lines it is expressed in an estrogen-dependent way (78). Additionally, TFF3 is highly expressed during the estrogen-driven period of the menstrual cycle in the epithelium of the human uterus (20, 104). During the phase of implantation, TFF3 is strongly downregulated in the human endometrium, probably leading to a reduced barrier function of the endometrium epithelium (88, 105). A recent study describes the influence of TFF3 expression and its interaction with ER in endometrial adenocarcinoma (106). The authors found TFF3 to be upregulated in endometrial adenocarcinoma in an estrogen-dependent manner correlating with a good prognosis in type I endometrial carcinomas (106).

Next to the ERE also a gastrin responsive element has been found in the human TFF1 promotor, and it could be shown that gastrin can induce human TFF1 and murine Tff1 expression in an ERK-dependent manner (107). Mice that cannot signal through SHP2/ras/ERK display reduced gastrin and Tff1 expression in gastric tumor tissue (108). In contrast to these findings, van de Bovenkamp and colleagues (109) reported that elevated gastrin levels occur along with reduced TFF1 expression in human Heliobacter pylori-mediated gastritis.

TFFs are aberrantly secreted during several inflammatory diseases. It has been shown that the secretion of EGF/urogaston by the so-called UACL, a type of cells found in the area of chronic GI ulceration, induces the expression of TFF1 and TFF2 in damaged GI tissues (59, 110). In a human bronchial epithelial cell line (BEAS-2B), TFF2 and TFF3 increase the tumor necrosis factor alpha (TNF-α)-induced secretion of interleukin (IL)-6 and IL-8 via signaling through protein kinase C (PKC) and ERK1/2 (111). It could also been shown that IL-1β and IL-6 themselves are regulators of TFF gene expression. They are able to decrease TFF1 promotor activity and gene expression via inhibition of NF-κB (nuclear factor ‘kappa-light-chain-enhancer’ of activated B cells) and C/EBPβ factors in GI cell lines (112). Soutto and colleagues (113) propose that loss of TFF1 in gastric cancer leads to an activation of the TNF receptor 1/IκB kinase pathway mediated by NF-κB transcription factors. In line with this hypothesis, TFF1 expression is downregulated and NF-κB was highly activated in human gastric tissue samples. Consistent with these data, a recent study demonstrates that IL-1β and TNF-α activate the NF-κB pathway resulting in decreased expression of TFF1 in human gastric carcinogenesis (114). By contrast, in vivo data showed that murine Tff1 gene expression is upregulated by IL-6, and this upregulation involves the activation of the SHP2/ERK/AP-1 pathway and signaling through the receptor gp130 (115).

In the pathogenesis of IBD, NF-κB is activated, and acting as a pro-inflammatory factor may contribute to the development of ulcerations, while both the expression of NF-κB and the toll-like receptor 4 (TLR4) is essential for expression of cytokines in intestinal epithelial cells. It could be shown that the expression of both factors induces downregulation of TFF3 by repressing its transcription in vitro (116). Conversely, the application of recombinant human TFF3 (rTFF3) leads to a downregulation of NF-κB/TLR4 expression, revealing that human TFF3 may have therapeutic potential by inhibiting TLR4/NF-κB pathways (117). In goblet cells, toll-like receptor 2 (TLR2) activation induces the expression of TFF3, while the loss of TFF3 induction leads to impaired wound healing. In addition, treatment with rTFF3 rescues TLR2-deficient mice from raised morbidity and mortality during acute colonic injury (118). In colon cancer cell lines, TFF3 expression is enhanced by IL-4 and IL-13 in a STAT6-dependent manner along with mucin core protein (MUC) 2, potentially directly mediated by the STAT6 binding site (119).

Regulation of TFFs by receptors and signaling pathways

The different biological functions of TFFs are expected to be mediated by cell surface receptor ligation. However, up to now no classical high-affinity binding receptor for TFF peptides has been found.

In 2009, the chemokine receptor CXCR4 has been described as a low-affinity receptor for TFF2 (120), and treatment of a CXCR4 expressing gastric cell line (AGS) with TFF2 leads to a distinct proliferative effect [for review, see (121)]. A more recent study suggests that TFF2 is involved in pancreatic β-cell proliferation through CXCR4-mediated ERK1/2 phosphorylation (122). Besides, several TFF binding proteins have been identified from membrane preparations of intestinal cells of different species: in the porcine gastric mucosa CRP-ductin was reported as a Tff2 binding protein (123), in mouse Tomasetto and co-workers (124) proposed binding of Tff1 to MUC2 and MUC5AC, and in the human gastric mucosa GKN2 is a putative candidate for a TFF1 receptor [for review, see (10)].

Several pathways are involved in TFF signaling, and they often play a major role in cancerous progression in human digestive mucosa and other organ types. Pathways that are correlated to biological actions of TFFs include the PI3-K/Akt pathway, the Rho-ROCK cascade, COX-2/TXA2-R/Gαq signaling, PLC/PKC, MAPK and EGF-R signaling (125). Additionally, many intermediates like src, Rho-like small GTPases, PI3Ks, COX-2, ERK1/2, JNK, Akt, NF-κB and EGF-R tyrosine kinase are included in these pathways (111, 125–128). It has long been suggested that TFF expression may be directly regulated via the EGF-R. TFFs, however, do not bind directly to the receptor but trigger its phosphorylation and downstream actions like initiation of the MAPK and PI3K pathways. In many normal and cancer tissues, various biological functions like growth and differentiation, angiogenesis, invasion and apoptosis are mediated via EGF-R signaling [for review, see (88)]. Along this line, in a cholangiocarcinoma cell line, recombinant TFF2 triggered phosphorylation of the EGF-R and downstream ERK, whereas a co-incubation with an EGF-R inhibitor blocked the EGF-R/ERK responses (129).

In cancer, the tumor suppressor function of TFF1 is triggered by different pathways. In human colon cancer cell lines, TFF1 (and TFF3) exerts growth-reducing effects through the activation of the MAPK/ERK pathway (80). In the colorectal cancer cell line, HCT8/S11 invasion is initiated by TFF1 in a COX-2- and TXA2-R-dependent way (127). A more recent study proposed that in gastric neoplastic transformation the downregulation of TFF1 expression is at least partially regulated through the activation of the NF-κB pathway via IL-1β and TNF-α activation (114).

TFF2-related effects on cell migration are dependent on the expression of E-cadherin and β-catenin (130), and EGF has been shown to enhance TFF2’s motogenic effects (131). In BEAS-2B cells, TFF2 causes motogenic effects through the activation of ERK1/2 and PKC (132) and the activation of Scr family tyrosine kinases (111). This pathway is PI3K/Akt-dependent with the participation of β-catenin/α-catenin complexes [for review, see (88)]. In gastric cells, TFF3 regulates migration in a Twist-dependent manner (133). In SGC7901 cells, the Twist pathway is activated by TFF3 with a coupled regulation of the migration markers CK-8 as well as ZO-1 and matrix metallopeptidase-9. Conversely, a siRNA-mediated knockdown of Twist prevents TFF3-induced cell migration in those cells.

TFF3 and the tumor angiogenesis regulator vascular endothelial growth factor induce cellular invasion and reduce growth in HCT8/S11 tumor xenografts in athymic mice through phosphorylation of STAT3 (134). In a recent study, the forced expression of TFF3 in a mammary carcinoma cell line was correlated with an increased STAT3 activity induced by the phosphorylation of c-Scr, which subsequently leads to a reduced expression of E-cadherin (135). Therefore, the authors concluded that TFF3 expression in mammary carcinomas stimulate cell invasion and metastasis with a poor survival outcome of patients (135).

Mechanisms conferring TFFs’ effects on cytoprotection and motility

There are various in vivo studies in rodents demonstrating the cytoprotective effect of all three TFFs after GI damage [for review, see (10, 15)]. The beneficial effects of TFFs have been correlated with a reduction in the vascular cell adhesion protein, IL-6, and TNF-α expression. Besides, a synergistic protective effect with EGF has been proposed [for review, see (12)].

It has been shown that the cytoprotective, anti-apoptotic effect of TFF1 is mediated via a decrease of caspase-3, -6, -8 and -9 activities (136, 137).

TFF3-induced resistance to anoikis in intestinal epithelial cells is linked to a signaling cascade involving NF-κB (138). Besides, TFF3 promotes mucosal cell integrity by activation of the PI3K/Akt signaling pathway (139). The protective effect of TFF3 on HCT116 and IEC-6 cells likewise seems to involve PI3K activation and Akt phosphorylation as well as activation of the EGF-R (65, 140). TFF3 has also been shown to activate the Akt protein kinase B survival pathway (8). Moreover, p-53-induced cell death in human gastric carcinoma cell lines after etoposide treatment is inhibited by TFF3 (8). However, no differences in gene expression of any proteins related to Fas or TNF receptor-mediated apoptotic pathways or apoptosis-associated proteins of the Bcl family like Bcl-2, Bax, Bad or Bcl-xL could be detected in Tff3-deficient mice (140).

To execute their role in wound healing and epithelial repair, TFF peptides stimulate cell survival, protect cells from apoptosis and, last but not least, increase cell motility (8, 21, 65). Migration is essential in the progression of cancer, e.g. tumor spreading, and motility is also essential for epithelial restitution. After superficial injury, migration of epithelial cells is observed, particularly in the GI mucosa (12), and TFFs have been shown to be essential for this rapid repair called ‘restitution’ (9, 11, 12). The first step in restitution is the reduction of cell-cell contacts. All TFF peptides have been shown to induce downregulation of E-cadherin/β-catenin complexes in adherent junctions, accounting for their pro-migratory effect (93, 141, 142).

For TFF1, Piezo 1 has been suggested as a new binding protein promoting gastric cancer cell motility (143).

TFF2 and TFF3 are motogens in wounded cell monolayer assays in a transforming growth factor beta (TGF-β)-independent manner (53). TGFα/EGF motogen factors seem to participate in wound healing in a TGF-β-dependent manner on the basolateral side of the wounded epithelium, whereas TFFs signal through a TGF-β-independent pathway at the apical side (53, 63, 144).

TFF3-mediated motogenic activity during restitution has been shown to be regulated through E-cadherin (142), independent of EGF-R activation, but via the MAPK pathway (126, 129) and MEK/ERK inhibition. In this context, it has been demonstrated that TFF3 modulates adherens and tight junctions by increasing the level of tightening claudin-1 and decreasing the amount of claudin-2, known to form cation-selective channels (145). Besides, TFF3 binds to the intestinal mucosa and increases NO production via type II or iNOS/NOS2 (146), and NO has been shown to mediate epithelial cell restitution during wound healing. Moreover, TFF3 activates the PI3K/Akt pathway to protect gastric mucosal epithelial cells from lipopolysaccharide-induced damage (147).

Epigenetic regulation of TFF expression

Epigenetic alterations, resulting in site-specific DNA methylation and histone deacetylation, are well-known mechanisms associated with transcriptional silencing of cancer-related genes (148–150). DNA methylation within the genome of vertebrates occurs at cytosines located 5′ to a guanosine called CpG dinucleotide. Short regions, known as CpG islands, are rich in CpG content and frequently found in the proximal promoter region of many human genes [for review, see (148, 151)].

Epigenetic mechanisms are also involved in the regulation of TFF expression in cancer (65, 88). The promotor regions of the TFFs are methylated or only partially demethylated in tissues where these genes are expressed. By contrast, in organs without TFF gene expression their promotors are methylated (152). In gut and pancreas (152) as well as human pancreatic ductal carcinomas (153), hepatocellular carcinoma (81), and mouse small intestine (152), TFF3 is strongly expressed with concomitant hypomethylation of its promotor region. A most recent study demonstrated that the methylation status of the CpG islands in the promoter region correlates with TFF1 expression levels in human gastric cancer cells, and DNA methylation is a key mechanism of silencing TFF1 in gastric carcinomas (154). Moreover, TFF1 has been described as a differentially methylated gene in Rb tumors (155) and as one of the upregulated genes in primary Rbs with a matching activating histone modification (150). In a recent study by our group, we could demonstrate that minimal methylation changes of certain CpGs in the TFF promoter of Rb cell lines results in changes of TFF expression levels (50). Our data are in good accordance with the finding that as little as 6–8% methylation can account for 67–90% downregulation of genes (156). Along this line, in human breast cancer cell lines, a correlation of DNA methylation and TFF1 expression was not observed at all CpG sites, since some CpGs were unmethylated in TFF1 non-expressing cell lines (157).

In prostate cancer cell lines, promoter hypomethylation of TFF1 and TFF3 has been shown to be closely related to increased expression of these genes (158). Additionally, it could be shown that the DNA methyltransferase inhibitor 5-Aza-2′deoxycytidine (5-Aza-dC) increases TFF expression in low-expressing prostate cancer cells and restores TFF1 expression in gastric carcinoma cell lines (154). In a study by our group, we observed a significant induction of TFF3 expression upon stimulation with 5-Aza-dC in all Rb cell lines exhibiting no or low endogenous TFF3 expression. TFF1 expression was, however, only slightly increased by 5-Aza-dC, suggesting a correlation of the extent of upregulation with endogenous basal expression level. No re-expression of TFF2 was observed in Rb cell lines, in which this gene seems to be silenced (50). A study from Sato and co-workers (153) reported that there is no clear correlation between TFF2 promoter methylation status and TFF2 expression, although hypomethylation of the TFF2 gene was observed in TFF2-overexpressing pancreatic ductal adenocarcinoma.

Histone modification is another epigenetic factor playing a key role in transcriptional regulation of gene expression. TFF1 is upregulated in Rbs with a matching activating histone modification, indicating an epigenetic regulation (150). A synergistic effect of DNA demethylation and the inhibition of histone deacetylation in the re-expression of silenced genes has been described (159). These findings match with data from our recent study showing that double treatment with 5-Aza-dC and the histone deacetylase inhibitor 4-phenylbutyric acid increases TFF mRNA levels in Rb cell lines (50).

Effects of miRNAs

MicroRNAs (miRNAs) are small non-coding RNA molecules (about 22 nucleotide in length) that play important roles in many pathways like differentiation, cell cycle progression, growth and apoptosis. The dysregulation of miRNAs may turn out to be crucial for various types of cancers and diseases. Lee and colleagues (160) published the first report on the function of miRNAs in 1993. Up to now little is known about the link between TFFs and miRNAs. There are a few studies reporting on a regulation of TFF1 in gastric cancer by miRNAs. MicroRNA-423-5p negatively regulates the expression of TFF1 by binding to its 3′UTR region and consequently influences proliferation and invasion-related processes in gastric cancer cells (161). Shi et al. (162) could show that the TFF1 coding DNA region is also a candidate for miR218-5p targeting and that TFF1 is downregulated by miR218-5p in gastric cancer cells. The reduced TFF1 expression regulates the progression of gastric cancer in an ERK1/2-dependent way. By in vitro and in vivo approaches, a recent study by Soutto and co-workers (163) demonstrated that in human gastric cancer, activation of p53 mediates TFF1 effects via downregulation of miR-504.

By whole miRNome profiling and in silico analysis of Tff2 KO mice vs. wild-type mice, Shah and colleagues (164) identified physiological regulated miRNAs. From this proof-of-concept study they suggested that the identified miRNAs may play a major role in regulatory processes of TFFs, particularly regulation of cancer cascades. In a more recent study of this group, the authors compared deregulated miRNAs in blood samples from human cancer patients with the expression pattern of a gastric carcinoma Tff1 KO mouse model. By subsequent in silico analysis of the identified subset of miRNAs, their involvement in targeting neoplastic and MAP-kinase pathways was demonstrated (165). Applying bioinformatic analyses on miRNA expression data of previous Tff2 KO studies, Yin and co-workers (166) set out to unravel pathways of genes involved in the TFF regulating mechanisms. Their KEGG pathway enrichment analyses revealed that the TGF-β signaling pathway as well as a cytokine-cytokine receptor interaction is enriched, both mediated via miRNAs (166).

Investigating the development of intestinal metaplasia in stomach cell lineages, a recent study proposed a miRNA to transcription factor network to be responsible for the expression of intestinal transcripts, identifying miR-30 and miR-194 as regulators for the transcription factors HNF4γ (hepatocyte nuclear factor 4 gamma) and NR2F2 (nuclear receptor subfamily 2) (167). The intestinal metaplasia markers TFF2 and TFF3 were also downregulated after overexpression of miR-30a in a HNF4γ-dependent way (167). Additionally, the ectopic expression of the caudal-related homeobox protein 2 (CDX2) is connected with the development of intestinal metaplasia in gastric carcinogenesis. By overexpression and knockdown experiments of the computationally predicted miR-9, Rotkrua and colleagues (168) found altered expression levels of the CDX2 protein and the corresponding downstream target genes including TFF3 in MKN45 and NUGC-3 cells (168).

Effects of TFF knockdown and overexpression in vivo and in vitro

Consequences of TFF knockout and overexpression in vivo

Tff KO mice are susceptible for gastric hyperplasia or dysplasia, ulceration, adenomas and carcinomas [reviewed in (10)].

Tff1-/- (and Tff3-/-) mice partly lack a functional mucus layer (54, 92). Mice deficient for Tff1 show increased gastric mucosal proliferation rates (92) and, compared to controls, display differences in the susceptibility for indomethacin (55). Conversely, Tff1 overexpressing mice show resistance to intestinal damage (55). In line with these findings, a most recent tumor xenograft mouse model of gastric cancer supports the notion of Tff1 as a protective tumor suppressor (169). By contrast, implantation of TFF1 overexpressing pancreatic ductal adenocarcinoma cells into nude mice did not induce primary tumor growth but increased metastasis (170). Along this line, constitutive expression of Tff1 potentiates the growth of colon and kidney tumor xenografts in athymic nude mice (171).

Tff2-/- mice show decreased gastric mucosal thickness and proliferation rates (172). It has been shown that compared to wild-type animals, Tff2-deficient mice display higher rates of non-steroidal anti-inflammatory drug-induced ulcers (172). In addition, analyses of Tff2-deficient mice revealed Tff2’s role in immune response (173, 174). To our knowledge, there are no reports on Tff2 overexpressing mice so far.

In Tff3-/- mice, re-epithelialization of corneal wounds is significantly prolonged compared to Tff3+/+ mice (48). Besides, Tff3-deficient mice display an increase in colonocyte apoptosis (140) and impaired mucosal healing (54). In addition, they are more susceptible to chemotherapy- and radiation-induced mucositis (175) and die from extensive colitis after oral exposure to dextran sulfate (54). Conversely, transgenic mice ectopically expressing rat Tff3 in their jejunum have been shown to be less susceptible to induced enteritis (176).

Implications of TFF overexpression and knockdown on prevention and induction of apoptosis in vitro

Forced expression of TFF1 has been demonstrated to promote both anchorage-independent growth in human colon carcinoma cells and transformation of premalignant colonic adenoma cells (171). In GI cell lines, application of recombinant TFF1 (rTFF1) protects the cells from chemical-, Bad- or anchorage-dependent apoptosis (80, 136). In addition, TFF1 protects gastric cancer cells from apoptosis after treatment with etoposide (177).

Lalani et al. (178) reported that TFF2 promotes the survival of MCF-7 human breast cancer cells via inhibition of apoptosis. Later, it had been reported that TFF2 likewise inhibits apoptosis in other breast cancer and colorectal cancer cell lines (179), and overexpression of TFF2 is associated with resistance to apoptosis (180). By contrast, in organotypic murine retinal cultures, Tff2 exerts a strong pro-apoptotic rather than an anti-apoptotic effect (49). In blockage experiments, our group was able to demonstrate that the pro-apoptotic effect of TFF2 is caspase-dependent (49). Western blot analyses revealed a significant reduction in the phosphorylation level of ERK and STAT3 proteins compared to basal conditions, suggesting that in the developing murine retina survival mechanisms are downregulated upon TFF2 administration (49).

It has been shown that exogenously applied TFF3 protects human colonic carcinoma-derived HCT116 cells and non-transformed rat intestinal epithelial cells from apoptosis (140). Along this line, cell lines overexpressing TFF3 (e.g. colonic HT-ITF1 cells) are resistant to serum starvation- and drug (e.g. ceramide)-induced apoptosis (126, 140). It has likewise been shown that TFF3 prevents IEC-18 cells from anchorage-dependent apoptosis (anoikis) (21, 138). Conversely, anti-sense TFF3-transfected human gastric cancer cells displayed an enhanced chemosensitivity and a marked increase in drug-induced apoptosis (181), and neutralization of secreted TFF3 by antibody promotes apoptosis in mammary carcinoma cells (182). However, evidence in support of the notion of a pro-apoptotic function of TFF3 was reported in articular cartilage during osteoarthritis (183). A pro-apoptotic function of TFF3 has already been suggested years ago: in a colorectal carcinoma cell line, rTFF3 induced DNA fragmentation and morphologic changes characteristic of apoptosis (141). To this end, murine Tff3 has been shown to interact with peptides contributing to apoptosis (184).

Effects of TFF overexpression and knockdown on cell proliferation and growth in vitro

In the human colon cancer cell line HCT116 and the human gastric adenocarcinoma cell line AGS, rTFF1 reduces cell proliferation (185), and transfected, TFF1-expressing HCT116 cells show reduced growth (136). Anti-proliferative effects were likewise documented when rTFF1 was applied or TFF1 was overexpressed in GI cell lines (136). In this context, it has been shown that TFF1 delays G1-S phase transition of the cell cycle (136). In agreement with these findings, a recent study by our group demonstrated that rTFF1 has a negative effect on the viability of Y-79 Rb cells and causes a reduction in cell proliferation (51). Contradicting these data, TFF1 has been shown to stimulate growth of pancreatic stellate cells (170).

Treatment with recombinant TFF2 (rTFF2) reduces cell proliferation in GI and carcinoma cell lines (178). By contrast, forced expression or application of rTFF2 has been shown to promote proliferation of pancreatic β-cells (122). Supporting these findings, our group showed that rTFF2 significantly upregulates cell proliferation in the developing murine retina (49). Along this line, Hoosein et al. (186) reported on a growth stimulatory effect of TFF2 on cultured human colon carcinoma (HCT116) and breast tumor cells (MCF-7) (186). In a later study, however, the authors could not confirm these initial data, reporting that the addition of TFF2 to human colon cancer-derived cell lines (HT-29 and CaCO2) other than HCT116 had no consistent stimulatory or inhibitory effect on cell proliferation (53).

TFF3 has been shown to suppress the growth of colorectal carcinoma cells (79). In the human colon carcinoma cell lines LoVo and SW837, overexpression of TFF3 significantly reduced cell growth (80). In human corneal epithelia cells, cell proliferation likewise decreases 24 h after stimulation with rTFF3 (86). By contrast, Sun et al. (147) demonstrated that TFF3 promotes proliferation of gastric mucosal epithelial cells by the activation of the PI3K/Akt pathway. Along this line, a most recent study likewise reported that rTFF3 enhances the proliferation of gastric endothelial cells (GES-1) through the activation of ERK1/2 (187). Moreover, forced expression of TFF3 in mammary carcinoma and prostate cancer cells significantly increases cell proliferation, viability and survival (182, 188).

Impact of TFF overexpression and knockdown on cell migration, invasiveness and metastasis in vitro

Migration and invasion of cells are crucial processes for epithelial restitution as well as tumor progression and metastasis. All TFF peptides tested so far are motogens enhancing the migration of epithelial cells in different systems (1, 10, 65). TFF2 and TFF3 have also been shown to enhance cell scattering, and all three TFFs induce invasion of transformed kidney and colonic epithelial cells (125).

TFF1 is a potent chemoattractant for human mammary carcinoma cells (65) and stimulates migration, invasiveness and metastasis of human pancreatic stellate (170) and breast cancer cells (67, 189). Besides, TFF1 mutations have been shown to enhance gastric carcinoma cell migration and invasion (177). Recently, Fu et al. (190) showed that compared to stationary rat gastric epithelial cells Tff1 expression is upregulated in migratory gastric epithelial cells (RGM-1), providing further evidence for a Tff1’s motogenic function.

TFF2 appears to stimulate cell migration and invasion as well (125). Overexpression of TFF2 is associated with increased cell migration and possibly increased gastric cancer invasion (180). Besides, a motogenic effect on bronchial epithelial cells (111) as well as the stimulation of migration of HT29 cells (63) have been reported for TFF2.

TFF3 likewise exerts a pro-migratory effect, e.g. on primary rabbit corneal epithelial cells (46, 86), oral keratinocytes (191) and human bronchial epithelial cells (131). Moreover, in wounded monolayers of intestinal epithelial cells (IEC-6), rTFF3 significantly increases the migration of cells into the wound (53). TFF3 has also been shown to promote the migration and invasiveness of rat fibroblasts (192) and the migration of gastric mucosal epithelial cells (139), gastric endothelial cells (GES-1) (187) and human colorectal cancer cells (193). Along this line, transfecting TFF3 into non-aggressive rat colorectal cancer cells has been shown to enhance their ability to migrate, invade and behave more aggressively (194). In line with these findings, siRNA-mediated knockdown of TFF3 in metastatic rat colon cancer cell lines significantly inhibited invasion (195). Further along this line, forced expression of TFF3 in prostate cancer and mammary carcinoma cells enhances anchorage-independent growth and 3-D colony formation and promotes cell migration and invasion (182, 188). Besides, overexpression of TFF3 in mammary carcinoma cells increased tumor size in xenograft models, whereas neutralization of secreted TFF3 in these cells arrested mammary carcinoma xenograft growth (182).

Summary and future challenges

Regarding the numerous functionalities of TFFs summarized in Table 1, these peptides have already been considered as useful targets for pharmacological intervention for several indications, e.g. mucosal or epithelial lesions, not only in the GI tract. Beyond that, changes in TFF expression seem to be a common feature of many types of tumors. In most tumor models studied so far, it has not yet been experimentally proven whether TFFs drive carcinogenesis or represent innocent bystanders. The data outlined above demonstrate that TFFs influence key functional characteristics of oncogenic processes by regulating cell survival, apoptosis, cell migration and invasion. The specific roles of TFFs in cancer are, however, not clear yet. Thus, with regard to the search for new diagnostic strategies, it will be challenging to reveal the potential of TFFs as general markers for a broader spectrum of cancers. Regarding a potential pivotal role in oncogenetic transformation, it will be challenging to further unravel the apparent contradiction between the double-faced tumor-promoting and tumor-suppressing functions of TFF peptides in various carcinomas. Recently, miRNAs attracted more and more attention with regard to their promising role in the regulation and dysregulation of TFFs in cancer development. Further investigations are needed to decipher the complex network between miRNAs, transcription factors and the expression of TFFs, which might provide helpful novel tools for future targeted cancer therapies.

Table 1

Summary of major confirmed effects of TFF peptides.

EffectTFF peptideModel systemReferences
CytoprotectionTFF1–3

Tff1

TFF2
Rodent models

Overexpressing mice

Rat model of GI damage
Reviewed in (10, 15)

(55)

(63)
Epithelial restitution/repair/wound healingTFF1–3Rodent models of GI tract damage(21, 22, 52–57) Reviewed in (9–12, 14, 15)
UlcerationTFF1–3

rITF
Rodent models of GI ulceration; UACL

Rat model of gastric ulceration
(52, 56, 59, 60) Reviewed in (10, 14)

(62)
Apoptosis ↑Tff2

TFF3

Tff3
Developing murine retina

Articular cartilage; colorectal carcinoma cells

Murine cochlea cDNA library;

Tff3-/- mice colonocytes
(49)

(141, 183)

(184, 140)
Apoptosis ↓TFF1

TFF2

TFF3
GI cells+gastric cancer cells

MCF-7; colorectal cancer cell lines

HCT116; transformed rat intestinal epithelial cells; HT-ITF1; IEC-18; gastric cancer+mammary carcinoma cells
(80, 124, 136, 177)

(178–180)

(21, 126, 138, 140, 182, 192)
Proliferation ↑Tff1

Tff2; TFF2

TFF3
Tff1-/- mice

Murine retina; pancreatic β-cells

GES-1; gastric mucosal epithelial+mammary carcinoma+prostate cancer cells
(92)

(49, 122)

(147, 182, 187, 188)
Proliferation ↓TFF1

TFF2

Tff2

TFF3
HCT116; AGS; Y-79, GI cell lines

GI cells and GI carcinoma cell lines

Tff2-/- mice

LoVo; SW837; colorectal carcinoma+corneal epithelia cells
(51, 136, 185)

(178)

(172)

(79, 80, 86)
Migration ↑TFF1–3

TFF1

TFF2

TFF3
Different epithelial cells

Pancreatic stellate+breast cancer+gastric carcinoma cells

HT29; gastric cancer cells; bronchial epithelial cells

Oral keratinocytes; IEC-6; GES-1; rat fibroblasts; rabbit corneal epithelial+bronchial epithelial+colorectal cancer+mammary carcinoma+prostate cancer cells
(1, 10, 65)

(67, 170, 177, 189)

(63, 111, 180)

(46, 53, 86, 131, 139, 182, 188, 191–193)
Invasion ↑TFF1–3

TFF1

TFF3
Transformed kidney+colonic epithelial cells

Pancreatic stellate+breast cancer+gastric carcinoma cells

Rat fibroblasts; mammary carcinoma+prostate cancer cells
(65)

(67, 170, 177, 189)

(182, 188, 192)
Cell scattering ↑TFF2, TFF3MDCKts.src(65)
Metastasis ↑TFF1Pancreatic stellate+breast cancer cells(67, 170, 189)
List of abbreviations
AP-1

activator protein 1

5-Aza-dC

5-Aza-2′deoxycytidine

CDX2

caudal-related homeobox protein 2

COX-2

cytochrome c oxidase assembly factor 2

EGF

epidermal growth factor

EGF-R

epidermal growth factor receptor

ER

estrogen receptor

ERE

estrogen-response element

ERK

extracellular signal-regulated kinase

FoxO3a

forkhead-box-protein O3

HNF

hepatocyte nuclear factor

IL

interleukin

JNK

c-Jun N-terminal kinase

MAPK

mitogen-activated protein kinase

MEK

MAPK kinase

MUC

mucin core protein

NF-κB

nuclear factor ‘kappa-light-chain-enhancer’ of activated B-cells

NR2F2

nuclear receptor subfamily 2

PI3-K

phosphatidylinositol 3-kinase

rITF

rat intestinal trefoil factor

STAT

signal transducer and activator of transcription

TGF-β

transforming growth factor beta

TLR

toll-like receptor

TNF-α

tumor necrosis factor alpha

TXA2-R

thromboxane receptor A2

UACL

ulcer-associated cell lineage

XBP1

X-box binding protein 1.


Corresponding author: Nicole Dünker, Medical Faculty, Institute for Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Hufelandstr. 55, D-45122 Essen, Germany, e-mail:

Acknowledgments

The authors would like to thank D. Gioè and S. Rotthus for proofreading of the manuscript and declare that there is no conflict of financial or other interests.

References

1. Wright NA, Hoffmann W, Otto WR, Rio MC, Thim L. Rolling in the clover: trefoil factor family (TFF)-domain peptides, cell migration and cancer. FEBS Lett 1997; 408: 121–3.10.1016/S0014-5793(97)00424-9Suche in Google Scholar

2. Thim L. A new family of growth factor-like peptides. ‘Trefoil’ disulphide loop structures as a common feature in breast cancer associated peptide (pS2), pancreatic spasmolytic polypeptide (PSP), and frog skin peptides (spasmolysins). FEBS Lett 1989; 250: 85–90.10.1016/0014-5793(89)80690-8Suche in Google Scholar

3. Tomasetto C, Rockel N, Mattei MG, Fujita R, Rio MC. The gene encoding the human spasmolytic protein (SML1/hSP) is in 21q22.3, physically linked to the homologous breast cancer marker gene BCEI/pS2. Genomics 1992; 13: 1328–30.10.1016/0888-7543(92)90059-2Suche in Google Scholar

4. Chinery R, Williamson J, Poulsom R. The gene encoding human intestinal trefoil factor (TFF3) is located on chromosome 21q22.3 clustered with other members of the trefoil peptide family. Genomics 1996; 32: 281–4.10.1006/geno.1996.0117Suche in Google Scholar

5. Thim L. Trefoil peptides: from structure to function. Cell Mol Life Sci 1997; 53: 888–903.10.1007/s000180050108Suche in Google Scholar

6. Hoffmann W, Hauser F. The P-domain or trefoil motif: a role in renewal and pathology of mucous epithelia? Trends Biochem Sci 1993; 18: 239–43.10.1016/0968-0004(93)90170-RSuche in Google Scholar

7. Thim L, May FE. Structure of mammalian trefoil factors and functional insights. Cell Mol Life Sci 2005; 62: 2956–73.10.1007/s00018-005-5484-6Suche in Google Scholar

8. Taupin D, Podolsky DK. Trefoil factors: initiators of mucosal healing. Nat Rev Mol Cell Biol 2003; 4: 721–32.10.1038/nrm1203Suche in Google Scholar

9. Ribieras S, Tomasetto C, Rio MC. The pS2/TFF1 trefoil factor, from basic research to clinical applications. Biochim Biophys Acta 1998; 1378: F61–77.10.1016/S0304-419X(98)00016-XSuche in Google Scholar

10. Kjellev S. The trefoil factor family – small peptides with multiple functionalities. Cell Mol Life Sci 2009; 66: 1350–69.10.1007/s00018-008-8646-5Suche in Google Scholar PubMed

11. Hoffmann W. Trefoil factors TFF (trefoil factor family) peptide-triggered signals promoting mucosal restitution. Cell Mol Life Sci 2005; 62: 2932–8.10.1007/s00018-005-5481-9Suche in Google Scholar

12. Hoffmann W. Chapter 182 – TFF peptides. In: Kastin AJ, editor. Handbook of biologically active peptides 2nd ed., Boston: Academic Press, 2013: 1338–45.Suche in Google Scholar

13. Perry JK, Kannan N, Grandison PM, Mitchell MD, Lobie PE. Are trefoil factors oncogenic? Trends Endocrinol Metab 2008; 19: 74–81.10.1016/j.tem.2007.10.003Suche in Google Scholar

14. Hoffmann W, Jagla W, Wiede A. Molecular medicine of TFF-peptides: from gut to brain. Histol Histopathol 2001; 16: 319–34.Suche in Google Scholar

15. Hoffmann W, Jagla W. Cell type specific expression of secretory TFF peptides: colocalization with mucins and synthesis in the brain. Int Rev Cytol 2002; 213: 147–81.10.1016/S0074-7696(02)13014-2Suche in Google Scholar

16. Jagla W, Wiede A, Hinz M, Dietzmann K, Gülicher D, Gerlach KL, Hoffmann W. Secretion of TFF-peptides by human salivary glands. Cell Tissue Res 1999; 298: 161–6.10.1007/s004419900087Suche in Google Scholar PubMed

17. Madsen J, Nielsen O, Tornøe I, Thim L, Holmskov U. Tissue localization of human trefoil factors 1, 2, and 3. J Histochem Cytochem 2007; 55: 505–13.10.1369/jhc.6A7100.2007Suche in Google Scholar PubMed

18. Tomasetto C, Rio MC, Gautier C, Wolf C, Hareuveni M, Chambon P, Lathe R. hSP, the domain-duplicated homolog of pS2 protein, is co-expressed with pS2 in stomach but not in breast carcinoma. EMBO J 1990; 9: 407–14.10.1002/j.1460-2075.1990.tb08125.xSuche in Google Scholar PubMed PubMed Central

19. Paulsen FP, Berry MS. Mucins and TFF peptides of the tear film and lacrimal apparatus. Prog Histochem Cytochem 2006; 41: 1–53.10.1016/j.proghi.2006.03.001Suche in Google Scholar PubMed

20. Wiede A, Hinz M, Canzler E, Franke K, Quednow C, Hoffmann W. Synthesis and localization of the mucin-associated TFF-peptides in the human uterus. Cell Tissue Res 2001; 303: 109–15.10.1007/s004410000297Suche in Google Scholar PubMed

21. Regalo G, Wright NA, Machado JC. Trefoil factors: from ulceration to neoplasia. Cell Mol Life Sci 2005; 62: 2910–5.10.1007/s00018-005-5478-4Suche in Google Scholar PubMed

22. Wong WM, Poulsom R, Wright NA. Trefoil peptides. Gut 1999; 44: 890–5.10.1136/gut.44.6.890Suche in Google Scholar PubMed PubMed Central

23. Hinz M, Schwegler H, Chwieralski CE, Laube G, Linke R, Pohle W, Hoffmann W. Trefoil factor family (TFF) expression in the mouse brain and pituitary: changes in the developing cerebellum. Peptides 2004; 25: 827–32.10.1016/j.peptides.2004.01.020Suche in Google Scholar

24. Hirota M, Awatsuji H, Sugihara Y, Miyashita S, Furukawa Y, Hayashi K. Expression of pS2 gene in rat brain. Biochem Mol Biol Int 1995; 35: 1079–84.Suche in Google Scholar

25. Probst JC, Skutella T, Müller-Schmid A, Jirikowski GF, Hoffmann W. Molecular and cellular analysis of rP1.B in the rat hypothalamus: in situ hybridization and immunohistochemistry of a new P-domain neuropeptide. Brain Res Mol Brain Res 1995; 33: 269–76.10.1016/0169-328X(95)00137-HSuche in Google Scholar

26. Belovari T, Bijelić N, Tolušić Levak M, Baus Lončar M. Trefoil factor family peptides TFF1 and TFF3 in the nervous tissues of developing mouse embryo. Bosn J Basic Med Sci 2015; 15: 33–7.10.17305/bjbms.2015.251Suche in Google Scholar

27. Jensen P, Heimberg M, Ducray AD, Widmer HR, Meyer M. Expression of trefoil factor 1 in the developing and adult rat ventral mesencephalon. PLoS One 2013; 8: e76592.10.1371/journal.pone.0076592Suche in Google Scholar

28. Hirota M, Miyashita S, Hayashi H, Furukawa Y, Hayashi K. pS2 gene especially expressed in the late G1/S phase of mouse astrocytes. Neurosci Lett 1994; 171: 49–51.10.1016/0304-3940(94)90601-7Suche in Google Scholar

29. Probst JC, Zetzsche T, Weber M, Theilemann P, Skutella T, Landgraf R, Jirikowski GF. Human intestinal trefoil factor is expressed in human hypothalamus and pituitary: evidence for a novel neuropeptide. FASEB J 1996; 10: 1518–23.10.1096/fasebj.10.13.8940297Suche in Google Scholar PubMed

30. Schwarz H, Jagla W, Wiede A, Hoffmann W. Ultrastructural co-localization of TFF3-peptide and oxytocin in the neural lobe of the porcine pituitary. Cell Tissue Res 2001; 305: 411–6.10.1007/s004410100412Suche in Google Scholar PubMed

31. Jagla W, Wiede A, Dietzmann K, Rutkowski K, Hoffmann W. Co-localization of TFF3 peptide and oxytocin in the human hypothalamus. FASEB J 2000; 14: 1126–31.10.1096/fasebj.14.9.1126Suche in Google Scholar PubMed

32. Paterson RW, Bartlett JW, Blennow K, Fox NC, Shaw LM, Trojanowski JQ, Zetterberg H, Schott JM. Cerebrospinal fluid markers including trefoil factor 3 are associated with neurodegeneration in amyloid-positive individuals. Transl Psychiatry 2014; 4: e419.10.1038/tp.2014.58Suche in Google Scholar PubMed PubMed Central

33. Bernstein H, Dobrowolny H, Trübner K, Steiner J, Bogerts B, Hoffmann W. Differential regional and cellular distribution of TFF3 peptide in the human brain. Amino Acids 2015; 47: 1053–63.10.1007/s00726-015-1938-9Suche in Google Scholar PubMed

34. Fu T, Stellmacher A, Znalesniak EB, Dieterich DC, Kalbacher H, Hoffmann W. Tff3 is expressed in neurons and microglial cells. Cell Physiol Biochem 2014; 34: 1912–9.10.1159/000366389Suche in Google Scholar

35. Schwarzberg H, Kalbacher H, Hoffmann W. Differential behavioral effects of TFF peptides: injections of synthetic TFF3 into the rat amygdala. Pharmacol Biochem Behav 1999; 62: 173–8.10.1016/S0091-3057(98)00137-3Suche in Google Scholar

36. Shi H, Zhu W, Liu J, Luo Y, Si J, Wang S, Xue Y, Ding Z, Shi J, Lu L. PI3K/Akt signaling pathway in the basolateral amygdala mediates the rapid antidepressant-like effects of trefoil factor 3. Neuropsychopharmacology 2012; 37: 2671–83.10.1038/npp.2012.131Suche in Google Scholar PubMed PubMed Central

37. Liu SQ, Roberts D, Zhang B, Ren Y, Zhang L, Wu YH. Trefoil factor 3 as an endocrine neuroprotective factor from the liver in experimental cerebral ischemia/reperfusion injury. PLoS ONE 2013; 8: e77732.10.1371/journal.pone.0077732Suche in Google Scholar PubMed PubMed Central

38. Wu P, Shi H, Luo Y, Zhang R, Li J, Shi J, Lu L, Zhu W. Neuropeptide trefoil factor 3 attenuates naloxone-precipitated withdrawal in morphine-dependent mice. Psychopharmacology (Berl) 2014; 231: 4659–68.10.1007/s00213-014-3615-1Suche in Google Scholar PubMed

39. Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR, Xie Z, Carrillo-Reid L, Auyeung G, Antonacci C, Buch A, Yang L, Beal MF, Surmeier DJ, Kordower JH, Tabar V, Studer L. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson’s disease. Nature 2011; 480: 547–51.10.1038/nature10648Suche in Google Scholar PubMed PubMed Central

40. Gardiner EJ, Cairns MJ, Liu B, Beveridge NJ, Carr V, Kelly B, Scott RJ, Tooney PA. Gene expression analysis reveals schizophrenia-associated dysregulation of immune pathways in peripheral blood mononuclear cells. J Psychiatr Res 2013; 47: 425–37.10.1016/j.jpsychires.2012.11.007Suche in Google Scholar PubMed PubMed Central

41. Langer G, Jagla W, Behrens-Baumann W, Walter S, Hoffmann W. Secretory peptides TFF1 and TFF3 synthesized in human conjunctival goblet cells. Invest Ophthalmol Vis Sci 1999; 40: 2220–4.Suche in Google Scholar

42. Langer G, Jagla W, Behrens-Baumann W, Walter S, Hoffmann W. Ocular TFF-peptides: new mucus-associated secretory products of conjunctival goblet cells. Adv Exp Med Biol 2002; 506: 313–6.10.1007/978-1-4615-0717-8_44Suche in Google Scholar PubMed

43. Jagla W, Wiede A, Hoffmann W. Localization of TFF3 peptide to porcine conjunctival goblet cells. Cell Tissue Res 1999; 296: 525–30.10.1007/s004410051313Suche in Google Scholar PubMed

44. Steven P, Schäfer G, Nölle B, Hinz M, Hoffmann W, Paulsen F. Distribution of TFF peptides in corneal disease and pterygium. Peptides 2004; 25: 819–25.10.1016/j.peptides.2004.01.023Suche in Google Scholar PubMed

45. Schulze U, Sel S, Paulsen FP. Trefoil factor family peptide 3 at the ocular surface. A promising therapeutic candidate for patients with dry eye syndrome? Dev Ophthalmol 2010; 45: 1–11.Suche in Google Scholar

46. Göke MN, Cook JR, Kunert KS, Fini ME, Gipson IK, Podolsky DK. Trefoil peptides promote restitution of wounded corneal epithelial cells. Exp Cell Res 2001; 264: 337–44.10.1006/excr.2000.5116Suche in Google Scholar

47. Paulsen FP, Hinz M, Schaudig U, Thale AB, Hoffmann W. TFF peptides in the human efferent tear ducts. Invest Ophthalmol Vis Sci 2002; 43: 3359–64.Suche in Google Scholar

48. Paulsen FP, Woon C, Varoga D, Jansen A, Garreis F, Jäger K, Amm M, Podolsky DK, Steven P, Barker NP, Sel S. Intestinal trefoil factor/TFF3 promotes re-epithelialization of corneal wounds. J Biol Chem 2008; 283: 13418–27.10.1074/jbc.M800177200Suche in Google Scholar

49. Paunel-Görgülü AN, Franke AG, Paulsen FP, Dünker N. Trefoil factor family peptide 2 acts pro-proliferative and pro-apoptotic in the murine retina. Histochem Cell Biol 2011; 135: 461–73.10.1007/s00418-011-0810-6Suche in Google Scholar

50. Philippeit C, Busch M, Dünker N. Epigenetic control of trefoil factor family (TFF) peptide expression in human retinoblastoma cell lines. Cell Physiol Biochem 2014; 34: 1001–14.10.1159/000366316Suche in Google Scholar

51. Weise A, Dünker N. High trefoil factor 1 (TFF1) expression in human retinoblastoma cells correlates with low growth kinetics, increased cyclin-dependent kinase (CDK) inhibitor levels and a selective down-regulation of CDK6. Histochem Cell Biol 2013; 139: 323–38.10.1007/s00418-012-1028-ySuche in Google Scholar

52. Wright NA, Poulsom R, Stamp G, van Noorden S, Sarraf C, Elia G, Ahnen D, Jeffery R, Longcroft J, Pike C. Trefoil peptide gene expression in gastrointestinal epithelial cells in inflammatory bowel disease. Gastroenterology 1993; 104: 12–20.10.1016/0016-5085(93)90830-6Suche in Google Scholar

53. Dignass A, Lynch-Devaney K, Kindon H, Thim L, Podolsky DK. Trefoil peptides promote epithelial migration through a transforming growth factor beta-independent pathway. J Clin Invest 1994; 94: 376–83.10.1172/JCI117332Suche in Google Scholar PubMed PubMed Central

54. Mashimo H, Wu DC, Podolsky DK, Fishman MC. Impaired defense of intestinal mucosa in mice lacking intestinal trefoil factor. Science 1996; 274: 262–5.10.1126/science.274.5285.262Suche in Google Scholar PubMed

55. Playford RJ, Marchbank T, Goodlad RA, Chinery RA, Poulsom R, Hanby AM. Transgenic mice that overexpress the human trefoil peptide pS2 have an increased resistance to intestinal damage. Proc Natl Acad Sci USA 1996; 93: 2137–42.10.1073/pnas.93.5.2137Suche in Google Scholar PubMed PubMed Central

56. Wright NA. Aspects of the biology of regeneration and repair in the human gastrointestinal tract. Philos Trans R Soc Lond B Biol Sci 1998; 353: 925–33.10.1098/rstb.1998.0257Suche in Google Scholar PubMed PubMed Central

57. Aamann L, Vestergaard EM, Grønbæk H. Trefoil factors in inflammatory bowel disease. World J Gastroenterol 2014; 20: 3223–30.10.3748/wjg.v20.i12.3223Suche in Google Scholar

58. Kim B, Kim K, Lee B, Maeng L, Choi H, Cho S, Chae H, Kim J, Choi K, Chung I. Expression of trefoil peptides in the subtypes of intestinal metaplasia. Peptides 2004; 25: 779–83.10.1016/j.peptides.2003.12.021Suche in Google Scholar

59. Wright NA, Poulsom R, Stamp GW, Hall PA, Jeffery RE, Longcroft JM, Rio MC, Tomasetto C, Chambon P. Epidermal growth factor (EGF/URO) induces expression of regulatory peptides in damaged human gastrointestinal tissues. J Pathol 1990; 162: 279–84.10.1002/path.1711620402Suche in Google Scholar

60. Poulsom R, Chinery R, Sarraf C, van Noorden S, Stamp GW, Lalani EN, Elia G, Wright NA. Trefoil peptide gene expression in small intestinal Crohn’s disease and dietary adaptation. J Clin Gastroenterol 1993; 17(Suppl 1): S78–91.10.1097/00004836-199312001-00016Suche in Google Scholar

61. Rio MC, Chenard MP, Wolf C, Marcellin L, Tomasetto C, Lathe R, Bellocq JP, Chambon P. Induction of pS2 and hSP genes as markers of mucosal ulceration of the digestive tract. Gastroenterology 1991; 100: 375–9.10.1016/0016-5085(91)90205-YSuche in Google Scholar

62. Alison MR, Chinery R, Poulsom R, Ashwood P, Longcroft JM, Wright NA. Experimental ulceration leads to sequential expression of spasmolytic polypeptide, intestinal trefoil factor, epidermal growth factor and transforming growth factor alpha mRNAs in rat stomach. J Pathol 1995; 175: 405–14.10.1002/path.1711750408Suche in Google Scholar

63. Playford RJ, Marchbank T, Chinery R, Evison R, Pignatelli M, Boulton RA, Thim L, Hanby AM. Human spasmolytic polypeptide is a cytoprotective agent that stimulates cell migration. Gastroenterology 1995; 108: 108–16.10.1016/0016-5085(95)90014-4Suche in Google Scholar

64. Faith DA, Isaacs WB, Morgan JD, Fedor HL, Hicks JL, Mangold LA, Walsh PC, Partin AW, Platz EA, Luo J, De Marzo, Angelo M. Trefoil factor 3 overexpression in prostatic carcinoma: prognostic importance using tissue microarrays. Prostate 2004; 61: 215–27.10.1002/pros.20095Suche in Google Scholar

65. Emami S, Rodrigues S, Rodrigue CM, Le Floch N, Rivat C, Attoub S, Bruyneel E, Gespach C. Trefoil factor family (TFF) peptides and cancer progression. Peptides 2004; 25: 885–98.10.1016/j.peptides.2003.10.019Suche in Google Scholar

66. Poulsom R, Hanby AM, Lalani EN, Hauser F, Hoffmann W, Stamp GW. Intestinal trefoil factor (TFF 3) and pS2 (TFF 1), but not spasmolytic polypeptide (TFF 2) mRNAs are co-expressed in normal, hyperplastic, and neoplastic human breast epithelium. J Pathol 1997; 183: 30–8.10.1002/(SICI)1096-9896(199709)183:1<30::AID-PATH1085>3.0.CO;2-KSuche in Google Scholar

67. Buache E, Etique N, Alpy F, Stoll I, Muckensturm M, Reina-San-Martin B, Chenard MP, Tomasetto C, Rio MC. Deficiency in trefoil factor 1 (TFF1) increases tumorigenicity of human breast cancer cells and mammary tumor development in TFF1-knockout mice. Oncogene 2011; 30: 3261–73.10.1038/onc.2011.41Suche in Google Scholar

68. Chenard M, Tomasetto C, Bellocq J, Rio M. Urinary pS2/TFF1 levels in the management of hormonodependent breast carcinomas. Peptides 2004; 25: 737–43.10.1016/j.peptides.2003.11.018Suche in Google Scholar

69. Dunnwald LK, Rossing MA, Li CI. Hormone receptor status, tumor characteristics, and prognosis: a prospective cohort of breast cancer patients. Breast Cancer Res 2007; 9: R6.10.1186/bcr1639Suche in Google Scholar

70. Mikhitarian K, Gillanders WE, Almeida JS, Hebert Martin R, Varela JC, Metcalf JS, Cole DJ, Mitas M. An innovative microarray strategy identities informative molecular markers for the detection of micrometastatic breast cancer. Clin Cancer Res 2005; 11: 3697–704.10.1158/1078-0432.CCR-04-2164Suche in Google Scholar

71. Weigelt B, Verduijn P, Bosma AJ, Rutgers EJ, Peterse HL, van’t Veer, LJ. Detection of metastases in sentinel lymph nodes of breast cancer patients by multiple mRNA markers. Br J Cancer 2004; 90: 1531–7.10.1038/sj.bjc.6601659Suche in Google Scholar

72. Hippo Y, Yashiro M, Ishii M, Taniguchi H, Tsutsumi S, Hirakawa K, Kodama T, Aburatani H. Differential gene expression profiles of scirrhous gastric cancer cells with high metastatic potential to peritoneum or lymph nodes. Cancer Res 2001; 61: 889–95.Suche in Google Scholar

73. Kirikoshi H, Katoh M. Expression of TFF1, TFF2 and TFF3 in gastric cancer. Int J Oncol 2002; 21: 655–9.10.3892/ijo.21.3.655Suche in Google Scholar

74. Leung WK, Yu J, Chan FK, To KF, Chan MW, Ebert MP, Ng EK, Chung SC, Malfertheiner P, Sung JJ. Expression of trefoil peptides (TFF1, TFF2, and TFF3) in gastric carcinomas, intestinal metaplasia, and non-neoplastic gastric tissues. J Pathol 2002; 197: 582–8.10.1002/path.1147Suche in Google Scholar

75. Taupin D, Pedersen J, Familari M, Cook G, Yeomans N, Giraud AS. Augmented intestinal trefoil factor (TFF3) and loss of pS2 (TFF1) expression precedes metaplastic differentiation of gastric epithelium. Lab Invest 2001; 81: 397–408.10.1038/labinvest.3780247Suche in Google Scholar

76. Suárez C, Vizoso F, Rodríguez JC, García I, Raigoso P, Allende MT, García-Muñíz JL, García-Morán M. Prognostic significance of cytosolic pS2 protein content in gastric cancer. Int J Biol Markers 2001; 16: 37–44.10.1177/172460080101600105Suche in Google Scholar

77. Yamachika T, Werther JL, Bodian C, Babyatsky M, Tatematsu M, Yamamura Y, Chen A, Itzkowitz S. Intestinal trefoil factor: a marker of poor prognosis in gastric carcinoma. Clin Cancer Res 2002; 8: 1092–9.Suche in Google Scholar

78. May FE, Westley BR. Expression of human intestinal trefoil factor in malignant cells and its regulation by oestrogen in breast cancer cells. J Pathol 1997; 182: 404–13.10.1002/(SICI)1096-9896(199708)182:4<404::AID-PATH875>3.0.CO;2-0Suche in Google Scholar

79. Uchino H, Kataoka H, Itoh H, Sekiya R, Onitsuka T, Koono M. Roles of intestinal trefoil factor (ITF) in human colorectal cancer: ITF suppresses the growth of colorectal carcinoma cells. Hum Cell 1999; 12: 181–8.Suche in Google Scholar

80. Uchino H, Kataoka H, Itoh H, Hamasuna R, Koono M. Overexpression of intestinal trefoil factor in human colon carcinoma cells reduces cellular growth in vitro and in vivo. Gastroenterology 2000; 118: 60–9.10.1016/S0016-5085(00)70414-8Suche in Google Scholar

81. Okada H, Kimura MT, Tan D, Fujiwara K, Igarashi J, Makuuchi M, Hui A, Tsurumaru M, Nagase H. Frequent trefoil factor 3 (TFF3) overexpression and promoter hypomethylation in mouse and human hepatocellular carcinomas. Int J Oncol 2005; 26: 369–77.10.3892/ijo.26.2.369Suche in Google Scholar

82. Terris B, Blaveri E, Crnogorac-Jurcevic T, Jones M, Missiaglia E, Ruszniewski P, Sauvanet A, Lemoine NR. Characterization of gene expression profiles in intraductal papillary-mucinous tumors of the pancreas. Am J Pathol 2002; 160: 1745–54.10.1016/S0002-9440(10)61121-2Suche in Google Scholar

83. Morito K, Nakamura J, Kitajima Y, Kai K, Tanaka T, Kubo H, Miyake S, Noshiro H. The value of trefoil factor 3 expression in predicting the long-term outcome and early recurrence of colorectal cancer. Int J Oncol 2015; 46: 563–8.10.3892/ijo.2014.2755Suche in Google Scholar

84. Khoury T, Chadha K, Javle M, Donohue K, Levea C, Iyer R, Okada H, Nagase H, Tan D. Expression of intestinal trefoil factor (TFF-3) in hepatocellular carcinoma. Int J Gastrointest Cancer 2005; 35: 171–7.10.1385/IJGC:35:3:171Suche in Google Scholar

85. Vestergaard EM, Borre M, Poulsen SS, Nexø E, Tørring N. Plasma levels of trefoil factors are increased in patients with advanced prostate cancer. Clin Cancer Res 2006; 12: 807–12.10.1158/1078-0432.CCR-05-1545Suche in Google Scholar PubMed

86. Schulze U, Hampel U, Sel S, Contreras-Ruiz L, Schicht M, Dieckow J, Diebold Y, Paulsen F. Trefoil factor family peptide 3 (TFF3) is upregulated under experimental conditions similar to dry eye disease and supports corneal wound healing effects in vitro. Invest Ophthalmol Vis Sci 2014; 55: 3037–42.10.1167/iovs.13-13423Suche in Google Scholar PubMed

87. Jakowlew SB, Breathnach R, Jeltsch JM, Masiakowski P, Chambon P. Sequence of the pS2 mRNA induced by estrogen in the human breast cancer cell line MCF-7. Nucleic Acids Res 1984; 12: 2861–78.10.1093/nar/12.6.2861Suche in Google Scholar PubMed PubMed Central

88. Baus-Loncar M, Giraud AS. Multiple regulatory pathways for trefoil factor (TFF) genes. Cell Mol Life Sci 2005; 62: 2921–31.10.1007/s00018-005-5480-xSuche in Google Scholar

89. Iwakiri D, Podolsky DK. A silencer inhibitor confers specific expression of intestinal trefoil factor in gobletlike cell lines. Am J Physiol Gastrointest Liver Physiol 2001; 280: G1114–23.10.1152/ajpgi.2001.280.6.G1114Suche in Google Scholar PubMed

90. Seib T, Blin N, Hilgert K, Seifert M, Theisinger B, Engel M, Dooley S, Zang KD, Welter C. The three human trefoil genes TFF1, TFF2, and TFF3 are located within a region of 55 kb on chromosome 21q22.3. Genomics 1997; 40: 200–2.10.1006/geno.1996.4511Suche in Google Scholar PubMed

91. Taupin D. The trefoil gene family are coordinately expressed immediate-early genes: EGF receptor- and MAP kinase-dependent interregulation. J Clin Invest 1999; 103: R31–8.10.1172/JCI3304Suche in Google Scholar PubMed PubMed Central

92. Lefebvre O, Chenard MP, Masson R, Linares J, Dierich A, LeMeur M, Wendling C, Tomasetto C, Chambon P, Rio MC. Gastric mucosa abnormalities and tumorigenesis in mice lacking the pS2 trefoil protein. Science 1996; 274: 259–62.10.1126/science.274.5285.259Suche in Google Scholar PubMed

93. Liu D, el-Hariry I, Karayiannakis AJ, Wilding J, Chinery R, Kmiot W, McCrea PD, Gullick WJ, Pignatelli M. Phosphorylation of beta-catenin and epidermal growth factor receptor by intestinal trefoil factor. Lab Invest 1997; 77: 557–63.Suche in Google Scholar

94. Brown AM, Jeltsch JM, Roberts M, Chambon P. Activation of pS2 gene transcription is a primary response to estrogen in the human breast cancer cell line MCF-7. Proc Natl Acad Sci USA 1984; 81: 6344–8.10.1073/pnas.81.20.6344Suche in Google Scholar PubMed PubMed Central

95. Rio MC, Bellocq JP, Gairard B, Rasmussen UB, Krust A, Koehl C, Calderoli H, Schiff V, Renaud R, Chambon P. Specific expression of the pS2 gene in subclasses of breast cancers in comparison with expression of the estrogen and progesterone receptors and the oncogene ERBB2. Proc Natl Acad Sci USA 1987; 84: 9243–7.10.1073/pnas.84.24.9243Suche in Google Scholar PubMed PubMed Central

96. Prud’homme JF, Fridlansky F, Le Cunff M, Atger M, Mercier-Bodart C, Pichon MF, Milgrom E. Cloning of a gene expressed in human breast cancer and regulated by estrogen in MCF-7 cells. DNA 1985; 4: 11–21.10.1089/dna.1985.4.11Suche in Google Scholar PubMed

97. Nunez AM, Berry M, Imler JL, Chambon P. The 5′ flanking region of the pS2 gene contains a complex enhancer region responsive to oestrogens, epidermal growth factor, a tumour promoter (TPA), the c-Ha-ras oncoprotein and the c-jun protein. EMBO J 1989; 8: 823–9.10.1002/j.1460-2075.1989.tb03443.xSuche in Google Scholar PubMed PubMed Central

98. Sun J, Spencer VA, Li L, Yu Chen H, Yu J, Davie JR. Estrogen regulation of trefoil factor 1 expression by estrogen receptor alpha and Sp proteins. Exp Cell Res 2005; 302: 96–107.10.1016/j.yexcr.2004.08.015Suche in Google Scholar PubMed

99. Lacroix M, Leclercq G. About GATA3, HNF3A, and XBP1, three genes co-expressed with the oestrogen receptor-alpha gene (ESR1) in breast cancer. Mol Cell Endocrinol 2004; 219: 1–7.10.1016/j.mce.2004.02.021Suche in Google Scholar PubMed

100. Morelli C, Lanzino M, Garofalo C, Maris P, Brunelli E, Casaburi I, Catalano S, Bruno R, Sisci D, Andò S. Akt2 inhibition enables the forkhead transcription factor FoxO3a to have a repressive role in estrogen receptor alpha transcriptional activity in breast cancer cells. Mol Cell Biol 2010; 30: 857–70.10.1128/MCB.00824-09Suche in Google Scholar PubMed PubMed Central

101. Lu D, Kiriyama Y, Lee KY, Giguère V. Transcriptional regulation of the estrogen-inducible pS2 breast cancer marker gene by the ERR family of orphan nuclear receptors. Cancer Res 2001; 61: 6755–61.Suche in Google Scholar

102. Campbell-Thompson ML. Estrogen receptor alpha and beta expression in upper gastrointestinal tract with regulation of trefoil factor family 2 mRNA levels in ovariectomized rats. Biochem Biophys Res Commun 1997; 240: 478–83.10.1006/bbrc.1997.7683Suche in Google Scholar PubMed

103. Singh S, Baker PR, Poulsom R, Wright NA, Sheppard MC, Langman MJ, Neoptolemos JP. Expression of oestrogen receptor and oestrogen-inducible genes in pancreatic cancer. Br J Surg 1997; 84: 1085–9.Suche in Google Scholar

104. Borthwick JM, Charnock-Jones DS, Tom BD, Hull ML, Teirney R, Phillips SC, Smith SK. Determination of the transcript profile of human endometrium. Mol Hum Reprod 2003; 9: 19–33.10.1093/molehr/gag004Suche in Google Scholar

105. Kao LC, Tulac S, Lobo S, Imani B, Yang JP, Germeyer A, Osteen K, Taylor RN, Lessey BA, Giudice LC. Global gene profiling in human endometrium during the window of implantation. Endocrinology 2002; 143: 2119–38.10.1210/endo.143.6.8885Suche in Google Scholar

106. Mhawech-Fauceglia P, Wang D, Samrao D, Liu S, DuPont NC, Pejovic T. Trefoil factor family 3 (TFF3) expression and its interaction with estrogen receptor (ER) in endometrial adenocarcinoma. Gynecol Oncol 2013; 130: 174–80.10.1016/j.ygyno.2013.03.030Suche in Google Scholar

107. Khan ZE, Wang TC, Cui G, Chi AL, Dimaline R. Transcriptional regulation of the human trefoil factor, TFF1, by gastrin. Gastroenterology 2003; 125: 510–21.10.1016/S0016-5085(03)00908-9Suche in Google Scholar

108. Judd LM, Alderman BM, Howlett M, Shulkes A, Dow C, Moverley J, Grail D, Jenkins BJ, Ernst M, Giraud AS. Gastric cancer development in mice lacking the SHP2 binding site on the IL-6 family co-receptor gp130. Gastroenterology 2004; 126: 196–207.10.1053/j.gastro.2003.10.066Suche in Google Scholar PubMed

109. Van De Bovenkamp JH, Korteland-Van Male AM, Büller HA, Einerhand AW, Dekker J. Infection with Helicobacter pylori affects all major secretory cell populations in the human antrum. Dig Dis Sci 2005; 50: 1078–86.10.1007/s10620-005-2708-4Suche in Google Scholar PubMed

110. Wright NA, Pike C, Elia G. Induction of a novel epidermal growth factor-secreting cell lineage by mucosal ulceration in human gastrointestinal stem cells. Nature 1990; 343: 82–5.10.1038/343082a0Suche in Google Scholar PubMed

111. Graness A, Chwieralski CE, Reinhold D, Thim L, Hoffmann W. Protein kinase C and ERK activation are required for TFF-peptide-stimulated bronchial epithelial cell migration and tumor necrosis factor-alpha-induced interleukin-6 (IL-6) and IL-8 secretion. J Biol Chem 2002; 277: 18440–6.10.1074/jbc.M200468200Suche in Google Scholar PubMed

112. Dossinger V, Kayademir T, Blin N, Gött P. Down-regulation of TFF expression in gastrointestinal cell lines by cytokines and nuclear factors. Cell Physiol Biochem 2002; 12: 197–206.10.1159/000066279Suche in Google Scholar PubMed

113. Soutto M, Belkhiri A, Piazuelo MB, Schneider BG, Peng D, Jiang A, Washington MK, Kokoye Y, Crowe SE, Zaika A, Correa P, Peek RM, El-Rifai W. Loss of TFF1 is associated with activation of NF-κB-mediated inflammation and gastric neoplasia in mice and humans. J Clin Invest 2011; 121: 1753–67.10.1172/JCI43922Suche in Google Scholar PubMed PubMed Central

114. Cobler L, Mejías-Luque R, Garrido M, Pera M, Badia-Garrido E, de Bolós C. Activation of the NF-kB pathway downregulates TFF-1 in gastric carcinogenesis. Virchows Arch 2013; 463: 497–507.10.1007/s00428-013-1469-2Suche in Google Scholar

115. Tebbutt NC, Giraud AS, Inglese M, Jenkins B, Waring P, Clay FJ, Malki S, Alderman BM, Grail D, Hollande F, Heath JK, Ernst M. Reciprocal regulation of gastrointestinal homeostasis by SHP2 and STAT-mediated trefoil gene activation in gp130 mutant mice. Nat Med 2002; 8: 1089–97.10.1038/nm763Suche in Google Scholar

116. Loncar MB, Al-azzeh E, Sommer PS, Marinovic M, Schmehl K, Kruschewski M, Blin N, Stohwasser R, Gött P, Kayademir T. Tumour necrosis factor alpha and nuclear factor kappaB inhibit transcription of human TFF3 encoding a gastrointestinal healing peptide. Gut 2003; 52: 1297–303.10.1136/gut.52.9.1297Suche in Google Scholar

117. Teng X, Xu L, Zhou P, Sun H, Sun M. Effects of trefoil peptide 3 on expression of TNF-alpha, TLR4, and NF-kappaB in trinitrobenzene sulphonic acid induced colitis mice. Inflammation 2009; 32: 120–9.10.1007/s10753-009-9110-xSuche in Google Scholar

118. Podolsky DK, Gerken G, Eyking A, Cario E. Colitis-associated variant of TLR2 causes impaired mucosal repair because of TFF3 deficiency. Gastroenterology 2009; 137: 209–20.10.1053/j.gastro.2009.03.007Suche in Google Scholar

119. Blanchard C, Durual S, Estienne M, Bouzakri K, Heim MH, Blin N, Cuber J. IL-4 and IL-13 up-regulate intestinal trefoil factor expression: requirement for STAT6 and de novo protein synthesis. J Immunol 2004; 172: 3775–83.10.4049/jimmunol.172.6.3775Suche in Google Scholar

120. Dubeykovskaya Z, Dubeykovskiy A, Solal-Cohen J, Wang TC. Secreted trefoil factor 2 activates the CXCR4 receptor in epithelial and lymphocytic cancer cell lines. J Biol Chem 2009; 284: 3650–62.10.1074/jbc.M804935200Suche in Google Scholar

121. Hoffmann W. Trefoil factor family (TFF) peptides and chemokine receptors: a promising relationship. J Med Chem 2009; 52: 6505–10.10.1021/jm9008136Suche in Google Scholar

122. Orime K, Shirakawa J, Togashi Y, Tajima K, Inoue H, Ito Y, Sato K, Nakamura A, Aoki K, Goshima Y, Terauchi Y. Trefoil factor 2 promotes cell proliferation in pancreatic β-cells through CXCR-4-mediated ERK1/2 phosphorylation. Endocrinology 2013; 154: 54–64.10.1210/en.2012-1814Suche in Google Scholar

123. Thim L, Mørtz E. Isolation and characterization of putative trefoil peptide receptors. Regul Pept 2000; 90: 61–8.10.1016/S0167-0115(00)00110-5Suche in Google Scholar

124. Tomasetto C, Masson R, Linares JL, Wendling C, Lefebvre O, Chenard MP, Rio MC. pS2/TFF1 interacts directly with the VWFC cysteine-rich domains of mucins. Gastroenterology 2000; 118: 70–80.10.1016/S0016-5085(00)70415-XSuche in Google Scholar

125. Emami S, Le Floch N, Bruyneel E, Thim L, May F, Westley B, Rio M, Mareel M, Gespach C. Induction of scattering and cellular invasion by trefoil peptides in src- and RhoA-transformed kidney and colonic epithelial cells. FASEB J 2001; 15: 351–61.10.1096/fj.00-0355comSuche in Google Scholar PubMed

126. Kinoshita K, Taupin DR, Itoh H, Podolsky DK. Distinct pathways of cell migration and antiapoptotic response to epithelial injury: structure-function analysis of human intestinal trefoil factor. Mol Cell Biol 2000; 20: 4680–90.10.1128/MCB.20.13.4680-4690.2000Suche in Google Scholar PubMed PubMed Central

127. Rodrigues S, Nguyen QD, Faivre S, Bruyneel E, Thim L, Westley B, May F, Flatau G, Mareel M, Gespach C, Emami S. Activation of cellular invasion by trefoil peptides and src is mediated by cyclooxygenase- and thromboxane A2 receptor-dependent signaling pathways. FASEB J 2001; 15: 1517–28.10.1096/fj.00-0802comSuche in Google Scholar PubMed

128. Régnauld K, Nguyen Q, Vakaet L, Bruyneel E, Launay J, Endo T, Mareel M, Gespach C, Emami S. G-protein alpha(olf) subunit promotes cellular invasion, survival, and neuroendocrine differentiation in digestive and urogenital epithelial cells. Oncogene 2002; 21: 4020–31.10.1038/sj.onc.1205498Suche in Google Scholar PubMed

129. Kosriwong K, Menheniott TR, Giraud AS, Jearanaikoon P, Sripa B, Limpaiboon T. Trefoil factors: tumor progression markers and mitogens via EGFR/MAPK activation in cholangiocarcinoma. World J Gastroenterol 2011; 17: 1631–41.10.3748/wjg.v17.i12.1631Suche in Google Scholar PubMed PubMed Central

130. Efstathiou JA, Liu D, Wheeler JM, Kim HC, Beck NE, Ilyas M, Karayiannakis AJ, Mortensen NJ, Kmiot W, Playford RJ, Pignatelli M, Bodmer WF. Mutated epithelial cadherin is associated with increased tumorigenicity and loss of adhesion and of responsiveness to the motogenic trefoil factor 2 in colon carcinoma cells. Proc Natl Acad Sci USA 1999; 96: 2316–21.10.1073/pnas.96.5.2316Suche in Google Scholar PubMed PubMed Central

131. Oertel M, Graness A, Thim L, Bühling F, Kalbacher H, Hoffmann W. Trefoil factor family-peptides promote migration of human bronchial epithelial cells: synergistic effect with epidermal growth factor. Am J Respir Cell Mol Biol 2001; 25: 418–24.10.1165/ajrcmb.25.4.4429Suche in Google Scholar PubMed

132. Chwieralski CE, Schnurra I, Thim L, Hoffmann W. Epidermal growth factor and trefoil factor family 2 synergistically trigger chemotaxis on BEAS-2B cells via different signaling cascades. Am J Respir Cell Mol Biol 2004; 31: 528–37.10.1165/rcmb.2003-0433OCSuche in Google Scholar PubMed

133. Zheng Q, Gao J, Li H, Guo W, Mao Q, Gao E, Zhu Y. Trefoil factor 3 peptide regulates migration via a Twist-dependent pathway in gastric cell. Biochem Biophys Res Commun 2013; 438: 6–12.10.1016/j.bbrc.2013.06.115Suche in Google Scholar PubMed

134. Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C, Darnell JE. Stat3 as an oncogene. Cell 1999; 98: 295–303.10.1016/S0092-8674(00)81959-5Suche in Google Scholar

135. Pandey V, Wu Z, Zhang M, Li R, Zhang J, Zhu T, Lobie PE. Trefoil factor 3 promotes metastatic seeding and predicts poor survival outcome of patients with mammary carcinoma. Breast Cancer Res 2014; 16: 429.10.1186/s13058-014-0429-3Suche in Google Scholar PubMed PubMed Central

136. Bossenmeyer-Pourié C, Kannan R, Ribieras S, Wendling C, Stoll I, Thim L, Tomasetto C, Rio M. The trefoil factor 1 participates in gastrointestinal cell differentiation by delaying G1-S phase transition and reducing apoptosis. J Cell Biol 2002; 157: 761–70.10.1083/jcb200108056Suche in Google Scholar PubMed PubMed Central

137. Tomasetto C, Rio M. Pleiotropic effects of Trefoil factor 1 deficiency. Cell Mol Life Sci 2005; 62: 2916–20.10.1007/s00018-005-5479-3Suche in Google Scholar PubMed

138. Chen YH, Lu Y, De Plaen IG, Wang LY, Tan XD. Transcription factor NF-kappaB signals antianoikic function of trefoil factor 3 on intestinal epithelial cells. Biochem Biophys Res Commun 2000; 274: 576–82.10.1006/bbrc.2000.3176Suche in Google Scholar PubMed

139. Sun Y, Wang L, Zhou Y, Mao X, Deng X. Cloning and characterization of the human trefoil factor 3 gene promoter. PLoS One 2014; 9: e95562.10.1371/journal.pone.0095562Suche in Google Scholar PubMed PubMed Central

140. Taupin DR, Kinoshita K, Podolsky DK. Intestinal trefoil factor confers colonic epithelial resistance to apoptosis. Proc Natl Acad Sci USA 2000; 97: 799–804.10.1073/pnas.97.2.799Suche in Google Scholar PubMed PubMed Central

141. Efstathiou JA, Noda M, Rowan A, Dixon C, Chinery R, Jawhari A, Hattori T, Wright NA, Bodmer WF, Pignatelli M. Intestinal trefoil factor controls the expression of the adenomatous polyposis coli-catenin and the E-cadherin-catenin complexes in human colon carcinoma cells. Proc Natl Acad Sci USA 1998; 95: 3122–7.10.1073/pnas.95.6.3122Suche in Google Scholar PubMed PubMed Central

142. Meyer zum Büschenfelde D, Hoschützky H, Tauber R, Huber O. Molecular mechanisms involved in TFF3 peptide-mediated modulation of the E-cadherin/catenin cell adhesion complex. Peptides 2004; 25: 873–83.10.1016/j.peptides.2003.11.024Suche in Google Scholar PubMed

143. Yang X, Lu Y, Liu J, Huang J, Liu Y, Xiao C, Jazag A, Ren J, Guleng B. Piezo1 is as a novel trefoil factor family 1 binding protein that promotes gastric cancer cell mobility in vitro. Dig Dis Sci 2014; 59: 1428–35.10.1007/s10620-014-3044-3Suche in Google Scholar PubMed

144. Sands BE, Podolsky DK. The trefoil peptide family. Annu Rev Physiol 1996; 58: 253–73.10.1146/annurev.ph.58.030196.001345Suche in Google Scholar

145. Meyer zum Büschenfelde D, Tauber R, Huber O. TFF3-peptide increases transepithelial resistance in epithelial cells by modulating claudin-1 and -2 expression. Peptides 2006; 27: 3383–90.10.1016/j.peptides.2006.08.020Suche in Google Scholar

146. Tan XD, Liu QP, Hsueh W, Chen YH, Chang H, Gonzalez-Crussi F. Intestinal trefoil factor binds to intestinal epithelial cells and induces nitric oxide production: priming and enhancing effects of mucin. Biochem J 1999; 338: 745–51.10.1042/bj3380745Suche in Google Scholar

147. Sun Z, Liu H, Yang Z, Shao D, Zhang W, Ren Y, Sun B, Lin J, Xu M, Nie S. Intestinal trefoil factor activates the PI3K/Akt signaling pathway to protect gastric mucosal epithelium from damage. Int J Oncol 2014; 45: 1123–32.10.3892/ijo.2014.2527Suche in Google Scholar

148. Bird A. The essentials of DNA methylation. Cell 1992; 70: 5–8.10.1016/0092-8674(92)90526-ISuche in Google Scholar

149. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet 2002; 3: 415–28.10.1038/nrg816Suche in Google Scholar

150. Zhang Y, Staal B, Dykema KJ, Furge KA, Vande Woude GF. Cancer-type regulation of MIG-6 expression by inhibitors of methylation and histone deacetylation. PLoS One 2012; 7: e38955.10.1371/journal.pone.0038955Suche in Google Scholar

151. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002; 16: 6–21.10.1101/gad.947102Suche in Google Scholar

152. Ribieras S, Lefèbvre O, Tomasetto C, Rio MC. Mouse Trefoil factor genes: genomic organization, sequences and methylation analyses. Gene 2001; 266: 67–75.10.1016/S0378-1119(01)00380-8Suche in Google Scholar

153. Sato N, Maitra A, Fukushima N, van Heek, NT, Matsubayashi H, Iacobuzio-Donahue CA, Rosty C, Goggins M. Frequent hypomethylation of multiple genes overexpressed in pancreatic ductal adenocarcinoma. Cancer Res 2003; 63: 4158–66.Suche in Google Scholar

154. Feng G, Zhang Y, Yuan H, Bai R, Zheng J, Zhang J, Song M. DNA methylation of trefoil factor 1 (TFF1) is associated with the tumorigenesis of gastric carcinoma. Mol Med Rep 2014; 9: 109–17.10.3892/mmr.2013.1772Suche in Google Scholar PubMed

155. Thériault BL, Dimaras H, Gallie BL, Corson TW. The genomic landscape of retinoblastoma: a review. Clin Experiment Ophthalmol 2014; 42: 33–52.10.1111/ceo.12132Suche in Google Scholar

156. Hsieh CL. Dependence of transcriptional repression on CpG methylation density. Mol Cell Biol 1994; 14: 5487–94.Suche in Google Scholar

157. Martin V, Ribieras S, Song-Wang X, Rio MC, Dante R. Genomic sequencing indicates a correlation between DNA hypomethylation in the 5′ region of the pS2 gene and its expression in human breast cancer cell lines. Gene 1995; 157: 261–4.10.1016/0378-1119(95)00096-OSuche in Google Scholar

158. Vestergaard EM, Nexø E, Tørring N, Borre M, Ørntoft TF, Sørensen KD. Promoter hypomethylation and upregulation of trefoil factors in prostate cancer. Int J Cancer 2010; 127: 1857–65.10.1002/ijc.25209Suche in Google Scholar

159. Cameron EE, Bachman KE, Myöhänen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet 1999; 21: 103–7.10.1038/5047Suche in Google Scholar

160. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75: 843–54.10.1016/0092-8674(93)90529-YSuche in Google Scholar

161. Liu J, Wang X, Yang X, Liu Y, Shi Y, Ren J, Guleng B. miRNA423-5p regulates cell proliferation and invasion by targeting trefoil factor 1 in gastric cancer cells. Cancer Lett 2014; 347: 98–104.10.1016/j.canlet.2014.01.024Suche in Google Scholar PubMed

162. Shi Y, Chen G, Huang Q, Chen X, Liu J, Xu W, Huang X, Liu Y, Xiao C, Wu D, Guleng B, Ren J. miR218-5p regulates the proliferation of gastric cancer cells by targeting TFF1 in an Erk1/2-dependent manner. Biochim Biophys Acta 2015; 1852: 970–9.10.1016/j.bbadis.2015.01.016Suche in Google Scholar PubMed

163. Soutto M, Chen Z, Saleh MA, Katsha A, Zhu S, Zaika A, Belkhiri A, El-Rifai W. TFF1 activates p53 through down-regulation of miR-504 in gastric cancer. Oncotarget 2014; 5: 5663–73.10.18632/oncotarget.2156Suche in Google Scholar PubMed PubMed Central

164. Shah AA, Leidinger P, Keller A, Wendschlag A, Meese E, Blin N. Altered miRNA expression patterns in Tff2 knock-out mice correlate with cellular pathways of neoplastic development and caloric metabolism. Int J Mol Med 2012; 29: 637–43.10.3892/ijmm.2012.881Suche in Google Scholar PubMed PubMed Central

165. Shah AA, Leidinger P, Backes C, Keller A, Karpinski P, Sasiadek MM, Blin N, Meese E. A set of specific miRNAs is connected with murine and human gastric cancer. Genes Chromosomes Cancer 2013; 52: 237–49.10.1002/gcc.22024Suche in Google Scholar PubMed

166. Yin Y, Shan HQ, Huang W, Wu YM, Lu H, Jin Y. Bioinformatic analysis of miRNA expression patterns in TFF2 knock-out mice. Genet Mol Res 2014; 13: 8502–10.10.4238/2014.October.20.26Suche in Google Scholar PubMed

167. Sousa JF, Nam KT, Petersen CP, Lee H, Yang H, Kim WH, Goldenring JR. miR-30-HNF4γ and miR-194-NR2F2 regulatory networks contribute to the upregulation of metaplasia markers in the stomach. Gut 2015. doi:10.1136/gutjnl-2014-308759.10.1136/gutjnl-2014-308759Suche in Google Scholar PubMed PubMed Central

168. Rotkrua P, Akiyama Y, Hashimoto Y, Otsubo T, Yuasa Y. MiR-9 downregulates CDX2 expression in gastric cancer cells. Int J Cancer 2011; 129: 2611–20.10.1002/ijc.25923Suche in Google Scholar PubMed

169. Soutto M, Peng D, Katsha A, Chen Z, Piazuelo MB, Washington MK, Belkhiri A, Correa P, El-Rifai W. Activation of β-catenin signalling by TFF1 loss promotes cell proliferation and gastric tumorigenesis. Gut 2014; 64: 1028–39.10.1136/gutjnl-2014-307191Suche in Google Scholar PubMed PubMed Central

170. Arumugam T, Brandt W, Ramachandran V, Moore TT, Wang H, May FE, Westley BR, Hwang RF, Logsdon CD. Trefoil factor 1 stimulates both pancreatic cancer and stellate cells and increases metastasis. Pancreas 2011; 40: 815–22.10.1097/MPA.0b013e31821f6927Suche in Google Scholar PubMed PubMed Central

171. Rodrigues S, Rodrigue CM, Attoub S, Fléjou JF, Bruyneel E, Bracke M, Emami S, Gespach C. Induction of the adenoma-carcinoma progression and Cdc25A-B phosphatases by the trefoil factor TFF1 in human colon epithelial cells. Oncogene 2006; 25: 6628–36.10.1038/sj.onc.1209665Suche in Google Scholar PubMed

172. Farrell JJ, Taupin D, Koh TJ, Chen D, Zhao C, Podolsky DK, Wang TC. TFF2/SP-deficient mice show decreased gastric proliferation, increased acid secretion, and increased susceptibility to NSAID injury. J Clin Invest 2002; 109: 193–204.10.1172/JCI0212529Suche in Google Scholar

173. Baus-Loncar M, Schmid J, Lalani E, Rosewell I, Goodlad RA, Stamp, Gordon WH, Blin N, Kayademir T. Trefoil factor 2 (TFF2) deficiency in murine digestive tract influences the immune system. Cell Physiol Biochem 2005; 16: 31–42.10.1159/000087729Suche in Google Scholar PubMed

174. Baus-Loncar M, Kayademir T, Takaishi S, Wang T. Trefoil factor family 2 deficiency and immune response. Cell Mol Life Sci 2005; 62: 2947–55.10.1007/s00018-005-5483-7Suche in Google Scholar PubMed

175. Beck PL, Wong JF, Li Y, Swaminathan S, Xavier RJ, Devaney KL, Podolsky DK. Chemotherapy- and radiotherapy-induced intestinal damage is regulated by intestinal trefoil factor. Gastroenterology 2004; 126: 796–808.10.1053/j.gastro.2003.12.004Suche in Google Scholar PubMed

176. Marchbank T, Cox HM, Goodlad RA, Giraud AS, Moss SF, Poulsom R, Wright NA, Jankowski J, Playford RJ. Effect of ectopic expression of rat trefoil factor family 3 (intestinal trefoil factor) in the jejunum of transgenic mice. J Biol Chem 2001; 276: 24088–96.10.1074/jbc.M101363200Suche in Google Scholar

177. Yio X, Diamond M, Zhang J, Weinstein H, Wang L, Werther L, Itzkowitz S. Trefoil factor family-1 mutations enhance gastric cancer cell invasion through distinct signaling pathways. Gastroenterology 2006; 130: 1696–706.10.1053/j.gastro.2006.01.040Suche in Google Scholar

178. Lalani EN, Williams R, Jayaram Y, Gilbert C, Chaudhary KS, Siu LS, Koumarianou A, Playford R, Stamp GW. Trefoil factor-2, human spasmolytic polypeptide, promotes branching morphogenesis in MCF-7 cells. Lab Invest 1999; 79: 537–46.Suche in Google Scholar

179. Siu LS, Romanska H, Abel PD, Baus-Loncar M, Kayademir T, Stamp GW, Lalani el-N. TFF2 (trefoil family factor2) inhibits apoptosis in breast and colorectal cancer cell lines. Peptides 2004; 25: 855–63.10.1016/j.peptides.2003.11.023Suche in Google Scholar

180. Tu SP, Chi AL, Ai W, Takaishi S, Dubeykovskaya Z, Quante M, Fox JG, Wang TC. p53 inhibition of AP1-dependent TFF2 expression induces apoptosis and inhibits cell migration in gastric cancer cells. Am J Physiol Gastrointest Liver Physiol 2009; 297: G385–96.10.1152/ajpgi.90620.2008Suche in Google Scholar

181. Chan MW, Chan VY, Leung WK, Chan KK, To KF, Sung JJ, Chan FK. Anti-sense trefoil factor family-3 (intestinal trefoil factor) inhibits cell growth and induces chemosensitivity to adriamycin in human gastric cancer cells. Life Sci 2005; 76: 2581–92.10.1016/j.lfs.2004.11.014Suche in Google Scholar

182. Kannan N, Kang J, Kong X, Tang J, Perry JK, Mohankumar KM, Miller LD, Liu ET, Mertani HC, Zhu T, Grandison PM, Liu DX, Lobie PE. Trefoil factor 3 is oncogenic and mediates anti-estrogen resistance in human mammary carcinoma. Neoplasia 2010; 12: 1041–53.10.1593/neo.10916Suche in Google Scholar

183. Rösler S, Haase T, Claassen H, Schulze U, Schicht M, Riemann D, Brandt J, Wohlrab D, Müller-Hilke B, Goldring MB, Sel S, Varoga D, Garreis F, Paulsen FP. Trefoil factor 3 is induced during degenerative and inflammatory joint disease, activates matrix metalloproteinases, and enhances apoptosis of articular cartilage chondrocytes. Arthritis Rheum 2010; 62: 815–25.10.1002/art.27295Suche in Google Scholar

184. Lubka M, Shah AA, Blin N, Baus-Loncar M. The intestinal trefoil factor (Tff3), also expressed in the inner ear, interacts with peptides contributing to apoptosis. J Appl Genet 2009; 50: 167–71.10.1007/BF03195669Suche in Google Scholar

185. Calnan DP, Westley BR, May FE, Floyd DN, Marchbank T, Playford RJ. The trefoil peptide TFF1 inhibits the growth of the human gastric adenocarcinoma cell line AGS. J Pathol 1999; 188: 312–7.10.1002/(SICI)1096-9896(199907)188:3<312::AID-PATH360>3.0.CO;2-PSuche in Google Scholar

186. Hoosein NM, Thim L, Jørgensen KH, Brattain MG. Growth stimulatory effect of pancreatic spasmolytic polypeptide on cultured colon and breast tumor cells. FEBS Lett 1989; 247: 303–6.10.1016/0014-5793(89)81357-2Suche in Google Scholar

187. Lin J, Sun Z, Zhang W, Liu H, Shao D, Ren Y, Wen Y, Cao L, Wolfram J, Yang Z, Nie S. Protective effects of intestinal trefoil factor (ITF) on gastric mucosal epithelium through activation of extracellular signal-regulated kinase 1/2 (ERK1/2). Mol Cell Biochem 2015; 404: 263–70.10.1007/s11010-015-2386-2Suche in Google Scholar PubMed

188. Perera O, Evans A, Pertziger M, MacDonald C, Chen H, Liu D, Lobie PE, Perry JK. Trefoil factor 3 (TFF3) enhances the oncogenic characteristics of prostate carcinoma cells and reduces sensitivity to ionising radiation. Cancer Lett 2015; 361: 104–11.10.1016/j.canlet.2015.02.051Suche in Google Scholar PubMed

189. Prest SJ, May FE, Westley BR. The estrogen-regulated protein, TFF1, stimulates migration of human breast cancer cells. FASEB J 2002; 16: 592–4.10.1096/fj.01-0498fjeSuche in Google Scholar PubMed

190. Fu T, Kalbacher H, Hoffmann W. TFF1 is differentially expressed in stationary and migratory rat gastric epithelial cells (RGM-1) after in vitro wounding: influence of TFF1 RNA interference on cell migration. Cell Physiol Biochem 2013; 32: 997–1010.10.1159/000354501Suche in Google Scholar PubMed

191. Storesund T, Hayashi K, Kolltveit KM, Bryne M, Schenck K. Salivary trefoil factor 3 enhances migration of oral keratinocytes. Eur J Oral Sci 2008; 116: 135–40.10.1111/j.1600-0722.2007.00516.xSuche in Google Scholar PubMed

192. Chan VY, Chan MW, Leung W, Leung P, Sung JJ, Chan FK. Intestinal trefoil factor promotes invasion in non-tumorigenic Rat-2 fibroblast cell. Regul Pept 2005; 127: 87–94.10.1016/j.regpep.2004.10.016Suche in Google Scholar PubMed

193. Rivat C, Rodrigues S, Bruyneel E, Piétu G, Robert A, Redeuilh G, Bracke M, Gespach C, Attoub S. Implication of STAT3 signaling in human colonic cancer cells during intestinal trefoil factor 3 (TFF3) – and vascular endothelial growth factor-mediated cellular invasion and tumor growth. Cancer Res 2005; 65: 195–202.10.1158/0008-5472.195.65.1Suche in Google Scholar

194. Yio X, Zhang J, Babyatsky M, Chen A, Lin J, Fan Q, Werther JL, Itzkowitz S. Trefoil factor family-3 is associated with aggressive behavior of colon cancer cells. Clin Exp Metastasis 2005; 22: 157–65.10.1007/s10585-005-6615-zSuche in Google Scholar PubMed

195. Babyatsky M, Lin J, Yio X, Chen A, Zhang J, Zheng Y, Twyman C, Bao X, Schwartz M, Thung S, Lawrence Werther J, Itzkowitz S. Trefoil factor-3 expression in human colon cancer liver metastasis. Clin Exp Metastasis 2009; 26: 143–51.10.1007/s10585-008-9224-9Suche in Google Scholar PubMed

Received: 2015-7-14
Accepted: 2015-8-20
Published Online: 2015-10-17
Published in Print: 2015-12-1

©2015 by De Gruyter

This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Heruntergeladen am 15.9.2025 von https://www.degruyterbrill.com/document/doi/10.1515/bmc-2015-0020/html
Button zum nach oben scrollen